<<

REVIEW ARTICLE published: 01 February 2013 doi: 10.3389/fphar.2013.00007 development for the : current challenges and future perspectives

Fabrizio De Ponti*

Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy

Edited by: are frequently used for the treatment of patients with the irritable bowel syn- Angelo A. Izzo, University of Naples drome (IBS), although their actual benefit is often debated. In fact, the recent progress in Federico II, Italy our understanding of the pathophysiology of IBS, accompanied by a large number of preclin- Reviewed by: Elisabetta Barocelli, University of ical and clinical studies of new , has not been matched by a significant improvement Parma, Italy of the armamentarium of medications available to treat IBS. The aim of this review is to Raffaele Capasso, University of outline the current challenges in drug development for IBS, taking advantage of what we Naples Federico II, Italy have learnt through the Rome process (Rome I, Rome II, and Rome III). The key questions *Correspondence: that will be addressed are: (a) do we still believe in the “magic bullet,” i.e., a very selective Fabrizio De Ponti, Pharmacology Unit, Department of Medical and Surgical drug displaying a single receptor mechanism capable of controlling IBS symptoms? (b) IBS Sciences, University of Bologna, Via is a “functional disorder” where complex neuroimmune and brain-gut interactions occur Irnerio, 48, 40126 Bologna, Italy. and minimal inflammation is often documented: do we need to target gut motility, vis- e-mail: [email protected] ceral sensitivity, or minimal inflammation? (c) are there validated biomarkers (accepted by regulatory agencies) for studies of sensation and motility with experimental medications in humans? (d) do animal models have predictive and translational value? (e) in the era of personalized medicine, does pharmacogenomics applied to these medications already play a role? Finally, this review will briefly outline medications currently used or in devel- opment for IBS. It is anticipated that a more focused interaction between basic science investigators, pharmacologists, and clinicians will lead to better treatment of IBS.

Keywords: drug targets, translational medical research, brain-gut interactions, drug selectivity, biomarkers, 5-hydroxytryptamine, transient receptor potential channels, neuroimmune intestinal interactions

INTRODUCTION sensitivity, or minimal inflammation? (c) are there validated bio- Irritable bowel syndrome (IBS) is a common functional gastroin- markers (accepted by regulatory agencies) for studies of sensation testinal disorder, characterized by recurrent abdominal pain or and motility with experimental medications in humans? (d) do discomfort in combination with disturbed bowel habits in the animal models have predictive and translational value? (e) in the absence of identifiable organic cause. Many medications are used era of personalized medicine, does pharmacogenomics applied for the treatment of patients with IBS, although their actual benefit to these medications already play a role? Finally, this review will is often a matter of debate. In particular, only a few are specif- briefly outline medications currently used or in development for ically labeled for IBS. In fact, notwithstanding great progress in IBS. It is anticipated that a more focused interaction between basic our understanding of the pathophysiology of IBS thanks to a large science investigators, pharmacologists, and clinicians will lead to number of preclinical and clinical studies of new drugs,the specific better treatment of IBS. armamentarium of medications available is scant. The aim of this review is to outline the current challenges in drug development for THE “MAGIC BULLET”: A CONCEPT THAT NEEDS IBS, taking advantage of what we have learned through the Rome RETHINKING process (from Rome I in the 1980s to Rome III published in 2006; The key pharmacodynamics, pharmacokinetic, and safety features Drossman, 2006). for drugs to be used in the treatment of IBS are outlined in Table 1. The key questions that will be addressed are: (a) do we still A selective drug is defined as a compound interacting only with believe in the “magic bullet,”i.e., a very selective drug displaying a one receptor subtype and leaving other receptors unaffected at single receptor mechanism capable of controlling IBS symptoms? concentrations achieved at therapeutic doses. The literature on (b) IBS is a “functional disorder” where complex neuroimmune the treatment of IBS has often resorted to the concept of the and brain-gut interactions occur and minimal inflammation is “magic bullet,” i.e., a very selective drug displaying a single recep- often documented: do we need to target gut motility, visceral tor mechanism capable of controlling IBS symptoms (Camilleri et al., 2006a). This was often considered the key to efficacy avoid- ing side-effects. This approach is no longer ideal because of several

Abbreviations: EMA, European Medicines Agency; ENS, enteric nervous system; important pitfalls. FDA, food and drug administration; 5-HT, 5-hydroxytryptamine; IBS, irritable First, drug selectivity is always a relative concept, which ignores bowel syndrome. the basic fact that most molecules, even at therapeutic doses,

www.frontiersin.org February 2013 | Volume 4 | Article 7 | 1 De Ponti New drugs for IBS

Table 1 | Key features for drugs to be used in the treatment of IBS. The third issue is that mechanisms underlying symptoms in IBS may differ among patients, hence the need to consider using Key features multiple therapies. With selective drugs, primary clinical end- points were achieved in less than 70% of patients with the approved Pharmacodynamics The drug should target a whole pathophysiological agents such as tegaserod or (Camilleri et al., 2000; mechanism rather than a single receptor Muller-Lissner et al., 2001; Cremonini et al., 2003). On the other Possible targets: motility, secretion, visceral hand, it seems reasonable to propose treatment with combination sensitivity, neuroimmune interactions/minimal therapy, which is the rule when treating medical conditions such as inflammation, brain-gut axis hypertension or ,when monotherapy is no longer adequate. The effect should be maintained over time during Because of the redundancy of mechanisms controlling neurosen- treatment sory, neuromuscular, and neuroimmune functions in the gut, it is conceivable that effective treatment of functional gut disorders Good oral (unless local action in the gut may require combination therapy. is specifically wanted) One example is provided by antagonists, Half-life allows once daily dosing which have so far given disappointing results because of inherent No metabolites with different or unwanted differences among animal models and humans: it has been sug- pharmacological actions gested that the analgesic efficacy of multi- or pan-tachykinin recep- Avoid CYP substrates with high likelihood of drug tor antagonists is superior to that of mono-receptor antagonists interactions (Holzer, 2004a). When drugs address a specific target (e.g., a symptom such as Consider interactions with food or herbal products visceral hypersensitivity or motility), heterogeneity in the patho- Safety Specificity cannot always avoid off-target effects physiology impacts negatively on the therapeutic gain, if patients because the same receptor/system also mediates are not carefully selected in a clinical trial. Indeed, some of the other effects disappointing results of the past can be ascribed to the lack of A drug can also hit antitargets (i.e., unwanted targets), understanding of pathophysiology: the same symptom (e.g., diar- another source of side-effects rhea) does not necessarily depend on the same pathways in all patients. Thus,new drugs should target a pathophysiological mechanism (provided that it is known!), rather than a specific receptor; on the may have several, sometimes disparate biological effects (i.e., hit other hand, recruiting carefully selected patient subgroups may a large number of targets in the pharmacological space; Garcia- significantly reduce the generalizability of the results of the trial. Serna et al., 2010; Kawasaki and Freire, 2011). These effects may Pharmacokinetics may help to achieve gut selectivity and depend on the fact that a single receptor/effector pathway plays a reduce side-effects. This approach is particularly relevant when role in different systems, so that even selective compounds have there are potential actions outside the gut, as it is indeed the case off-target effects (Table 1). In addition, there are many instances with peripherally restricted receptor antagonists (such as when the compound is endowed with additional pharmacolog- and ), which do not cross the blood ical properties that hit the so-called antitargets (i.e., unwanted brain barrier and, in addition, have very low oral bioavailabil- targets), responsible for side-effects, which are clarified only after ity (De Ponti, 2002; Holzer, 2004b). An example of minimally the compound has undergone clinical trials. The classical example absorbed compound is also the guanylate cyclase-C lina- is provided by the cardiac side-effects due to hERG K+ chan- clotide (Wensel and Luthin, 2011; Busby et al., 2013), which is now nel blockade by the early 5-HT4 receptor (Tonini et al., FDA- and European Medicines Agency (EMA)-approved for IBS 1999). with constipation. The second issue is that the multifactorial pathophysiology Another important pharmacokinetic property is the lack of of IBS (with multiple brain-gut and neuroimmune interactions) interactions with food or other drugs. Significant interactions with makes it unrealistic to expect that drugs acting on a single receptor CYP2D6 and CYP3A4 should be predicted in early drug discovery may achieve substantial therapeutic gain over placebo in an area because of their involvement in drug metabolism with important where the placebo response rate is substantial (approaching 40% pharmacogenetic aspects. across all randomized controlled trials; Ford and Moayyedi, 2010). Finally, as regards safety aspects, apart from the standard safety As in other fields (Morphy et al., 2004), evidence suggests that a evaluations, two issues deserve special attention following the balanced modulation of multiple targets can provide a superior experience with (torsades de pointes associated with therapeutic effect and side effect profile compared to the action of QT prolongation; De Ponti et al., 2001) and alosetron (ischemic a selective ligand. Designed multiple ligands that hit a large vari- colitis; Moynihan, 2002). It is clear that even very rare events ety of targets have been produced through rational approaches may negatively impact the risk/benefit balance of drugs that are in which structural features from selective ligands are combined used to provide symptom improvement of non-serious (though (Morphy et al., 2004). A key challenge in the design of multiple troublesome) diseases such as IBS (De Ponti et al., 2002; Tack ligands is attaining a balanced activity at each target of interest et al., 2012). It is remarkable that in IBS with diarrhea, Shah with a suitable pharmacokinetic profile. et al.(2012) found that one adverse event resulting in study

