* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download Options for patients with irritable bowel syndrome
Survey
Document related concepts
Transcript
Neurogastroenterol Motil (2004) 16, 701–711 doi: 10.1111/j.1365-2982.2004.00550.x Options for patients with irritable bowel syndrome: contrasting traditional and novel serotonergic therapies J. F. JOHANSON Department of Medicine, University of Illinois College of Medicine, Rockford, IL, USA pation and diarrhoea [IBS-A]).4 Well-established diagnostic criteria are used to identify patients for clinical trials,4 but, in practice, a global definition of abdominal discomfort associated with altered bowel habits is more appropriate and clinically useful.1 The economic burden inflicted by IBS on both society and the health care system is tremendous, compared with that imposed by asthma, hypertension and stroke.5,6 The chronic and episodic nature of IBS symptoms accounts for up to 3.7 million doctor visits7 and more than two million prescriptions annually.8 Direct (e.g. doctor visits, outpatient care, diagnostic tests) and indirect (e.g. work absenteeism and decreased productivity) costs associated with IBS are estimated at $30 billion (adjusted for 1999 and 2000 dollars, respectively) per year.7,8 Symptoms experienced by persons with IBS significantly impact quality-of-life.9 Studies using general or disease-specific quality-of-life instruments have consistently shown that the health status of those with IBS is poorer than that of the general population.10–12 Quality-of-life scores of IBS patients are directly correlated with their responses to therapy,12 and management of multiple IBS symptoms is important for overall improvement in patient well-being. Until recently, treatment options for IBS have been limited by poor efficacy, poorly tolerated adverse effects or both. Research into the pathophysiology of IBS, which has uncovered the potential role of serotonin (5-HT), has led to the development of two serotonergic agents for the treatment of patients with IBS. This paper reviews the traditional single symptom-based treatment options for IBS and the novel serotonergic agents that target multiple IBS symptoms through pharmacological action in serotonin receptors. Abstract This article reviews the efficacy and tolerability of traditional therapies for irritable bowel syndrome (IBS) and concludes that they are limited by both poor efficacy and adverse effects. Serotonin, a neurotransmitter found mainly in the gut, appears to represent a link in IBS pathophysiological processes – altered gut motility, abnormal intestinal secretion and visceral hypersensitivity. Recently, available treatments for IBS have targeted serotonin receptors that are involved in motor, sensory and secretory functions of the gut. Serotonergic agents, such as alosetron (a 5-HT3 receptor antagonist) and tegaserod (a selective 5-HT4 receptor partial agonist), provide global relief of the multiple symptoms of IBS with diarrhoea and IBS with constipation, respectively, and represent important additions to the IBS treatment armamentarium. Keywords alosetron, constipation, irritable bowel syndrome, serotonin, tegaserod, treatment. OVERVIEW Irritable bowel syndrome (IBS) is one of the most common gastrointestinal (GI) disorders, with a prevalence of up to 20% in the United States.1 It is the most common diagnosis made by gastroenterologists (28% of total) and accounts for 12% of diagnoses made by primary care doctors.2,3 In North America, IBS affects twice as many women as men, and patients are generally between the ages of 30 and 50 years when they first consult a doctor.2 Irritable bowel syndrome is considered a functional bowel disorder characterized by abdominal discomfort or pain associated with altered bowel function (constipation [IBS-C], diarrhoea [IBS-D] or alternating constiAddress for correspondence John F. Johanson MSc, MD, Rockford Gastroenterology Associates, 401 Roxbury Road, Rockford, IL 61107, USA. Tel: 815-397-7340; fax: 815-397-2156; e-mail: [email protected] Received: 30 September 2003 Accepted for publication: 1 April 2004 2004 Blackwell Publishing Ltd TRADITIONAL IBS THERAPIES Despite their limitations, many patients continue to rely on traditional pharmacological and non-pharmacological treatments that target individual IBS 701 J. F. Johanson Neurogastroenterology and Motility examined the efficacy of these compounds for relieving the pain and discomfort associated with IBS.16–18 The two meta-analyses found that five16 and six17 smooth muscle relaxants have proven clinical efficacy in IBS without significant adverse reactions. In the systematic review, 13 studies, including seven high-quality trials, found these smooth muscle relaxants to be effective for abdominal pain;18 significant improvement in constipation was reported in two of the highquality trials. However, these agents are not available in the United States. Although dicyclomine and hyoscyamine are available in the United States for the treatment of IBS patients, clinical trials examining these agents have had numerous methodological flaws. Moreover, they have demonstrated inconsistent efficacy, making evidence-based assessment of efficacy difficult.1,18 Of the three trials evaluating dicyclomine, only one (40 mg four times daily) demonstrated significant symptom improvement compared with placebo. At this high dose, however, close to 70% of patients experienced adverse effects.20 In their systematic review of all evidence available for these agents, the ACG FGID Task Force concluded that the data are insufficient to allow a meaningful evaluation of the effectiveness of these agents.1 Tricyclic antidepressants have been used in low doses (i.e. lower than those used for depression) to alleviate the abdominal pain or discomfort associated with IBS.19,21 The mechanistic evidence that they alter motor or sensory function is limited and their effects may reflect a central action. A meta-analysis of eight clinical trials in which five TCAs were used to treat IBS patients showed these agents to be effective in reducing pain-related symptoms. However, these trials were not adequately blinded, included small numbers of patients and were of relatively short duration (averaging only 46 patients and 6 weeks). In addition, many were crossover trials, which are suboptimal in that the effects of the drug may not be completely washed out before the patient is Ôcrossed overÕ to the control.13 A systematic review of