Scolaris Content Display Scolaris Content Display

Mu‐опиоидные антагонисты при опиоид‐индуцированной дисфункции кишечника у людей с раком и людей, получающих паллиативную помощь

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

Ahmedzai 2012 {published data only}

Ahmedzai SH, Nauck F, Bar‐Sela G, Bosse B, Leyendecker P, Hopp M. A randomized, double‐blind, active‐ controlled, double‐dummy, parallel‐group study to determine the safety and efficacy of oxycodone/naloxone prolonged‐release tablets in patients with moderate/severe, chronic cancer pain. Palliative Medicine 2012;26:50‐60. CENTRAL
Center for Drug Evaluation and Research, Medical Review. Application number: 205777Orig1s000. www.accessdata.fda.gov/drugsatfda_docs/nda/2014/205777Orig1s000MedR.pdf (accessed 17 November 2016). CENTRAL
US Food, Drug Administration. Center for Drug Evaluation and Research. Risk Assessment and Risk Mitigation Review. Product oxycodone/naloxone extended‐release tablets. www.accessdata.fda.gov/drugsatfda_docs/nda/2014/205777Orig1s000RiskR.pdf (assessed 17 November 2016). CENTRAL

Bull 2015 {published data only}

Bull J, Wellman CV, Israel RJ, Barrett AC, Paterson C, Forbes WP. Fixed‐dose subcutaneous methylnaltrexone in patients with advanced illness and opioid‐induced constipation: results of a randomized, placebo‐controlled study and open‐label extension. Journal of Palliative Medicine 2015;18:593‐600. CENTRAL

Dupoiron 2017 {published data only}

Dupoiron D, Stachowiak A, Loewenstein O, Ellery A, Kremers W, Bosse B, et al. A phase III randomized controlled study on the efficacy and improved bowel function of prolonged‐release (PR) oxycodone/naloxone (up to 160/80 mg daily) vs oxycodone PR. European Journal of Pain 2017;21:1‐10. CENTRAL

Katakami 2017 {published data only}

Katakami N, Oda K, Tauchi K, Nakata K, Shinozaki K, Yokota T, et al. Phase IIb randomized, double‐blind, placebo‐controlled study of naldemedine for the treatment of opioid‐induced constipation in patients with cancer. Journal of Clinical Oncology 2017;35(17):1921‐1928. CENTRAL

Portenoy 2008 {published data only}

Portenoy RK, Thomas J, Moehl Boatwright ML, Galasso FL, Stambler N, Von Gunten CF, et al. Subcutaneous methylnaltrexone for the treatment of opioid‐induced constipation in patients with advanced illness: a double‐blind, randomised, parallel group, dose‐ranging study. Journal of Pain and Symptom Management 2008;35:458‐68. CENTRAL

Slatkin 2009 {published data only}

Center for Drug Evaluation and Research. Statistical review. Application 21‐964. Food and Drug Administration. www.accessdata.fda.gov (assessed 01/04/16)2008. CENTRAL
European Medicines Agency. Assessment report for Relistor. Procedure No. EMEA/H/C/870. www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Public_assessment_report/human/000870/WC500050566.pdf (accessed 1 April 2016). CENTRAL
Slatkin N, Thomas J, Lipman AG, Wilson G, Boatwright ML, Wellman C, et al. Methylnaltrexone for treatment of opioid‐induced constipation in advanced illness patients. Journal of Supportive Oncology 2009;7:39‐46. CENTRAL

Sykes 1996 {published data only}

Sykes NP. An investigation of the ability of oral naloxone to correct opioid‐related constipation in patients with advanced cancer. Palliative Medicine 1996;10:135‐44. CENTRAL

Thomas 2008 {published data only}

Chamberlain BH, Cross K, Winston JL, Thomas J, Wang W, Su C, et al. Methylnaltrexone treatment of opioid‐induced constipation in patients with advanced illness. Journal of Pain and Symptom Management 2009;38:683‐90. CENTRAL
Thomas J, Karver S, Cooney GA, Chamberlain BH, Watt CK, Slatkin NE, et al. Methylnaltrexone for opioid‐induced constipation in advanced illness. New England Journal of Medicine 2008;358:2332‐4. CENTRAL

Meissner 2009 {published data only}

Meissner W, Leyendecker P, Mueller‐Lissner S, Nadstawek J, Hopp M, Ruckes C, et al. A randomised controlled trial with prolonged‐release oral oxycodone and naloxone to prevent and reverse opioid‐induced constipation. European Journal of Pain 2009;13:55‐64. CENTRAL

Mori 2017 {published data only}

Mori M, Ji Y, Kumar S, Ashikaga T, Ades S. Phase II trial of subcutaneous methylnaltrexone in the treatment of severe opioid‐induced constipation (OIC) in cancer patients: an exploratory study. International Journal of Clinical Oncology2017; Vol. 22, issue 2:397‐404. CENTRAL

Nadstawek 2008 {published data only}

Nadstawek J, Leyendecker P, Hopp M, Ruckes C, Wirz S, Fleischer W, et al. Patient assessment of a novel therapeutic approach for the treatment of severe, chronic pain. International Journal of Clinical Practice 2008;62:1159‐67. CENTRAL

Poelaert 2015 {published data only}

Poelaert J, Koopmans‐Klein G, Dioh A, Louis F, Gorissen M, Loge D, et al. Treatment with prolonged‐release oxycodone/naloxone improves pain relief and opioid‐induced constipation compared with prolonged‐release oxycodone in patients with chronic severe pain and laxative‐refractory constipation. Clinical Therapeutics 2015;37:784‐92. CENTRAL