Frontiers in Pharmacology | Gastrointestinal Pharmacology February 2013 | Volume 4 | Article 7| 2 De Ponti New drugs for IBS

discontinuation occurred for every 2.3 and 2.6 patients who ben- Several studies have reported the onset of IBS-like symptoms efited, respectively, from tricyclic antidepressants and alosetron, following established bacterial or viral infections of the GI tract i.e., the number needed to harm was approximately 3. This is (Barbara et al., 2009). This so-called“postinfectious”IBS occurs in quite low, considering the numbers needed to treat reported in approximately 10% of patients undergoing a documented infec- the literature for drugs in IBS (Brandt et al., 2009; Camilleri and tious gastroenteritis, and risk factors to develop symptom per- Mayer, 2009; Menees et al., 2012). Shah et al.(2012) conclude sistence are longer duration of the gastroenteritis, female sex, that, rather than simply focusing on the number needed to treat, psychosocial stressors at the time of the infection, and psycho- clinicians should be aware of harm when using pharmacotherapy logical factors such as anxiety or depression. Although a causal for IBS. relationship between abdominal pain and acute or chronic infec- tions cannot be established most of the times, it is tempting to IBS AS A “FUNCTIONAL DISORDER”: NEW PERSPECTIVES speculate that host-microbial interactions in vulnerable individu- AND GLOBAL REGULATORY FRAMEWORK als during the early phase of the disorder may lead to permanently The classical concept of IBS as a functional disorder derives from altered immune response,which then continues to play a role when the fact that no organic cause can be identified and the diagnosis of symptoms persist in the absence of the infectious organism. IBS is one of exclusion after other disorders have been ruled out. In The participation of the gut microenvironment in the patho- addition, the precise mechanisms underlying symptom generation physiology of IBS is suggested by studies indicating an interplay are unknown. between luminal factors including the microbiome, the epithe- However, research of the past 20 years has provided signifi- lial barrier, and the mucosal immune system (Stanghellini et al., cant advances in the understanding of the pathophysiology of 2010; Camilleri et al., 2012). In an animal model (McVey Neufeld IBS, with an emerging consensus that the various clinical man- et al., 2013), microbiota were shown to be necessary for normal ifestations (including non-gastrointestinal comorbid symptoms) excitability of gut sensory neurons and this provides a potential of chronic abdominal pain can best be viewed as a dysregula- mechanism for the transfer of information between the microbiota tion in the complex interplay between events occurring in the and the nervous system. gut lumen (including microbiota), the mucosa, the enteric ner- In postinfectious IBS and in IBS with diarrhea, decreased vous system (ENS), and the central nervous system (Mayer and expression and structural rearrangement of tight junction pro- Tillisch, 2011; Matricon et al., 2012). This dysregulaton leads to teins in the small bowel and colon may lead to increased intestinal alterations in sensation, motility, brain-gut interactions, and neu- permeability. These abnormalities might contribute to the outflow roimmune interactions. Considerable evidence documents that of antigens through the epithelium, causing overstimulation of the sensitizing proinflammatory mediators, mast cells and their prod- mucosal immune system. Accordingly, subgroups of patients with ucts, tryptases, are increased in tissues of patients with colorectal IBS show higher numbers and activation of mucosal immuno- hypersensitivity (Cenac et al., 2007; Balestra et al., 2012; Buhner cytes, especially mast cells. Immune factors, released by these cells, et al., 2012). including proteases,histamine,and prostanoids,might also partic- It has been shown that colonic mast cell infiltration and media- ipate in maintaining the permeability dysfunction and contribute tor release in proximity to mucosal innervation may contribute to the activation of abnormal neural responses, which, in turn, to abdominal pain perception in IBS patients (Barbara et al., are involved in abdominal pain perception and changes in bowel 2004). Indeed, mucosal mast cell mediators from IBS patients habits. excite rat nociceptive visceral sensory nerves (Barbara et al., 2007). All these mechanisms represent new therapeutic targets in IBS. In a recent study (Balestra et al., 2012), mucosal biopsies were Here,it is important to remember that probiotics are also currently obtained from the descending colon of patients with IBS and viewed as an attractive therapeutic option in IBS because of their controls. Mucosal mast cells were identified immunohistochemi- recognized safety and of their documented biological effects on the cally. The impact of spontaneously released mucosal mediators on host. Preclinical studies have shown that some probiotic strains guinea pig electrically stimulated longitudinal muscle myenteric exhibit potentially useful properties including anti-inflammatory plexus (LMMP) preparations was assessed in vitro by means of effects, improvement of mucosal barrier homeostasis, beneficial selective receptor antagonists and inhibitors. Patients with IBS effects on intestinal microbiota, and a reduction of visceral hyper- showed an increased mast cell count compared with controls. sensitivity. However, it remains to be determined to what extent Application of mucosal mediators of IBS to LMMPs potentiated a beneficial effect on these parameter translates to a significant cholinergic twitch contractions, an effect directly correlated with effect on clinical outcomes: although the effect of probiotics on mast cell counts and mediated by activation of prostanoid recep- IBS is positive in some randomized, controlled studies, the gain tors, TRPV1, and P2X receptors. These results support the role over placebo is small and identification of a tailored probiotic of mucosal inflammatory mediators and mast cell activation in approach for subgroups of patients represents a future challenge. altered motor function of IBS. The complex neuroimmune and brain-gut interactions some- It is also intriguing that,in patients with IBS,5-HT spontaneous times associated with minimal mucosal inflammation (Ford and release was significantly increased irrespective of bowel habit and Talley, 2011) and neuroplastic changes in the ENS (Giaroni et al., correlated with mast cell counts and the severity of abdominal 1999) pose several questions as regards potential targets for phar- pain. This suggests that increased 5-HT release contributes to macological intervention: should the therapeutic focus be pri- development of abdominal pain in IBS, probably through mucosal marily gut motility, visceral sensitivity, or minimal inflammation? immune activation (Cremon et al., 2011). Assessment of these parameters in humans can be undertaken by

www.frontiersin.org February 2013 | Volume 4 | Article 7| 3 De Ponti New drugs for IBS

using a variety of invasive and non-invasive techniques, some well ongoing regulatory discussion will certainly help all those involved established and others requiring further validation. By using these in clinical trials to plan future research. techniques, alterations in both sensory and motor function have Linaclotide may serve as an example of the current regulatory been reported in IBS and our understanding of sensorimotor dys- situation, with differences in endpoints recommended by the FDA function has indeed increased. Thus, inflammatory, immunologic, and the EMA: the efficacy and safety of this agent in patients with and other processes, as well as psychosocial factors such as stress, IBS-C was evaluated in two randomized, placebo-controlled Phase can alter the normal patterns of sensitivity and motility through 3 trials. These trials were designed according to both FDA and alterations in local reflex activity or via altered neural processing EMA guidelines and findings based on FDA-recommended end- along the brain-gut axis. A firm relationship between sensorimo- points were reported in two recent studies (Chey et al., 2012; Rao tor dysfunction and the production of symptoms, however, has et al., 2012), whereas the findings of a planned, separate analysis been difficult to show. Thus, the clinical relevance of the former of both trials based on the distinct efficacy parameters prespeci- requires further research. fied for EMA submission were published separately (Quigley et al., In this context, it is important to remember that in 2003 the 2013). EMA adopted a document produced by the Efficacy Working Party In closing this section, it should be remembered that, over the on the “Points to consider on the evaluation of medicinal products past 25 years, the Rome process has insisted on clinical features to for the treatment of the irritable bowel syndrome”(EMA, 2003). diagnose IBS on the assumption that grouping of patients with Although the document now needs to be updated (it still refers similar features facilitates identification of patients most likely to to Rome II criteria), a key statement is that “The patient’s global respond to a given pharmacological agent, but this is not nec- assessment of symptoms and abdominal discomfort/pain should be essarily so because the same symptom (pain) may have several used as the two primary endpoints. Statistically significant changes underlying pathophysiological mechanisms. This explains the crit- must be found in both parameters.” Thus, clinical efficacy must icism raised by some investigators against the Rome criteria (Dang rely on clinical endpoints in the patient’s perspective, for instance et al., 2012). On the other hand, it must be acknowledged that through the global assessment of multiple symptoms. Mecha- there are several reasons to establish an accurate diagnosis of IBS: nistic (pathophysiological) studies provide a rationale for drug to relieve patient uncertainty and initiate the most appropriate development, but do not generally predict symptomatic success treatment, avoiding the burden of unnecessary medications or and do not necessarily identify the most appropriate dose for diagnostic procedures and surgeries (Mearin and Lacy, 2012). In clinical trials. An important goal is to develop non-invasive tests other words, the Rome criteria try to transform the diagnosis of that identify important pathophysiological mechanisms and assess IBS from one of exclusion into a positive diagnosis based on his- symptom pattern in short-term (4 weeks) therapeutic trials that tory, physical examination, use of precise diagnostic criteria in the pave the way to longer trials. Notably, the EMA document care- absence of specific alarm features. fully considers the duration of efficacy trials, stating that they must be long enough to determine whether the response is sus- BIOMARKERS FOR IBS tained and to determine the effects of treatment withdrawal. A Biomarkers are objectively measurable indicators of normal or duration of 6 months of active treatment is considered neces- pathological processes or pharmacological responses to a ther- sary considering the cyclic and non-life threatening nature of the apeutic intervention (Anonymous, 2001). In order to improve disease. development and usage of biomarkers, a score system of differ- For inclusion and exclusion criteria for IBS, the current EMA ent types of biomarkers has been proposed, depending on their “Points to consider” document refers to Rome II criteria (current impact on drug development (Wehling, 2009). Rome III criteria differ from Rome II and, notably, the Rome IV Positive modifications of biomarkers which imply improve- process is expected to start in 2013). A revised EMA guideline is ment of the disease can be taken as endpoints during drug devel- awaited soon and should come into force by the end of 2013. A opment. To provide a more complete picture, a therapeutic target key issue that needs to be addressed, apart from the update to can coincide with the biomarker (e.g., TNFα) or, as a component Rome III, is the discrepancy with the FDA guidance issued in 2012 of the disease mechanism, modulate it (e.g., NF-κB). (FDA, 2012). Indeed, the EMA document recommends the two Unfortunately, as stated in the 2012 FDA document, no vali- co-primary endpoints indicated above, whereas the FDA guid- dated and accepted biomarker exists in IBS. In addition,the limited ance recommends a primary endpoint that measures the effect of repertoire of clinical manifestations of sensorimotor disorders of treatment on two major IBS signs and symptoms: abdominal pain the gut such as IBS can actually derive from multiple mechanisms, and abnormal defecation (stool frequency or stool consistency, leading in turn to similar symptoms. In many clinical programs of depending on subtype of IBS). The FDA guidance also acknowl- new drugs for IBS, the emphasis was primarily on symptom assess- edges that patient-reported outcome (PRO) measures of the signs ment of broad groups of patients identified by the Rome criteria. and symptoms of the condition are the only currently available As already discussed above, this approach was not ideal and it not measures that can adequately define a treatment effect in a clinical surprising that drugs of potential value have been abandoned. trial. In addition, because of the limitations of using a single-item Certain biomarkers can, in a limited fashion, be used to pre- patient-reported rating of overall change as a primary endpoint, dict the success of a drug in IBS or to understand its mode of the FDA document recommends the development of a multi-item action. These studies may be incorporated in the recommended PRO instrument. The PRO measure(s) should capture all the clin- steps for drug development, but should be viewed only as prelim- ically important signs and symptoms of the target population. The inary/complementary steps of the development program, which

