111
Views
13
CrossRef citations to date
0
Altmetric
Review

Methylnaltrexone in the treatment of opioid-induced constipation

&
Pages 49-58 | Published online: 14 Dec 2008

Abstract

Constipation is a significant problem related to opioid medications used to manage pain. This review attempts to outline the latest findings related to the therapeutic usefulness of a μ opioid receptor antagonist, methylnaltrexone in the treatment of opioid-induced constipation. The review highlights methylnaltrexone bromide (Relistor™; Progenics/Wyeth) a quaternary derivative of naltrexone, which was recently approved in the United States, Europe and Canada. The Food and Drug Administration in the United States approved a subcutaneous injection for the treatment of opioid bowel dysfunction in patients with advanced illness who are receiving palliative care and when laxative therapy has been insufficient. Methylnaltrexone is a peripherally restricted, μ opioid receptor antagonist that accelerates oral–cecal transit in patients with opioid-induced constipation without reversing the analgesic effects of morphine or inducing symptoms of opioid withdrawal. An analysis of the mechanism of action and the potential benefits of using methylnaltrexone is based on data from published basic research and recent clinical studies.

Introduction

Morphine and other opioid agonists are potent analgesics that represent the mainstay of therapy in the treatment of acute and chronic severe pain. Opioid analgesics work by predominantly stimulating μ opioid receptors in the central nervous system (CNS). However, constipation is a significant problem in patients taking opioid agonists for pain relief due primarily to their effect on μ opioid receptors located in the periphery within the gut itself. This review will focus on one of the currently available μ opioid receptor antagonists, specifically methylnaltrexone bromide (Relistor™; Progenics/Wyeth), a quaternary derivative of naltrexone which was recently approved by the Food and Drug Administration (FDA) as a subcutaneous injection for the treatment of opioid bowel dysfunction in patients with advanced illness who are receiving palliative care and when laxative therapy has been insufficient.Citation1 The review will provide an overview of methylnaltrexone’s ability to promote gastrointestinal (GI) motility in patients with opioid-induced constipation without compromising the analgesic effects of morphine or stimulating symptoms of opioid withdrawal. The later sections of the review will discuss the potential use of methylnaltrexone for the treatment of constipation associated with post-operative ileus.

Effect of opioids on gastrointestinal motility

In the CNS the μ opioid receptor is the primary opioid receptor involved in pain transmission. Although there is limited evidence to suggest that centrally located μ opioid receptors may be involved in the control of GI transit, the dominant effect of the μ opioid receptor agonist, morphine, on the GI tract appears to occur via μ opioid receptors located peripherally within the gut wall.Citation2,Citation3 Recent evidence has demonstrated μ opioid receptors in enteric nervous system, specifically the submucosal and myenteric plexus in association with interstitial cells of Cajal. The muscle layers of the small and large intestine also have shown μ opioid receptor immunoreactivity. For excellent reviews of the anatomical distribution and function of the μ opioid receptor in the GI tract the reader is referred to Sternini and colleagues.Citation4Citation6 Within the GI tract classical animal experiments demonstrated that morphine, fentanyl and met-enkephalin cause inhibition of both the longitudinal and circular muscle layers.Citation7 More recent studies using a vascularly perfused intestinal segment demonstrated that morphine, dermorphin, D-AlaCitation2-D-MetCitation5-enkephalin, FK 33–824 and dynorphin reduced the frequency of peristaltic waves and the maximal ejection pressure.Citation8 From multiple studies and clinical experience, a delay in GI transit is a well known characteristic of μ opioid receptor agonists including morphine, diphenoxylate and loperamide.Citation9 The effects of opioids in delaying intestinal transit is species-dependent and interspecies differences must be taken into account when examining the contribution of opioid agonists on GI motility. The delay in GI transit with opioid agonists occurs through either an inhibition of propulsive motility (rat) or a stimulation of non-propulsive or segmental patterns of motility in dog and man.Citation10 The in vivo effects of opioid agonists to delay GI transit is due to an inhibition of the release of acetylcholine as well as to the release of nonadrenergic-noncholinergic (NANC) neurotransmitters from enteric nerves.Citation11 These effects are mediated at least in part via μ opioid receptors present on circular muscle motor neurons.Citation12 Moreover, in the rat ileum, selective agonists of μ (PLO17) and κ (U-50488) receptors inhibit neurotransmitter release along the ascending excitatory reflex pathway.Citation13 A detailed review of the literature reveals that δ opioid receptors also are involved in the effects of opioids on motility. Delta receptors do not regulate the activity of myenteric excitatory motor neurons, since selective δ receptor agonists (DPDPE) or antagonists (ICI174864) are ineffective.Citation14 However, in the circular muscle of the guinea pig and human colon, NANC inhibitory motor responses are reduced by activation of δ receptors.Citation15,Citation16

In summary, morphine or morphine-like opioid agonists induce a delay in GI transit and are involved in the development of opioid bowel dysfunction through a mechanism involving predominantly μ opioid receptors located within the GI tract.