the six published clinical trials with TCAs indicated that, in general, these trials were not sufficiently powered for statistically valid conclusions to be drawn, and they suffered from suboptimal methodologies (e.g. not using Rome criteria for enrolling patients, short durations of therapy and high dropout rates); thus, the validity of their results is in question. Overall, the data are inadequate to conclude whether TCAs are more effective than placebo in providing global relief of IBS symptoms.1 A recent trial of desipramine did not show benefit over placebo in the intent-to-treat analysis.22 symptoms (Table 1). Traditional IBS management approaches often rely on combinations of dietary changes, antispasmodics, antidiarrhoeals, laxatives, antidepressants and analgesics. However, the efficacy (Table 2),1,13–18 safety and tolerability of these treatments have not been examined in well-controlled clinical trials. The American College of Gastroenterology (ACG) Functional Gastrointestinal Disorders (FGID) Task Force, performed a systematic review of the efficacy and safety of traditional therapies and found none of these agents demonstrated level I evidence (i.e. randomized, controlled trials with P-values <0.05, adequate sample sizes and appropriate methodology) to support their use in the treatment of IBS.1 It is important to note that the levels of evidence used by the Task Force were defined to ensure that level I randomized, controlled trials had few limitations, thereby minimizing type I errors, and were adequately powered to minimize type II errors. Thus, if a trial did show a significant difference between treatment and placebo, it is unlikely that this finding was due to biased study design. Moreover, if the trial did not show a significant difference between treatment and placebo, it would be unlikely that this finding was due to an inadequate sample size of patients. In addition, few studies of traditional therapies measure global relief of symptoms as an efficacy variable. Global symptom relief was identified by the Rome Committee to be the primary outcome measure incorporating the key symptoms of IBS, allowing the patient to integrate the contributions of multiple symptoms into a single global clinical rating. To date, there has been no compelling evidence that combining multiple modalities is effective in treating patients with IBS. Furthermore, the adverse effects of some treatments for IBS may outweigh their potential benefits. Antispasmodics and antidepressants Traditional medications evaluated in published clinical trials for the treatment of IBS-related abdominal pain or discomfort include antispasmodics and tricyclic antidepressants (TCAs). Some antispasmodics (e.g. cimetropium bromide) directly relax intestinal smooth muscle; others (e.g. dicyclomine) act indirectly via anticholinergic effects.19 Smooth muscle relaxants have been evaluated in a number of double-blind, randomized, placebo-controlled clinical trials with IBS patients. Three systematic reviews – two meta-analyses of 26 and 23 randomized, controlled trials, respectively, and a systematic review of 16 randomized, controlled trials – 702 2004 Blackwell Publishing Ltd 2004 Blackwell Publishing Ltd Abdominal pain/ discomfort Traditional Antispasmodics Dicyclomine Hyoscyamine 703 Constipation (stool frequency, stool consistency) 1. Urgency 2. Diarrhoea Serotonergic agonists/antagonists Tegaserod 1. Abdominal pain/ discomfort 2. Constipation 3. Bloating Bulking agents/fibre Psyllium Calcium polycarbophil Wheat bran Ispaghula/poloxamer Corn fibre Wheat fibre Ispaghula husk Antidiarrhoeal Loperamide Tricyclic antidepressants Desipramine Abdominal pain/ Trimipramine discomfort Doxepin Amitriptyline Targeted symptom(s) Therapy 1. Global IBS symptoms 2. Individual symptoms (e.g. abdominal pain/ discomfort, bloating, stool frequency, satisfaction with bowel habits) 1. Global IBS symptomsà 2. GI symptoms (e.g. abdominal pain, bowel movements) 1. SGA 2. Abdominal pain/ distension 3. Diarrhoea 4. Urgency GI symptoms (e.g. abdominal pain, vomiting, mucus in stool, belching) 1. Individual IBS symptoms 2. Symptom improvement 3. Global IBS 4. symptoms Study assessment(s) Table 1 Traditional and recently approved IBS pharmacotherapies 1. Acceptable duration (12 weeks) 2. Large samples (520–1519) 3. Optimal study design (12–13) 1. Short duration (3–12 weeks, most <8 weeks) 2. Small samples (most <30) 3. Suboptimal study design (5–9) 1. Short duration (3–5 weeks) 2. Small samples (30–90) 3. Suboptimal study design (5–7) 1. Short duration (4–8 weeks) 2. Small samples (most £31) 3. Suboptimal study design (quality score: 5–6) 1. Short duration (£8 weeks) 2. Small samples (29–96) 3. Suboptimal study design (quality score : 5–7) 4. Inconsistent effectiveness Study assessment Grade A recommendation: more effective than placebo in providing global relief of IBS symptoms in female patients with IBS-C Grade B recommendation: not more effective than placebo in providing global relief of IBS symptoms Grade B recommendation: improves diarrhoea; not more effective than placebo in providing global relief of IBS symptoms Grade B recommendation: not more effective than placebo in providing global relief of IBS symptoms. TCAs improve abdominal pain in IBS patients Grade B recommendation: insufficient data for recommendation about effectiveness of antispasmodic agents available in the United States Evidence-based classification*1 May cause mild, transient diarrhoea Limited adverse effect data, precluding evidence-based conclusions Increased fibre intake may worsen bloating and abdominal pain/ discomfort in patients with IBS Limited adverse effects data, precluding evidencebased conclusions Constipation and fatigue have been documented May cause anticholinergic adverse effects at higher doses Should be used cautiously in patients with constipation Evidence-based assessment of adverse effects1 Volume 16, Number 6, December 2004 Efficacy and tolerability of IBS therapies Serious, sometimes fatal, constipation-associated complications (e.g. ischaemic colitis) Neurogastroenterology and Motility The most common adverse effects among patients using low-dose TCAs for IBS-related abdominal pain include fatigue, dry mouth, sedation, weight gain, difficulty with urination and constipation.2 TCAs should, therefore, be used with caution in IBS-C patients and may be most beneficial for patients with IBS-D.19 Other limitations of these agents include a possible 