277
Views
0
CrossRef citations to date
0
Altmetric
Review

Advancements in drug development for diarrhea-predominant irritable bowel syndrome

, , , &
Pages 251-263 | Received 05 Oct 2017, Accepted 14 Feb 2018, Published online: 22 Feb 2018

References

  • Lacy BE, Mearin F, Chang L, et al. Bowel disorders. Gastroenterology. 2016;150:1393–1407e5.
  • Canavan C, West J, Card T. The epidemiology of irritable bowel syndrome. Clin Epidemiol. 2014;6:71–80.
  • Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel syndrome: a meta-analysis. Clin Gastroenterol Hepatol. 2012;10:712–721.e4.
  • Enck P, Aziz Q, Barbara G, et al. Irritable bowel syndrome. Nat Rev Dis Prim. 2016;2:16014.
  • Mearin F, Lacy BE, Chang L, et al. Bowel disorders. Gastroenterology. 2016;150:1393–1407e5.
  • Wong RK, Palsson OS, Turner MJ, et al. Inability of the Rome III criteria to distinguish functional constipation from constipation-subtype irritable bowel syndrome. Am J Gastroenterol. 2010;105:2228–2234.
  • Ford AC, Bercik P, Morgan DG, et al. Characteristics of functional bowel disorder patients: a cross-sectional survey using the Rome III criteria. Aliment Pharmacol Ther. 2014;39:312–321.
  • Palsson OS, Whitehead WE, Van Tilburg MAL, et al. Development and validation of the Rome IV diagnostic questionnaire for adults. Gastroenterology. 2016;150:1481–1491.
  • Dothel G, Barbaro MR, Boudin H, et al. Nerve fiber outgrowth is increased in the intestinal mucosa of patients with irritable bowel syndrome. Gastroenterology. 2015;148:1002-1011.
  • Öhman L, Simrén M. Pathogenesis of IBS: role of inflammation, immunity and neuroimmune interactions. Nat Rev Gastroenterol Hepatol. 2010;7:163–173.
  • Barbara G, Stanghellini V, De Giorgio R, et al. Activated mast cells in proximity to colonic nerves correlate with abdominal pain in irritable bowel syndrome. Gastroenterology. 2004;126:693–702.
  • Piche T, Barbara G, Aubert P, et al. Impaired intestinal barrier integrity in the colon of patients with irritable bowel syndrome: involvement of soluble mediators. Gut. 2009;58:196–201.
  • Carroccio A, Brusca I, Mansueto P, et al. A cytologic assay for diagnosis of food hypersensitivity in patients with irritable bowel syndrome. Clin Gastroenterol Hepatol. 2010;8:254–260.
  • Jeffery IB, Quigley EMM, Öhman L, et al. The microbiota link to irritable bowel syndrome an emerging story. Gut Microbes. 2012;3:572–576.
  • Tap J, Derrien M, Törnblom H, et al. Identification of an intestinal microbiota signature associated with severity of irritable bowel syndrome. Gastroenterology. 2017;152:111–123.e8.
  • Atkinson W, Lockhart S, Whorwell PJ, et al. Altered 5-hydroxytryptamine signaling in patients with constipation- and diarrhea-predominant irritable bowel syndrome. Gastroenterology. 2006;130:34–43.
  • Zucchelli M, Camilleri M, Nixon Andreasson A, et al. Association of TNFSF15 polymorphism with irritable bowel syndrome. Gut. 2011;60:1671–1677.
  • Camilleri M. Genetics of human gastrointestinal sensation. Neurogastroenterol Motil. 2013;25:458–466.
  • Tack J, Corsetti M, Camilleri M, et al. Plausibility criteria for putative pathophysiological mechanisms in functional gastrointestinal disorders: a consensus of experts. Gut. 2017. doi: 10.1136/gutjnl-2016–312230.
  • Camilleri M. Bile acid diarrhea: prevalence, pathogenesis, and therapy. 2015;9:332–339.