Opioid bowel dysfunction

Opioid analgesics represent an important therapy for pain management, however opioids also have significant effects on GI motility to delay GI transit and cause constipation that may be so severe that it can limit pain management. Opioids delay GI transit via an inhibition of gastric emptying, a slowing of small and large bowel transit and an increase in anal sphincter tone due to activation of μ opioid receptors located in the GI tract.Citation17Citation19 As early as 1917, Trendelenburg demonstrated in an isolated preparation of guinea-pig small intestine that morphine inhibits peristalsis.Citation7 Since then many studies have confirmed that morphine and related opioids delay transit throughout the GI tract via a peripheral mechanism. Although peripherally restricted μ opioid receptor agonists such as loperamide have been shown to slow GI transit and are useful for the treatment of diarrheaCitation9, the constipating effects of morphine that acts at both central and peripheral μ opioid receptors is a significant problem in patients receiving morphine for the relief of pain. Stimulation of the μ opioid receptors in the GI tract by morphine frequently results in unwanted effects termed opioid bowel dysfunction. Opioid bowel dysfunction is characterized by severe constipation, hard stools, straining, incomplete evacuation, bloating, abdominal distension, and increased gastroesophageal reflux.Citation20Citation22 It is estimated that about 40% of patients taking chronic opioids for pain develop opioid bowel dysfunction.Citation20 Clearly, the mechanisms of opioid bowel dysfunction are complex; however studies have shown that gut hypomotility correlates with opioid concentration in the enteric nervous systemCitation21 and the morphine-induced inhibition of GI transit results from activation of μ opioid receptors in the gut.Citation22 Interestingly, although patients develop tolerance to opioid-induced nausea, vomiting and sedation, they rarely develop tolerance to the delayed GI transit and resulting constipation induced by opioids.Citation23 In summary, chronic opioid use for severe pain is associated with significant adverse effects including opioid-induced bowel dysfunction characterized by constipation that is often not relieved by laxatives.Citation24Citation26

Therapeutic approaches for treating opioid-induced constipation

Laxatives and promotility agents

Until recently the management of opioid-induced bowel dysfunction was limited to use of a stimulate laxative such as bisacodyl or senna with or without addition of stool softeners such as docusate sodium or lactuloseCitation24Citation26 as well as increasing dietary fiber, increasing fluid intake and/or suggesting daily exercise whenever possible to treat the constipation. Osmotic laxatives and bulk forming laxatives also have been employed in patients with opioid-induced bowel dysfunction but they must be used cautiously in patients requiring fluid restriction, bedridden patients or those with strictures or partial bowel obstruction.Citation26,Citation27 Often increased fiber intake or use of bulk laxatives increase pressure in the gut, worsening the patient’s pain and increasing their discomfort. Promotility agents such as metaclopramide, a dopamine D2 receptor antagonist, also are used to accelerate GI transit in a subset of patients with delayed GI transit suffering from GI autonomic dysfunction.Citation27 Although laxatives are beneficial in some patients, they are poorly effective in most and opioid-induced bowel dysfunction persists despite aggressive laxative therapy. Until recently laxatives represented the mainstay of therapy for the treatment of opioid-induced constipation, despite their serious limitations.

Opioid receptor antagonists

In very severe cases of opioid-induced constipation patients reduce their use of opioids to alleviate the constipation despite the resulting loss of adequate pain relief. The first competitive opioid antagonists that were used to treat opioid bowel dysfunction included naloxone, naltrexone and nalmefene. While these antagonists were selective for opioid receptors, they were not selective for the periphery, and have both central and peripheral activity due to their ability to cross the blood brain barrier. Although such compounds were able to increase laxation in patients with opioid-induced constipation, they also were associated with symptoms of opioid withdrawal and a marked decrease in adequate pain relief due to the effects of the compounds on central μ opioid receptors. Thus the search was initiated for an opioid antagonist that possessed the capability of reversing opioid-induced constipation without reducing the level of analgesia or stimulating opioid withdrawal. It was suggested in the latter part of the twentieth century that quaternary narcotic antagonists might be useful to treat opioid-induced peripheral side effects since some of these agents failed to cross the blood brain barrier readily.Citation28,Citation29 Quaternary opioid antagonists were developed that had increased polarity and decreased lipid solubility and a reduced ability to cross the blood brain barrier following systemic administration. As a result, these compounds bound only to peripheral μ receptors unless administered directly into the brain.Citation28,Citation29 Today peripheral μ opioid receptor antagonism offers a newly approved class of therapeutics for the treatment of constipation associated with the long-term use of opioids while preserving centrally mediated analgesia.Citation1 A major focus of the subsequent section of this review will be the use of methylnaltrexone for the treatment of opioid-induced bowel dysfunction, specifically severe constipation in patients receiving opioid therapy for pain management.