6-week lag before therapeutic effects are seen 23 and the requirement of continual use for maintenance of effect.19 IBS, irritable bowel syndrome; IBS-C, irritable bowel syndrome with constipation; IBS-D, irritable bowel syndrome with diarrhoea; SGA, Subject’s Global Assessment; TCA, tricyclic antidepressant. *Evidence-based recommendations: Grade A – evidence based on a minimum of two randomized, controlled trials with P < 0.05, adequate sample size, and appropriate methodology. Grade B – evidence based on randomized, controlled trials with P > 0.05 and/or inadequate sample size and/or inappropriate methodology. Range 0–13. àDefinition of global symptoms was poorly defined in many studies. Alosetron 1. Abdominal pain/ discomfort 2. Fecal urgency 3. Diarrhoea 1. Global IBS symptoms 2. Individual symptoms (e.g. abdominal pain, fecal urgency, stool consistency, stool frequency, satisfaction with bowel habits) 1. Acceptable duration (12 weeks) 2. Large samples (370–801) 3. Optimal study design (12) Grade A recommendation: more effective than placebo in providing global relief of IBS symptoms in female patients with IBS-D May cause constipation J. F. Johanson Antidiarrhoeals The antidiarrhoeal agent most commonly used for IBSD patients is loperamide, which works by binding to opiate receptors.19 Loperamide improves diarrhoea, urgency, and fecal soiling in IBS-D patients by slowing intestinal transit, enhancing absorption of intestinal water and ions, and strengthening the resting tone of the anal sphincter.19,21,24 An evidence-based review of loperamide indicated that suboptimal trial design has limited assessment of its efficacy.1 Although loperamide appears to be an effective treatment for diarrhoea, it is not more effective than placebo in treating IBS-associated abdominal pain or in providing global relief of IBS symptoms.1 Fibre and laxatives Patients with IBS-C are often advised to increase dietary fibre intake or to take commercially available fibre products such as psyllium or calcium polycarbophil.24 Fibre has been shown to decrease whole-gut transit time in persons with mild transit delays, thereby alleviating constipation.19 However, in most instances of moderate-to-severe constipation, fibre provides little therapeutic benefit. Although fibre can be effective in alleviating constipation, its value for IBS patients remains unclear. Randomized crossover trials of fibre use in IBS patients did not show differences in symptom improvement between active treatment and control groups.25,26 The benefits of fibre for constipation have been achieved mainly with high doses (20–30 g day)1), which are associated with an increased incidence of adverse effects.19 Moreover, fibre frequently aggravates the bloating, distension and abdominal pain associated with IBS.19 In fact, a recent systematic review of 17 studies concluded that the benefits of fibre in the treatment of global IBS symptoms are marginal and fibre has no effect on the relief of abdominal pain.27 Thus, the benefit of fibre in IBS patients is dubious – fibre and bulking agents do not appear to be more 704 2004 Blackwell Publishing Ltd Volume 16, Number 6, December 2004 Efficacy and tolerability of IBS therapies Table 2 Efficacy of traditional therapies for IBS Therapy No. of trials NNT* Agents tested Outcome measurements Antispasmodics 26 trials16 23 trials17 16 trials18 3.7 5.6 1.6–6.7 Global improvement, individual symptom improvement (abdominal pain/discomfort, abdominal distension, constipation, transit) Antidepressants 11 trials13 3.2 (95% CI, 2.1–6.5) Antidiarrhoeal agents One trial18 3.6 Cimetropium Mebeverine Dicyclomine Trimebutine Rociverine Diltiazem Pinaverium Otilonium Pirenzepine Pirifinium Amitriptyline Cloipramine Desipramine Doxepin Trimipramine Mianserin Loperamide Bulking agents Three trials18 2.2–3.5 Tegaserod Four trials (phase III)15 10 (95% CI, 7.1–20) Alosetron Six trials14 7 (95% CI, 5.7–9.4) Psyllium Coarse bran Concentrated fibre Corn fibre Polycarbophil Ispaghula husk 6 mg b.i.d. tegaserod Alosetron Global improvement, individual symptom improvement (abdominal pain/discomfort, diarrhoea/constipation, ability to work) Global improvement (stool frequency, stool consistency, abdominal pain/discomfort), diarrhoea improvement Global improvement, individual symptom improvement (stool passage, satisfaction with bowel movements, constipation, stool frequency, abdominal pain, bloating) Global IBS symptom improvement (abdominal pain/discomfort, bloating, constipation), individual symptom improvement (abdominal pain/discomfort, bloating, stool frequency) Global IBS symptom improvement (abdominal pain, diarrhoea, urgency), individual symptom improvement *Global assessment. effective than placebo in providing global relief of IBS symptoms.1 Laxatives prescribed for IBS-C patients include osmotic laxatives (e.g. polyethylene glycol, lactulose and magnesium-containing products), stimulant laxatives (e.g. bisacodyl or cascara) and emollients (e.g. docusate). With the exception of polyethylene glycol, the efficacy of laxatives has not been established in randomized, controlled trials in the United States, although their effectiveness in relieving constipation is generally well accepted. However, some medications in this class (e.g. lactulose) can worsen symptoms common to IBS, such as bloating and cramping, and excessive use can lead to diarrhoea and dehydration. Long-term laxative use, especially of stimulant laxatives, generally is not recommended.19 Finally, laxatives do not treat IBS-associated abdominal pain or discomfort.19 2004 Blackwell Publishing Ltd Behavioural therapy Increasing evidence indicates that behavioural therapy may benefit IBS patients, particularly those with co-morbid psychiatric conditions (e.g. depression or severe anxiety) – estimated to be approximately 18% of IBS patients in the community.28 In one study, 23 patients with IBS were assessed before and after 12 weeks of hypnotherapy focused on the GI system; the results suggest that hypnotherapy improves abnormal sensory perception in IBS patients, while leaving normal sensation unchanged.29 The benefits of psychotherapy were evaluated by the FGID Task Force, whose members examined clinical trials that compared behavioural treatments (e.g. psychotherapy, hypnotherapy and relaxation) with placebo.1 Most clinical trials published to date suggest that behavioural therapy is more effective than placebo in alleviating 705 J. F. Johanson Neurogastroenterology