Frontiers in Pharmacology | Gastrointestinal Pharmacology February 2013 | Volume 4 | Article 7| 4 De Ponti New drugs for IBS

must comply with the regulatory guidance quoted in the previous suggest that the colonic marker transit time (<15 or >60 h) accu- section. rately predicts the extremes of stool consistency, with significant Currently established tests that can be used as potential bio- overlap for transit times between those extremes (Degen et al., markers for clinically relevant endpoints in IBS include the 2001). following. RADIOSCINTIGRAPHIC MARKERS FOR COLONIC TRANSIT INTRALUMINAL MEASUREMENTS OF COLONIC OR RECTAL MOTILITY Scintigraphic transit measurements are sufficiently well charac- AND SENSATION terized to allow meaningful pharmacodynamic conclusions on Intraluminal measurements may serve as biomarkers for motor the effect of therapeutic agents (Cremonini et al., 2002; Camil- or sensory modulation in IBS. Manometry has long been used leri, 2010a; Vazquez-Roque et al., 2012). Namely, several examples for pathophysiological investigations (Camilleri et al., 2008) and support the use of detailed colonic transit measurement in the can be used as a useful technique to study the effects of drugs development of medications for IBS-associated changes in bowel on colonic motility (De Schryver et al., 2002; Dinning and Scott, function. First,alosetron (a 5-HT3 ) slows over- 2011). Another possible test is intracolonic measurement of post- all colonic transit and, on average, increases the time for emptying prandial tone using a barostat, which, in healthy subjects (von der the ascending colon by 50% (Viramontes et al., 2001a). Second, Ohe et al., 1994), showed the potential of 5-HT3 receptor antag- tegaserod (a 5-HT4 receptor agonist) accelerates overall colonic onists to prevent diarrhea and other postprandial symptoms in transit and, on average, halves the time for emptying the ascend- diseases including IBS and carcinoid diarrhea (von der Ohe et al., ing colon (Prather et al., 2000), and several studies showed that 1993). This indicates that measuring tone intraluminally may be a this was effective in the treatment of IBS with con- useful biomarker for preliminary tests before subsequent trials for stipation (Muller-Lissner et al., 2001; Novick et al., 2002; Kellow efficacy. A recent study in healthy subjects (Sweetser et al., 2009) et al., 2003). However, tegaserod was withdrawn by the manufac- investigated the effects of lubiprostone on colonic sensation and turer for safety issues in 2007 and was made available only under a motility with the following endpoints: colonic compliance, fasting restricted access program (Al-Judaibi et al., 2010). Finally, a more and postprandial tone and motility indexes, pain thresholds, and recent example of use of scintigraphy to assess transit includes sensory ratings to distensions. This investigation well exemplifies linaclotide (Andresen et al., 2007). the potential of pharmacodynamic studies in drug development. Although testing visceral sensitivity may provide useful mech- URINE SUGARS FOR IN VIVO GUT PERMEABILITY anistic insights when developing new medications, results always Because of the possible role of disruption of intestinal mucosal require careful interpretation and are sometimes disappointing. barrier function in the pathophysiology of IBS, recently a urine For instance, alosetron was shown to alter colonic compliance, but sugar (lactulose and mannitol) excretion test was validated in not colonic sensitivity to isobaric distension (Delvaux et al., 1998). patients with IBS and diarrhea (Rao et al., 2011). Urine sugars Previously, the κ agonist was shown to at 0–2 and 8–24 h reflect small bowel and colonic permeability, decrease sensitivity to colonic distension, but the therapeutic gain respectively and are increased in patients with IBS and diarrhea vs. in placebo-controlled studies in IBS was found to be of insuffi- controls. This method can be applied to study the effects of agents cient magnitude for further development (Dapoigny et al., 1995; directed at mucosal pathophysiology, such as mast cell stabilizers Delvaux et al., 1999; Ness, 1999). is another exam- or modulators of microbial flora. ple of drug tested for its effect on visceral sensitivity in humans (Delgado-Aros et al., 2003). ANIMAL MODELS: PREDICTIVITY ISSUES AND CURRENT One disadvantage of sensation biomarkers is that the sample ROLE size required to avoid a type 2 error while assessing clinically mean- Translational medicine defines conditions and prerequisites to ingful effect sizes is higher than with transit endpoints in healthy transfer more reliable results obtained from preclinical biomed- volunteers and probably even higher in patients. Nevertheless, ical research to clinical applications, thus improving patient care these sample sizes (12–20 per treatment arm) are still more prac- through timely and efficient promotion of clinical innovation. tical than testing symptom endpoints, which require much larger A disease model should mimic the clinical disease condition as samples. Thus, a 25–30% effect size can be demonstrated with ∼20 much as possible and allow the investigation of unclear pathophys- subjects per treatment arm in sensation-based studies and ∼12 per iological mechanisms toward the development of new potential treatment arm in studies of transit, on the basis of the variability therapeutic options. In spite of undeniable differences among reported in published studies (Camilleri et al., 2006b). species, animal models still represent the major source of infor- mation about biological system and provide an invaluable means RADIOPAQUE MARKERS FOR COLONIC TRANSIT to study complex physiological and biochemical interactions. The radiopaque marker test for colonic transit is a widely avail- The predictivity of the disease model itself will substantially able test, as shown by early studies with for diarrhea depend on the efforts toward optimization and scientific validation and fiber for constipation, where radiopaque markers were used of the model, both in terms of resemblance and transferability to to assess whole gut transit time (Cann et al., 1984a,b). The over- humans. Pharmacological targets and biomarkers must be repro- all effects of drugs for IBS can be predicted by the marker transit duced and be susceptible to pharmacological modulation in the test, although transit times are not characteristic of IBS (Horikawa animal model, to ensure that the model is predictive of a therapeu- et al., 1999) and other studies addressing more specific endpoints tic effect in humans. The real translational value of a biomarker is

www.frontiersin.org February 2013 | Volume 4 | Article 7| 5 De Ponti New drugs for IBS