Effect of methylnaltrexone in preclinical experimental models

An extensive series of studies were designed to characterize the pharmacological profile of methylnaltrexone. Until recently, opioid receptor affinity was assessed in whole rat brain or guinea pig ileum tissue by displacing the binding of non-selective opioid antagonists, 3H-etorphine or 3H-diprenorphine. These studies revealed that the quaternary derivative of naltrexone exhibited only 1% to 3% of the affinity for the μ opioid receptor as naltrexone itself.Citation30 Assessment of the affinities and selectivity of methylnaltrexone for μ, δ and κ opioid receptors recently was performed in cells expressing recombinant human opioid receptors as well as in animal tissues expressing endogenous opioid receptors. These studies demonstrated that the compound displaced opioid binding to μ opioid receptors with an affinity of 10 nM, and a 3-fold lower affinity for κ opioid receptors (Ki 30 nM).Citation31 These studies also demonstrated that the affinity of methylnaltrexone for δ opioid receptors is much less (Ki 15.8 μM).Citation31 While initial reports indicated that methylnaltrexone had no intrinsic opioid agonist activity,Citation28Citation30 more recent studies comparing it against a variety of opioid antagonists demonstrated that the compound exhibited weak partial agonist activity at recombinant μ and κ opioid receptors (intrinsic activity 10 and 12, respectively).Citation31 Using in vitro and in vivo models the effect of methylnaltrexone on the GI tract was systematically studied. Methylnaltrexone reversed morphine-induced inhibition of gut contractility in isolated guinea-pig ileum and human small intestine.Citation31,Citation32 A limited number of studies have also suggested that there may be endogenous inhibitory opioid tone of the GI musculature since methylnaltrexone alone enhanced muscle contractility in tissue isolated from the humanCitation32 or equineCitation33 GI tract. However, effects of methylnaltrexone on electrically-evoked contraction of guinea pig ileum muscle strips are mixed, with methylnaltrexone inhibiting contractions in one studyCitation31 and exacerbating contractions in another.Citation32 Some of these differences may result from differences in electrical current applied to the tissue in the two experiments, as well as to different levels of endogenous opioid inhibitory tone between species. However, whether methylnaltrexone has therapeutic potential in other GI disorders characterized by hypomotility requires further research. The effect of methylnaltrexone on GI transit in vivo demonstrated that methylnaltrexone at doses of 1, 3 and 10 mg/kg, administered subcutaneously, effectively antagonized morphine-induced inhibition of GI transit in rats.Citation34 To test the hypothesis that antagonism of the effects of opioids in the gut can be accomplished without compromising analgesia, experimental models demonstrated that subcutaneous administration of methylnaltrexone had no effect on morphine-induced analgesia whereas if the methylnaltrexone was administered directly into the brain there was a marked reduction in analgesia.Citation34,Citation35

In summary, the efficacy of methylnaltrexone to antagonize the GI-mediated events effects of morphine occurred at doses that failed to antagonize morphine-induced analgesia. Early preclinical experiments were also conducted with methylnaltrexone to ensure that following intravenous administration there were no signs or symptoms of withdrawal in opioid-tolerant dogs.Citation28 The study found that in response to doses of methylnaltrexone as high at 50 mg/kg there were no symptoms of withdrawal whereas signs of withdrawal were noted with doses of naltrexone as low as 0.5 mg/kg.Citation28,Citation36

Clinical pharmacology of methylnaltrexone

A summary of the efficacy and pharmacokinetic characteristics of methylnaltrexone in healthy human subjects, as well as patients with advanced illness or on chronic methadone treatment are provided in . With iv or sc delivery, methylnaltrexone is rapidly absorbed in a dose-dependent manner, with a peak concentration (Cmax) generally reached within 20 to 30 minutes (Tmax) and τ1/2 of elimination is 100 to 130 minutes.Citation37Citation39 While orally administered methylnaltrexone is absorbed in a dose-dependent manner, far less is absorbed in general, maximal blood levels are not achieved until almost 2 hours, and the τ1/2 is around 3 hours.Citation40 The primary path-ways of metabolism are the conversion to methyl-6-naltrexol isomer (5% of the total) and methylnaltrexone sulphate (1.3% of the total). N-methylation of methylnaltrexone to naltrexone is not a significant issue.Citation41 The excretion of methylnaltrexone is via the urine and feces and approximately 40% to 50% of the compound is excreted unchanged in the urine following sc or iv. administration.Citation37Citation39,Citation42,Citation43 Interestingly, following oral administration, only a tiny fraction of methylnaltrexone is excreted unchanged (0.3% or less).Citation40,Citation44 Almost a 100-fold higher dose of oral methylnaltrexone (19.2 mg/kg)Citation40 was required to produce maximal plasma concentrations equivalent to those produced by 0.1 mg/kg sc.Citation39 or 0.16 mg/kg iv methylnaltrexone.Citation37 Since only a fraction of the methylnaltrexone was excreted unchanged yet maintained its ability to antagonize morphine-induced oral–cecal transit delay (), this suggests that the majority of the compound remained in the GI tract instead of being absorbed into the bloodstream.Citation40,Citation44 An enteric-coated formulation of methylnaltrexone was effective orally at only 3.2 mg/kg.Citation44

Table 1 Clinical pharmacokinetic and efficacy profiles of methylnaltrexone (MNTX) in healthy subjects, patients with advanced illness and chronic methadone patients

Clinical efficacy studies of methylnaltrexone for opioid-induced constipation

In early clinical studies, methylnaltrexone was used as a pharmacological tool to examine the relative importance of peripheral opioid receptor antagonism in modulating opioid-induced delay in gastric emptyingCitation45 as well as oral–cecal transit.Citation38 In the first human study to demonstrate that opioids affect gastric emptying via a peripheral mechanism distinct from the central analgesic effects of opioids, 11 healthy controls were given placebo (saline) plus morphine or methylnaltrexone plus morphine in a randomized double-blind crossover controlled trial.Citation45 The results showed that morphine prolonged gastric emptying and that methylnaltrexone prevented the morphine-induced gastric emptying. In 1996, Yuan et al reported that methylnaltrexone blocked morphine-induced oral–cecal transit delay with no effect on morphine analgesia, demonstrating for the very first time in human that opioid effects on the gut are mediated through peripheral μ opioid receptors distinct from those receptors located centrally that mediate analgesia.Citation38