and Motility individual IBS symptoms.1 However, numerous limitations in study design make difficult the determination of efficacy of behavioural therapy in the treatment of IBS patients. A recently reported, well designed trial of cognitive behavioural therapy (CBT) vs education demonstrated that CBT was efficacious.22 PATHOPHYSIOLOGICAL BASIS FOR SEROTONERGIC AGENTS IN IRRITABLE BOWEL SYNDROME A number of discoveries over the past several years have shed new light on the pathophysiology of IBS and have focused attention on processes that regulate bowel sensorimotor responses and visceral pain perception. A unifying theory is that IBS involves dysregulation of the brain–gut axis. The enteric nervous system (ENS) operates semi autonomously in controlling intrinsic bowel functions (both motility and secretion) and is modified centrally by the central nervous system (CNS), which supplies extrinsic sympathetic and parasympathetic innervation to the GI tract via the autonomic nervous system.31,32 Disruptions along the brain–gut communication path likely cause the various symptoms of IBS, such as altered gut motility, abnormal intestinal secretion and visceral hypersensitivity.33,34 The neurotransmitter serotonin is a common link among these three processes – GI motility (peristaltic reflex), secretion and perception of visceral pain. Serotonin is an important signalling molecule in the ENS,31 and the GI tract is the largest source of serotonin in mammals; 95% is synthesized, stored and secreted in the gut.33,34 Serotonin acts on several receptors, including the 5-HT1P, 5-HT3and 5-HT4 receptor subtypes localized in the gut, mediating numerous GI tract functions,33,34 such as stimulation of secretory and peristaltic reflexes (via intrinsic sensory neurones) and modulation of visceral sensation (via extrinsic sensory neurones and vagal spinal afferent neurones).34 Its diverse activity in modulating these processes is believed to be pivotal in the normalization of GI function, making 5-HT receptors and their associated neuronal pathways important targets for IBS-specific drugs. WHAT DO PATIENTS THINK OF TRADITIONAL IBS THERAPIES It is not surprising that IBS patients are often dissatisfied with the efficacy and tolerability of traditional treatment options that address single symptoms. In a survey conducted by the International Foundation for Functional Gastrointestinal Disorders, 350 IBS patients (chosen randomly from a database of IBS patients in the United States) responded to questions regarding their IBS symptoms, the impact of these symptoms on their lifestyles and their satisfaction with treatment.9 Eighty-eight per cent of the respondents reported using prescription medications (antispasmodics and antidepressants being among the most common) at some point; 78 and 64% acknowledged using over-the-counter laxatives or antidiarrhoeals, respectively. Less than half (45%) of patients surveyed believed their prescription IBS medications were effective, and 40% reported being dissatisfied with their current IBS medications or remedies. Dissatisfaction was related primarily to lack of effectiveness, although adverse effects, likely resulting from long-term use and the interaction of multiple medications, also contributed to patient dissatisfaction. Of those persons taking prescription medications, 62% reported adverse effects.9 The results of a recent study reviewing utilization trends of GI medications by IBS patients support the hypothesis that patient dissatisfaction leads to the use of multiple medications, switching of medications, and repeated doctor visits. In this study, 5960 outpatient service claims records from 1995 to 1997 for the 12 months following an initial diagnosis of IBS were analysed.30 The results indicated that 53% of patients used prescription GI medications continuously throughout the year following IBS diagnosis – 36% used monotherapy and 64% used polytherapy. Among IBS patients using multiple IBS therapies, 22% switched from one drug to another, and 42% augmented the originally prescribed drug with another GI medication from a different drug class. In the year after the index diagnosis, expenditures related to outpatient services and pharmaceuticals were higher for polytherapy users who switched and augmented treatment than for monotherapy patients.30 GI motility and secretion Irritable bowel syndrome patients have demonstrated increased intestinal motility in response to both intrinsic (e.g. food intake) and environmental factors (e.g. stress).35 In general, 5-HT3 receptors are associated with excitation in the GI tract, resulting in increased motility, secretion and sensation.33 5-HT3 antagonists, therefore, slow colonic transit and increase fluid absorption, thus improving symptoms of IBS-D. 5-HT4 receptors mediate both excitatory or inhibitory effects on gut function.33,34 In vitro studies indicate that 5-HT4 receptors are essential mediators of the peristaltic reflex and synthetic 5-HT4 receptor agonists 706 2004 Blackwell Publishing Ltd Volume 16, Number 6, December 2004 Efficacy and tolerability of IBS therapies have been found to increase gut motility.36 Although the mechanism underlying the development of IBS is unknown, one hypothesis is that patients with different bowel symptoms differ in colonic 5-HT concentration 37 and/or perhaps in 5-HT receptor composition.38,39 Intestinal secretion, which is also mediated by 5-HT3 and 5-HT4 receptors, can be modulated by synthetic agonists or antagonists.33 Antagonism of 5-HT3 receptors leads to decreased fluid secretion and 5-HT4 receptor agonism results in increased intestinal fluid and electrolyte secretion.33,40,41 Alosetron Alosetron hydrochloride is currently indicated for use in women with severe diarrhoea-predominant IBS in whom traditional therapy has failed. Initial trials demonstrated that alosetron decreased gut transit in non-IBS and IBS subjects,50 increased fluid absorption by normal human intestine43 and reduced pain perception upon rectal distension in IBS patients.51 These pharmacodynamic effects are believed to be the reason for alosetron’s effective alleviation of multiple IBS symptoms, including abdominal pain and discomfort, diarrhoea, and urgency. Two large, randomized, placebo-controlled, clinical trials comprising 1273 women assessed the efficacy of alosetron 1 mg twice daily for 12 weeks among patients with diarrhoea-predominant IBS.52,53 A comparator trial of 623 women with IBS assessed alosetron 1 