102
Views
10
CrossRef citations to date
0
Altmetric
Review

Naloxegol in opioid-induced constipation: a new paradigm in the treatment of a common problem

&
Pages 1265-1271 | Published online: 24 Jul 2017

Abstract

Opioid-induced constipation (OIC) imposes a significant burden for patients taking pain medications, often resulting in decreased quality of life. Treatment of OIC with traditional medications for functional constipation can be incompletely effective, leading to nonadherence with opioid treatment and undertreated pain. An emerging class of medications that counteract the adverse effects of opioids in the gastrointestinal tract while preserving central nervous system-based pain relief may represent a paradigm shift in the prevention and treatment of OIC. One of these medications, naloxegol, is a once-daily, oral opioid antagonist that is effective, well-tolerated, and approved for treatment of OIC in patients with noncancer pain. More studies are needed to demonstrate this same utility in patients with cancer-related pain.

Introduction: opioid-induced constipation (OIC) and its burden

Opioids, the most frequently prescribed class of drugs in the US, are an essential tool in the treatment of pain. They function through their agonistic effects on mu receptors in the central nervous system (CNS). Agonism of mu receptors outside of the CNS can lead to undesired adverse effects. In the gastrointestinal (GI) system, it decreases peristalsis and muscular contractions, decreases secretion of water and electrolytes, and increases rectal sphincter tone. This collection of effects is known as opioid-induced bowel dysfunction (OIBD). Symptoms of OIBD include abdominal pain, gastroesophageal reflux, anorexia, bloating, xerostomia, hard feces, incomplete evacuation, and constipation. Unfortunately, patients do not typically develop tolerance to these GI side effects, leading to patient dissatisfaction with opioid therapy.Citation1,Citation2

Reported prevalence of OIC varies widely between studies, as 60%–94% and 40%–60% of patients take opioids for malignant and nonmalignant pain, respectively.Citation3,Citation4 This makes OIC the most common symptom of OIBD. Interestingly, until recently, there has been no consensus definition for OIC, which perhaps contributed to some of the variation in reported OIC prevalence.Citation5 Most trials of anticonstipation agents do not define OIC in the study and often rely on a history of opioid use along with the following traditional measures of functional constipation:

  • Defecation frequency of <3 complete spontaneous bowel movements per week

  • Hard lumpy stools

  • Incomplete evacuation

  • Infrequent spontaneous bowel movements without the use of a laxative within 24 hours of defecation.Citation6

A working group of gastroenterologists, palliative care specialists, and neurologists proposed the following consensus definition in their 2014 Neurogastroenterology and Motility review article:

A change when initiating opioid therapy from baseline bowel habits that is characterized by any of the following: reduced bowel movement frequency, development or worsening of straining to pass bowel movements, a sense of incomplete rectal evacuation, harder stool consistency.Citation4

More recent consensus recommendations from the American Academy of Pain Management in 2015 proposed the 3-item Bowel Function Index (BFI) as a validated assessment tool to detect OIC and provide a threshold for pharmacologic intervention. The BFI asks patients to consider, over the preceding 7 days on a scale of 0–100, the following three parameters:

  • Ease of defecation

  • Feeling of incomplete defecation

  • Personal judgment of constipation.

The panel further suggests that patients on chronic opioids using typical first-line therapies for OIC with scores ≥30 on the BFI be considered for prescription medication intervention.Citation7

Despite the lack of consensus definition until recently, multiple studies suggest that OIC is perhaps the most debilitating side effect of opioid therapy and a significant burden for patients. Nearly 1/3 of individuals on opioids state that constipation is their most bothersome symptom,Citation6 suggesting that OIC may be even more distressing than the underlying chronic pain.Citation2 OIC impairs patients’ ability to carry out their activities of daily living and results in decreased productivity at work, quality of life, sense of well-being, and level of health in general.Citation2,Citation8Citation10 Compared to patients without OIC, patients suffering from chronic OIC are more likely to miss work or feel impaired in their performance.Citation2 Even among patients who also take traditional laxatives, 81% of patients still reported OIC as their most bothersome side effect.Citation10

As a result of the prevalence and severity of OIC, many chronic pain patients are often forced to choose between adequate pain relief and comfortable bowel function. Multiple studies illustrate this dilemma.Citation4,Citation8,Citation11 Studies have found 8%–33% of patients report altering or stopping their opioid use due to OIC, thereby severely limiting the clinical benefit of opioids.Citation8,Citation11 In the PROBE study,Citation4 92% of patients who altered their opioid regimen due to OIC subsequently experienced increased pain, and 86% of those experiencing increased pain reported that it moderately to greatly reduced their quality of life and activities of daily living.