represented by its capacity of being thoroughly informative after effective drugs for further development. Since pain, though a its validation in clinical studies which provide the bedside to bench major problem, is not the only symptom affecting quality of life, approach. animal models detailing the effects of drugs on motility and vis- An animal model used as a screening tool in drug development ceral sensitivity may add a further dimension to the assessment of should ideally reproduce different biomarkers providing multi- new compounds. ple endpoints to fully assess the spectrum of activity of a test compound. Accurate selection of endpoints plays a pivotal role to THE ROLE OF PHARMACOGENOMICS assure the translational value of disease models and should take Pharmacogenomics refers to the variability of the expression of into account the sensitivity and specificity of each endpoint toward individual genes relevant to disease susceptibility as well as drug prediction of activity. response at cellular, tissue, individual, or population level. Phar- Although development of new drugs for treatment of IBS can macogenetics refers more specifically to the study of individual be facilitated by preclinical animal models (Bulmer and Grundy, variations in DNA sequence related to drug response. The growing 2011; Holschneider et al., 2011), it must be acknowledged that a interest in this field is due to the fact that risk/benefit evaluations of single sufficiently predictive model of efficacy still does not exist drugs are not fully appreciated if one does not fully consider indi- and investigators have the option to use multiple models to assess vidual variations that may significantly affect pharmacokinetics different aspects of the pathophysiology of IBS. This section is and pharmacodynamics. only intended as a brief outline of the most commonly used ani- mal models of visceral pain and disturbed gastrointestinal motility, POLYMORPHISMS AS MARKERS OF DISEASE which are reviewed elsewhere (Camilleri et al., 2006a). As an example, patients with IBS had significantly reduced fre- quencies of the high producer genotype for interleukin 10 than MOTILITY controls (21 vs. 32%; p = 0.003): this suggests a genetic predispo- The techniques used to record motility or measure transit in ani- sition in at least some patients with IBS to produce lower amounts mals may differ from techniques used in humans but the endpoints of the anti-inflammatory cytokine interleukin 10 (Gonsalkorale are identical to those mentioned in the previous section. et al., 2003) and lends support to the hypothesis that there may be an inflammatory or genetic component in some cases of IBS VISCERAL PAIN (Bashashati et al., 2012). There are several forms of stimulation and endpoints to measure visceral pain: (a) mechanical stimuli: experiments can be per- POLYMORPHISMS AFFECTING DRUG RESPONSE formed in awake or anesthetized rats, and the most frequently Genetic variations can affect drug response in at least three dif- used stimulus of pain in animals is distension of a gut segment ferent ways (Camilleri and Katzka, 2012): (a) changes in drug with a balloon connected to a barostat to measure simultaneously metabolism, e.g., through functional CYP450 2D6 genes, which compliance and the response to the painful stimulus (Rouzade determine the pharmacokinetics and plasma levels of many et al., 1998). Poor standardization of methods across laboratories commonly used agents such as antidepressants; (b) changes in is a drawback of these investigations. (b) Chemical stimuli: infus- drug transporters, which may affect the response to medica- ing glycerol into the rat colon through a chronically implanted tions: for instance, polymorphisms in the promoter for synthesis catheter induces abdominal cramps. This model is considered rel- of serotonin transporter (SERT-P) influence response to sero- evant because intracolonic glycerol induces abdominal pain in tonergic medications in depressed individuals. SERT polymor- humans and mimics pain reported by patients with IBS (Louvel phisms were associated with a greater colonic transit response et al., 1996). (c) Other stimuli: the significance of other models of in those with long homozygous than those with heterozygous visceral pain,such as the“writhing test”(consisting of an intraperi- polymorphisms in D-IBS (Camilleri et al., 2002); (c) genetic toneal injection of an irritant compound such as acetic acid), is polymorphism in drug targets. Several examples are provided by questionable. recent studies (Camilleri and Katzka, 2012; Vazquez-Roque et al., In the aforementioned models, it is important to distinguish 2012). between evaluation of allodynia (decrease in the threshold of In summary, pharmacogenetics is a rapidly growing field which sensitivity to distension) and hyperalgesia (enhanced response to may provide important pieces of information to fully understand painful stimulus). A commonly used endpoint is the contraction the variability of drug action in patients and cannot be ignored in of abdominal muscles induced by rectal or colorectal distension in drug development programs, although its exploitation probably the rat; the contractions are recorded by electromyography, where still needs some time. the number of spike bursts correlates with the intensity of the stimulus applied (Morteau et al., 1994). CLASSES OF DRUGS USED OR UNDER DEVELOPMENT IN IBS Visceral distension also induces viscero-visceral reflexes, such as A detailed discussion of all the classes of drugs proposed as poten- relaxation of anal sphincters during rectal distension or change in tial therapeutic agents in IBS is beyond the scope of this review. blood pressure, which is a pseudoaffective response used to assess The main pharmacological approaches to IBS are summarized in visceral pain. Table 2, which does not include agents traditionally used in IBS, In summary, selection of one or more definitive animal models such as laxatives and antidiarrheal agents, respectively for con- of visceral hyperalgesia is not possible and using results from more stipation and diarrhea, and probiotics. The gut microbiome as a than one animal model may enhance the probability of selecting therapeutic target is covered elsewhere (Floch et al., 2011; Simren

Frontiers in Pharmacology | Gastrointestinal Pharmacology February 2013 | Volume 4 | Article 7| 6 De Ponti New drugs for IBS

Table 2 | Main pharmacological approaches in IBS.

Drug class Examples Pharmacodynamics

5-HT4 receptor agonists Prucalopride Enteric neurons, smooth muscle cells Naronapride Increased motility/bowel frequency Velusetrag TD-8954

5-HT3-receptor antagonists Inhibition of secretion, motility, nociception

TPH1 blocker LX-1031 Inhibits 5-HT synthesis by blocking tryptophane hydroxylase 1

Cl-C2 channel activator Lubiprostone Increased intestinal water and electrolyte secretion Accelerates transit

Guanylate cyclase-C agonist Linaclotide Increased intestinal water and electrolyte secretion Accelerates transit

PAR2 blockers GB88 Inflammation

TRPV1 blockers Capsazepine Inflammation

TRPV4 blockers RN1734

Mast cell stabilizers Ketotifen Inflammation

µ-Opioid receptor agonists Loperamide Enterocyte, enteric neurons, afferent neurons, and inflammation

µ-Opioid receptor antagonists Enteric neurons, afferent neurons, and inflammation Methylnaltrexone alvimopan

κ-Opioid receptor agonists Asimadoline Enteric neurons and afferent neurons Increase in sensory threshold

β3-Adrenoceptor agonists Solabegron Smooth muscle

α2-Adrenoceptor agonists Clonidine Enteric neurons and enterocytes

NK1 receptor antagonists Enteric neurons, ICC, smooth muscle, immune cells

NK2 receptor antagonists Reduced smooth muscle contractility

NK3 receptor antagonists Role in nociception not confirmed in clinical trials in patients with IBS

CCK1 receptor antagonists Loxiglumide Afferent vagal nerves and enteric neurons

Antibiotics Rifaximin Poorly absorbed with virtually no systemic effects et al.,2013). The reader is also referred to a recent detailed review of smooth muscle and mediate a number of different actions (De current and potential pharmacological approaches in IBS (Camil- Ponti, 2004). Actions of 5-HT are terminated by a reuptake sys- leri, 2012). Herbal preparations used in IBS are covered by another tem, which is inhibited by antidepressants (Gershon and Jonakait, review (Rahimi and Abdollahi, 2012). Brain-gut interactions and 1979). Selective serotonin reuptake inhibitors (SSRIs) alter motility possible sites/mechanisms of pharmacological intervention along in the stomach, small bowel, and colon (Gorard et al., 1994), but the brain-gut axis are discussed in a recent review in this Journal no convincing beneficial therapeutic effects have been reported in (Fichna and Storr, 2012). IBS. Interestingly, the tryptophane hydroxylase inhibitor LX-1031 Several meta-analyzes of pharmacological treatments for IBS was recently reported to have therapeutic potential in IBS (Tack have been published in recent years and there is ongoing debate et al., 2011). on their interpretation (Lesbros-Pantoflickova et al., 2004; Brandt 5-HT3 receptor antagonists include alosetron and ramosetron et al., 2009; Camilleri and Mayer, 2009). (Camilleri et al.,2000; Hirata et al.,2007; Lee et al.,2011); alosetron delays orocecal and colonic transit times, and reduce colonic com- SEROTONERGIC AGENTS pliance but not sensitivity to isobaric distension (Gore et al., 1990; 5-Hydroxytryptamine (5-HT) plays a key role in the control Talley et al., 1990; Scolapio et al., 1995). Shortly after its intro- of gastrointestinal motility, sensitivity, and secretion (De Ponti, duction, alosetron was withdrawn due to suspected side-effects of 2004). Several 5-HT receptor types are present on both nerves and colonic ischemia (Moynihan, 2002).

www.frontiersin.org February 2013 | Volume 4 | Article 7| 7 De Ponti New drugs for IBS