mg twice daily vs the antispasmodic mebeverine.54 Significantly more subjects treated with alosetron reported adequate relief of abdominal pain and discomfort for all 3 months of treatment compared with those receiving placebo/comparator treatment.52–54 Alosetron also improved stool consistency and decreased stool frequency and bowel urgency compared with placebo. However, in the single study that examined the effects of alosetron on bloating, no statistically significant difference was observed between the two groups during the 12-week treatment course.53 Constipation was the most common adverse effect reported in these trials 52–54 and may be expected to occur in up to 25% of patients treated continually. Approximately 10% of patients taking alosetron withdrew prematurely because of constipation-related adverse events.53 Alosetron 1 mg twice daily was also assessed in a placebo-controlled clinical trial of 801 women with severe diarrhoea-predominant IBS – enrolled patients experienced symptoms of urgency on at least 50% of days at study entry.55 Compared with placebo, subjects taking alosetron had a significantly greater proportion of days with satisfactory control of urgency, as well as greater global improvement in IBS symptoms. Alosetron-treated subjects also showed improvement in bowel symptoms compared with those receiving placebo. The most common adverse effects, which were reported in at least 1% of IBS patients taking alosetron in clinical trials, included constipation, abdominal discomfort/pain and nausea. In patients receiving alosetron 1 mg twice daily, constipation was generally mildly to moderately severe and transient (typically occurring as a single episode during the first month of treatment); it resolved on its own or with an interruption in Visceral hypersensitivity Irritable bowel syndrome patients also appear to differ from controls in the way the CNS processes colorectal pain stimulation.42,43 It is interesting to note that this hypersensitivity to distension is not restricted to the distal bowel – IBS patients experience hypersensitivity to distension throughout the entire GI tract. Therefore, abnormal neurosensory function may occur in both the CNS and the peripheral nervous system in IBS patients.30 Alosetron, a 5HT3 antagonist, changes the pattern of blood flow during noxious rectal distension activating centres that downregulate pain sensation.44,45 Serotonin appears to be involved in modulating visceral hypersensitivity in IBS patients. Results from animal studies suggest that 5-HT3 receptors mediate visceral nocioceptive signals via spinal vagal afferent neurones.46 Painful colonic distension from visceral stimuli leads to increased CNS activation that is inhibited in a dose-dependent manner by 5-HT3 receptor antagonism.46 Similarly, 5-HT4 receptors are believed to mediate visceral sensation and perception in experimental animals. The firing rate of rectal spinal afferents decreases in response to slow pressure distension when a 5-HT4 receptor agonist is applied.47 Moreover, 5-HT4 receptor agonism inhibits the visceral pain responses and afferent signalling induced by rectal distension.48 Data from human studies also suggest they alter reflex sensory responses.49 SEROTONERGIC AGENTS: ALOSETRON AND TEGASEROD The preclinical studies described previously led to the clinical development and approval of two medications for the treatment of IBS: alosetron, a 5-HT3 receptor antagonist, and tegaserod, a selective 5-HT4 receptor agonist. 2004 Blackwell Publishing Ltd 707 J. F. Johanson Neurogastroenterology and Motility well as in the single symptoms of abdominal pain/ discomfort, number of bowel movements and stool consistency.62,63 Significant improvement in daily bloating scores was also observed in two of these trials,61,62 and one study showed that tegaserod-treated patients experienced a significant reduction in the number of days with straining, compared with controls.61 More recently, the efficacy of tegaserod was evaluated in randomized controlled trials among Asian-Pacific and Nordic populations with IBS whose primary bowel habits were not diarrhoea. In both studies, patients received either tegaserod 6 mg b.i.d. or placebo for 12 weeks. The results showed that the mean proportion of patients with overall satisfactory relief was statistically significantly greater in the tegaserod group compared with placebo for the majority of the study period.64,65 Reductions in the number of days with at least moderate abdominal pain/discomfort, bloating, no bowel movements and hard/lumpy stools were also greater in the tegaserod group.64 Tegaserod was well tolerated, with diarrhoea and headache being the most frequently reported adverse effects.40,61,62 In the 12-week trials, diarrhoea occurred in 9 and 4% of patients treated with tegaserod and placebo, respectively. The diarrhoea was typically mild and transient, causing less than 2% of subjects to withdraw prematurely. Headaches occurred more frequently among those taking tegaserod (15% vs 12%), but the incidence of migraine headaches was similar between the two treatment groups. The long-term safety of tegaserod has been assessed in a multicentre, open-label, clinical trial of patients with IBS-C.66 The most common adverse effects related to tegaserod were mild and transient diarrhoea (10.1%), headache (8.3%), abdominal pain (7.4%) and flatulence (5.5%). The adverse effects profile, clinical laboratory evaluations, vital signs and electrocardiogram recordings revealed no evidence of any unexpected adverse effects and suggested that tegaserod treatment was safe over a 12-month period.66 Because IBS patients may alternate between constipation and diarrhoea, the safety of tegaserod in patients with IBS-D was also examined. 