This and other studies performed in healthy volunteers measured the ability of methylnaltrexone to reverse morphine-induced oral to cecal transit time using the lactulose hydrogen breath test; Citation38,Citation39,Citation44,Citation46 pain intensity in response to the cold pressor test was assessed in one study to measure levels of pain reporting in the same subjects.Citation38 Methylnaltrexone administered intravenously, orally or subcutaneously reversed the delay in oral–cecal transit induced by morphineCitation38,Citation39,Citation44,Citation46 without any effect on levels of analgesia.Citation38 Furthermore, in a randomized placebo-controlled trial, methylnaltrexone administered intravenously at a cumulative low dose ranging from 0.015–0.365 mg/kg to 11 subjects with chronic methadone-induced constipation reversed the opioid-induced increase in GI transit time and produce immediate laxation in 91% of patients on day 1 and 100% of the patients by the morning of the second day of dosingCitation43 (). More recently in one small study and then two larger pivotal studies, the effectiveness of methylnaltrexone was investigated in patients with advanced illness with a life expectancy of less than 6 months, who were receiving palliative opioid therapy and had opioid-induced constipation (defined as fewer than 3 bowel movements in the preceding week or no bowel movement for 2 days). Rescue laxatives were prohibited from 4 hours before until 4 hours after receiving the injection of methylnaltrexone. A double-blind, randomized parallel group, dose ranging study was conducted by Portenoy and colleagues in 33 adult patients with advanced illness defined as terminal or end stage diseases such as advanced metastatic cancer or AIDS.Citation47 Methylnaltrexone at doses of 5, 12.5 and 20 mg (0.05–0.5 mg/kg sc) induced a bowel movement within 4 hours in 33% to 60% of patients; this was reproducible with repeated dosing and occurred in the absence of opioid withdrawal or loss of adequate pain control. In a larger Phase III study where the primary end point was the proportion of patients with a rescue-free laxation within 4 hours of receiving the medication, the goal was to compare a single, double blind subcutaneous dose of methylnaltrexone (0.15 mg/kg or 0.3 mg/kg) versus placebo. The double blind dose was followed by an open-label 4 week dosing period during which time methylnaltrexone could be used as needed but no more that 1 dose in a 24-hour period. In the double blind period 154 patients were enrolled (47 patients received methylnaltrexone 0.15 mg/kg, 55 methylnaltrexone 0.3 mg/kg and 52 placebo). The results showed that 62% of the patients receiving 0.15 mg/kg methylnaltrexone and 58% of those receiving 0.3 mg/kg methylnaltrexone had a significantly higher rate of laxation within 4 hours of the double-blind dose compared to 14% treated with placebo.Citation41 Similar results were report in another Phase III study published by Thomas and colleagues in 2008.Citation48 A total of 133 patients who were using opioids for pain relief for at least 2 weeks and were taking laxatives without relief for their opioid-induced constipation were randomly assigned to receive either subcutaneous methylnaltrexone (0.15 mg/kg, 62 patients) or placebo (71 patients) every other day for a week. During the second week the dose of the compound could be increased 0.30 mg/kg if the patient had 2 or less rescue-free laxations up to day 8. The study found that 48% of patients receiving methylnaltrexone had a bowel movement within 4 hours of the first dose while only 15% of patients receiving placebo had a bowel movement within 4 hours. However, when the patients were asked to self assess improvement in their bowel status using a Global Clinical Impression of Change (GCIC) Scale after 7 and 14 days, most patients (73%) reported increased satisfaction following methylnaltrexone therapy compared to only 35% of patients in the placebo group, suggesting that methylnaltrexone improved the constipating symptoms in over 70% of patients. An important component of the study was that no significant changes in pain scores were observed and there were no signs of opioid withdrawal.Citation48 This and other clinical studies have also assessed the occurrence of methylnaltrexone-related adverse effects.Citation23,Citation37,Citation43,Citation45,Citation47,Citation48 There was no dose-dependence associated with the adverse effects, which were generally gastrointestinal in nature (abdominal pain, diarrhea, flatulence and nausea) and tolerable. In fact, these side effects are common to existing treatments for opioid-induced constipation. In a dose-escalating study in which methylnaltrexone was iv infused, a few subjects reported transient orthostatic hypotension, that resolved quickly.Citation37 The rates of discontinuation due to adverse events during the double-blind placebo controlled clinical trials described above were comparable for methylnaltrexone (1.2%) and placebo (2.4%); no serious adverse effects were attributed to methylnaltrexone.

In summary, the results showed that in these specific patient groups with advanced illness, methylnaltrexone administered subcutaneously, relieved opioid-induced constipation but most importantly did not reduce analgesia or cause any symptomatology associated with opioid withdrawal. Methylnaltrexone was effective in almost 70% of those patients studied, and it is hoped that further clinical studies will be performed in patients with less advanced disease. In addition to the currently approved subcutaneous route of administration for methylnaltrexone, its future development involves multiple routes of administration including oral/oral enteric-coated oral forms of methylnaltrexone for opioid-induced constipation and an intravenous formulation for post-operative ileus.