Not only does OIC impact patients’ quality of life, it also results in increased health care costs. Compared to patients who did not alter their opioid therapy, those who modified their regimen due to OIC had greater health care resource use as evidenced by increased number of surgeries, ER visits, and hospitalizations in the previous 6 months due to pain (odds ratio [OR] of 3.72 for having surgery, OR of 1.88 for ER visits, and OR of 2.47 for hospitalization). They also had more pain-related and OIC-related drug and health care provider out-of-pocket costs.Citation12 Another cost analysis in Sweden showed an increased burden of cost for patients with severe OIC. Severe OIC was associated with increased direct and indirect costs when compared to patients with no or mild constipation. Sick leave was found to be the largest cost. Use of outpatient facilities and total medication costs were also increased. Depending on the patient’s work status, the difference in monthly total cost could be as much as EUR 1,525 for those with severe OIC versus EUR 1,034 for those without OIC.Citation13 These quality of life and health care cost data suggest a significant unmet need in the diagnosis and treatment of OIC.

OIC treatment: historical and peripherally acting mu-opioid-receptor antagonists (PAMORAs)

Until recently, there has been little differentiation between treatment of functional constipation and that for OIC. Traditionally, treatment includes lifestyle modifications such as increased fiber intake, liquid intake, and physical activity.Citation2 As these nonpharmacologic measures often do not adequately relieve OIC, treatment then turns toward the use of laxatives. Laxatives function as stimulants, which increase smooth muscle contraction, and as stool softeners, which act as surfactants, lubricants, and osmotics. Stimulants include senna and bisacodyl. Stool softeners include docusate, mineral oil, lactulose, and polyethylene glycol.Citation2

Interestingly, some of these traditional remedies have been proven ineffective or harmful in recent years. Docusate has not demonstrated efficacy in randomized controlled studies for OIC compared to placebo,Citation14 and the use of bulk-forming laxatives (psyllium or fiber) was found to worsen constipation or even cause bowel obstruction the setting of opioid use.Citation2 No laxative has been found to be superior to any other.Citation15 Currently, no GI society has published treatment guidelines specifically for managing OIC,Citation6 although the need for such guidelines has been outlined recently by Camilleri et al.Citation4

Numerous professional societies including the European Association of Palliative Care and the National Comprehensive Cancer Network recommend prophylactic laxatives at the onset of opioid therapy despite a lack of randomized trials to validate such a practice.Citation6 Even with such prophylaxis, studies suggest that the majority of patients do not achieve adequate relief from OIC. Coyne et alCitation8 found that among patients who took recommended doses of laxatives, 94% failed to have adequate response. Kumar et alCitation2 found that 54% of patients prescribed concurrent laxatives with their opioids did not achieve the desired level of symptomatic (constipation) improvement more than half the time. The relative lack of efficacy of traditional approaches to constipation for OIC may be due at least in part to the distinct pathophysiology of OIC.Citation6

More recently, medications aimed specifically at the GI tract’s opioid receptors have been employed to combat OIC. These include a combined, prolonged-release, oral naloxone/oxycodone product that has been shown in a randomized, double-blind study to provide a statistically significant improvement in BFI scores with no significant difference in pain control in patients with both cancer-related and noncancer-related chronic pain. Naloxone, typically utilized for its ability to cross the blood–brain barrier to reverse life-threatening opioid overdoses, has low systemic bioavailability (<3%) in a prolonged-release formulation.Citation16

PAMORAs are a newer class of medications that are unable to cross the blood–brain barrier allowing them to counteract opioids’ GI side effects while preserving CNS-mediated analgesia. PAMORAs include methylnaltrexone, alvimopan, and naloxegol.

Methylnatrexone has been shown to be superior to placebo for treatment of OIC in patients who failed traditional management.Citation15,Citation17 Its limitations include subcutaneous administration and restricted indication, ie, use only in patients with advanced disease.Citation1 Alvimopan is available in oral formulation, but is restricted to short-term use for post-operative ileus as clinical trials showed increased incidence of myocardial infarction with long-term use.Citation1,Citation18 Naloxegol, (Movantik™, AstraZeneca, Cambridge, England) is an oral, once-daily PAMORA currently approved by the US Food and Drug Administration (FDA) for treatment of OIC in noncancer pain.

