Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Dec 1.
Published in final edited form as: Curr Opin Pharmacol. 2008 Sep 26;8(6):697–703. doi: 10.1016/j.coph.2008.08.009

Visceral analgesics: drugs with a great potential in functional disorders?

Sylvie Bradesi 1,2, Jeremy Herman 1, Emeran A Mayer 1
PMCID: PMC2651816  NIHMSID: NIHMS94862  PMID: 18786654

Abstract

Irritable bowel syndrome remains an incompletely understood, common syndrome with significant unmet medical needs. In IBS patients, abdominal pain is a primary factor related to quality of life impairment, symptom severity and health care utilization, and chronic visceral hyperalgesia has been identified as an important aspect of IBS pathophysiology. However, the development of therapies aimed at reducing this hyperalgesia (visceral analgesics) has been only partially successful despite preclinical evidence supporting the potential usefulness of several preclinical compounds aimed at peripheral as well as central targets.

Introduction

Abdominal pain and discomfort in the absence of detectable organic disease are the hallmark of functional GI disorders (FGIDs). When such symptoms are referred to the lower abdomen and are associated with altered bowel habits, they make up the symptom complex of irritable bowel syndrome (IBS). Efforts during the past decade to develop highly effective drugs to treat patients with IBS have largely been disappointing. Although several novel treatments aimed at normalizing bowel movements have been developed recently, the overall effect of such therapies on patients’ well being and quality of life is relatively small, with an effect size over placebo generally not above 15%.

An alternative approach in the drug development efforts for FGID has targeted the abdominal pain component of the syndrome. Visceral hypersensitivity, determined experimentally in patients (as increased perceptual ratings of controlled visceral distension) and in animal models (as pseudo affective reflex responses), has been considered as a reliable marker of the disease [1••]. Even though there is only a modest correlation between experimentally determined colorectal sensitivity in rodent models and perceptual sensitivity to rectal distension in human subjects, and between the latter and IBS symptoms [2], the traditional approach to the development of new visceral analgesics has used a translational strategy to take candidate compounds from rodent models to human sensitivity testing. Candidate compounds are either molecules which had been developed for other targets (e.g. motility modulators such as tegaserod or alosetron; or centrally acting drugs such as serotonin-specific reuptake inhibitors [SSRIs]), or molecules specifically developed to target membrane receptors or ion channels on visceral afferent pathways which play a role in visceral mechanotransduction and which may be upregulated in models of visceral hyperalgesia. Ideally, candidate drugs should have no analgesic effects (e.g. not affect normal afferent sensitivity) but only antihyperalgesic effects (e.g. normalize enhanced visceral sensitivity). One major challenge in the development of visceral analgesics for IBS therapy is the fact that three major questions remain unanswered: Firstly, is the primary abnormality that leads to the observed enhanced perception of visceral signals, first, increased peripheral encoding and transduction of stimuli, second, central pain amplification or third, both? Secondly, are drugs specifically targeted at peripheral afferent mechanisms (analgesics or antihyperalgesics) relevant for effective IBS therapy? Thirdly, are drugs aimed at reducing central pain amplification relevant for IBS therapy?

Drugs and treatments in clinical development

Serotonin receptor modulators

The serotonin (5-HT) signaling system is widely distributed within the brain–gut axis and with potential effects on visceral afferent pathways and central pain modulation mechanisms [3••]. Several agents targeting various 5-HT receptors have been developed as modulatory agents of gut functions, including viscerosensory functions, though the precise roles of the various 5-HT-related mechanisms in IBS pathophysiology remain to be established.

5-HT3-receptor antagonists

5-HT3Rs are expressed on subsets of neurons intrinsic to the enteric nervous system including intrinsic primary afferent neurons (IPANs), as well as on extrinsic primary afferents (EPANs; both spinal and vagal afferents). Several 5-HT3Rs antagonists have been developed for the treatment of diarrhea-predominant IBS (IBS-D), and unequivocal evidence for their clinical effectiveness in treating several IBS symptoms, including diarrhea and abdominal pain has been reported (reviewed in Ref. [4]). The mechanism(s) by which abdominal pain and discomfort are reduced remains to be determined, but are unlikely due to a peripheral visceral analgesic effect as originally suggested. They may involve attenuating effects on central targets in the brain and spinal cord [5], while peripheral receptors on vagal afferents may actually mediate pronociceptive effects. Owing to rare but potentially serious side effects (ischemic colitis and severe constipation), the first such drug approved for use in female IBS patients with diarrhea (alosetron) is only available within a restricted access program. Two newer compounds, a selective 5-HT3R antagonist (DPP-733) and a combined norepinephrine (NE) reuptake inhibitor and 5-HT3R antagonist (NARI) have been evaluated in small clinical trials in IBS and preliminary results have been presented in abstract form [6,7]. Efforts aimed at understanding the mechanisms underlying the side effects of ischemic colitis and severe constipation, and the role of several receptor subtypes located on different intestinal and neuronal cells have only been partially successful.

5-HT4R agonists

Although there is extensive clinical and preclinical evidence that serotonin [8], via 5-HT4Rs, plays a pivotal role in the modulation of gastrointestinal motor function, a possible role of 5-HT4Rs in the modulation of visceral afferent function remains controversial. Some preclinical [8] and clinical evidence [9] suggest that the partial 5-HT4R agonist tegaserod may exert a modulatory effect on visceral afferent pathways. It remains to be determined if these effects are mediated by 5-HT4 receptors or other 5-HT receptor subtypes, such as the 5-HT2 receptors. The marketing of tegaserod, the first commercially available 5-HT4 receptor agonist, was suspended in March 2007, when an analysis of the data from clinical trials identified a significant increase in the number of cardiovascular ischemic events (myocardial infarction, stroke, and unstable angina) in patients taking the drug [10]. The possible visceral analgesic effect of more specific and more potent, full agonists of the 5-HT4R receptors as well as mixed 5-HT4R agonist and 5-HT3R antagonist activities have not been reported.