67 In a randomized, placebo-controlled, double-blind trial comparing patients with IBS-D receiving either placebo or 4 or 12 mg of tegaserod per day, no complications of diarrhoea or serious adverse effects were reported. When data from the two tegaserod groups were pooled, the overall frequency of diarrhoea was similar among patients receiving tegaserod (33%) and placebo (35%).67 Tegaserod has also been shown to be devoid of untoward electrocardiographic effects. Recent studies have evaluated its cardiac safety profile. One analysed treatment. However, serious complications such as constipation requiring hospitalization or ischaemic colitis have been reported in clinical studies and in postmarketing experience.41 These serious complications occurred in approximately one of 1000 patients. The cumulative incidence of ischaemic colitis in women taking alosetron was two of 1000 patients during 3 months and three of 1000 patients during 6 months. Long-term safety data have yet to be established.41 Alosetron was reintroduced in the United States in November 2002 after it had been voluntarily withdrawn from the marketplace in 2001 and is now indicated only for women with severe diarrhoeapredominant IBS for whom conventional IBS therapies have failed. A number of restrictions have been imposed on those prescribing alosetron, including mandatory doctor training and certification. Patients must also sign consent forms confirming the severity of their symptoms and their understanding of the potential risks associated with alosetron therapy. While undergoing treatment, patients should be monitored for the development of severe constipation or ischaemic colitis. In addition, alosetron should be used cautiously in patients with hepatic insufficiency because of its extensive metabolism by the liver.41 Tegaserod The selective 5-HT4 receptor agonist tegaserod is indicated for women with IBS whose primary bowel symptom is constipation. Tegaserod is the first in a novel class of drugs (aminoguanidine indoles) designed to be similar in structure to serotonin and to act selectively at 5-HT4 receptors in the GI tract. Tegaserod accelerates overall GI transit in healthy subjects 56 and promotes gastric emptying, small bowel transit and colonic transit in IBS-C patients.57,58 Tegaserod was found to reduce the firing rate of spinal afferents in vivo,47,59 supporting its role in the modulation of visceral sensitivity. Tegaserod also increased fecal water and intestinal secretion in female subjects.60 The efficay of tegaserod has been evaluated in several large, multicentre, randomized, double-blind, placebo-controlled trials.40,61,62 In these trials, patients with IBS-C received placebo or tegaserod (2 or 6 mg twice daily) for 12 weeks. Two of the trials included 4-week follow-up without medication.40,61 When compared with placebo, tegaserod 6 mg twice daily produced statistically significant improvement in the Subject’s Global Assessment (a validated 5-point ordinal scale) of relief (overall symptom improvement), as 708 2004 Blackwell Publishing Ltd Volume 16, Number 6, December 2004 Efficacy and tolerability of IBS therapies electrocardiographic data (11 535 electrocardiograms) from 2516 IBS patients participating in various clinical trials (receiving tegaserod 2 or 6 mg twice daily or placebo); the other analysed data from a study of 36 healthy male patients (receiving tegaserod 0.8–20 mg day)1 intravenously). Clinical trial results revealed that tegaserod was no more likely than placebo to result in an electrocardiographic abnormality, and no serious dysrhythmias were reported. In healthy volunteers, no cardiac effects were noted, even when drug plasma concentrations reached 100 times those observed after a typical therapeutic dose (12 mg day)1).68 Tegaserod is classified as pregnancy category B.40 Although animal studies have indicated that tegaserod does not impair fertility or cause fetal harm at concentrations 15 to 51 times those expected at recommended doses, no adequate, well-controlled studies have been undertaken in pregnant women.40 Because experience drawn from clinical trials regarding tegaserod and pregnancy has been limited (15 unintended pregnancies on tegaserod and five on placebo), tegaserod is not recommended during pregnancy.69 Use in nursing mothers must be evaluated carefully as well; it is not known whether tegaserod is excreted in human milk, but it is excreted in the milk of lactating rats with high milk-to-plasma ratios.40 Adding alosetron and tegaserod to the armamentarium of treatments for patients with IBS offers patients the option of therapies that target IBS-specific pathophysiological pathways. Although this focus may help to provide global relief of the multiple symptoms of IBS for patients, it is a first step. These therapies do not address all the needs of all patients with IBS and more work in the area of IBS management needs to be done in order to provide all IBS patients with optimal treatment of their symptoms. It is important to note, however, that even in the clinical trials that show these medications to be superior to placebo or other comparators, the proportion of patients with global relief rarely reaches 70%. It is clear, therefore, that there is still room for improvement in IBS therapy. SUMMARY AND CONCLUSIONS Until recently, no drugs targeted IBS-specific pathophysiological pathways or the multiple symptoms of IBS. Patients are often dissatisfied with the efficacy and adverse effect profiles of traditional therapies. This leads to multiple doctor visits and frequent medication switching or augmentation – factors that often lead to increased medical costs. Alosetron and tegaserod represent a step in the right direction for the treatment of patients with IBS by providing these patients with options for treating the multiple and varied symptoms of IBS, resulting in global symptom improvement. The future management of IBS appears promising in that agents now in clinical development will follow the lead of serotonergic modulators in targeting the multiple symptoms of IBS. SUPPORT FOR THE USE OF SEROTONERGIC AGENTS IN THE TREATMENT OF PATIENTS WITH IBS In addition to evaluating traditional IBS therapies, the FGID Task Force used an evidence-based approach to evaluate the published tegaserod and alosetron clinical trials.1 Based on study methodologies and clinical trial evidence, tegaserod and alosetron were the only IBS therapies to receive grade A recommendations from the Task Force. This is not surprising given the fact that criteria for determining optimal study design were published relatively recently. Nevertheless, the Task Force concluded that, when compared with placebo, these agents provided significant global relief of symptoms of IBS-C and IBS-D, respectively. Results of a recent meta-analysis of six alosetronrandomized, placebo-controlled clinical trials are analogous with the conclusions of the ACG FGID Task Force, providing further evidence to support the efficacy of alosetron in women with diarrhoea-predominant IBS.14 The meta-analysis used the Quality of Reports of Meta-analyses statement guidelines for ensuring quality of the analysis.70 2004 Blackwell Publishing Ltd REFERENCES 1 Brandt LJ, Locke GR, Olden K et al. An evidence-based approach to the management of irritable bowel syndrome in North America. Am J Gastroenterol 2002; 97(Suppl.): S1–28. 