  • Chumpitazi BP, Cope JL, Hollister EB, et al. Randomised clinical trial: gut microbiome biomarkers are associated with clinical response to a low FODMAP diet in children with the irritable bowel syndrome. Aliment Pharmacol Ther. 2015;42:418–427.
  • Bafutto M, de Almeida JR, Leite NV, et al. Treatment of postinfectious irritable bowel syndrome and noninfective irritable bowel syndrome with mesalazine. Arq Gastroenterol. 2011;48:36–40.
  • Camilleri M, Boeckxstaens G. Dietary and pharmacological treatment of abdominal pain in IBS. Gut. 2017;66:966–974.
  • Mayer EA, Bradesi S, Chang L, et al. Functional GI disorders: from animal models to drug development. Gut. 2008;57:384–404.
  • European Medical Agen. Guideline on the evaluation of medicinal products for the treatment of irritable bowel syndrome - CPMP/EWP/785/97 Rev. 1 [Internet]. [cited 2017 Jul 18].
  • Food and Drug administration. Guidance for Industry Irritable Bowel Syndrome — Clinical Evaluation of Drugs for Treatment. [cited 2017 Jul 18].
  • Fukudo S, Kinoshita Y, Okumura T, et al. Effect of ramosetron in female patients with irritable bowel syndrome with diarrhea : a phase III long-term study. J Gastroenterol. 2016;51:874–882.
  • Food and Drug Administration. HIGHLIGHTS OF PRESCRIBING INFORMATION - XIFAXAN® (rifaximin) tablets, for oral use. [cited 20 Dec 2017]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/021361s012lbledt.pdf
  • Hartmann G, Honikel KO, Knüsel F, et al. The specific inhibition of the DNA-directed RNA synthesis by rifamycin. Biochim Biophys Acta. 1967;145:843–844.
  • Calanni F, Renzulli C, Barbanti M, et al. Rifaximin: beyond the traditional antibiotic activity. J Antibiot (Tokyo). 2014;667–670.
  • Bajaj JS, Heuman DM, Sanyal AJ, et al. Modulation of the metabiome by rifaximin in patients with cirrhosis and minimal hepatic encephalopathy. PLoS One. 2013;8.
  • Ma X, Shah YM, Guo GL, et al. Rifaximin is a gut-specific human pregnane X receptor activator. J Pharmacol Exp Ther. 2007;322:391–398.
  • Dring MM, Goulding CA, Trimble VI, et al. The pregnane X receptor locus is associated with susceptibility to inflammatory bowel disease. Gastroenterology. 2006;130:341–348.
  • Food and Drug Administration. FDA approves two therapies to treat IBS-D.[cited 2017 Nov 28]. Available from: https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm448328.htm
  • Pimentel M, Lembo A, Chey WD, et al. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N Engl J Med. 2011;364:22–32.
  • Lembo A, Pimentel M, Rao SS, et al. Repeat treatment with rifaximin is safe and effective in patients with diarrhea-predominant irritable bowel syndrome. Gastroenterology. 2016;151:1113–1121.
  • Pimentel M. Review article: potential mechanisms of action of rifaximin in the management of irritable bowel syndrome with diarrhoea. Aliment Pharmacol Ther. 2016;43(Suppl 1):37–49.
  • Gershon MD. Review article: serotonin receptors and transporters - roles in normal and abnormal gastrointestinal motility. Aliment Pharmacol Ther. 2004;20:3–14.
  • Garcia-Olmo D, Guadalajara H, Rubio-Perez I, et al. Recurrent anal fistulae: limited surgery supported by stem cells. World J Gastroenterol. 2015;21:3330–3336.
  • Galligan JJ, Pan H, Messori E. Signalling mechanism coupled to 5-hydroxytryptamine4 receptor-mediated facilitation of fast synaptic transmission in the guinea-pig ileum myenteric plexus. Neurogastroenterol Motil. 2003;15:523–529.