5-HT4 receptor agonists, after withdrawal of tegaserod, now OPIOID RECEPTOR LIGANDS include prucalopride, naronapride, velusetrag (Camilleri, 2010b), Three types of opioid receptors, µ-, δ-, and κ-receptors, located in and TD-8954 (Beattie et al., 2011). These agents are thought to act the ENS as well as on nociceptive pathways, have effects on human on intrinsic neurons to stimulate gastric, small bowel, and colonic gastrointestinal function. Opioid receptor activation reduces vis- transit in health, in constipation and in IBS with constipation ceral pain through peripheral (spinal afferents) and central mech- (Bouras et al., 1999; Poen et al., 1999; Degen et al., 2001). In the anisms, and inhibits motility through decreased acetylcholine stomach, 5-HT4 receptor agonists enhance (postprandial) prox- release. κ-Opioid receptor agonists have been proposed as a phar- imal gastric volumes in health, but have no effects on sensation macological approach to the treatment of altered visceral sen- (Tack et al., 2003). Prucalopride was also shown to be effective in sitivity. Acute studies with fedotozine and asimadoline showed the treatment of constipation (Emmanuel et al., 2002). decreased sensitivity to gastric or colonic distension (Coffin et al., 1996; Delvaux et al., 1999, 2002; Delgado-Aros et al., 2003). How- LINACLOTIDE ever, therapeutic studies in IBS and FD with fedotozine have This is an example of guanylate cyclase-C agonist (Busby et al., been disappointing (Dapoigny et al., 1995; Read et al., 1997). The 2013), which has been shown to reduce visceral hypersensitivity µ-opioid receptor agonist loperamide, used in the treatment of in preclinical studies and to improve abdominal pain and consti- diarrhea, inhibits secretion, reduces colonic transit, and increases pation symptoms in phase 2 and 3 clinical trials of patients with resting anal sphincter tone (Corazziari, 1999). Peripherally act- IBS and constipation (Johnston et al., 2013). ing µ-opioid receptor antagonists (e.g., N-methylnaltrexone and alvimopan) normalize bowel function in -treated patients LUBIPROSTONE without compromising central opioid analgesia (Holzer, 2004b). This is an oral bicyclic fatty acid selectively activating type 2 , a neutral endopeptidase (NEP) inhibitor, increases chloride channels in the apical membrane of the intestinal epithe- the exposure to NEP substrates including : it was found lial cells, hence stimulating chloride secretion, along with passive consistently effective in animal models and patients with vari- secretion of sodium and water, inducing peristalsis and laxation, ous forms of acute diarrhea by inhibiting secretion from the gut without stimulating gastrointestinal smooth muscle (Schey and without changing gastrointestinal transit time or motility (Eber- Rao, 2011). It is indicated in IBS with constipation. Considering lin et al., 2012). In direct comparative studies with loperamide, the importance of epithelial barrier function and cell integrity and racecadotril was at least as effective, but exhibited fewer adverse the known impact of stressors, the observation that lubiprostone events in most studies, particularly less rebound constipation exhibits the additional distinct property of effective protection or (Eberlin et al., 2012). However, its potential in IBS remains to repair of the epithelial barrier and cell function after stress suggests be established. potential clinical importance for patients with impaired barrier function, which might occur in IBS (Cuppoletti et al., 2012). MISCELLANEOUS AGENTS CCK has a large number of effects on gastrointestinal contractility TACHYKININ RECEPTOR ANTAGONISTS and secretion (Walsh, 1994). CCK1 receptor antagonists like lox- Three distinct receptors, NK1, NK2, and NK3, mediate the biolog- iglumide and dexloxiglumide enhance gastric emptying in health ical effects of endogenous tachykinins SP, NKA, and NKB, in the and in IBS with constipation, though effects on colonic motil- gastrointestinal tract. Through the locations of NK receptors on ity are unclear (De Ponti and Malagelada, 1998; Scarpignato and intrinsic nerves, extrinsic nerves, inflammatory cells, and smooth Pelosini, 1999) and clinical usefulness has not been established. muscle, inhibition of tachykinin receptors has the potential to The transient receptor potential ion channel of the vanilloid inhibit motility, sensitivity, secretion, and inflammation in the type 1 (TRPV1), expressed by primary afferent neurons, is viewed gastrointestinal tract (Holzer, 2004a; Lecci et al., 2004). NK1 recep- as a trigger for chemonociception and may be upregulated in some tor antagonists also have properties (Holzer, 2004a). functional gut disorders (Chan et al., 2003). TRPV1 and TRPV4 Several tachykinin receptor antagonists have been developed so blockade are areas of current investigation (Holzer, 2011; Fichna far, but the results, in general, have been disappointing. Recently, et al., 2012). chronic treatment with AV608 (NK1 receptor antagonist) in IBS Muscarinic receptor antagonists and smooth muscle relaxants are has been reported to be associated with improved mood and pain used in some countries for the treatment of IBS. Meta-analysis ratings and activity of emotional arousal related brain regions suggests they are superior to placebo in IBS-related pain (Poynard (Tillisch et al., 2012). et al., 2001), though the quality of trials is often questionable. CB1 receptors are expressed on nociceptive affer- ADRENOCEPTOR AGONIST ents and ENS neurons while CB2 receptors are expressed on The α2-adrenoceptor agonist clonidine was shown to reduce immune cells (Schicho and Storr, 2011). Activation of CB1 recep- colonic tone and pain sensation in response to distension tors slows gastrointestinal transit in animals through inhibi- (Bharucha et al., 1997; Malcolm et al., 2000; Viramontes et al., tion of acetylcholine release. The non-specific agonist delta-9- 2001b). A preliminary study of clonidine in IBS with diarrhea has strong antiemetic properties and delays suggested therapeutic potential for clonidine, but clinical applica- gastric emptying in humans (Frytak et al., 1979; McCallum et al., tion is hampered by dose-limiting side-effects like somnolence or 1999). The observation that, in an animal model of intestinal hypotension. Among β3-adrenoceptor agonists, solabegron is an inflammation, CB1 and CB2 receptor subtypes are upregulated example (Grudell et al., 2008). opens a new perspective on the possible use of CB1 or CB2 receptor

Frontiers in Pharmacology | Gastrointestinal Pharmacology February 2013 | Volume 4 | Article 7| 8 De Ponti New drugs for IBS

agonists in postinfectious IBS with diarrhea (Kimball et al., 2010). CONCLUSION Indeed, , a non-selective CB agonist, reduces fasting From the above overview, it is clear that research in the treat- colonic motility in patients with IBS with diarrhea or alternating ment of IBS is still very active. Although in the past decade some (Wong et al., 2011). innovative pharmacological agents have not fulfilled their promise Finally, α2δ ligands such as gabapentin and pregabalin (Gale because of unexpected side-effects, it is likely that new pathophys- and Houghton, 2011) have undergone a number of preclinical and iological concepts as well as the publication of new regulatory clinical tests for their potential in disorders with visceral hyper- documents by the FDA and the EMA will be of great help for drug sensitivity. In fact, voltage-sensitive Ca2+ channels are involved developers. in neural function and have an α2δ binding site to which the aforementioned ligands bind potently, reducing Ca2+ influx at the ACKNOWLEDGMENTS nerve terminals. Pregabalin was effective in several animal models The original research of the author is supported by a grant from of visceral pain (Gale and Houghton, 2011). the University of Bologna (Ricerca Fondamentale Orientata 2011).

REFERENCES Beattie, D. T., Armstrong, S. R., Vickery, Camilleri, M. (2010b). Review article: Camilleri, M., Lasch, K., and Zhou, W. Al-Judaibi, B., Chande, N., and Gre- R. G., Tsuruda, P. R., Campbell, C. new receptor targets for medical (2012). Irritable bowel syndrome: gor, J. (2010). Safety and efficacy of B., Richardson, C., et al. (2011). The therapy in irritable bowel syndrome. methods, mechanisms, and tegaserod therapy in patients with pharmacology of TD-8954, a potent Aliment. Pharmacol. Ther. 31, pathophysiology. The conflu- irritable bowel syndrome or chronic and selective 5-HT4 receptor ago- 35–46. ence of increased permeability, constipation. Can. J. Clin. Pharma- nist with gastrointestinal prokinetic Camilleri, M. (2012). Pharmacology inflammation, and pain in irritable col. 17, e194–e200. properties. Front. Pharmacol. 2:25. of the new treatments for lower bowel syndrome. Am. J. Physiol. Andresen, V., Camilleri, M., Busciglio, doi:10.3389/fphar.2011.00025 gastrointestinal motility disorders Gastrointest. Liver Physiol. 303, I. A., Grudell, A., Burton, D., McK- Bharucha, A. E., Camilleri, M., Zins- and irritable bowel syndrome. Clin. G775–G785. inzie, S., et al. (2007). Effect of meister, A. R., and Hanson, R. B. Pharmacol. Ther. 91, 44–59. Camilleri, M., and Mayer, E. A. (2009). 5 days linaclotide on transit and (1997). Adrenergic modulation of Camilleri, M., Atanasova, E., Carl- Developing irritable bowel syn- bowel function in females with human colonic motor and sensory son, P. J., Ahmad, U., Kim, drome guidelines through meta- constipation-predominant irritable function. Am. J. Physiol. 273, G997– H. J., Viramontes, B. E., et al. analyses: does the emperor really bowel syndrome. Gastroenterology G1006. (2002). Serotonin-transporter have new clothes? Gastroenterology 133, 761–768. Bouras, E. P., Camilleri, M., Burton, D. polymorphism pharmacogenetics 137, 766–769. Anonymous. (2001). Biomarkers and D., and McKinzie, S. (1999). Selec- in diarrhea-predominant irritable Camilleri, M., Northcutt,A. R., Kong, S., surrogate endpoints: preferred def- tive stimulation of colonic transit bowel syndrome. Gastroenterology Dukes,G. E.,McSorley,D.,and Man- initions and conceptual framework. by the benzofuran 5HT4 agonist, 123, 425–432. gel, A. W. (2000). Efficacy and safety Clin. Pharmacol. Ther. 69, 89–95. prucalopride, in healthy humans. Camilleri, M., Bharucha, A. E., Di, L. C., of alosetron in women with irrita- Balestra, B., Vicini, R., Cremon, C., Zec- Gut 44, 682–686. Hasler, W. L., Prather, C. M., Rao, S. ble bowel syndrome: a randomised, chi, L., Dothel, G., Vasina, V., et al. Brandt, L. J., Chey, W. D., Foxx- S., et al. (2008). American Neurogas- placebo-controlled trial. Lancet 355, (2012). Colonic mucosal mediators Orenstein, A. E., Schiller, L. R., troenterology and Motility Society 1035–1040. from patients with irritable bowel Schoenfeld, P. S., Spiegel, B. M., et consensus statement on intralumi- Cann, P. A., Read, N. W., and syndrome excite enteric cholinergic al. (2009). An evidence-based posi- nal measurement of gastrointestinal Holdsworth, C. O. (1984a). What motor neurons. Neurogastroenterol. tion statement on the management and colonic motility in clinical prac- is the benefit of coarse wheat bran Motil. 24, 1118–e570. of irritable bowel syndrome. Am. tice. Neurogastroenterol. Motil. 20, in patients with irritable bowel syn- Barbara, G., Cremon, C., Pallotti, F., J. Gastroenterol. 104(Suppl. 1), S1– 1269–1282. drome? Gut 25, 168–173. De, G. R., Stanghellini, V., and Cori- S35. Camilleri, M., Bueno, L., De Ponti, Cann, P.A., Read, N. W.,Holdsworth, C. naldesi, R. (2009). Postinfectious Buhner, S., Li, Q., Berger, T., Vignali, S., F., Fioramonti, J., Lydiard, R. D., and Barends, D. (1984b). Role of irritable bowel syndrome. J. Pedi- Barbara, G., De, G. R., et al. (2012). B., and Tack, J. (2006a). Phar- loperamide and placebo in manage- atr. Gastroenterol. Nutr. 48(Suppl. 2), Submucous rather than myenteric macological and pharmacokinetic ment of irritable bowel syndrome. S95–S97. neurons are activated by mucosal aspects of functional gastrointesti- Dig. Dis. Sci. 29, 239–247. Barbara, G., Stanghellini, V., De, G. R., biopsy supernatants from irritable nal disorders. Gastroenterology 130, Cenac, N., Andrews, C. N., Holzhausen, Cremon, C., Cottrell, G. S., Santini, bowel syndrome patients. Neurogas- 1421–1434. M., Chapman, K., Cottrell, G., D., et al. (2004). Activated mast cells troenterol. Motil. 24, 1134–e572. Camilleri, M., Bueno, L., De Ponti, Andrade-Gordon, P., et al. (2007). in proximity to colonic nerves cor- Bulmer, D. C., and Grundy, D. (2011). F., Fioramonti, J., Lydiard, R. B., Role for protease activity in visceral relate with abdominal pain in irrita- Achieving translation in models of and Tack, J. (2006b). “Pharmaco- pain in irritable bowel syndrome. J. ble bowel syndrome. Gastroenterol- visceral pain. Curr. Opin. Pharmacol. logical and pharmacokinetic aspects Clin. Invest. 117, 636–647. ogy 126, 693–702. 11, 575–581. of functional gastrointestinal disor- Chan, C. L., Facer, P.,Davis, J. B., Smith, Barbara, G., Wang, B., Stanghellini, V., Busby, R. W., Kessler, M. M., Bar- ders,” in ROME III, The Functional G. D., Egerton, J., Bountra, C., et De, G. R., Cremon, C., Di, N. G., et tolini, W. P., Bryant, A. P., Han- Gastrointestinal Disorders, Chap. 4, al. (2003). Sensory fibres express- al. (2007). Mast cell-dependent exci- nig, G., Higgins, C. S., et al. (2013). eds D. A. Drossman, E. Corazziari, ing capsaicin receptor TRPV1 in tation of visceral-nociceptive sen- Pharmacologic properties, metabo- M. Delvaux, R. C. Spiller, N. J. Tal- patients with rectal hypersensitiv- sory neurons in irritable bowel syn- lism, and disposition of linaclotide, a ley, W. G. Thompson, and W. E. ity and faecal urgency. Lancet 361, drome. Gastroenterology 132, 26–37. novel therapeutic approved Whitehead (Lawrence: Allen Press), 385–391. Bashashati, M., Rezaei, N., Bashashati, for the treatment of irritable bowel 161–230. Chey, W. D., Lembo, A. J., Lavins, B. J., H., Shafieyoun, A., Daryani, N. E., syndrome with constipation and Camilleri, M., and Katzka, D. A. (2012). Shiff, S. J., Kurtz, C. B., Currie, M. G., Sharkey,K. A.,et al. (2012). Cytokine chronic idiopathic constipation. J. Irritable bowel syndrome: methods, et al. (2012). Linaclotide for irritable gene polymorphisms are associated Pharmacol. Exp. Ther. 344, 196–206. mechanisms, and pathophysiology. bowel syndrome with constipation: a with irritable bowel syndrome: a sys- Camilleri, M. (2010a). Scintigraphic Genetic epidemiology and pharma- 26-week, randomized, double-blind, tematic review and meta-analysis. biomarkers for colonic dysmotil- cogenetics in irritable bowel syn- placebo-controlled trial to evaluate Neurogastroenterol. Motil. 24, 1102– ity. Clin. Pharmacol. Ther. 87, drome. Am. J. Physiol. Gastrointest. efficacy and safety. Am. J. Gastroen- e566. 748–753. Liver Physiol. 302, G1075–G1084. terol. 107, 1702–1712.