Probiotics

Beneficial effects of probiotics on gastrointestinal functions have been proposed based on their ability to modulate pathogenic bacteria adherence, enhance barrier function of the epithelium, alter mucosal response to stress as well as from their immunomodulatory properties. Evidence for possible antihyperalgesic effect of probiotics has been reported in several animal models. For example, the administration of Escherichia coli Nissle 1917 (EcN) was found to reduce TNBS (2,4,4-trinitrobenzenesulfonic acid) colitis associated with visceral hyperalgesia in rats [11], while Lactobacillus paracasei reduced the visceral hyperalgesia associated with antibiotic-induced inflammation in healthy mice [12]. A modulatory role of L. paracasei on the synthesis of neuro-peptides involved in nociception was suggested in this effect. Similarly, L. paracasei was found to reduce visceral hyperalgesia in response to restraint stress in rats [13]. Interestingly, oral administration of Lactobacillus acidophilus produced both visceral analgesic and antihyperalgesic effects in rats, and these behavioral changes were associated with the increased expression of mu-opioid and cannabinoid receptors in intestinal epithelial cells, suggesting a mechanism of action different from anti-inflammatory properties [14•]. At the clinical level, whereas increasing data have emerged on the potential beneficial role of probiotics in IBS [15•,1618], inadequate reporting of trial methodology and the assessment of poorly defined composite endpoint generally used in these studies, which includes nonpainful discomfort such as bloating, makes it difficult to reach definitive conclusions on true visceroanalgesic effects of probiotics in human patients.

Adrenoceptor (AR) modulators

Similar to 5-HT receptors, αARs are widely distributed within the brain–gut axis and have the potential to modulate sensitivity of visceral afferents, spinal cord transmission, and central pain modulation [19]. There is evidence of sympathetic nervous system dysfunction in a subgroup of patients with IBS [20]. Converging evidence suggest a role for abnormal noradrenergic signaling involving αARs in patients with chronic functional pain, including IBS.

Alpha2-adrenoceptor agonists

The α2ARs agonist clonidine reduced the perception of colonic or rectal balloon distension in healthy volunteers. As this effect was associated with colonic and rectal wall relaxation, no direct visceroanalgesic effect was demonstrated [21]. In an exploratory RCT of clonidine in IBS-D patients, clonidine was associated with satisfactory symptoms relief compared to placebo [22]. No data on more selective agonists for treatment in IBS have been reported till date.

Beta3-adrenoceptor agonists

Beta3-adrenoceptors (β3-AR) have been under investigation as novel targets for functional gastrointestinal disorders in which abdominal discomfort and pain are key features. In a rat model of visceral pain induced by intracolonic mustard oil, the β3-AR agonist elicited somatostatin-dependent visceral analgesia [23]. In addition, in view of several older reports on the visceroanalgesic effect of the somatostatin analog octreotide in human subjects [24] and the preliminary results about a favorable effect on IBS symptoms [25], this class of drug holds promise for IBS therapy.

CRF1R antagonists

Extensive preclinical evidence supports an important role of the brain CRF-CRF1R signaling system in mediating the endocrine, autonomic, behavioral, and pain modulatory responses to stress, suggesting that these receptors might be an ideal target in the context of functional bowel disorders [26]. Specifically, consistent antihyperalgesic effects of CRF1R antagonism on stress-induced visceral hyperalgesia has been demonstrated in different stress models in rats [2730]. In humans, a recent study from Sagami et al. reported an inhibitory effect of intravenous injection of the CRF-receptor antagonist α-helical CRF9–41 on abdominal pain and anxiety scores in a model of colonic distension and electrical stimulation of the rectal mucosa in IBS-D patients [31]. As this compound is not thought to cross the blood–brain barrier, the findings suggest the possibility that antagonism of peripheral CRF1R may have therapeutic effects in IBS patients. Additional evidence for a peripheral role of CRF in IBS comes from a recent study in which peripheral administration of a CRF antagonist, α-helical CRH9–41 (αhCRF) in IBS patients improved decreased alpha power spectra and increased beta power spectra of electroencephalogram (EEG) in response to colonic distension, compared with controls [32]. By contrast, a recent preliminary report on a clinical trial in female IBS-D patients with the selective CRF1R antagonist Bms-562086 did not show any significant effects on IBS symptoms (GI transit and bowel habits) [33]. It remains to be determined if the strong preclinical evidence supporting the usefulness of CRF1R antagonists in reversing stress-induced visceral hyperalgesia in rodents will translate into therapeutic efficacy in IBS patients, reducing symptoms of abdominal pain and discomfort.

Kappa opioid receptor agonists

Fedotozine is a κ opioid receptor (KOR) preferring agonist for which a peripheral antinociceptive mechanism of action had been proposed based on several studies in rodent models [34]. However, a series of elegant studies from G. Gebhart’s laboratory demonstrated that the apparent visceroanalgesic effect of these compounds was mediated by a combination of inhibition of Na channels on primary afferents together with central effects on MOR and KORs [35]. Even though a small number of phase IIa studies in IBS patients suggested a possible visceral analgesic effect [36], the majority of human studies, including two well-designed phase IIb studies were negative and further development of this compound was discontinued. Asimadoline, a different KOR agonist, failed to reduce the severity of abdominal pain in IBS patients with on-demand dosing schedule [37]. However, in a recent large (596 patients), randomized, placebo-controlled, 12-week, dose-ranging (0.15, 0.5, or 1.0 mg tablets, b.i.d.) study [38], asimadoline (0.5 mg) produced significant improvement on total number of months with adequate relief of IBS pain or discomfort (46.7% versus 20.0%), pain scores (week 12: −1.6 versus −0.7), and pain free days (42.9% versus 18.0%), in IBS-D patients with at least average moderate pain. Despite the encouraging recent clinical data, several questions remain: First, is the beneficial effect of asimadoline mediated by an effect on peripheral KOR on visceral afferents, or does it involve effects on other peripheral targets, such as Na channels? Second, is there a central component to the drug’s effectiveness and is it mediated by central opioid receptors? Third, what is the therapeutic window, between antagonism of peripheral KOR and the development of side effects mediated by central KOR, in particular dysphoria and diuresis.