  • Suzuki R, Rygh LJ, Dickenson AH. Bad news from the brain: descending 5-HT pathways that control spinal pain processing. Trends Pharmacol Sci. 2004;25:613–617.
  • Stasi C, Bellini M, Bassotti G, et al. Serotonin receptors and their role in the pathophysiology and therapy of irritable bowel syndrome. Tech Coloproctol. 2014;18:613–621.
  • Zheng Y, Yu T, Tang Y, et al. Efficacy and safety of 5-hydroxytryptamine 3 receptor antagonists in irritable bowel syndrome: a systematic review and metaanalysis of randomized controlled trials. PLoS One. 2017. doi 10.1371/journal.pone.0172846.
  • Ford AC, Moayyedi P, Lacy BE, et al. American college of gastroenterology monograph on the management of irritable bowel syndrome and chronic idiopathic constipation. Am J Gastroenterol. 2014;109:S2–S26.
  • Lucak S, Chang L, Halpert A, et al. Current and emergent pharmacologic treatments for irritable bowel syndrome with diarrhea: evidence-based treatment in practice. Therap Adv Gastroenterol. 2017;10:253–275.
  • Miller JL. FDA draws patients into alosetron risk management. Am J Heal Pharm. 2000;57:1736.
  • Garvey TQI, Hyman PE, Hyman PE, et al. Return of alosetron. Expert Opin Drug Saf. 2017;1:1-4.
  • Chang L, Tong K, Ameen V. Ischemic colitis and complications of constipation associated with the use of alosetron under a risk management plan : clinical characteristics, outcomes, and incidences. Am J Gastroenterol. 2010;105:866–875.
  • Cremonini F, Nicandro JP, Atkinson V, et al. Randomised clinical trial: alosetron improves quality of life and reduces restriction of daily activities in women with severe diarrhoea-predominant IBS. Aliment Pharmacol Ther. 2012;36:437–448.
  • Fukudo S, Kinoshita Y, Okumura T, et al. Ramosetron reduces symptoms of irritable bowel syndrome with diarrhea and improves quality of life in women. Gastroenterology. 2016;150:258–366e8.
  • Matsueda SFK, Ida KHM, Akiho HHH, et al. Optimal dose of ramosetron in female patients with irritable bowel syndrome with diarrhea : a randomized, controlled phase II study. Neurogastroenterol Motil. 2017;29:1–10.
  • Shiotani A, Kusunoki H, Ishii M, et al. Pilot study of biomarkers for predicting effectiveness of ramosetron in diarrhea-predominant irritable bowel syndrome: expression of S100A10 and polymorphisms of TPH1. Neurogastroenterol Motil. 2015;27:82–91.
  • Grover M, Camilleri M. Ramosetron in irritable bowel syndrome with diarrhea: new hope or the same old story? Clin Gastroenterol Hepatol. 2014;12:960–962.
  • Katsumata R, Shiotani A, Murao T, et al. Gender differences in serotonin signaling in patients with diarrhea-predominant irritable bowel syndrome. Intern Med. 2017;56:993–999.
  • Garsed K, Chernova J, Hastings M, et al. A randomised trial of ondansetron for the treatment of irritable bowel syndrome with diarrhoea. Gut. 2014;63:1617–1625.
  • Miles TF, Lester HA, Dougherty DA. Allosteric activation of the 5-HT3AB receptor by mCPBG. Neuropharmacology. 2015;91:103–108.
  • Brown PM, Drossman DA, Wood AJJ, et al. The tryptophan hydroxylase inhibitor LX1031 shows clinical benefit in patients with nonconstipating irritable bowel syndrome. Gastroenterology. 2011;141:507–516.