www.frontiersin.org February 2013 | Volume 4 | Article 7| 9 De Ponti New drugs for IBS

Coffin, B., Bouhassira, D., Chollet, R., Montanaro, N. (2002). Safety of W. E. Whitehead (Lawrence: Allen and a placebo. Ann. Intern. Med. 91, Fraitag, B., De Meynard, C., Gen- non-antiarrhythmic drugs that Press), 1–29. 825–830. eve, J., et al. (1996). Effect of the prolong the QT interval or induce Eberlin, M., Muck, T., and Michel, M. Gale, J. D., and Houghton, L. A. kappa agonist fedotozine on percep- torsade de pointes: an overview. C. (2012). A comprehensive review (2011). Alpha 2 delta (α(2)δ) lig- tion of gastric distension in healthy Drug Saf. 25, 263–286. of the pharmacodynamics, pharma- ands, gabapentin and pregabalin: humans. Aliment. Pharmacol. Ther. De Ponti, F.,Poluzzi, E., and Montanaro, cokinetics, and clinical effects of what is the evidence for potential use 10, 919–925. N. (2001). Organising evidence on the neutral endopeptidase inhibitor of these ligands in irritable bowel Corazziari, E. (1999). Role of opi- QT prolongation and occurrence racecadotril. Front. Pharmacol. 3:93. syndrome. Front. Pharmacol. 2:28. oid ligands in the irritable bowel of Torsades de Pointes with non- doi:10.3389/fphar.2012.00093 doi:10.3389/fphar.2011.00028 syndrome. Can. J. Gastroenterol. antiarrhythmic drugs: a call for con- EMA. (2003). Points to Consider on the Garcia-Serna, R., Ursu, O., Oprea, T. 13(Suppl. A), 71A–75A. sensus. Eur. J. Clin. Pharmacol. 57, Evaluation of Medicinal Products I., and Mestres, J. (2010). iPHACE: Cremon,C.,Carini,G.,Wang,B.,Vasina, 185–209. for the Treatment of the Irritable integrative navigation in pharma- V., Cogliandro, R. F., De Giorgio, De Schryver, A. M., Andriesse, G. I., Bowel Syndrome. Available at: http:// cological space. Bioinformatics 26, R., et al. (2011). Intestinal sero- Samsom, M., Smout, A. J., Gooszen, www.ema.europa.eu/docs/en_GB/ 985–986. tonin release, sensory neuron activa- H. G., and Akkermans, L. M. (2002). document_library/Scientific_ Gershon, M. D., and Jonakait, G. tion, and abdominal pain in irritable The effects of the specific 5HT4 guideline/2009/09/WC500003187. M. (1979). Uptake and release bowel syndrome. Am. J. Gastroen- receptor agonist, prucalopride, on pdf [accessed December 13, 2012]. of 5-hydroxytryptamine by enteric terol. 106, 1290–1298. colonic motility in healthy volun- Emmanuel, A. V., Roy, A. J., Nicholls, 5-hydroxytryptaminergic neurones: Cremonini, F., Delgado-Aros, S., and teers. Aliment. Pharmacol. Ther. 16, T. J., and Kamm, M. A. (2002). effects of fluoxetine (Lilly 110140) Camilleri, M. (2003). Efficacy of 603–612. Prucalopride, a systemic enteroki- and chlorimipramine. Br. J. Pharma- alosetron in irritable bowel syn- Degen, L., Matzinger, D., Merz, M., netic, for the treatment of constipa- col. 66, 7–9. drome: a meta-analysis of random- Appel-Dingemanse, S., Osborne, tion. Aliment. Pharmacol. Ther. 16, Giaroni, C., De Ponti, F., Cosentino, ized controlled trials. Neurogastroen- S., Luchinger, S., et al. (2001). 1347–1356. M., Lecchini, S., and Frigo, G. terol. Motil. 15, 79–86. Tegaserod, a 5-HT4 receptor par- FDA. (2012). Irritable Bowel Syndrome: (1999). Plasticity in the enteric ner- Cremonini, F., Mullan, B. P., Camilleri, tial agonist, accelerates gastric Clinical Evaluation of Drugs for vous system. Gastroenterology 117, M., Burton, D. D., and Rank, M. emptying and gastrointestinal Treatment. Available at: http://www. 1438–1458. R. (2002). Performance characteris- transit in healthy male subjects. fda.gov/downloads/Drugs/Guidance Gonsalkorale, W. M., Perrey, C., Pravica, tics of scintigraphic transit measure- Aliment. Pharmacol. Ther. 15, ComplianceRegulatoryInformation/ V., Whorwell, P. J., and Hutchinson, ments for studies of experimental 1745–1751. Guidances/UCM205269.pdf I. V. (2003). Interleukin 10 geno- therapies. Aliment. Pharmacol. Ther. Delgado-Aros, S., Chial, H. J., Camil- [accessed December 13, 2012]. types in irritable bowel syndrome: 16, 1781–1790. leri, M., Szarka, L. A., Weber, F. T., Fichna, J., Mokrowiecka, A., evidence for an inflammatory com- Cuppoletti, J., Blikslager, A. T., Jacob, J., et al. (2003). Effects of a Cygankiewicz, A. I., Zakrzewski, ponent? Gut 52, 91–93. Chakrabarti, J., Nighot, P. K., kappa-opioid agonist, asimadoline, P. K., Malecka-Panas, E., Janecka, Gorard, D. A., Libby, G. W., and and Malinowska, D. H. (2012). on satiation and GI motor and sen- A., et al. (2012). Transient receptor Farthing, M. J. (1994). 5- Contrasting effects of linaclotide sory functions in humans. Am. J. potential vanilloid 4 blockade Hydroxytryptamine and human and lubiprostone on restitution of Physiol. 284, G558–G566. protects against experimental col- small intestinal motility: effect of epithelial cell barrier properties and Delvaux, M., Jacob, J., Beck, A., Bouza- itis in mice: a new strategy for inhibiting 5-hydroxytryptamine cellular homeostasis after exposure mondo, H., Weber, F. T., and Frex- inflammatory bowel diseases treat- reuptake. Gut 35, 496–500. to cell stressors. BMC Pharmacol. inos, J. (2002). Effect of asimado- ment? Neurogastroenterol. Motil. 24, Gore, S., Gilmore, I. T., Haigh, C. 12:3. doi:10.1186/1471-2210-12-3 line, a new agonist of kappa opiate e557–e560. G., Brownless, S. M., Stockdale, Dang, J., Ardila-Hani, A., Amichai, M. receptors on pain induced by rectal Fichna, J., and Storr, M. A. (2012). H., and Morris, A. I. (1990). M., Chua, K., and Pimentel, M. distension in IBS patients. Gastroen- Brain-gut interactions in Colonic transit in man is slowed by (2012). Systematic review of diag- terology 122, A221. IBS. Front. Pharmacol. 3:127. (GR38032F), a selec- nostic criteria for IBS demonstrates Delvaux, M., Louvel, D., Lagier, E., doi:10.3389/fphar.2012.00127 tive 5-hydroxytryptamine receptor poor validity and utilization of Scherrer, B., Abitbol, J. L., and Frex- Floch, M. H., Walker, W. A., Madsen, (type 3) antagonist. Aliment. Phar- Rome III. Neurogastroenterol. Motil. inos, J. (1999). The k-agonist fedo- K., Sanders, M. E., MacFarlane, G. macol. Ther. 4, 139–144. 24, 853–e397. tozine relieves hypersensitivity to T., Flint, H. J., et al. (2011). Rec- Grudell, A. B., Camilleri, M., Jensen, K. Dapoigny, M., Abitbol, J. L., and colonic distention in patients with ommendations for probiotic use- L., Foxx-Orenstein, A. E., Burton, D. Fraitag,B. (1995). Efficacy of periph- irritable bowel syndrome. Gastroen- 2011 update. J. Clin. Gastroenterol. D., Ryks, M. D., et al. (2008). Dose- eral kappa agonist fedotozine ver- terology 116, 38–45. 45(Suppl.), S168–S171. response effect of a beta3-adrenergic sus placebo in treatment of irrita- Delvaux, M., Louvel, D., Mamet, J. P., Ford, A. C., and Moayyedi, P. (2010). receptor agonist, solabegron, on gas- ble bowel syndrome: a multicenter Campos-Oriola, R., and Frexinos, Meta-analysis: factors affect- trointestinal transit, bowel function, dose-response study. Dig. Dis. Sci. J. (1998). Effect of alosetron on ing placebo response rate in and somatostatin levels in health. 40, 2244–2248. reponses to colonic distension in the irritable bowel syndrome. Am. J. Physiol. Gastrointest. Liver De Ponti, F. (2002). Methylnaltrex- patients with irritable bowel syn- Aliment. Pharmacol. Ther. 32, Physiol. 294, G1114–G1119. one progenics. Curr. Opin. Investig. drome. Aliment. Pharmacol. Ther. 144–158. Hirata, T., Funatsu, T., Keto, Y., Nakata, Drugs 3, 614–620. 12, 849–855. Ford, A. C., and Talley, N. J. (2011). M., and Sasamata, M. (2007). Phar- De Ponti, F. (2004). Pharmacology of Dinning, P. G., and Scott, S. M. (2011). Mucosal inflammation as a potential macological profile of ramosetron, serotonin: what a clinician should Novel diagnostics and therapy of etiological factor in irritable bowel a novel therapeutic agent for IBS. know. Gut 53, 1520–1535. colonic motor disorders. Curr. Opin. syndrome: a systematic review. J. Inflammopharmacology 15, 5–9. De Ponti, F., and Malagelada, J. R. Pharmacol. 11, 624–629. Gastroenterol. 46, 421–431. Holschneider, D. P., Bradesi, S., and (1998). Functional gut disorders: Drossman, D. A. (2006). “The func- Frytak, S., Moertel, C. G., O’Fallon, Mayer, E. A. (2011). The role of from motility to sensitivity disor- tional gastrointestinal disorders and J. R., Rubin, J., Creagan, E. T., experimental models in developing ders. A review of current and inves- the Rome III process,” in Rome O’Connell, M. J., et al. (1979). new treatments for irritable bowel tigational drugs for their manage- III. The Functional Gastrointestinal Delta-9-tetrahydrocannabinol syndrome. Expert Rev. Gastroenterol. ment. Pharmacol. Ther. 80, 49–88. Disorders, eds D. A. Drossman, E. as an antiemetic for patients Hepatol. 5, 43–57. De Ponti, F., Poluzzi, E., Cav- Corazziari, M. Delvaux, R. C. Spiller, receiving cancer chemotherapy. A Holzer, P.(2004a).“Tachykinin receptor alli, A., Recanatini, M., and N. J. Talley, W. G. Thompson, and comparison with antagonists: silencing neuropeptides