Antidepressants

Preclinical and clinical evidence supports the effectiveness of tricyclic antidepressants (TCAs) in neuropathic pain [39]. In addition, the effect of SSRIs and particularly norepinephrine (NE)–serotonin (5-HT) reuptake inhibitor (NSRI) on enhancing the effectiveness of endogenous pain inhibition systems has been suggested. Despite the attractive rationale for using these centrally acting drugs in IBS patients, strong supportive evidence from well-designed clinical trials in IBS patients is currently not available. This is in contrast to the well-documented clinical effectiveness of both TCAs and NSRIs in the treatment of other chronic pain conditions [40•].

Low-dose tricyclic antidepressants

TCAs, although not FDA approved for IBS, are frequently used to treat IBS and while several randomized, placebo-controlled trials have supported the use of low-dose TCAs in the treatment of IBS patients [41], a systematic review of TCA trials for IBS failed to observe a beneficial effect of TCA on global IBS symptoms or abdominal pain, mainly because of insufficient statistical power (although they were effective against depressive and anxiety symptoms) [42]. In a large randomized 12-week placebo-controlled trial including 431 female adult patients, it was found that desipramine (150 mg/day) was not superior to placebo in the intention-to-treat analysis (probably linked to a high dropout rate due to side effects) [43]. It is unclear whether TCAs, in particular at doses greater than 50 mg qd act by influencing mood or anxiety or through an analgesic effect. It is also unclear whether their efficacy in IBS is owing to their effect not only on reuptake inhibition but also on their postsynaptic receptors. Chronic intake of low-dose amitriptyline has been shown to alter CNS processing of visceral sensory information during stressful conditions [44].

Selective and nonselective monoamine uptake inhibitors

SSRIs may have beneficial effect in IBS patients through central effects by reducing both anxiety and pain, yet, their efficacy for IBS remains to be confirmed. Both animal and human studies have indicated an analgesic effect of SSRIs in chronic pain conditions and it has been suggested that the improvement seen in IBS trials (only few randomized controlled trials) mainly relates to increase in overall well being and improvement of extra-intestinal symptoms rather than specific gut-related dysfunctions [45]. In a wide randomized, multicenter, controlled study comparing the effect of paroxetine, with psychotherapy and routine treatment care, the severity and frequency of abdominal pain improved in both the paroxetine and psychotherapy groups; but showed no statistically significant improvement when compared with routine treatment of care [46]. Newer monoamine reuptake inhibitors, such as the 5-HT and NE reuptake inhibitors (SNRIs) duloxetine and venlafaxine have been proposed as more effective treatments for chronic pain conditions associated with depression, and while they have been evaluated in patients with painful diabetic neuropathy [47] and fibromyalgia [48], their effect in IBS remain to be evaluated.

In summary, although a visceral analgesic effect of antidepressants has been demonstrated in animal models [49] supportive evidence from well-designed clinical trials in IBS patients is currently not available.

Pregabalin

Pregabalin (Lyrica), is a second-generation α2δ ligand that is approved for the treatment of neuropathic pain and epilepsy. Although its mechanism of action for pain relief remains unclear, it is believed to bind potently to the two auxiliary proteins associated with voltage-gated calcium channels, reducing depolarization-induced calcium influx at the nerve terminals, and consequently reducing the release of several excitatory neurotransmitters [50]. In animal models, pregabalin has been shown to be effective at reducing TNBS or LPS (lipopolysaccharide)-induced visceral hyperalgesia [51,52]. In a recent randomized, double-blind, placebo-controlled clinical trial in IBS patients, three weeks of oral treatment with pregabalin normalized the perception threshold for rectal distension in patients with rectal hypersensitivity [53]. A concomitant increase in rectal compliance was thought to be unrelated to the reduction in sensitivity.

Drugs in preclinical development

TRP channel antagonists

The transient receptor potential (TRP) family of ion channels are molecular sensory transducers involved in a wide range of processes including osmoregulation and sensing of thermal, chemical, and mechanical stimuli. [54•] The TRP family comprises five main members (TRPA, TRPC, TRPM, TRPP, and TRPV) among which TRP vanilloid TRPV1 and TRPV4 are involved in the encoding of chemical and mechanical stimuli on visceral afferents, and thus have attracted interest as possible new targets in the development of drugs for visceral pain.

TRPV1

Considerable preclinical evidence supports the potential role of TRPV1 in visceral hyperalgesia. First, TRPV1 is expressed on primary afferent fibers (as well as on epithelial cells lining the esophagus and the urinary bladder) and can become sensitized by proalgesic and inflammatory mediators. Second, there is evidence for an upregulation of the TRVP1 in animal models of postinflammatory visceral hyperalgesia as well as human GI syndromes characterized by enhanced visceral sensitivity, including idiopathic rectal hypersensitivity and fecal urgency (reviewed in [55•]). An upregulation of TRPV1 immunoreactivity has also been described in several visceral conditions where pain is a prominent symptom: on colonic biopsies of patients with active Crohn’s disease [56], on esophageal biopsies of patients with gastroesophageal reflux disease [57], on bladder biopsies of patients with interstitial cystitis [58] and on rectosigmoid biopsies of IBS patients [59]. In the latter study, the IBS patients also exhibited increased CD3+ cells as well as increased mast cells number in the mucosa, and showed evidence for a correlation between TRPV1 expression and IBS pain severity rating on a visual analog scale [59]. In this study, the role of prior infectious gastroenteritis and the possible role of altered mucosal immune activation as a potential trigger in the upregulation of TRPV1 have been suggested. Despite the potential of agents targeting TRPV1 for the treatment of visceral hyperalgesia states, the incomplete understanding of the role of TRPV1 in mucosal homeostatis, including thermosensing and protection of the GI mucosa constitutes a challenge to a safe therapeutic effect.