  • Clinicaltrial.gov. Study of LX1031 in Subjects With Non-Constipating Irritable Bowel Syndrome. [cited 2017 Jul 25]. Available from: https://clinicaltrials.gov/ct2/show/NCT00813098?term=NCT00813098&rank=1
  • Moore NA, Sargent BJ, Manning DD, et al. Partial agonism of 5-HT3 receptors: A novel approach to the symptomatic treatment of IBS-D. ACS Chem Neurosci. 2013;4:43–47.
  • Manning DD, Guo C, Zhang Z, et al. The discovery of diazepinone-based 5-HT3 receptor partial agonists. Bioorg Med Chem Lett. 2014;24:2578–2581.
  • Jarvis GE, Barbosa R, Thompson AJ. Noncompetitive inhibition of 5-HT3 receptors by citral, linalool, and eucalyptol revealed by nonlinear mixed-effects modeling. J Pharmacol Exp Ther. 2016;356:549–562.
  • Khanna R, MacDonald JK, Levesque BG. Peppermint oil for the treatment of irritable bowel syndrome: a systematic review and meta-analysis. J Clin Gastroenterol. 2014;48:505–512.
  • Kariv R, Tiomny E, Grenshpon R, et al. Low-dose naltreoxone for the treatment of irritable bowel syndrome : a pilot study. Dig Dis Sci. 2006;2128–2133.
  • Kest B, Lee CE, McLemore GL, et al. An antisense oligodeoxynucleotide to the delta opioid receptor (DOR-1) inhibits morphine tolerance and acute dependence in mice. Brain Res Bull. 1996;39:185–188.
  • Zhu Y, King MA, Schuller AGP, et al. Retention of supraspinal delta-like analgesia and loss of morphine tolerance in δ opioid receptor knockout mice. Neuron. 1999;24:243–252.
  • Zielińska M, Chen C, Mokrowiecka A, et al. Orally administered novel cyclic pentapeptide P-317 alleviates symptoms of diarrhoea-predominant irritable bowel syndrome. J Pharm Pharmacol. 2015;67:244–254.
  • Sobczak M, Cami-Kobeci G, Sałaga M, et al. Novel mixed NOP/MOP agonist BU08070 alleviates pain and inhibits gastrointestinal motility in mouse models mimicking diarrhea-predominant irritable bowel syndrome symptoms. Eur J Pharmacol. 2014;736:63–69.
  • Al-Hasani R, Bruchas MR. Molecular mechanisms of opioid receptor-dependent signaling and behavior. Anesthesiology. 2011;115:1363-1381.
  • Fujita W, Gomes I, Devi LA. Heteromers of μ-δ opioid receptors: new pharmacology and novel therapeutic possibilities. Br J Pharmacol. 2015;172:375–387.
  • Hojo M, Sudo Y, Ando Y, et al. Mu-opioid receptor forms a functional heterodimer with cannabinoid CB receptor: electrophysiological and FRET assay analysis. J Pharmacol Sci. 2008;108:308–319.
  • Rios C, Gomes I, Devi LA. mu opioid and CB1 cannabinoid receptor interactions: reciprocal inhibition of receptor signaling and neuritogenesis. Br J Pharmacol. 2006;148:387–395.
  • Pfeiffer M, Koch T, Schröder H, et al. Heterodimerization of somatostatin and opioid receptors cross-modulates phosphorylation, internalization, and desensitization. J Biol Chem. 2002;277:19762–19772.
  • Schröder H, Wu DF, Seifert A, et al. Allosteric modulation of metabotropic glutamate receptor 5 affects phosphorylation, internalization, and desensitization of the micro-opioid receptor. Neuropharmacology. 2009;56:768–778.
  • Le Naour M, Lunzer MM, Powers MD, et al. Putative kappa opioid heteromers as targets for developing analgesics free of adverse effects. J Med Chem. 2014;57:6383–6392.
  • Shippenberg TS. The dynorphin/kappa opioid receptor system: a new target for the treatment of addiction and affective disorders? Neuropsychopharmacology. 2009;34:247.