Potential use of methylnaltrexone for the treatment of post-operative ileus

Although quite different from opioid bowel dysfunction in the setting of chronic opioid administration, another common condition termed post-operative ileus is a transient impairment of GI motility that routinely develops as a consequence of abdominal surgery. Although the pathophysiology of post-operative ileus is very complex involving inflammatory, neural and hormonal mechanisms, there is a significant pharmacological component to post-operative ileus. Specifically the use of opioid drugs, such as morphine for the management of post-operative pain, is well known to exacerbate the severity of post-operative ileus (see reviewCitation49). In fact a retrospective study of post-operative ileus patients identified that the use of opioid-based analgesics represent a key risk factor for the development of ileus following surgery.Citation50 Usually patients recover from post-operative ileus in 1 to 2 days, particularly after laparoscopic bowel surgery. However, prolonged and untreated post-operative ileus may require nasogastric intubation and sometimes even parenteral nutrition. The duration of ileus following surgery is dependent on which part of the GI tract is most affected, with the small intestine recovering within the first 24 hours whereas the stomach (24–48 hours) and the colon require longer (48–72 hours) recovery periods. Certain patients who are less mobile post-surgery have been found to be more susceptible to post-operative ileus. Delayed gastric emptying and intestinal transit are the main factors leading to symptoms of post-operative ileus which include abdominal bloating and pain, nausea and vomiting, anorexia, and reduced defecation.

In addition to postoperative opioid dosage, the duration of post-operative ileus following colorectal surgery also is positively correlated with the amount of blood loss and surgery time.Citation51 Although the analgesic effects of opioids, such as morphine, are predominantly mediated by μ opioid receptors in the CNS, the action of morphine to delay GI transit involves predominantly activation of μ opioid receptors in the periphery to inhibit enteric reflexes and suppress GI transit.Citation16 Despite the fact that post-operative abnormalities in GI transit are common and can significantly delay a patient’s recovery, the pathophysiological mechanisms causing post-operative ileus are incompletely understood.Citation52 Research from animal studies has shown abdominal surgery stimulates capsaicin-sensitive afferent fibers that lead to activation of inhibitory efferent pathways and disrupt coordinated patterns of GI motility. Evidence is greatest for adrenergic receptor mediated pathways that involve alpha2 receptors located on vagal nerve terminal and postganglionic cholinergic nerves in the myenteric plexus that inhibit the release of acetylcholine in post-operative ileus.Citation53 Thus neural pathways producing post-operative ileus are complex and dependent on a number of factors including the magnitude and intensity of the stimulus. For example, incision of the skin and laparotomy stimulate adrenergic inhibitory neuronal pathways,Citation54,Citation55 whereas abdominal surgery with handling of the intestine activates supra-spinal pathways that involve stimulation of the hypothalamic pituitary adrenal (HPA) stress axis and release of corticotrophin-releasing factor (CRF).Citation56Citation58 In addition, non-adrenergic, non-cholinergic (NANC) neuronal pathways may also inhibit GI motility. Multiple inhibitory NANC neurotransmitters such as nitric oxide (NO), calcitonin gene-related peptide (CGRP) and vasoactive intestinal polypeptide (VIP) are present within the enteric nervous systems, and they too may play an important role in the pathogenesis of post-operative ileus.Citation59Citation61 Recent findings have also shown that manipulation of the bowel during surgery induces a marked intestinal inflammation in rodents.Citation62Citation65 These studies showed that specific inflammatory cells such as monocytes, neutrophils and mast cells increase in response to bowel manipulation, and there was a marked induction within the GI musculature of inducible nitric oxide synthase (iNOS) and cyclo-oxygenase-2 (COX-2) mRNA. The inflammatory response induced by bowel manipulation also leads to the secretion of a series of pro-inflammatory cytokines, including interleukin-1β, inter-leukin-6, tissue necrosis factor-α and monocyte chemotactic protein-1. There is also evidence that bowel manipulation up-regulates adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1), and that the ICAM-1 antisense oligonucleotide ISIS 3082 prevents the development of ileus in mice.Citation66 Interestingly, in an animal model of post-operative ileus produced by bowel manipulation, the magnitude of the inflammatory response appeared to be proportional to the decrease in GI transit measured in vivo and the loss of smooth muscle contractility observed in an organ bath preparation.Citation67,Citation68

The development of new pharmacological strategies to accelerate the recovery from POI are urgently needed because post-surgical GI dysmotility represents a major health problem contributing to patient morbidity, prolonged hospital stays and increased health care costs. Recently positive clinical efficacy data obtained with a peripherally acting antagonist of the μ opioid receptor, alvimopan, in treating the delay in GI transit following surgery led to its recent approval by the FDA for treatment of post-operative ileus. However, clinical reports related to the efficacy of methylnaltrexone for the treatment of post-operative ileus have been inconclusive to date. In a Phase II trial performed in 65 patients with post-operative ileus induced by colonic resection, administration of methylnaltrexone (0.3 mg/kg, intravenous) after surgery every 6 hours for 24 hours recovered from post-operative ileus 1 day faster than those receiving placebo as assessed by time to first bowel movement, ability to tolerate food and hospital discharge.Citation69 However, those findings were not supported by preliminary results from a Phase III trial of 542 patients with post-operative ileus; methylnaltrexone administered at 12 or 24 mg every 6 hr did not show efficacy over placebo.Citation70 At the time of this review, a second Phase III trial is underway with preliminary results suggesting that methylnaltrexone was not different from placebo in treating post-operative ileus.Citation70 The reason for these differences in clinical efficacy between alvimopan and methylnaltrexone for the treatment of post-operative ileus remains to be determined but while both alvimopan and methylnaltrexone share many pharmacological properties, there are a few notable differences. Evidence suggests that alvimopan has inverse agonist activity in the guinea pig ileum assay while methylnaltrexone does not.Citation8 If the various conditions associated with post-operative ileus described above contribute to impaired GI transit, one can see how an antagonist with inverse agonist properties might more effectively increase gut motility than one with very weak partial agonist activity. Alvimopan also exhibits a slightly greater μ/κ opioid receptor selectivity ratio.Citation30