TRPV4

TRPV4 has been implicated in mechanosensation and pain and has been shown to play an important role in somatic pain [60]. In a recent study, TRPV4 was localized on colonic sensory afferents (in both mice and humans) where they are thought to be specifically involved in the transmission of nociceptive visceral stimuli. In tissue obtained from patients with active colitis, they observed that serosal blood vessels were more densely innervated by TRPV4-positive fibers [61]. In a different report, the role of TRPV4 in visceral nociception was confirmed by the observations in a mouse model, where intracolonic administration of a TRPV4 agonist was able to induce visceral hyperalgesia, and this effect was blocked by intervertebral knockdown of TRPV4 expression by siRNA. Similar siRNA treatment reduced basal visceral nociception and decreased visceral hypersensitivity induced by a PAR2 agonist [62]. Viewed together with a previous report suggesting a key role of PAR2 in IBS-related pain symptoms [63], these data suggest a potential role of TRPV4 in increased abdominal pain in IBS patients. Till date, there are no reports about TRPV4 expression in patients with visceral pain, or about the potential effectiveness of TRPV4 antagonists in such patients.

PAR

Protease-activated receptor type 2 (PAR2) is a member of the G-protein-coupled seven transmembrane-domain PAR receptors family that can be activated by trypsin and mast cell tryptase. The role of PAR2 activation in inflammation and its involvement in visceral nociceptive responses has been established in various animal models [64]. Elevated colonic luminal serine protease activity has been observed in IBS-D patients [63]. The involvement of proteases and PAR2 activation in the generation of pain symptoms has been suggested by the observation that mice injected with mediators released from colonic biopsies of patients with IBS exhibit enhanced nociceptive responses to colorectal distension whereas transgenic mice without PAR2 failed to show such visceral hyperalgesia [63].

CRF2R agonists

CRF has been shown to have receptor-based bimodal effects on multiple physiological responses, such as gastrointestinal motility and stress. Similarly, a divergent role of the CRF receptor subtypes has been suggested in the modulation of visceral pain. While the CRF1 receptor is involved in a pronociceptive effect of CRF, the CRF2 receptor can exert antinociceptive effects at both the peripheral and spinal level [65]. Additional evidence supporting an antinociceptive effect of CRF2R was provided by M. Mulugeta et al. showing that the activation of CRF2R blunted visceral pain and inhibited sensitization to colorectal distension in awake rats. These effects were completely blocked by a selective CRF2-receptor antagonist [66].

Compounds with potential visceral analgesic effects

Preliminary reports indicate that Na(v) channels [67] or acid-sensing ion channels (ASICS) [68] may be involved in visceral pain transduction, though their implication in the physiopathology of visceral hypersensitivity remains to be studied.

Conclusions

Despite the well-documented phenomenon of visceral hypersensitivity (enhanced perception of visceral signals) in IBS patients, the clinical results with drugs presumed to have visceral analgesic effects have largely been disappointing. One reason for the discrepant results between preclinical and clinical evaluations may be due to the fact that although many of these compounds are effective in reducing nociceptive reflex responses in rodents, this effectiveness may not necessarily translate into normalizing an abnormal human perception of visceral signals, which is influenced by many other factors besides visceral afferent input to the brain (discussed in [2]). A better characterization of normal as well as abnormal visceral pain perception (e.g. central pain amplification) and underlying mechanisms (sensory, cognitive, and emotional) in human patients is required before more effective drug development is possible.

Footnotes

Supported by NIH grants P50 DK64539 (EAM), R24 AT00281 (EAM), and R21 DK071767 (SB).

Publisher's Disclaimer: This article appeared in a journal published by Elsevier. The attached copy is furnished to the author for internal non-commercial research and education use, including for instruction at the authors institution and sharing with colleagues.

Other uses, including reproduction and distribution, or selling or licensing copies, or posting to personal, institutional or third party websites are prohibited.

In most cases authors are permitted to post their version of the article (e.g. in Word or Tex form) to their personal website or institutional repository. Authors requiring further information regarding Elsevier’s archiving and manuscript policies are encouraged to visit: http://www.elsevier.com/copyright