  • Fujita W, Gomes I, Dove LS, et al. Molecular characterization of eluxadoline as a potential ligand targeting mu-delta opioid receptor heteromers. Biochem Pharmacol. 2014;92:448–456.
  • National Institute for Health and Clinical Excellence. Resource impact report: Eluxadoline for treating irritable bowel syndrome with diarrhoea (TA471). [cited 2017 Sept 9]. Available from: https://www.nice.org.uk/guidance/ta471/resources/resource-impact-report-pdf-4595718349
  • Lembo AJ, Lacy BE, Zuckerman MJ, et al. Eluxadoline for irritable bowel syndrome with diarrhea. N Engl J Med. 2016;374:242–253.
  • R V F, Henningfield JE, Cash BD, et al. Eluxadoline demonstrates a lack of abuse potential in phase 2 and 3 studies of patients with irritable bowel syndrome with diarrhea. Clin Gastroenterol Hepatol. 2017;15:1021–1029.e6.
  • Marbury TC, Berg JK, Dove LS, et al. Effect of hepatic impairment on eluxadoline pharmacokinetics. J Clin Pharmacol. 2017;57:1454-1459.
  • Food and Drug Administration. FDA Drug Safety Communication: FDA warns about increased risk of serious pancreatitis with irritable bowel drug Viberzi (eluxadoline) in patients without a gallbladder Available from: https://www.fda.gov/Drugs/DrugSafety/ucm546154.htm [cited 2017 Sep 28]
  • Gawron AJ, Bielefeldt K. Risk of pancreatitis following treatment of irritable bowel syndrome with eluxadoline. Clin Gastroenterol Hepatol.2018;16:378-384
  • European Medical Association. List of medicinal products under additional monitoring. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Medicines_under_additional_monitoring/2013/04/WC500142453.pdf [cited 2017 Aug].
  • Piomelli D, Sasso O. Peripheral gating of pain signals by endogenous lipid mediators. Nat Neurosci. 2014;17:164–174.
  • Barann M, Molderings G, Brüss M, et al. Direct inhibition by cannabinoids of human 5-HT3A receptors: probable involvement of an allosteric modulatory site. Br J Pharmacol. 2002;137:589–596.
  • Guindon J, Hohmann AG. Cannabinoid CB2 receptors: a therapeutic target for the treatment of inflammatory and neuropathic pain. Br J Pharmacol. 2008;153: 319–334.
  • Howlett AC. Pharmacology of cannabinoid receptors. Annu Rev Pharmacol Toxicol. 1995;35:607–634.
  • Yao B, Mackie K. Endocannabinoid receptor pharmacology. Curr Top Behav Neurosci. 2009;1:37–63.
  • Abalo R, Chen C, Vera G, et al. In vitro and non-invasive in vivo effects of the cannabinoid-1 receptor agonist AM841 on gastrointestinal motor function in the rat. Neurogastroenterol Motil. 2015;27:1721–1735.
  • Keenan CM, Storr MA, Thakur GA, et al. AM841, a covalent cannabinoid ligand, powerfully slows gastrointestinal motility in normal and stressed mice in a peripherally restricted manner. Br J Pharmacol. 2015;172:2406–2418.
  • Picone RP, Khanolkar AD, Xu W. (-)-7'-Isothiocyanato-11-hydroxy-1',1'-dimethylheptylhexahydrocannabinol (AM841), a high-affinity electrophilic ligand interacts covalently with a cysteine in helix six and activates the cb1 cannabinoid receptor. Mol Pharmacol. 2005;68:1623-1635.
  • Reichenbach ZW, Schey R. Cannabinoids and GI disorders: endogenous and exogenous. Curr Treat Options Gastroenterol. 2016;14:461–477.
  • Sanson M, Bueno L, Fioramonti J. Involvement of cannabinoid receptors in inflammatory hypersensitivity to colonic distension in rats. Neurogastroenterol Motil. 2006;18:949–956.