Finally, it has been proposed that P-glycoprotein (P-gp) expression in the small intestine may affect the response of the small intestine to chronic drug treatment.Citation71 For instance, while tolerance does not develop to the constipating effects of morphine, tolerance does develop to the inhibitory GI transit effects of loperamide. This difference may be attributed to the fact that loperamide is a better substrate for P-gp than morphine. To date, there are no reports on the affinity of either methylnaltrexone or alvimopan for P-gp or other drug transporters, but expression of these transporters is altered (generally reduced) by acute activation of inflammatory mediators such as iNOS, interleukin-1β, interleukin-6 and tumor necrosis factor-α.72–75 Reduced expression of P-gp or other drug efflux transporters could increase the level of drug in the gut tissue and increase its efficacy. Finally, the lack of efficacy of methylnaltrexone for the treatment of post-operative ileus may relate to the design of the clinical trails, dosage and route of administration, and such factors are currently under investigation.

Conclusions and future directions

Although the actions of opioids in the gut have been well documented, major gaps remain in our understanding of the precise mechanisms underlying these effects, and of the potential role of opioid systems in GI diseases. Opioid analgesic use is commonly associated with GI side effects suggesting a role for opioid systems in both GI function and pathophysiology. The use of μ opioid receptor antagonists with activity limited to the periphery has proven to be of benefit in the clinic, where peripherally acting μ opioid antagonists, such as methylnaltrexone, reverse the unwanted peripheral side effect of constipation associated with opioid therapy while preserving centrally mediated opioid analgesia. Taken together, the preclinical data on methylnaltrexone are consistent with the clinical reports confirming the ability of methylnaltrexone to antagonize opioid-induced constipation without reversing analgesia or precipitating withdrawal symptoms. Additional research is required to determine whether methylnaltrexone also will be useful for the treatment of post-operative ileus.

Disclosures

The authors have no conflicts of interest to disclose.