2 Drossman DA, Camilleri M, Mayer EA, Whitehead WE. AGA technical review on irritable bowel syndrome. Gastroenterology 2002; 123: 2108–31. 3 Mitchell CM, Drossman DA. Survey of the AGA membership relating to patients with functional gastrointestinal disorders. Gastroenterology 1987; 92: 1282–4. 4 Thompson WG, Longstreth GF, Drossman DA, Heaton KW, Irvine EJ, Muller-Lissner SA. Functional bowel disorders and functional abdominal pain. Gut 1999; 45(Suppl. II): II43–7. 5 American Heart Association. 2002 Heart and Stroke Statistical Update Dallas, TX: American Heart Association, 2002: 1–35. 6 Druss BG, Marcus SC, Olfson M, Tanielian T, Elinson L, Pincus HA. Comparing the national economic burden of 709 J. F. Johanson 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 Neurogastroenterology and Motility 24 Rosemore JG, Lacy BE. Irritable bowel syndrome: basis of clinical management strategies. J Clin Gastroenterol 2002; 35(Suppl.): S37–44. 25 Cook IJ, Irvine EJ, Campbell D, Shannon S, Reddy SN, Collins SM. Effect of dietary fiber on symptoms and rectosigmoid motility in patients with irritable bowel syndrome: a controlled, crossover study. Gastroenterology 1990; 98: 66–72. 26 Lucey MR, Clark ML, Lowndes J, Dawson AM. Is bran efficacious in irritable bowel syndrome? A double blind placebo controlled crossover study. Gut 1987; 28: 221–5. 27 Bijkerk CJ, Muris JVM, Knottnerus JA, Hoes AW, De Wit NJ. Systematic review: the role of different types of fibre in the treatment of irritable bowel syndrome. Aliment Pharmacol Ther 2004; 19: 245–51. 28 Walker EA, Katon WJ, Jemelka RP, Roy-Bryne PP. Comorbidity of gastrointestinal complaints, depression, and anxiety in the Epidemiologic Catchment Area (ECA) Study. Am J Med 1992; 92: 26S–30. 29 Lea R, Houghton LA, Calvert EL et al. Gut-focused hypnotherapy normalizes disordered rectal sensitivity in patients with irritable bowel syndrome. Aliment Pharmacol Ther 2003; 17: 635–42. 30 Zacker C, Albers LA, Chawla A, Wang S.Drug utilization patterns in patients with irritable bowel syndrome. Presented at: Drug Information Association Annual Meeting, June 22, 2000, San Diego, CA. 31 Hunt RH. Evolving concepts in the pathophysiology of functional gastrointestinal disorder. J Clin Gastroenterol 2002; 35(Suppl.): S2–6. 32 Wood JD. Neuropathophysiology of irritable bowel syndrome. J Clin Gastroenterol 2002; 35(Suppl.): S11–22. 33 Crowell MD. The role of serotonin in the pathophysiology of irritable bowel syndrome. Am J Manag Care 2001; 7(Suppl. 8): S252–60. 34 Gershon MD. Review article: roles played by 5-hydroxytryptamine in the physiology of the bowel. Aliment Pharmacol Ther 1999; 13(Suppl. 2): 15–30. 35 Ringel Y, Drossman DA. Irritable bowel syndrome: classification and conceptualization. J Clin Gastroenterol 2002; 35(Suppl.): S7–10. 36 Grider JR, Foxx-Orenstein AE, Jin JG. 5-Hydroxytryptamine4 receptor agonists initiate the peristaltic reflex in human, rat, and guinea pig intestine. Gastroenterology 1998; 115: 370–80. 37 Miwa J, Echizen H, Matsueda K, Umeda N. Patients with constipation-predominant irritable bowel syndrome (IBS) may have elevated serotonin concentrations in colonic mucosa as compared with diarrhea-predominant patients and subjects with normal bowel habits. Digestion 2001; 63: 188–94. 38 Spiller RC. Role of nerves in enteric infection. Gut 2002; 51: 759–62. 39 Linden DR, Chen JX, Gershon MD, Sharkey KA, Mawe GM. Serotonin availability is increased in mucosa of guinea pigs with TNBS-induced colitis. Am J Physiol Gastrointest Liver Physiol 2003; 285: G207–16. 40 Novartis Pharmaceuticals Corporation. Zelnorm (tegaserod maleate) Prescribing Information East Hanover, NJ: Novartis Pharmaceuticals Corporation, 2002. 41 GlaxoSmithKline. Lotronex (alosetron hydrochloride) Prescribing Information Research Triangle Park, NC: GlaxoSmithKline, 2002. five chronic conditions. Health Aff (Milwood) 2001; 20: 233–41. American Gastroenterological Association. The Burden of Gastrointestinal Diseases Bethesda, MD: American Gastroenterological Association, 2001: 1–86 (available at: http://www.gastro.org/pdf/burden-report.pdf). Martin R, Barron JJ, Zacker C. Irritable bowel syndrome: toward a cost-effective management approach. Am J Manag Care 2001; 7(Suppl. 8): S268–75. International Foundation for Functional Gastrointestinal Disorders. IBS in the Real World Survey Milwaukee, WI: International Foundation for Functional Gastrointestinal Disorders, 2002: 1–19. Gralnek IM. Health care utilization and economic issues in irritable bowel syndrome. Eur J Surg 1998; 164(Suppl.): 73–6. Frank L, Kleinman L, Rentz A, Ciesla G, Kim JJ, Zacker C. Health-related quality of life associated with irritable bowel syndrome: comparison with other chronic diseases. Clin Ther 2002; 24: 675–89. El Serag HB, Olden K, Bjorkman D. Health-related quality of life among persons with irritable bowel syndrome: a systematic review. Aliment Pharmacol Ther 2002; 16: 1171–85. Jackson JL, O’Malley PG, Tomkins G, Balden E, Santoro J, Kroenke K. Treatment of functional gastrointestinal disorders with antidepressant medications: a meta-analysis. Am J Med 2000; 108: 65–72. Cremonini F, Delgado-Aros S, Camilleri M. Efficacy of alosetron in irritable bowel syndrome: a meta-analysis of randomized controlled trials. Neurogastroenterol Motil 2003; 15: 79–86. Schoenfeld P, Chey WD, Drossman D, Kim HM, Thompson WG. Effectiveness and safety of tegaserod in the treatment of irritable bowel syndrome: a meta-analysis of randomized control trials. Gastroenterology 2002; 122(Suppl.): A465. Poynard T, Naveau S, Mory B, Chaput JC. Meta-analysis of smooth muscle relaxants in the treatment of irritable bowel syndrome. Aliment Pharmacol Ther 1994; 8: 499– 510. Poynard T, Regimbeau C, Benhamou Y. Meta-analysis of smooth muscle relaxants in the treatment of irritable bowel syndrome. Aliment Pharmacol Ther 2001; 15: 355– 61. Jailwala J, Imperiale TF, Kroenke K. Pharmacologic treatment of the irritable bowel syndrome: a systematic review of randomized, controlled trials. Ann Intern Med 2000; 133: 136–47. Lembo T, Rink R. Current pharmacologic treatments of irritable bowel syndrome. Participate 2002; 11: 1–4. Page JG, Dirnberger GM. Treatment of the irritable bowel syndrome with bentyl (dicyclomine hydrochloride). J Clin Gastroenterol 1981; 3: 153–6. Camilleri M. Review article: clinical evidence to support current therapies of irritable bowel syndrome. Aliment Pharmacol Ther 1999; 13(Suppl. 