  • Kikuchi A, Ohashi K, Sugie Y, et al. Pharmacological evaluation of a novel cannabinoid 2 (CB2) ligand, PF-03550096, in vitro and in vivo by using a rat model of visceral hypersensitivity. J Pharmacol Sci. 2008;106:219–224.
  • Wong BS, Camilleri M, Busciglio I, et al. Pharmacogenetic trial of a cannabinoid agonist shows reduced fasting colonic motility in patients with nonconstipated irritable bowel syndrome. Gastroenterology. 2011;141:1638–1647.
  • Fichna J, Sałaga M, Stuart J, et al. Selective inhibition of FAAH produces antidiarrheal and antinociceptive effect mediated by endocannabinoids and cannabinoid-like fatty acid amides. Neurogastroenterol Motil. 2014;26:470–481.
  • Fichna J, Wood JAT, Papanastasiou M, et al. Endocannabinoid and cannabinoid-like fatty acid amide levels correlate with pain-related symptoms in patients with IBS-D and IBS-C: a pilot study. PLoS One. 2013; doi: 10.1371/journal.pone.0085073
  • Aloe L, Leon A, Levi-Montalcini R. A proposed autacoid mechanism controlling mastocyte behaviour. Agents Actions. 1993;39:145–147.
  • Mazzari S, Canella R, Petrelli L, et al. N-(2-Hydroxyethyl) hexadecanamide is orally active in reducing edema formation and inflammatory hyperalgesia by down-modulating mast cell activation. Eur J Pharmacol. 1996;300:227–236.
  • Bortolini M, Wright MB, Bopst M, et al. Examining the safety of PPAR agonists - current trends and future prospects. Expert Opin Drug Saf. 2013;12:65–79.
  • Di Cesare Mannelli L, D’Agostino G, Pacini A, et al. Palmitoylethanolamide is a disease-modifying agent in peripheral neuropathy: pain relief and neuroprotection share a PPAR-alpha-mediated mechanism. Mediators Inflamm. 2013;2013.
  • Guida F, Luongo L, Boccella S, et al. Palmitoylethanolamide induces microglia changes associated with increased migration and phagocytic activity: involvement of the CB2 receptor. Sci Rep. 2017;7:375.
  • Li JJ, Cao JJ, Yang C, et al. Polydatin attenuated food allergy via store-operated calcium channels in mast cell. World J Gastroenterol. 2013;19:3980–3989.
  • Cremon C, Stanghellini V, Barbaro MR, et al. Randomised clinical trial: the analgesic properties of dietary supplementation with palmitoylethanolamide and polydatin in irritable bowel syndrome. Aliment Pharmacol Ther. 2017;45:1–14.
  • Eb R. Clinical endocannabinoid deficiency (CECD): can this concept explain therapeutic benefits of. Eur PMC. 2004;25:31–34.
  • Rousseaux C, Thuru X, Gelot A, et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med. 2007;13:35–37.
  • Camilleri M, Carlson P, McKinzie S, et al. Genetic variation in endocannabinoid metabolism, gastrointestinal motility, and sensation. Am J Physiol Gastrointest Liver Physiol. 2008;294:G13–9.
  • Wong BS, Camilleri M, Carlson P, et al. Increased bile acid biosynthesis is associated with irritable bowel syndrome with diarrhea. Clin Gastroenterol Hepatol. 2012;10:1009–1015.
  • 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;42:3–11.
  • Vijayvargiya P, Camilleri M, Shin A, et al. Diagnostic methods for bile acid malabsorption in clinical practice. Clin Gastroenterol Hepatol. 2013;11:1232–1239.
  • Wedlake L, Thomas K, Lalji A, et al. Effectiveness and tolerability of colesevelam hydrochloride for bile-acid malabsorption in patients with cancer : a retrospective chart review and patient questionnaire. Clin Ther. 2009;31:2549–2558.