Frontiers in Pharmacology | Gastrointestinal Pharmacology February 2013 | Volume 4 | Article 7| 10 De Ponti New drugs for IBS

with a role in the disturbed gut,” injection of glycerol: a model for constipation. Aliment. Pharmacol. peripheral kappa agonist fedotozine in Pathophysiology of the Enteric abdominal pain in irritable bowel Ther. 15, 1655–1666. versus placebo in the treatment Nervous System, eds R. Spiller and syndrome? Gastroenterology 110, Ness, T. J. (1999). Kappa opioid recep- of functional dyspepsia. Gut 41, D. Grundy (London: Blackwell), 351–361. tor agonists differentially inhibit two 664–668. 212–227. Malcolm, A., Camilleri, M., Kost, L., classes of rat spinal neurons excited Rouzade, M. L., Fioramonti, J., and Holzer, P. (2004b). and opi- Burton, D. D., Fett, S. L., and Zins- by colorectal distention. Gastroen- Bueno, L. (1998). A model for evalu- oid receptors in the enteric nervous meister, A. R. (2000). Towards iden- terology 117, 388–394. ation of gastric sensitivity in awake system: from a problem in opioid tifying optimal doses for alpha-2 Novick, J., Miner, P., Krause, R., Gle- rats. Neurogastroenterol. Motil. 10, analgesia to a possible new proki- adrenergic modulation of colonic bas, K., Bliesath, H., Ligozio, G., et 157–163. netic therapy in humans. Neurosci. and rectal motor and sensory func- al. (2002). A randomized, double- Scarpignato, C., and Pelosini, I. (1999). Lett. 361, 192–195. tion. Aliment. Pharmacol. Ther. 14, blind, placebo-controlled trial of Management of irritable bowel syn- Holzer, P. (2011). Transient receptor 783–793. tegaserod in female patients suffer- drome: novel approaches to the potential (TRP) channels as drug Matricon, J., Meleine, M., Gelot, A., ing from irritable bowel syndrome pharmacology of gut motility. Can. targets for diseases of the diges- Piche, T., Dapoigny, M., Muller, E., with constipation. Aliment. Pharma- J. Gastroenterol. 13(Suppl. A), 50A– tive system. Pharmacol. Ther. 131, et al. (2012). Review article: asso- col. Ther. 16, 1877–1888. 65A. 142–170. ciations between immune activa- Poen,A. C., Felt-Bersma, R. J.,Van Don- Schey, R., and Rao, S. S. (2011). Horikawa, Y., Mieno, H., Inoue, M., and tion, intestinal permeability and the gen, P. A., and Meuwissen, S. G. Lubiprostone for the treatment of Kajiyama, G. (1999). Gastrointesti- irritable bowel syndrome. Aliment. (1999). Effect of prucalopride, a new adults with constipation and irrita- nal motility in patients with irri- Pharmacol. Ther. 36, 1009–1031. enterokinetic agent,on gastrointesti- ble bowel syndrome. Dig. Dis. Sci. 56, table bowel syndrome studied by Mayer, E. A., and Tillisch, K. (2011). nal transit and anorectal function 1619–1625. using radiopaque markers. Scand. J. The brain-gut axis in abdominal in healthy volunteers. Aliment. Phar- Schicho, R., and Storr, M. (2011). Gastroenterol. 34, 1190–1195. pain syndromes. Annu. Rev. Med. 62, macol. Ther. 13, 1493–1497. Alternative targets within the endo- Johnston, J. M., Shiff, S. J., and Quigley, 381–396. Poynard, T., Regimbeau, C., and Ben- cannabinoid system for future treat- E. M. (2013). A review of the clinical McCallum, R. W., Soykan, I., Sridhar, hamou, Y. (2001). Meta-analysis of ment of gastrointestinal diseases. efficacy of linaclotide in IBS-C. Curr. K. R., Ricci, D. A., Lange, R. C., smooth muscle relaxants in the Can. J. Gastroenterol. 25, 377–383. Med. Res. Opin. 29, 149–160. and Plankey, M. W. (1999). Delta- treatment of irritable bowel syn- Scolapio, J. S., Camilleri, M., der Kawasaki,Y.,and Freire, E. (2011). Find- 9-tetrahydrocannabinol delays the drome. Aliment. Pharmacol. Ther. Ohe, M. R., and Hanson, R. B. ing a better path to drug selectivity. gastric emptying of solid food in 15, 355–361. (1995). Ascending colon response Drug Discov. Today 16, 985–990. humans: a double-blind, random- Prather, C. M., Camilleri, M., Zinsmeis- to feeding: evidence for a 5- Kellow,J.,Lee,O.Y.,Chang,F.Y.,Thong- ized study. Aliment. Pharmacol. Ther. ter, A. R., McKinzie, S., and Thom- hydroxytryptamine-3 mechanism. sawat, S., Mazlam, M. Z., Yuen, 13, 77–80. forde, G. (2000). Tegaserod acceler- Scand. J. Gastroenterol. 30, 562–567. H., et al. (2003). An Asia-Pacific, McVey Neufeld, K. A., Mao, Y. K., ates orocecal transit in patients with Shah, E., Kim, S., Chong, K., Lembo, double blind, placebo controlled, Bienenstock, J., Foster, J. A., and constipation-predominant irritable A., and Pimentel, M. (2012). Evalu- randomised study to evaluate the Kunze, W. A. (2013). The micro- bowel syndrome. Gastroenterology ation of harm in the pharmacother- efficacy, safety, and tolerability of biome is essential for normal gut 118, 463–468. apy of irritable bowel syndrome. Am. tegaserod in patients with irritable intrinsic primary afferent neuron Quigley, E. M., Tack, J., Chey, W. D., J. Med. 125, 381–393. bowel syndrome. Gut 52, 671–676. excitability in the mouse. Neurogas- Rao, S. S., Fortea, J., Falques, M., Simren, M., Barbara, G., Flint, H. J., Kimball, E. S., Wallace, N. H., Schnei- troenterol. Motil. 25, 183–189. et al. (2013). Randomised clinical Spiegel, B. M., Spiller, R. C., Vanner, der, C. R., D’Andrea, M. R., Mearin, F., and Lacy, B. E. (2012). trials: linaclotide phase 3 studies S., et al. (2013). Intestinal micro- and Hornby, P. J. (2010). Small Diagnostic criteria in IBS: useful or in IBS-C – a prespecified further biota in functional bowel disorders: intestinal cannabinoid receptor not? Neurogastroenterol. Motil. 24, analysis based on European Med- a Rome foundation report. Gut 62, changes following a single colonic 791–801. icines Agency-specified endpoints. 159–176. insult with oil of mustard in Menees, S. B., Maneerattannaporn, M., Aliment. Pharmacol. Ther. 37, 49–61. Stanghellini, V., Barbara, G., Cremon, mice. Front. Pharmacol. 1:132. Kim, H. M., and Chey, W. D. (2012). Rahimi, R., and Abdollahi, M. (2012). C., Cogliandro, R., Antonucci, A., doi:10.3389/fphar.2010.00132 The efficacy and safety of rifaximin Herbal medicines for the manage- Gabusi, V., et al. (2010). Gut micro- Lecci, A., Capriati, A., and Maggi, C. for the irritable bowel syndrome: a ment of irritable bowel syndrome: biota and related diseases: clini- A. (2004). Tachykinin NK2 recep- systematic review and meta-analysis. a comprehensive review. World J. cal features. Intern. Emerg. Med. tor antagonists for the treatment Am. J. Gastroenterol. 107, 28–35. Gastroenterol. 18, 589–600. 5(Suppl. 1), S57–S63. of irritable bowel syndrome. Br. J. Morphy, R., Kay, C., and Rankovic, Rao, A. S., Camilleri, M., Eckert, D. J., Sweetser, S., Busciglio, I. A., Camilleri, Pharmacol. 141, 1249–1263. Z. (2004). From magic bullets to Busciglio, I., Burton, D. D., Ryks, M., Bharucha, A. E., Szarka, L. A., Lee, K. J., Kim, N. Y., Kwon, J. K., designed multiple ligands. Drug Dis- M., et al. (2011). Urine sugars for Papathanasopoulos, A., et al. (2009). Huh, K. C., Lee, O. Y., Lee, J. S., et cov. Today 9, 641–651. in vivo gut permeability: valida- Effect of a chloride channel acti- al. (2011). Efficacy of ramosetron Morteau, O., Hachet, T., Caussette, M., tion and comparisons in irritable vator, lubiprostone, on colonic sen- in the treatment of male patients and Bueno, L. (1994). Experimental bowel syndrome-diarrhea and con- sory and motor functions in healthy with irritable bowel syndrome with colitis alters visceromotor response trols. Am. J. Physiol. Gastrointest. subjects. Am. J. Physiol. Gastrointest. diarrhea: a multicenter, randomized to colorectal distension in awake Liver Physiol. 301, G919–G928. Liver Physiol. 296, G295–G301. clinical trial, compared with mebev- rats. Dig. Dis. Sci. 39, 1239–1248. Rao, S., Lembo, A. J., Shiff, S. J., Lavins, Tack, J., Camilleri, M., Chang, L., Chey, erine. Neurogastroenterol. Motil. 23, Moynihan, R. (2002). Alosetron: a case B. J., Currie, M. G., Jia, X. D., et W. D., Galligan, J. J., Lacy, B. E., et 1098–1104. study in regulatory capture, or a vic- al. (2012). A 12-week, randomized, al. (2012). Systematic review: car- Lesbros-Pantoflickova, D., Michetti, P., tory for patients’ rights? BMJ 325, controlled trial with a 4-week ran- diovascular safety profile of 5-HT(4) Fried, M., Beglinger, C., and Blum, 592–595. domized withdrawal period to eval- agonists developed for gastrointesti- A. L. (2004). Meta-analysis: the Muller-Lissner, S. A., Fumagalli, I., uate the efficacy and safety of lina- nal disorders. Aliment. Pharmacol. treatment of irritable bowel syn- Bardhan, K. D., Pace, F., Pecher, E., clotide in irritable bowel syndrome Ther. 35, 745–767. drome. Aliment. Pharmacol. Ther. Nault, B., et al. (2001). Tegaserod, with constipation. Am. J. Gastroen- Tack, J., Janssen, P., Wouters, M., and 20, 1253–1269. a 5-HT4 receptor partial ago- terol. 107, 1714–1724. Boeckxstaens, G. (2011). Targeting Louvel, D., Delvaux, M., Staumont, nist, relieves symptoms in irri- Read, N. W., Abitbol, J. L., Bardhan, serotonin synthesis to treat irrita- G., Camman, F., Fioramonti, J., table bowel syndrome patients K. D., Whorwell, P. J., and Fraitag, ble bowel syndrome. Gastroenterol- Bueno, L., et al. (1996). Intracolonic with abdominal pain, bloating and B. (1997). Efficacy and safety of the ogy 141, 420–422.