References

  • LangLThe Food and Drug Administration approves methylnaltrexone bromide for opioid-induced constipationGastroenterology200813516
  • ManaraLBianchettiAThe central and peripheral influence of opioids on gastrointestinal propulsionAnnu Rev Pharmacol Toxicol1985252492733890704
  • ManaraLBianchiGFerrettiPTavaniAInhibition of gastrointestinal transit by morphine in rats results primarily from direct drug action on the gut opioid sitesJ Pharmacol Exp Ther198623739459493012075
  • SterniniCReceptors and transmission in the brain-gut axis: potential for novel therapies. III. Mu-opioid receptors in the enteric nervous systemAm J Physiol Gastrointest Liver Physiol20012811G8G1511408250
  • SterniniCPatiernoSSelmerISKirchgessnerAThe opioid system in the gastrointestinal tractNeurogastroenterol Motil200416Suppl 231615357847
  • DeHaven-HudkinsDLDeHavenRNLittlePJTechnerLMThe involvement of the mu-opioid receptor in gastrointestinal patho-physiology: therapeutic opportunities for antagonism at this receptorPharmacol Ther2008117116218718022696
  • TrendelenburgP[Physiological and pharmacological investigations of small intestinal peristalsis]Naunyn Schmiedebergs Arch Pharmacol191781155129
  • SangerGJTuladharBRThe role of endogenous opioids in the control of gastrointestinal motility: prediction from in vitro modellingNeurogastroenterol Motil200416Suppl 2384515357850
  • OomsLADegryseADJanssenPAMechanisms of action of loperamideScand J Gastroenterol Suppl1984961451556382576
  • BuenoLFioramontiJAction of opiates on gastrointestinal functionBaillieres Clin Gastroenterol1988211231392838107
  • WoodJDGalliganJJFunction of opioids in the enteric nervous systemNeurogastroenterol Motil200416Suppl 2172815357848
  • ToniniMWatermanSACanduraSMCocciniTCostaMSites of action of morphine on the ascending excitatory reflex in the guinea-pig small intestineNeurosci Lett19921441–21951981331912
  • AllescherHDStorrMBrechmannCHahnASchusdziarraVModulatory effect of endogenous and exogenous opioids on the excitatory reflex pathway of the rat ileumNeuropeptides2000341626810688971
  • KojimaYTakahashiTFujinaMOwyangCInhibition of cholinergic transmission by opiates in ileal myenteric plexus is mediated by kappa receptor. Involvement of regulatory inhibitory G protein and calcium N-channelsJ Pharmacol Exp Ther199426829659708114012
  • HoyleCHKammMABurnstockGLennard-JonesJEEnkephalins modulate inhibitory neuromuscular transmission in circular muscle of human colon via delta-opioid receptorsJ Physiol19904314654781966052
  • ZagorodnyukVMaggiCAElectrophysiological evidence for different release mechanism of ATP and NO as inhibitory NANC transmitters in guinea-pig colonBr J Pharmacol19941124107710827952866
  • De LucaACouparIMInsights into opioid action in the intestinal tractPharmacol Ther19966921031158984506
  • FriedmanJDDello BuonoFAOpioid antagonists in the treatment of opioid-induced constipation and pruritusAnn Pharmacother2001351859111197589
  • Greenwood-Van MeerveldBGardnerCJLittlePJHicksGADeHaven-HudkinsDLPreclinical studies of opioids and opioid antagonists on gastrointestinal functionNeurogastroenterol Motil200416Suppl 2465315357851
  • PappagalloMIncidence, prevalence and management of opioid bowel dysfunctionAm J Surg20011825A Suppl11S18S11755892
  • ThomasJOpioid-induced bowel dysfunctionJ Pain Symptom Manage200835110311317981003
  • FossJFA review of the potential role of methylnaltrexone in opioid bowel dysfunctionAm J Surg20011825A Suppl19S26S11755893
  • McNicolEDBoyceDSchumannRCarrDBMu-opioid antagonists for opioid-induced bowel dysfunctionCochrane Database Syst Rev20082CD00633218425947
  • WalshTDPrevention of opioid side effectsJ Pain Symptom Manage1990563623671980127
  • GlarePLickissJNUnrecognized constipation in patients with advanced cancer: a recipe for therapeutic disasterJ Pain Symptom Manage1992763693711517653
  • SwegleJMLogemannCManagement of common opioid-induced adverse effectsAm Fam Physician20067481347135417087429
  • LagmanRLDavisMPLeGrandSBWalshDCommon symptoms in advanced cancerSurg Clin North Am200585223725515833469
  • RussellJBassPGoldbergLISchusterCRMerzHAntagonism of gut, but not central effects of morphine with quaternary narcotic antagonistsEur J Pharmacol19827832552617200037
  • BrownDRGoldbergLIThe use of quaternary narcotic antagonists in opiate researchNeuropharmacology19852431811912986035
  • ValentinoRJHerlingSWoodsJHMedzihradskyFMerzHQuaternary naltrexone: evidence for the central mediation of discriminative stimulus effects of narcotic agonists and antagonistsJ Pharmacol Exp Ther198121736526596262493
  • BeattieDTCheruvuMMaiNThe in vitro pharmacology of the peripherally restricted opioid receptor antagonists, alvimopan, ADL 08-0011 and methylnaltrexoneNaunyn Schmiedebergs Arch Pharmacol2007375320522017340127
  • YuanCSFossJFMossJEffects of methylnaltrexone on morphine-induced inhibition of contraction in isolated guinea-pig ileum and human intestineEur J Pharmacol19952761–21071117781680
  • Van HoogmoedLMBoscanPLIn vitro evaluation of the effect of the opioid antagonist N-methylnaltrexone on motility of the equine jejunum and pelvic flexureEquine Vet J200537432532816028621
  • GmerekDECowanAWoodsJHIndependent central and peripheral mediation of morphine-induced inhibition of gastrointestinal transit in ratsJ Pharmacol Exp Ther198623618133941402
  • WalkerMJLeADPoulosCXCappellHRole of central versus peripheral opioid receptors in analgesia induced repeated administration of opioid antagonistsPsychopharmacology (Berl)199110421641661652143
  • ValentinoRJKatzJLMedzihradskyFWoodsJHReceptor binding, antagonist, and withdrawal precipitating properties of opiate antagonistsLife Sci19833225288728966304445
  • FossJFO’ConnorMFYuanCSMurphyMMossJRoizenMFSafety and tolerance of methylnaltrexone in healthy humans: a randomized, placebo-controlled, intravenous, ascending-dose, pharmacokinetic studyJ Clin Pharmacol199737125309048269
  • YuanCSFossJFO’ConnerMToledanoARoizenMFMossJMethylnaltrexone prevents morphine-induced delay in oral-cecal transit time without affecting analgesia: a double-blind randomized placebo-controlled trialClin Pharmacol Ther19965944694758612393
  • YuanCSWeiGFossJFO’ConnorMKarrisonTOsinskiJEffects of subcutaneous methylnaltrexone on morphine-induced peripherally mediated side effects: a double-blind randomized placebo-controlled trialJ Pharmacol Exp Ther2002300111812311752106