  • Peleman C, Camilleri M, Busciglio I, et al. Colonic transit and bile acid synthesis or excretion in patients with irritable bowel syndrome – diarrhea without bile acid malabsorption. Clin Gastroenterol Hepatol. 2017;15:720–727.e1.
  • Camilleri M, Acosta A, Busciglio I, et al. Effect of colesevelam on faecal bile acids and bowel functions in diarrhoea-predominant irritable bowel syndrome. Aliment Pharmacol Ther. 2015;41:438–448.
  • Camilleri M, Vazquez-Roque MI, Carlson P, et al. Association of bile acid receptor TGR5 variation and transit in health and lower functional gastrointestinal disorders. Neurogastroenterol Motil. 2011;23:995.
  • Camilleri M, Carlson P, Acosta A, et al. RNA sequencing shows transcriptomic changes in rectosigmoid mucosa in patients with irritable bowel syndrome-diarrhea: a pilot case-control study. AJP Gastrointest Liver Physiol. 2014;306:G1089–G1098.
  • Mroz MS, Keating N, Ward JB, et al. Farnesoid X receptor agonists attenuate colonic epithelial secretory function and prevent experimental diarrhoea in vivo. Gut. 2014;63:808–817.
  • Walters JRF, Johnston IM, Nolan JD, et al. The response of patients with bile acid diarrhoea to the farnesoid x receptor agonist obeticholic acid. Aliment Pharmacol Ther. 2015;41:54–64.
  • Gavva NR, Treanor JJS, Garami A, et al. Pharmacological blockade of the vanilloid receptor TRPV1 elicits marked hyperthermia in humans. Pain. 2008;136:202–210.
  • Kaneko Y, Szallasi A. Transient receptor potential (TRP) channels: a clinical perspective. Br J Pharmacol. 2014;171:2474–2507.
  • Szallasi A, Cortright DN, Blum CA, et al. The vanilloid receptor TRPV1: 10 years from channel cloning to antagonist proof-of-concept. Nat Rev Drug Discov. 2007;6:357–372.
  • Zhou Q, Yang L, Larson S, et al. Decreased miR-199 augments visceral pain in patients with IBS through translational upregulation of TRPV1. Gut. 2016;65:797–805.
  • Lehto SG, Tamir R, Deng H, et al. Antihyperalgesic effects of (R,E)-N-(2-hydroxy-2,3-dihydro-1H-inden-4-yl)-3-(2-(piperidin-1-yl)-4-(trifluoromethyl)phenyl)-acrylamide (AMG8562), a novel transient receptor potential vanilloid type 1 modulator that does not cause hyperthermia in rats. J Pharmacol Exp Ther. 2008;326:218–229.
  • Arendt-Nielsen L, Harris S, Whiteside GT, et al. A randomized, double-blind, positive-controlled, 3-way cross-over human experimental pain study of a TRPV1 antagonist (V116517) in healthy volunteers and comparison with preclinical profile. Pain. 2016;157:2057–2067.
  • Karrer T, Bartoshuk L. Effects of capsaicin desensitization on taste in humans. Physiol Behav. 1995;57:421–429.
  • Aniwan S, Gonlachanvit S. Effects of chili treatment on gastrointestinal and rectal sensation in diarrhea-predominant irritable bowel syndrome: a randomized, double-blinded, crossover study. J Neurogastroenterol Motil. 2014;20:400–406.
  • Harrington AM, Hughes PA, Martin CM, et al. A novel role for TRPM8 in visceral afferent function. Pain. 2011;152:1459–1468.
  • Cenac N, Bautzova T, Le Faouder P, et al. Quantification and potential functions of endogenous agonists of transient receptor potential channels in patients with irritable bowel syndrome. Gastroenterology. 2015;149:433–444.e7.
  • Salari P, Abdollahi M. Systematic review of modulators of benzodiazepine receptors in irritable bowel syndrome: is there hope? 2011;17:4251–4257.