Naloxegol: brief pharmacology

Naloxegol reduces OIC by functioning as a PAMORA. Chemically, it is a PEGylated form of naloxone.Citation19,Citation20 It does not reverse pain relief primarily due to its inability to cross the blood–brain barrier.Citation3

Pharmacokinetic summary

Naloxegol reaches two peaks in concentration following oral administration. The first peak occurs in less than two hours, and the second peak occurs at anywhere from 24 minutes to 3 hours later due to enterohepatic recycling. Fatty meals and grapefruit juice can increase naloxegol plasma concentrations; therefore, it is recommended to take it on an empty stomach and avoid grapefruit juice.Citation19

Because metabolism is primarily through the CYP3A4 enzyme, naloxegol is contraindicated if there is concurrent use of strong CYP3A4 inhibitors or inducers. A dose reduction of 50% is recommended with concurrent use of moderate CYP3A4 inhibitors.Citation19

The half-life of naloxegol is between 6 and 11 hours. Excretion is primarily in the feces.Citation19 No dose adjustment is recommended for mild-to-moderate hepatic impairment. Naloxegol use is not recommended for patients with severe hepatic impairment due to lack of investigation in this population.Citation19

Renal clearance is only a minor route of elimination for naloxegol. It is not removed with standard hemodialysis.Citation21 For patients with creatinine clearance less than 60 mL/min, the recommended starting dose is 12.5 mg daily, with an increase of 25 mg daily if tolerated and if OIC persists.Citation21

Clinical studies of naloxegol: efficacy, safety, and patients’ perspectives

Efficacy

Key Phase I, II, and III trials evaluating pharmacokinetics, efficacy, and safety of naloxegol are summarized in . Two of the Phase III trials compared response rates between placebo and naloxegol doses of 12.5 mg daily and 25 mg daily. Response was defined as at least three spontaneous bowel movements per week that was at least 1 greater than baseline. These changes needed to be seen in at least 9 of the 12 trial weeks and at least 3 of the last 4 weeks of the trial. The KODIAC 04 trial found significant response at both doses, while the KODIAC 05 trial found significant response at the 25 mg dose.

Table 1 Details of relevant naloxegol studies

Secondary end points evaluated in these trials included time to first spontaneous bowel movement, mean number of spontaneous bowel movements per week, mean number of days per week with at least one spontaneous bowel movement, severity of straining, hardness of stool, and need for rescue laxative use.Citation22 Although results were variable for the 12.5 mg dose, all of the secondary end points had positive responses at the 25 mg dose. In an analysis of the subgroup of patients with moderate-to-very-severe OIBD who failed prior use of laxatives, significant response to both doses was found in KODIAC 04 and to the 25 mg dose in the KODIAC 05 trial.Citation22

Safety and tolerability

Recorded adverse events were mostly GI in nature and occurred more commonly at the 25 mg dose of naloxegol.Citation22,Citation23 Approximately 10% of participants taking the 25 mg dose discontinued use due to adverse events, with diarrhea the most common reason in KODIAC 04 and abdominal pain being the most common reason in KODIAC 05.Citation22 Overall pain scores and mean daily opioid doses remained unchanged. Possible symptoms of withdrawal were rare and felt to be artifacts caused by the symptom scoring system.Citation22

A third Phase III trial followed patients taking naloxegol over 1 year. As in KODIAC 04 and 05, an approximately 10% discontinuation rate was found due to GI adverse effects.Citation23 Most participants reported an unwanted adverse effect from naloxegol; however, in general, they were considered as being mild to moderate in severity. No change in reported pain, opioid use, or symptoms of withdrawal was observed.Citation23

No major events or bowel perforation was reported. However, the studies excluded participants in whom there was evidence of bowel obstruction or conditions that increased risk of bowel perforation.Citation22,Citation23 An additional study in healthy men showed that QT/QTc intervals did not increase more than 30 ms with doses up to 150 mg.Citation24

Patients’ perspectives

Unfortunately, clinical trials of naloxegol have included relatively little patient-oriented outcomes data. Webster et al’s Phase II trial,Citation25 however, did assess patients’ perceptions of their OIC symptoms and quality of life on naloxegol. Webster et alCitation25 used three common, validated questionnaires to assess patients’ perspectives: the Patient Assessment of Constipation-Symptom Questionnaire (PAC-SYM), qualifying abdominal, rectal, and stool symptoms; the Patient-Assessment of Constipation Quality of Life Questionnaire (PAC-QoL), assessing physical discomfort, worries/concerns, psychosocial discomfort, and satisfaction; and the Short Form Health Survey (SF-36), which yields an 8-scale profile of functional health and well-being scores.