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

• of special interest

•• of outstanding interest

  • 1••.Azpiroz F, Bouin M, Camilleri M, Mayer EA, Poitras P, Spiller RC. Mechanisms of hypersensitivity in IBS and functional disorders. Neurogastroenterol Motil. 2007;19(Suppl 1):62–88. doi: 10.1111/j.1365-2982.2006.00875.x. A comprehensive review summarizing the evidence of visceral hypersensitivity as a biological marker of functional gut disorders, the peripheral and central mechanisms involved, and the role of inflammation on hypersensitivity. It also discusses the importance of peripheral mechanisms, like motor disturbances and alterations of the intraluminal milieu and genetics in visceral hypersensitivity. [DOI] [PubMed] [Google Scholar]
  • 2.Mayer EA, Bradesi S, Chang L, Spiegel BM, Bueller JA, Naliboff BD. Functional GI disorders: from animal models to drug development. Gut. 2008;57:384–404. doi: 10.1136/gut.2006.101675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3••.Gershon MD, Tack J. The serotonin signaling system: from basic understanding to drug development for functional GI disorders. Gastroenterology. 2007;132:397–414. doi: 10.1053/j.gastro.2006.11.002. The different aspects of serotonin production, signaling, and degradation are reviewed in normal conditions and abnormal GI physiology. The rationale for serotonin as a drug target in functional GI disease is covered with a comprehensive review of specific clinical and preclinical agents. [DOI] [PubMed] [Google Scholar]
  • 4.Andresen V, Montori VM, Keller J, West CP, Layer P, Camilleri M. Effects of 5-hydroxytryptamine (serotonin) type 3 antagonists on symptom relief and constipation in nonconstipated irritable bowel syndrome: a systematic review and meta-analysis of randomized controlled trials. Clin Gastroenterol Hepatol. 2008;6:545–555. doi: 10.1016/j.cgh.2007.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.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. doi: 10.1016/j.tips.2004.10.002. [DOI] [PubMed] [Google Scholar]
  • 6.Paterson WG, Ford D, Ganguli SC, Reynolds RP, Pliamm L, O’Mahony M, Pare P, Nurbhai S, Feagan B, Landau SB. A novel, oral 5HT3 partial agonist, DDP-733, improves overall response in patients with irritable bowel syndrome and constipation (IBS-C): a randomized, double-blind, placebo-controlled, parallel-group, dose-ranging study. Gastroenterology. 2008;134:A546–A547. [Google Scholar]
  • 7.Paterson WG, Springer J, O’Mahony M, Reynolds RP, Ganguli SC, Feagan B, Nurbhai S, Landau SB. A randomized, double-blind, placebo-controlled trial with a novel dual noradrenergic reuptake inhibitor (NARI) and 5-HT3 antagonist: results of a phase II 8-week study in female patients with diarrhea predominant irritable bowel syndrome (D-IBS) Gastroenterology. 2008;134:A50. [Google Scholar]
  • 8.Greenwood-Van Meerveld B, Venkova K, Hicks G, Dennis E, Crowell MD. Activation of peripheral 5-HT receptors attenuates colonic sensitivity to intraluminal distension. Neurogastroenterol Motil. 2006;18:76–86. doi: 10.1111/j.1365-2982.2005.00723.x. [DOI] [PubMed] [Google Scholar]
  • 9.Sabate JM, Bouhassira D, Poupardin C, Wagner A, Loria Y, Coffin B. Sensory signalling effects of tegaserod in patients with irritable bowel syndrome with constipation. Neurogastroenterol Motil. 2008;20:134–141. doi: 10.1111/j.1365-2982.2007.01030.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Pasricha PJ. Desperately seeking serotonin. A commentary on the withdrawal of tegaserod and the state of drug development for functional and motility disorders. Gastroenterology. 2007;132:2287–2290. doi: 10.1053/j.gastro.2007.04.057. [DOI] [PubMed] [Google Scholar]
  • 11.Liebregts T, Adam B, Bertel A, Jones S, Schulze J, Enders C, Sonnenborn U, Lackner K, Holtmann G. Effect of E. coli Nissle 1917 on post-inflammatory visceral sensory function in a rat model. Neurogastroenterol Motil. 2005;17:410–414. doi: 10.1111/j.1365-2982.2005.00638.x. [DOI] [PubMed] [Google Scholar]
  • 12.Verdu EF, Bercik P, Verma-Gandhu M, Huang XX, Blennerhassett P, Jackson W, Mao Y, Wang L, Rochat F, Collins SM. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut. 2006;55:182–190. doi: 10.1136/gut.2005.066100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Eutamene H, Lamine F, Chabo C, Theodorou V, Rochat F, Bergonzelli GE, Corthesy-Theulaz I, Fioramonti J, Bueno L. Synergy between Lactobacillus paracasei and its bacterial products to counteract stress-induced gut permeability and sensitivity increase in rats. J Nutr. 2007;137:1901–1907. doi: 10.1093/jn/137.8.1901. [DOI] [PubMed] [Google Scholar]
  • 14•.Rousseaux C, Thuru X, Gelot A, Barnich N, Neut C, Dubuquoy L, Dubuquoy C, Merour E, Geboes K, Chamaillard M, et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med. 2007;13:35–37. doi: 10.1038/nm1521. Interesting report studying the effect of the probiotic L. acidophilus and other strains, on intestinal pain. The role of opioid and cannabinoid receptors in this effect are studied both in vitro with human cells and in vivo in rats. [DOI] [PubMed] [Google Scholar]