www.frontiersin.org February 2013 | Volume 4 | Article 7| 11 De Ponti New drugs for IBS

Tack, J., Vos, R., Janssens, J., Salter, Vazquez-Roque, M. I., Camilleri, M., carcinoid syndrome and diarrhea. N. Conflict of Interest Statement: The J., Jauffret, S., and Vandeplassche, Smyrk, T., Murray, J. A., O’Neill, J., Engl. J. Med. 329, 1073–1078. author declares that the research was G. (2003). Influence of tegaserod Carlson, P.,et al. (2012). Association von der Ohe, M. R., Hanson, R. B., conducted in the absence of any com- on proximal gastric tone and on of HLA-DQ gene with bowel transit, and Camilleri, M. (1994). Sero- mercial or financial relationships that the perception of gastric disten- barrier function, and inflammation tonergic mediation of postprandial could be construed as a potential con- sion. Aliment. Pharmacol. Ther. 18, in irritable bowel syndrome with colonic tonic and phasic responses flict of interest. 1031–1037. diarrhea. Am. J. Physiol. Gastrointest. in humans. Gut 35, 536–541. Talley, N. J., Phillips, S. F., Haddad, Liver Physiol. 303, G1262–G1269. Walsh, J. H. (1994). Gastrointestinal Received: 13 December 2012; paper A., Miller, L. J., Twomey, C., Zins- Viramontes, B., Camilleri, M., McK- Hormones. Physiology of the Gas- pending published: 20 December 2012; meister, A. R., et al. (1990). GR inzie, S., Pardi, D. S., Burton, D., trointestinal Tract. New York: Raven, accepted: 10 January 2013; published 38032F (ondansetron), a selective and Thomforde, G. M. (2001a). 1–128. online: 01 February 2013. 5HT3 receptor antagonist, slows Gender-related differences in slow- Wehling, M. (2009). Assessing the trans- Citation: De Ponti F (2013) Drug devel- colonic transit in healthy man. Dig. ing colonic transit by a 5-HT3 latability of drug projects: what opment for the irritable bowel syn- Dis. Sci. 35, 477–480. antagonist in subjects with diarrhea- needs to be scored to predict success? drome: current challenges and future Tillisch, K., Labus, J., Nam, B., Bueller, J., predominant irritable bowel syn- Nat. Rev. Drug Discov. 8, 541–546. perspectives. Front. Pharmacol. 4:7. doi: Smith, S., Suyenobu, B., et al. (2012). drome. Am. J. Gastroenterol. 96, Wensel, T. M., and Luthin, D. R. (2011). 10.3389/fphar.2013.00007 Neurokinin-1-receptor antagonism 2671–2676. Linaclotide: a novel approach to This article was submitted to Frontiers decreases anxiety and emotional Viramontes, B. E., Malcolm, A., Camil- the treatment of irritable bowel in Gastrointestinal Pharmacology, a spe- arousal circuit response to noxious leri, M., Szarka, L. A., McKinzie, S., syndrome. Ann. Pharmacother. 45, cialty of Frontiers in Pharmacology. visceral distension in women with Burton, D. D., et al. (2001b). Effects 1535–1543. Copyright © 2013 De Ponti. This is an irritable bowel syndrome: a pilot of an a2-adrenergic agonist on gas- Wong, B. S., Camilleri, M., Busciglio, open-access article distributed under the study. Aliment. Pharmacol. Ther. 35, trointestinal transit, colonic motil- I., Carlson, P., Szarka, L. A., Bur- terms of the Creative Commons Attribu- 360–367. ity, and sensation in humans. Am. J. ton, D., et al. (2011). Pharmaco- tion License, which permits use, distrib- Tonini, M., De Ponti, F., Di Nucci, A., Physiol. 281, G1468–G1476. genetic trial of a cannabinoid ago- ution and reproduction in other forums, and Crema, F. (1999). Review article: von der Ohe, M. R., Camilleri, M., Kvols, nist shows reduced fasting colonic provided the original authors and source cardiac adverse effects of gastroin- L. K., and Thomforde, G. M. (1993). motility in patients with noncon- are credited and subject to any copy- testinal prokinetics. Aliment. Phar- Motor dysfunction of the small stipated irritable bowel syndrome. right notices concerning any third-party macol. Ther. 13, 1585–1591. bowel and colon in patients with the Gastroenterology 141, 1638–1647. graphics etc.

Frontiers in Pharmacology | Gastrointestinal Pharmacology February 2013 | Volume 4 | Article 7| 12