The study found statistically significant improvements in the aforementioned questionnaires for the group of patients taking 25 mg of naloxegol daily. These patients reported lower PAC-SYM scores for rectal symptoms, lower PAC-QoL scores for physical discomfort, and improved SF-36 scores for physical functioning, mental health, social functioning, and vitality.Citation25 Additionally, Webster et alCitation25 found that most adverse events (nausea, diarrhea, and abdominal pain) were mild or moderate and self-limiting in nature. Importantly, the results also suggested that patients did not alter their opioid use in order to relieve bowel symptoms as no statistically significant change in opioid usage was seen in patients taking naloxegol.

In their Phase III clinical trial of naloxegol, Webster et alCitation23 also assessed patients’ use of rescue medication (bisacodyl) for those patients randomized to treatment with naloxegol at month 1, months 1–3, months 3–6, months 6–9, and months 9–12. Use of rescue medication was considered part of the usual care arm and was not tracked. Bisacodyl use for rescue among naloxegol users was low – median use was 1.1 mg per week during the first month and no use for the remainder of the 52-week study period. This suggests that naloxegol alone was sufficient for acceptable bowel function among the opioid users studied.

Clinical use and follow-up

Current recommended dosage of naloxegol is 25 mg once daily in the morning, on an empty stomach (>30 minutes before the first meal of the day or >2 hours afterward).Citation19,Citation26 A lower starting dose of 12.5 mg may be indicated in patients with tolerability issues or renal impairment; naloxegol is not recommended in patients with severe liver dysfunction.Citation19 Clinical trials have required patients to stop other constipation treatments prior to starting naloxegol. These same trials have suggested that the need for rescue medication in addition to naloxegol is low.Citation23 Prescribing guidelines recommend stopping other bowel medications and restarting them only if there is an inadequate response to naloxegol after 3 days.Citation19

To assess patient responses to naloxegol treatment, Camilleri et alCitation4 proposed two validated outcome tools specifically for OIC to determine whether particular treatments improve OIC symptoms:

  • A 3-item BFI in which patients rate on a 0–100 scale their perception of:

    Ease of defecation

    Feeling of incomplete bowel evacuation

    Personal judgment of constipation based on the previous 7 days.

  • A bowel function diary that requires patient assessment via:

    A 4-item module after each bowel movement

    A 5-item module completed each evening.Citation4

In the US, naloxegol is only currently approved for use in noncancer patients with OIC. In the European Union, however, it is approved for any patient with OIC and an inadequate response to treatment with more than one laxative for at least 4 days in the previous 2 weeks.Citation19 Additional investigation is needed to establish naloxegol’s efficacy, safety, and tolerability specifically in cancer patients on chronic opioid therapy.

Conclusion

OIC is a burdensome problem for patients on chronic opioids, negatively affecting their quality of life and increasing health care costs. Traditional treatments for functional constipation often have insufficient efficacy in OIC. Naloxegol is a once-daily, oral PAMORA specifically designed to address the unique pathophysiology of OIC by inhibiting mu-opioid-receptor binding in the GI tract without reversing CNS-mediated pain relief. Clinical experience is limited, but efficacy and safety studies in patients with noncancer pain suggest a daily dose of 25 mg naloxegol is clinically effective and generally well-tolerated. Patients on naloxegol experienced improved quality of life, adherence to opioid therapy, and reduced reliance on other laxatives for regular bowel function. Naloxegol use led to statistically significant lower levels of physical discomfort as well as statistically significant improvement in physical functioning, mental health, social functioning, and vitality. There were no statistically significant changes in opioid use for patients on naloxegol, suggesting that patients did not need to alter their opioid use to relieve constipation symptoms, nor did they experience any withdrawal symptoms. Finally, studies showed a low use of rescue medication use by patients on naloxegol, suggesting that naloxegol alone was sufficient for acceptable bowel function among opioid users studied. These data, however, are currently limited to patients with nonmalignant pain, but future investigations seek to study the use of naloxegol in cancer patients with OIC.

Disclosure

The authors report no conflicts of interest in this work.