  • 15•.Quigley EM, Flourie B. Probiotics and irritable bowel syndrome: a rationale for their use and an assessment of the evidence to date. Neurogastroenterol Motil. 2007;19:166–172. doi: 10.1111/j.1365-2982.2006.00879.x. Overview of the rationale that probiotics may alter pathophysiologic variables of IBS such as type and number of flora, immune activation, motility, and brain–gut dysregulation. The major clinical evidence is summarized. [DOI] [PubMed] [Google Scholar]
  • 16.Guyonnet D, Chassany O, Ducrotte P, Picard C, Mouret M, Mercier CH, Matuchansky C. Effect of a fermented milk containing Bifidobacterium animalis DN-173 010 on the health-related quality of life and symptoms in irritable bowel syndrome in adults in primary care: a multicentre, randomized, double-blind, controlled trial. Aliment Pharmacol Ther. 2007;26:475–486. doi: 10.1111/j.1365-2036.2007.03362.x. [DOI] [PubMed] [Google Scholar]
  • 17.Kajander K, Myllyluoma E, Rajilic-Stojanovic M, Kyronpalo S, Rasmussen M, Jarvenpaa S, Zoetendal EG, de Vos WM, Vapaatalo H, Korpela R. Clinical trial: multispecies probiotic supplementation alleviates the symptoms of irritable bowel syndrome and stabilizes intestinal microbiota. Aliment Pharmacol Ther. 2008;27:48–57. doi: 10.1111/j.1365-2036.2007.03542.x. [DOI] [PubMed] [Google Scholar]
  • 18.O’Mahony L, McCarthy J, Kelly P, Hurley G, Luo F, Chen K, O’Sullivan GC, Kiely B, Collins JK, Shanahan F, et al. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology. 2005;128:541–551. doi: 10.1053/j.gastro.2004.11.050. [DOI] [PubMed] [Google Scholar]
  • 19.Pertovaara A. Noradrenergic pain modulation. Prog Neurobiol. 2006;80:53–83. doi: 10.1016/j.pneurobio.2006.08.001. [DOI] [PubMed] [Google Scholar]
  • 20.Aggarwal A, Cutts TF, Abell TL, Cardoso S, Familoni B, Bremer J, Karas J. Predominant symptoms in irritable bowel syndrome correlate with specific autonomic nervous system abnormalities. Gastroenterology. 1994;106:945–950. doi: 10.1016/0016-5085(94)90753-6. [DOI] [PubMed] [Google Scholar]
  • 21.Malcolm A, Phillips SF, Camilleri M, Hanson RB. Pharmacological modulation of rectal tone alters perception of distension in humans. Am J Gastroenterol. 1997;92:2073–2079. [PubMed] [Google Scholar]
  • 22.Camilleri M, Kim DY, McKinzie S, Kim HJ, Thomforde GM, Burton DD, Low PA, Zinsmeister AR. A randomized, controlled exploratory study of clonidine in diarrhea-predominant irritable bowel syndrome. Clin Gastroenterol Hepatol. 2003;1:111–121. doi: 10.1053/cgh.2003.50019. [DOI] [PubMed] [Google Scholar]
  • 23.Cellek S, Thangiah R, Bassil AK, Campbell CA, Gray KM, Stretton JL, Lalude O, Vivekanandan S, Wheeldon A, Winchester WJ, et al. Demonstration of functional neuronal beta3-adrenoceptors within the enteric nervous system. Gastroenterology. 2007;133:175–183. doi: 10.1053/j.gastro.2007.05.009. [DOI] [PubMed] [Google Scholar]
  • 24.Schwetz I, Naliboff B, Munakata J, Lembo T, Chang L, Matin K, Ohning G, Mayer EA. Anti-hyperalgesic effect of octreotide in patients with irritable bowel syndrome. Aliment Pharmacol Ther. 2004;19:123–131. doi: 10.1111/j.1365-2036.2004.01774.x. [DOI] [PubMed] [Google Scholar]
  • 25.Klooker TK, Kuiken SD, Lei A, Boeckxstaens GE. Effect of long-term treatment with octreotide on rectal sensitivity in patients with non-constipated irritable bowel syndrome. Aliment Pharmacol Ther. 2007;26:605–615. doi: 10.1111/j.1365-2036.2007.03398.x. [DOI] [PubMed] [Google Scholar]
  • 26.Taché Y, Martinez V, Million M, Maillot C. Role of corticotropin releasing factor receptor subtype 1 in stress-related functional colonic alterations: implications in irritable bowel syndrome. Eur J Surg Suppl. 2002;587:16–22. [PubMed] [Google Scholar]
  • 27.Schwetz I, McRoberts JA, Coutinho SV, Bradesi S, Mulugeta M, Miller JC, Zhou H, Ohning G, Mayer EA. CRF1 receptor mediates acute and sustained visceral hyperalgesia following an acute stressor in maternally separated Long Evans rats. Gastroenterology. 2003;124(Suppl 1):A612. [Google Scholar]
  • 28.Larauche M, Bradesi S, Million M, McLean P, Tache Y, Mayer EA, McRoberts JA. Corticotropin-releasing factor type 1 receptors mediate the visceral hyperalgesia induced by repeated psychological stress in rats. Am J Physiol Gastrointest Liver Physiol. 2008;294:G1033–G1040. doi: 10.1152/ajpgi.00507.2007. [DOI] [PubMed] [Google Scholar]
  • 29.Schwetz I, Bradesi S, McRoberts JA, Sablad M, Miller JC, Zhou H, Ohning G, Mayer EA. Delayed stress-induced colonic hypersensitivity in male Wistar rats: role of neurokinin-1 and corticotropin-releasing factor-1 receptors. Am J Physiol: Gastrointest Liver Physiol. 2004;286:G683–G691. doi: 10.1152/ajpgi.00358.2003. [DOI] [PubMed] [Google Scholar]
  • 30.Schwetz I, McRoberts JA, Coutinho SV, Bradesi S, Gale G, Fanselow M, Million M, Ohning G, Taché Y, Plotsky PM, et al. Corticotropin-releasing factor receptor 1 mediates acute and delayed stress-induced visceral hyperalgesia in maternally separated Long Evans rats. Am J Physiol: Gastrointest Liver Physiol. 2005;289:G704–G712. doi: 10.1152/ajpgi.00498.2004. [DOI] [PubMed] [Google Scholar]