  • YuanCSFossJFO’ConnorMOsinskiJRoizenMFMossJEfficacy of orally administered methylnaltrexone in decreasing subjective effects after intravenous morphineDrug Alcohol Depend19985221611659800145
  • Full Prescribing Information [linked PDF, cited 2008 Nov 14]. Available from: http://www.wyeth.com/hcp/relistor/landing.
  • YuanCSFossJFO’ConnorMOsinskiJRoizenMFMossJEffects of intravenous methylnaltrexone on opioid-induced gut motility and transit time changes in subjects receiving chronic methadone therapy: a pilot studyPain199983363163510568873
  • YuanCSFossJFO’ConnorMMethylnaltrexone for reversal of constipation due to chronic methadone use: a randomized controlled trialJAMA2000283336737210647800
  • YuanCSFossJFO’ConnorMEffects of enteric-coated methylnaltrexone in preventing opioid-induced delay in oral-cecal transit timeClin Pharmacol Ther200067439840410801249
  • MurphyDBSuttonJAPrescottLFMurphyMBOpioid-induced delay in gastric emptying: a peripheral mechanism in humansAnesthesiology19978747657709357876
  • YuanCSFossJFOsinskiJToledanoARoizenMFMossJThe safety and efficacy of oral methylnaltrexone in preventing morphine-induced delay in oral-cecal transit timeClin Pharmacol Ther19976144674759129564
  • PortenoyRKThomasJMoehl BoatwrightMLSubcutaneous methylnaltrexone for the treatment of opioid-induced constipation in patients with advanced illness: a double-blind, randomized, parallel group, dose-ranging studyJ Pain Symptom Manage200835545846818440447
  • ThomasJKarverSCooneyGAMethylnaltrexone for opioid-induced constipation in advanced illnessN Engl J Med2008358222332234318509120
  • Greenwood-Van MeerveldBEmerging drugs for postoperative ileusExpert Opin Emerg Drugs200712461962617979603
  • SenagoreAJOverview of POI and its impact on surgical outcomesContemporary Surg20063Suppl47
  • ArtinyanANunoo-MensahJWBalasubramaniamSProlonged postoperative ileus-definition, risk factors and predictors after surgeryWorld J Surg20083271495150018305994
  • BauerAJBoeckxstaensGEMechanisms of postoperative ileusNeurogastroenterol Motil200416Suppl 2546015357852
  • SagradaAFargeasMJBuenoLInvolvement of alpha-1 and alpha-2 adrenoceptors in the postlaparotomy intestinal motor disturbances in the ratGut19872889559592889649
  • BoeckxstaensGEHirschDPKoddeAActivation of an adrenergic and vagally mediated NANC pathway in surgery-induced fundic relaxation in the ratNeurogastroenterol Motil199911646747410583854
  • HolzerPLippeITAmannRParticipation of capsaicin-sensitive afferent neurons in gastric motor inhibition caused by laparotomy and intraperitoneal acidNeuroscience19924837157221603337
  • BarquistEBonazBMartinezVRivierJZinnerMJTacheYNeuronal pathways involved in abdominal surgery-induced gastric ileus in ratsAm J Physiol19962704 Pt 2R888R8948967419
  • BonazBTacheYCorticotropin-releasing factor and systemic capsaicin-sensitive afferents are involved in abdominal surgery-induced Fos expression in the paraventricular nucleus of the hypothalamusBrain Res19977481–212209067440
  • TacheYMonnikesHBonazBRivierJRole of CRF on stress-related alterations of gastric and colonic motor functionAnn N Y Acad Sci19936972332438257013
  • De WinterBYBoeckxstaensGEDe ManJGMoreelsTGHermanAGPelckmansPAEffect of adrenergic and nitrergic blockade on experimental ileus in ratsBr J Pharmacol199712034644689031750
  • De WinterBYRobberechtPBoeckxstaensGERole of VIP1/PACAP receptors in postoperative ileus in ratsBr J Pharmacol19981246118111869720789
  • BoeckxstaensGEHollmannMHeisterkampSHEvidence for VIP(1)/PACAP receptors in the afferent pathway mediating surgery-induced fundic relaxation in the ratBr J Pharmacol2000131470571011030719
  • KälffJCTürlerASchwarzNTIntra-abdominal activation of a local inflammatory response within the human muscularis externa during laparotomyAnn Surg2003237330131512616113
  • SchwarzNTBeer-StolzDSimmonsRLBauerAJPathogenesis of paralytic ileus: intestinal manipulation opens a transient pathway between the intestinal lumen and the leukocytic infiltrate of the jejunal muscularisAnn Surg20022351314011753040
  • De JongeWJTheFOvan der ZandenEPvan den WijngaardRMBoeckxstaensGEInflammation and gut motility; neural control of intestinal immune cell activationJ Pediatr Gastroenterol Nutr200541Suppl 1S10S1116131950
  • KälffJCCarlosTMSchrautWHBilliarTRSimmonsRLBauerAJSurgically induced leukocytic infiltrates within the rat intestinal muscularis mediate postoperative ileusGastroenterology1999117237838710419919
  • TheFOde JongeWJBenninkRJvan der WijngaardRMBoeckxstaensGEThe ICAM-1 antisense oligonucleotide ISIS-3082 prevents the development of postoperative ileus in miceBr J Pharmacol2005146225225815997238
  • KälffJCBuchholzBMEskandariMKBiphasic response to gut manipulation and temporal correlation of cellular infiltrates and muscle dysfunction in ratsSurgery1999126349850910486602
  • TurlerAMooreBAPezzoneMAOverhausMKalffJCBauerAJColonic postoperative inflammatory ileus in the ratAnn Surg20022361566612131086
  • KreissCToegelSBauerAJAlpha2-adrenergic regulation of NO production alters postoperative intestinal smooth muscle dysfunction in rodentAm J Physiol Gastrointest Liver Physiol20042873G65866615331355
  • ViscusiERathmellJFicheraAGanTLIsraelRJA double-blind, randomized, placebo-controlled trial of methylnaltrexone (MNTX) for post-operative bowel dysfunction in segmental colectomy patients [abstract]Am Soc Anesth2005103A893
  • KraftMDMethylnaltrexone, a new peripherally acting mu-opioid receptor antagonist being evaluated for the treatment of postoperative ileusExpert Opin Investig Drugs200817913651377
  • Tan-NoKNiijimaFNakagawasaiOSatoTSatohSTadanoTDevelopment of tolerance to the inhibitory effect of loperamide on gastrointestinal transit in miceEur J Pharm Sci200320335736314592702
  • HoEAPiquette-MillerMRegulation of multidrug resistance by pro-inflammatory cytokinesCurr Cancer Drug Targets20066429531116848721
  • IizasaHGendaNKitanoTAltered expression and function of P-glycoprotein in dextran sodium sulfate-induced colitis in miceJ Pharm Sci200392256957612587118
  • HartmannGKimHPiquette-MillerMRegulation of the hepatic multidrug resistance gene expression by endotoxin and inflammatory cytokines in miceInt Immunopharmacol20011218919911360920
  • HeemskerkSvan KoppenAvan den BroekLNitric oxide differentially regulates renal ATP-binding cassette transporters during endotoxemiaPflugers Arch2007454232133417285300