References

  • PoulsenJLBrockCOlesenAENilssonMDrewesAMClinical potential of naloxegol in the management of opioid-induced bowel dysfunctionClin Exp Gastroenterol2014734535825278772
  • KumarLBarkerCEmmanuelAOpioid-induced constipation: pathophysiology, clinical consequences, and managementGastroenterol Res Pract2014201416
  • TackJCorsettiMNaloxegol for the treatment of opioid-induced constipationExpert Rev Gastroenterol Hepatol20148885586125220391
  • CamilleriMDrossmanGWebsterLRDaviesANMaweGMEmerging treatment in neurogastroenterology: a multidisciplinary working group consensus statement on opioid-induced constipationNeurogastroenterol Motil2014261386139525164154
  • GaertnerJSiemensWCamilleriMDefinitions and outcome measures of clinical trials regarding opioid-induced constipation: a systematic reviewJ Clin Gastroenterol201549191625356996
  • DavisMGamierPNew options in constipation managementCurr Oncol Rep2015175526449843
  • ArgoffCEBrennanMJCamilleriMConsensus recommendations of initiating prescription therapies for opioid-induced constipationPain Med2015162324233726582720
  • CoyneKSLoCasaleRJDattoCJSextonCCYeomanKTackJOpioid-induced constipation in patients with chronic noncancer pain in the USA, Canada, Germany and the UK: descriptive analysis of baseline patient-reported outcomes and retrospective chart reviewClinicoecon Outcomes Res2014626928124904217
  • AbramowitzLBéziaudNCausséCChuberreBAllaertFPerrotSFurther validation of the psychometric properties of the bowel function index for evaluating opioid-induced constipation (OIC)J Med Econ201316121423144124102123
  • BellTPanchalSMiaskowskiCBolgeSMilanovaTWilliamsonRThe prevalence, severity, and impact of opioid-induced bowel dysfunction: results of a US and European patient surveyPain Med2009101354218721170
  • OlesenADrewesAValidated tools for evaluating opioid-induced bowel dysfunctionAdv Ther201128427929421437762
  • GuptaSPatelJScopelJModyRImpact of constipation on opioid therapy management among long-term opioid users, based on a patient surveyJ Opioid Manag201511432533826312960
  • HjalteFBerggrenABergendahlHHjortsbergCThe direct and indirect costs of opioid-induced constipationJ Pain Symptom Manage201040569670320727708
  • TarumiYWilsonMPSzafranOSpoonerGRRandomized, double-blind, placebo-controlled trial of oral docusate in the management of constipation in hospice patientsJ Pain Symptom Manage201345121322889861
  • CandyBJonesLGoodmanMLDrakeRTookmanALaxatives or methylnaltrexone for the management of constipation in palliative care patientsCochrane Database Syst Rev20111CD003448
  • AhmedzaiSHLeppartWJaneckiMLong-term safety and efficacy of oxycodone/naloxone prolonged-release tablets in patients with moderate-to-severe chronic cancer painSupport Care Cancer20152382383025218610
  • ThomasJKarverSCooneyGAMethylnaltrexone for opioid-induced constipation in advanced illnessN Engl J Med2008358222332234318509120
  • Entereg (alvimopan) [package insert]Lexington, MACubist Pharmaceuticals, Inc.2013
  • Movantik™ (naloxegol) tablets [package insert]Product informationWilmington, DEAstraZeneca Pharmaceuticals LP92014
  • EldonMASongDNeumannTANKTR-118 (oral PEG-naloxol), a PEGylated derivative of naloxone: demonstration of selective peripheral opioid antagonism after oral administration in preclinical modelsPoster presented at: 18th Annual Clinical Meeting of the American Academy of Pain Management927–302007Las Vegas, NV
  • BuiKSheFSostekMThe effects of renal impairment on the pharmacokinetics, safety, and tolerability of naloxegolJ Clin Pharmacol201454121375138224946021
  • CheyWDWebsterLSostekMLappalainenJBarkerPNTackJNaloxegol for opioid-induced constipation in patients with noncancer painN Engl J Med2014370252387239624896818
  • WebsterLCheyWDTackJLappalainenJDivaUSostekMRandomised clinical trial: the long-term safety and tolerability of naloxegol in patients with pain and opioid-induced constipationAliment Pharmacol Ther201440777177925112584
  • GottfridssonCCarlsonGLappalainenJSostekMEvaluation of the effect of Naloxegol on cardiac repolarization: a randomized, placebo- and positive-controlled crossover thorough QT/QTc study in healthy volunteersClin Ther201335121876188324238792
  • WebsterLDharSEldonMMasoukaLLappaliaininJSostekMA Phase 2, double-blind, randomized, placebo-controlled, dose-escalation study to evaluate the efficacy, safety, and tolerability of naloxegol in patient with opioid-induced constipationPain20131541542155023726675
  • HolzerPNaloxegol increases frequency of bowel movements and combats inadequate response to laxativesEvid Based Med2015201525209526
  • BuiKSheFSostekMThe effects of mild or moderate hepatic impairment on the pharmacokinetics, safety, and tolerability of naloxegolJ Clin Pharmacol201454121368137424945932
  • BrunerHCAtayeeRSEdmondsKPBuckholzGTClinical utility of naloxegol in the treatment of opioid-induced constipationJ Pain Res2015828929426109876