2): 48–53. Drossman DA, Toner BB, Whitehead WE et al. Cognitivebehavioral therapy versus education and desipramine versus placebo for moderate to severe functional bowel disorders. Gastroenterology 2003; 125: 19–31. Wald A. Psychotrophic agents in irritable bowel syndrome. J Clin Gastroenterol 2002; 35(Suppl.): S53–7. 710 2004 Blackwell Publishing Ltd Volume 16, Number 6, December 2004 Efficacy and tolerability of IBS therapies 57 Prather CM, Camilleri M, Zinsmeister AR, Thomforde G, McKinzie S. Tegaserod accelerates orocecal transit in patients with constipation-predominant irritable bowel syndrome. Gastroenterology 2000; 118: 463–8. 58 Petrig C, Templeton J, Matzinger D, Ruegg P, Beglinger C, Degen L. Effect of tegaserod on gastrointestinal transit in male and female subjects. Gut 2002; 51(Suppl. III): A138. 59 Schikowski A, Mathis C, Thewiben M, Ross HG, Pak MA, Enck P. Dose-dependent modulation of rectal afferent sensitivity by a 5-HT4 receptor agonist. Gastroenterology 1999; 116(Suppl. 2): A643. 60 Petrig C, Templeton J, Matzinger D, Ruegg P, Beglinger C, Degen L. Effect of tegaserod on faecal water and electrolyte secretion in male and female subjects. Gut 2002; 51(Suppl. III): A249. 61 Novick J, Miner P, Krause R et al. A randomized, doubleblind, placebo-controlled trial of tegaserod in female patients suffering from irritable bowel syndrome with constipation. Aliment Pharmacol Ther 2002; 16: 1877– 88. 62 Muller-Lissner SA, Fumagalli I, Bardhan KD et al. Tegaserod, a 5-HT4receptor partial agonist, relieves symptoms in irritable bowel syndrome patients with abdominal pain, bloating and constipation. Aliment Pharmacol Ther 2001; 15: 1655–66. 63 Lefkowitz MP, Ruegg P, Shi Y, Dunger-Baldauf C. Relief of overall gastrointestinal symptoms and abdominal pain and discomfort as outcome measures in a clinical trial of irritable bowel syndrome with tegaserod. Gastroenterology 1999; 116: A1027. 64 Kellow J, Lee OY, Chang FY et al. An Asia-Pacific, double blind, placebo controlled, randomized study to evaluate the efficacy, safety, and tolerability of tegaserod in patients with irritable bowel syndrome. Gut 2003; 52: 671–6. 65 Nyhlin H, Bang C, Elsborg L et al. Tegaserod is an effective and safe therapy for irritable bowel syndrome in a Nordic population. Gastroenterology 2003; 124(Suppl. 1): A389. 66 Tougas G, Snape WJ, Otten MH et al. Long-term safety of tegaserod in patients with constipation-predominant irritable bowel syndrome. Aliment Pharmacol Ther 2002; 16: 1701–8. 67 Fidelholtz J, Smith W, Rawls J et al. Safety and tolerability of tegaserod in patients with irritable bowel syndrome and diarrhea symptoms. Am J Gastroenterol 2002; 97: 1176– 81. 68 Morganroth J, Ruegg PC, Dunger-Baldauf C, AppelDingemanse S, Bliesath H, Lefkowitz M. Tegaserod, a 5-hydroxytryptamine type 4 receptor partial agonist, is devoid of electrocardiographic effects. Am J Gastroenterol 2002; 97: 2321–7. 69 Appel-Dingemanse S. Clinical pharmacokinetics of tegaserod, a serotonin 5-HT(4) receptor partial agonist with promotile activity. Clin Pharmacokinet 2002; 41: 1021– 42. 70 Moher D, Cook DJ, Eastwood S, Olkin I, Rennie D, Stroup DF. Improving the quality of reports of meta-analyses of randomized controlled trials: the QUOROM statement. Onkologie 2000; 23: 597–602. 42 Silverman DH, Munakata JA, Ennes H, Mandelkern MA, Hoh CK, Mayer EA. Regional cerebral activity in normal and pathological perception of visceral pain. Gastroenterology 1997; 112: 64–72. 43 Mertz H, Morgan V, Tanner G et al. Regional cerebral activation in irritable bowel syndrome and control subjects with painful and nonpainful rectal distention. Gastroenterology 2000; 118: 842–8. 44 Chang L, Berman S, Mayer EA et al. Brain responses to visceral and somatic stimuli in patients with irritable bowel syndrome with and without fibromyalgia. Am J Gastroenterol 2003; 98: 1354–61. 45 Naliboff BD, Berman S, Chang L et al. Sex-related differences in IBS patients: central processing of visceral stimuli. Gastroenterology 2003; 124: 1738–47. 46 Kozlowski CM, Green A, Grundy D, Boissonade FM, Bountra C. The 5-HT3 receptor antagonist alosetron inhibits the colorectal distention induced depressor response and spinal c-fos expression in the anaesthetised rat. Gut 2000; 46: 474–80. 47 Mathis C, Schikowski A, Thewiben M, Ross HG, Pak MA, Enck P. Modulation of rectal spinal afferent sensitivity via 5-HT4 receptors. Neurogastroenterol Motil 1998; 10: 459. 48 Schikowski A, Thewissen M, Mathis C, Ross HG, Enck P. Serotonin type-4 receptors modulate the sensitivity of intramural mechanoreceptive afferents of the cat rectum. Neurogastroenterol Motil 2002; 14: 221–7. 49 Coffin B, Farmachidi JP, Rueegg P, Bastie A, Bouhassira D. Tegaserod, a 5-HT4 receptor partial agonist, decreases sensitivity to rectal distension in healthy subjects. Aliment Pharmacol Ther 2003; 17: 577–85. 50 Houghton LA, Foster JM, Whorwell PJ. Alosetron, a 5-HT3 receptor antagonist, delays colonic transit in patients with irritable bowel syndrome and healthy volunteers. Aliment Pharmacol Ther 2000; 14: 775–82. 51 Delvaux M, Louvel D, Mamet JP, Campos-Oriola R, Frexinos J. Effect of alosetron on responses to colonic distension in patients with irritable bowel syndrome. Aliment Pharmacol Ther 1998; 12: 849–55. 52 Camilleri M, Northcutt AR, Kong S, Dukes GE, McSorley D, Mangel AW. Efficacy and safety of alosetron in women with irritable bowel syndrome: a randomized, placebocontrolled trial. Lancet 2000; 355: 1035–40. 53 Camilleri M, Chey WY, Mayer EA et al. A randomized controlled clinical trial of the serotonin type 3 receptor antagonist alosetron in women with diarrhea-predominant irritable bowel syndrome. Arch Intern Med 2001; 161: 1733–40. 54 Jones RH, Holtmann G, Rodrigo L et al. Alosetron relieves pain and improves bowel function compared with mebeverine in female nonconstipated irritable bowel syndrome patients. Aliment Pharmacol Ther 1999; 13: 1419–27. 55 Lembo T, Wright RA, Bagby B et al. Alosetron controls bowel urgency and provides global symptom improvement in women with diarrhea-predominant irritable bowel syndrome. Am J Gastroenterol 2001; 96: 2662–70. 56 Degen L, Matzinger D, Maecke H et al. Tegaserod (HTF 919), a partial 5-HT4 receptor agonist, accelerates gastrointestinal (GI) transit. Gastroenterology 2000; 118: A845. 2004 Blackwell Publishing Ltd 711