  • Leventer SM, Raudibaugh K, Frissora CL, et al. Clinical trial: dextofisopam in the treatment of patients with diarrhoea-predominant or alternating irritable bowel syndrome. Aliment Pharmacol Ther. 2008;27:197–206.
  • Manocha M, Khan WI. Serotonin and GI disorders: an update on clinical and experimental studies. Clin Transl Gastroenterol. 2012;3:e13.
  • Reiter RJ, Tan DX, Mayo JC, et al. Neurally-mediated and neurally-independent beneficial actions of melatonin in the gastrointestinal tract. J Physiol Pharmacol. 2003;54 Suppl 4:113–125.
  • Matheus N, Mendoza C, Iceta R, et al. Melatonin inhibits serotonin transporter activity in intestinal epithelial cells. J Pineal Res. 2010;48:332–339.
  • Buendia I, Gómez-Rangel V, González-Lafuente L, et al. Neuroprotective mechanism of the novel melatonin derivative Neu-P11 in brain ischemia related models. Neuropharmacology. 2015;99:187–195.
  • Carocci A, Catalano A, Sinicropi MS. Melatonergic drugs in development. Clin Pharmacol Adv Appl. 2014;6:127–137.
  • Mosińska P, Socała K, Nieoczym D, et al. Anticonvulsant activity of melatonin, but not melatonin receptor agonists Neu-P11 and Neu-P67, in mice. Behav Brain Res. 2016;307:199–207.
  • He P, Ouyang X, Zhou S, et al. A novel melatonin agonist Neu-P11 facilitates memory performance and improves cognitive impairment in a rat model of Alzheimer’ disease. Horm Behav. 2013;64:1–7.
  • Tian S, Laudon M, Han L, et al. Antidepressant- and anxiolytic effects of the novel melatonin agonist Neu-P11 in rodent models. Acta Pharmacol Sin. 2010;31:775–783.
  • Huang L, Zhang C, Hou Y, et al. Blood pressure reducing effects of piromelatine and melatonin in spontaneously hypertensive rats. Eur Rev Med Pharmacol Sci. 2013;17:2449–2456.
  • Storr L-M-U of M. The Effect of Neu-P11 on Symptoms in Patients With D-IBS (Neu-P11) Available from: [cited 2017 Sep 1]. https://clinicaltrials.gov/ct2/show/NCT01558284?term=NCT01558284&rank=1
  • Papadopoulos V, Baraldi M, Guilarte TR, et al. Translocator protein (18 kDa): new nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol Sci. 2006;27:402–409.
  • Rupprecht R, Papadopoulos V, Rammes G, et al. Translocator protein (18 kDa) (TSPO) as a therapeutic target for neurological and psychiatric disorders. Nat Rev Drug Discov. 2010;9:971–988.
  • Whitehead WE, Duffy K, Sharpe J, et al. Randomised clinical trial: exploratory phase 2 study of ONO-2952 in diarrhoea-predominant irritable bowel syndrome. Aliment Pharmacol Ther. 2016;45:14-26.
  • EvaluatePharma. EvaluatePharma – USA sales by indication (IBS-D). [cited 2017 Sep 9].
  • Beckers AB, Weerts ZZRM, Helyes Z, et al. Review article: transient receptor potential channels as possible therapeutic targets in irritable bowel syndrome. Aliment Pharmacol Ther. 2017;46:938-952.
  • Balestra B, Vicini R, Cremon C, et al. Colonic mucosal mediators from patients with irritable bowel syndrome excite enteric cholinergic motor neurons. Neurogastroenterol Motil. 2012;24:1118-e570.
  • Barbaro MR, Di Sabatino A, Cremon C, et al. Interferon-γ is increased in the gut of patients with irritable bowel syndrome and modulates serotonin metabolism. Am J Physiol Gastrointest Liver Physiol. 2016;310:G439–G447.

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.