  • 31.Sagami Y, Shimada Y, Tayama J, Nomura T, Satake M, Endo Y, Shoji T, Karahashi K, Hongo M, Fukudo S. Effect of a corticotropin releasing hormone receptor antagonist on colonic sensory and motor function in patients with irritable bowel syndrome. Gut. 2004;53:958–964. doi: 10.1136/gut.2003.018911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tayama J, Sagami Y, Shimada Y, Hongo M, Fukudo S. Effect of alpha-helical CRH on quantitative electroencephalogram in patients with irritable bowel syndrome. Neurogastroenterol Motil. 2007;19:471–483. doi: 10.1111/j.1365-2982.2007.00903.x. [DOI] [PubMed] [Google Scholar]
  • 33.Sweetser SR, Linker Nord SJ, Burton DD, Grudell A, Eckert DJ, Manini ML, Busciglio I, Tong G, Dockens RC, Zinsmeister AR, et al. Effects of a novel corticotrophin releasing factor receptor-1 antagonist, Bms-562086, on gastrointestinal and colonic transit and bowel habits in patients with diarrhea-predominant irritable bowel syndrome (D-IBS) DDW 2008 Presented Abstract. 2008 [Google Scholar]
  • 34.Ozaki N, Sengupta JN, Gebhart GF. Differential effects of mu-, delta-, and kappa-opioid receptor agonists on mechanosensitive gastric vagal afferent fibers in the rat. J Neurophysiol. 2000;83:2209–2216. doi: 10.1152/jn.2000.83.4.2209. [DOI] [PubMed] [Google Scholar]
  • 35.Su X, Wachtel RE, Gebhart GF. Inhibition of calcium currents in rat colon sensory neurons by K- but not mu- or delta-opioids. J Neurophysiol. 1998;80:3112–3119. doi: 10.1152/jn.1998.80.6.3112. [DOI] [PubMed] [Google Scholar]
  • 36.Dapoigny M, Abitbol JL, Fraitag B. Efficacy of peripheral k-agonist fedotozine versus placebo in treatment of irritable bowel syndrome: a multicenter dose–response study. Digest Dis Sci. 1995;40:2244–2249. doi: 10.1007/BF02209014. [DOI] [PubMed] [Google Scholar]
  • 37.Szarka LA, Camilleri M, Burton D, Fox JC, McKinzie S, Stanislav T, Simonson J, Sullivan N, Zinsmeister AR. Efficacy of on-demand asimadoline, a peripheral kappa-opioid agonist, in females with irritable bowel syndrome. Clin Gastroenterol Hepatol. 2007;5:1268–1275. doi: 10.1016/j.cgh.2007.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Mangel AW, Bornstein JD, Hamm LR, Buda J, Wang J, Irish W, Urso D. Clinical trial: asimadoline in the treatment of patients with irritable bowel syndrome. Aliment Pharmacol Ther. 2008;28:239–249. doi: 10.1111/j.1365-2036.2008.03730.x. [DOI] [PubMed] [Google Scholar]
  • 39.Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev. 2007:CD005454. doi: 10.1002/14651858.CD005454.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40•.Jann MW, Slade JH. Antidepressant agents for the treatment of chronic pain and depression. Pharmacotherapy. 2007;27:1571–1587. doi: 10.1592/phco.27.11.1571. A comprehensive review discussing the potential beneficial effect of antidepressants in medical conditions associated with chronic pain other than depression and anxiety disorders. [DOI] [PubMed] [Google Scholar]
  • 41.Clouse RE. Antidepressants for irritable bowel syndrome. Gut. 2003;52:598–599. doi: 10.1136/gut.52.4.598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Brandt LJ, Bjorkman D, Fennerty MB, Locke GR, Olden K, Peterson W, Quigley E, Schoenfeld P, Schuster M, Talley N. Systematic review on the management of irritable bowel syndrome in North America. Am J Gastroenterol. 2002;97(11 Suppl):S7–S26. doi: 10.1016/s0002-9270(02)05657-5. [DOI] [PubMed] [Google Scholar]
  • 43.Drossman DA, Toner BB, Whitehead WE, Diamant NE, Dalton CB, Duncan S, Emmott S, Proffitt V, Akman D, Frusciante K, et al. Cognitive-behavioral therapy versus education and desipramine versus placebo for moderate to severe functional bowel disorders. Gastroenterology. 2003;125:19–31. doi: 10.1016/s0016-5085(03)00669-3. [DOI] [PubMed] [Google Scholar]
  • 44.Morgan V, Pickens D, Gautam S, Kessler R, Mertz H. Amitriptyline reduces rectal pain related activation of the anterior cingulate cortex in patients with irritable bowel syndrome. Gut. 2005;54:601–607. doi: 10.1136/gut.2004.047423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Pae CU, Masand PS, Ajwani N, Lee C, Patkar AA. Irritable bowel syndrome in psychiatric perspectives: a comprehensive review. Int J Clin Pract. 2007;61:1708–1718. doi: 10.1111/j.1742-1241.2007.01409.x. [DOI] [PubMed] [Google Scholar]
  • 46.Creed F, Fernandes L, Guthrie E, Palmer S, Ratcliffe J, Read N, Rigby C, Thompson D, Tomenson B North of England IBSRG. The cost-effectiveness of psychotherapy and paroxetine for severe irritable bowel syndrome. Gastroenterology. 2003;124:303–317. doi: 10.1053/gast.2003.50055. [DOI] [PubMed] [Google Scholar]
  • 47.Raskin J, Pritchett YL, Wang F, D’Souza DN, Waninger AL, Iyengar S, Wernicke JF. A double-blind, randomized multicenter trial comparing duloxetine with placebo in the management of diabetic peripheral neuropathic pain. Pain Med (Malden, MA) 2005;6:346–356. doi: 10.1111/j.1526-4637.2005.00061.x. [DOI] [PubMed] [Google Scholar]
  • 48.Arnold LM. Duloxetine and other antidepressants in the treatment of patients with fibromyalgia. Pain Med. 2007;8(Suppl 2):S63–S74. doi: 10.1111/j.1526-4637.2006.00178.x. [DOI] [PubMed] [Google Scholar]
  • 49.Leventhal L, Smith V, Hornby G, Andree TH, Brandt MR, Rogers KE. Differential and synergistic effects of selective norepinephrine and serotonin reuptake inhibitors in rodent models of pain. J Pharmacol Exp Ther. 2007;320:1178–1185. doi: 10.1124/jpet.106.109728. [DOI] [PubMed] [Google Scholar]
  • 50.Ben-Menachem E. Pregabalin pharmacology and its relevance to clinical practice. Epilepsia. 2004;45(Suppl 6):13–18. doi: 10.1111/j.0013-9580.2004.455003.x. [DOI] [PubMed] [Google Scholar]
  • 51.Diop L, Raymond F, Fargeau H, Petoux F, Chovet M, Doherty AM. Pregabalin (CI-1008) inhibits the trinitrobenzene sulfonic acid-induced chronic colonic allodynia in the rat. J Pharmacol Exp Ther. 2002;302:1013–1022. doi: 10.1124/jpet.302.3.1013. [DOI] [PubMed] [Google Scholar]
  • 52.Eutamene H, Coelho AM, Theodorou V, Toulouse M, Chovet M, Doherty A, Fioramonti J, Bueno L. Antinociceptive effect of pregabalin in septic shock-induced rectal hypersensitivity in rats. J Pharmacol Exp Ther. 2000;295:162–167. [PubMed] [Google Scholar]
  • 53.Houghton LA, Fell C, Whorwell PJ, Jones I, Sudworth DP, Gale JD. Effect of a second-generation alpha2delta ligand (pregabalin) on visceral sensation in hypersensitive patients with irritable bowel syndrome. Gut. 2007;56:1218–1225. doi: 10.1136/gut.2006.110858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54•.Levine JD, Alessandri-Haber N. TRP channels: targets for the relief of pain. Biochim Biophys Acta. 2007;1772:989–1003. doi: 10.1016/j.bbadis.2007.01.008. A short review focusing on the contribution of transient receptor potential (TRP) channels to pain hypersensitivity associated with peripheral inflammatory and neuropathic pain states. [DOI] [PubMed] [Google Scholar]
  • 55•.Holzer P. TRPV1: a new target for treatment of visceral pain in IBS? Gut. 2008;57:882–884. doi: 10.1136/gut.2008.149724. A short review discussing the potential role of TRPV1 in the modulation of visceral pain and the potential use of agents modulating TRPV1 activity in the treatment of visceral pain in IBS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yiangou Y, Facer P, Dyer NH, Chan CL, Knowles C, Williams NS, Anand P. Vanilloid receptor 1 immunoreactivity in inflamed human bowel. Lancet. 2001;357:1338–1339. doi: 10.1016/s0140-6736(00)04503-7. [DOI] [PubMed] [Google Scholar]
  • 57.Bhat YM, Bielefeldt K. Capsaicin receptor (TRPV1) and non-erosive reflux disease. Eur J Gastroenterol Hepatol. 2006;18:263–270. doi: 10.1097/00042737-200603000-00006. [DOI] [PubMed] [Google Scholar]
  • 58.Mukerji G, Yiangou Y, Agarwal SK, Anand P. Transient receptor potential vanilloid receptor subtype 1 in painful bladder syndrome and its correlation with pain. J Urol. 2006;176:797–801. doi: 10.1016/j.juro.2006.03.074. [DOI] [PubMed] [Google Scholar]
  • 59.Akbar A, Yiangou Y, Facer P, Walters JR, Anand P, Ghosh S. Increased capsaicin receptor TRPV1-expressing sensory fibres in irritable bowel syndrome and their correlation with abdominal pain. Gut. 2008;57:923–929. doi: 10.1136/gut.2007.138982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Suzuki M, Mizuno A, Kodaira K, Imai M. Impaired pressure sensation in mice lacking TRPV4. J Biol Chem. 2003;278:22664–22668. doi: 10.1074/jbc.M302561200. [DOI] [PubMed] [Google Scholar]
  • 61.Brierley SM, Page AJ, Hughes PA, Adam B, Liebregts T, Cooper NJ, Holtmann G, Liedtke W, Blackshaw LA. Selective role for TRPV4 ion channels in visceral sensory pathways. Gastroenterology. 2008;134:2059–2069. doi: 10.1053/j.gastro.2008.01.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Cenac N, Altier C, Chapman K, Liedtke W, Zamponi G, Vergnolle N. Transient receptor potential vanilloid-4 has a major role in visceral hypersensitivity symptoms. Gastroenterology. 2008 May 10; doi: 10.1053/j.gastro.2008.05.024. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 63.Cenac N, Andrews CN, Holzhausen M, Chapman K, Cottrell G, Andrade-Gordon P, Steinhoff M, Barbara G, Beck P, Bunnett NW, et al. Role for protease activity in visceral pain in irritable bowel syndrome. J Clin Invest. 2007;117:636–647. doi: 10.1172/JCI29255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Coelho AM, Vergnolle N, Guiard B, Fioramonti J, Bueno L. Proteinases and proteinase-activated receptor 2: a possible role to promote visceral hyperalgesia in rats. Gastroenterology. 2002;122:1035–1047. doi: 10.1053/gast.2002.32387. [DOI] [PubMed] [Google Scholar]
  • 65.Nijsen M, Ongenae N, Meulemans A, Coulie B. Divergent role for CRF1 and CRF2 receptors in the modulation of visceral pain. Neurogastroenterol Motil. 2005;17:423–432. doi: 10.1111/j.1365-2982.2005.00644.x. [DOI] [PubMed] [Google Scholar]
  • 66.Million M, Wang L, Wang Y, Adelson DW, Yuan PQ, Maillot C, Coutinho SV, McRoberts JA, Bayati A, Mattsson H, et al. CRF2 receptor activation prevents colorectal distension induced visceral pain and spinal ERK1/2 phosphorylation in rats. Gut. 2006;55:172–181. doi: 10.1136/gut.2004.051391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Martinez V, Melgar S. Lack of colonic inflammation-induced acute visceral hypersensitivity to colorectal distension in Na(v)1.9 knockout mice. Eur J Pain. 2008;12:934–944. doi: 10.1016/j.ejpain.2007.12.011. [DOI] [PubMed] [Google Scholar]
  • 68.Page AJ, Brierley SM, Martin CM, Hughes PA, Blackshaw LA. Acid sensing ion channels 2 and 3 are required for inhibition of visceral nociceptors by benzamil. Pain. 2007;133:150–160. doi: 10.1016/j.pain.2007.03.019. [DOI] [PubMed] [Google Scholar]

RESOURCES