Vondrackova 2008 {published data only}

Vondrackova D, Leyendecker P, Meissner W, Hopp M, Szombati I, Hermanns K, et al. Analgesic efficacy and safety of oxycodone in combination with naloxone as prolonged release tablets in patients with moderate to severe chronic pain. Journal of Pain 2008;9:1144‐54. CENTRAL

Webster 2013 {published data only}

Webster L, Dhar S, Eldon M, Masuoka L, Lappalainen J, Sostek M. A phase 2, double‐blind, randomized, placebo‐controlled, dose‐escalation study to evaluate the efficacy, safety, and tolerability of naloxegol in patients with opioid‐induced constipation. Pain 2013;154:1542‐50. CENTRAL

Dimitroulis 2014 {published data only}

Dimitroulis I A. Methylnaltrexone bromide in the treatment of opioid‐induced constipation in lung cancer patients. Is it effective?. Journal of Thoracic Oncology 2014;9:S204. CENTRAL

JAPIC‐CTI‐132340 {published data only}

Harada T, Katakami N, Murata T, Shinozakl K, Tsutsumi M, Yokota T, et al. Phase 3 study to evaluate the efficacy and safety of naldemedine for the treatment of opioid‐induced constipation (OIC) in cancer patients. Journal of Clinical Oncology 2016;34 (Suppl):10016. CENTRAL

NCT00135577 {unpublished data only}

NCT00135577. Study 767905/008 extension study: alvimopan for treatment of opioid‐induced bowel dysfunction in cancer pain subjects. clinicaltrials.gov/ct2/show/NCT00135577 Date first received: 26 August 2005. CENTRAL

NCT00331045/00101998 {unpublished data only}

NCT00101998. Study of alvimopan drug for treatment of constipation due to prescription pain medication. clinicaltrials.gov/ct2/show/NCT00101998 Date first received: 19 January 2005. CENTRAL
NCT00331045. Clinical evaluation of alvimopan (SB767905) on constipation and related symptoms associated with opioid. clinicaltrials.gov/ct2/show/NCT00331045 Date first received: 29 May 2006. CENTRAL

NCT01438567 {unpublished data only}

NCT01438567. A study to demonstrate improvement in symptoms of constipation in subjects that require around‐the‐clock opioid pain killer therapy. clinicaltrials.gov/ct2/show/NCT01438567 Date first received: 22 September 2011. CENTRAL

NCT02321397 {published data only}

NCT02321397. To demonstrate equivalence in analgesic efficacy & bowel function between OXN PR higher dose & lower dose tablet strengths in subjects with non‐cancer or cancer pain. clinicaltrials.gov/ct2/show/NCT02321397 Date first received: 22 December 2014. CENTRAL

NCT02574819 {published data only}

NCT02574819. Study of methylnaltrexone in opioid‐induced constipation patients. clinicaltrials.gov/ct2/show/NCT02574819 Date first received: 14 October 2015. CENTRAL

NCT02745353 {published data only}

NCT02745353. Naloxegol in cancer opioid‐induced constipation. clinicaltrials.gov/ct2/show/NCT02745353 Date first received: 20 April 2016. CENTRAL

NCT02839889 {published data only}

NCT02839889. Tolerability, safety, and feasibility of naloxegol in patients with cancer and OIC (opioid induced constipation). clinicaltrials.gov/ct2/show/NCT02839889 Date first received: 21 July 2016. CENTRAL

Neefjes 2014 {published data only}

Neefjes EC, van der Vorst MJ, Boddaert MS, Zuurmond WW, van der Vliet HJ, Beeker A, et al. Clinical evaluation of the efficacy of methylnaltrexone in resolving constipation induced by different opioid subtypes combined with laboratory analysis of immunomodulatory and antiangiogenic effects of methylnaltrexone. BMC Palliative Care 2014;13:42. CENTRAL

Peppin 2013 {unpublished data only}

Peppin. RCT: methylnaltrexone in patients with advanced illness with opioid induced constipation. Clinicaltrials.gov2013. CENTRAL

Bader 2012

Bader S, Weber M, Becker G. Is the pharmacological treatment of constipation in palliative care evidence based? A systematic literature review. Der Schmerz 2012;26:568‐86.

Bader 2013

Bader S, Durk T, Becker G. Methylnaltrexone for the treatment of opioid‐induced constipation. Expert Review of Gastroenterology & Hepatology 2013;7:13‐26.

Brown 1985

Brown DR, Goldberg LI. The use of quaternary narcotic antagonists in opiate research. Neuropharmacology 1985;24:181‐91.

Camilleri 2011

Camilleri 2011. Opioid‐induced constipation: challenges and therapeutic opportunities. American Journal of Gastroenterology 2011;106:835‐42.

Camilleri 2014

Camilleri M, Drossman DA, Becker G, Webster LR, Davies AN, Mawe GM. Emerging treatments in neurogastroenterology: a multidisciplinary working group consensus statement on opioid‐induced constipation. Neurogastroenterology & Motility 2014;26:1386‐95.

Candy 2015

Candy B, Jones L, Larkin PJ, Vickerstaff V, Tookman A, Stone P. Laxatives for the management of constipation in people receiving palliative care. Cochrane Database of Systematic Reviews 2015, Issue 5. [DOI: 10.1002/14651858.CD003448.pub4]

Caraceni 2012

Caraceni A, Hanks G, Kaasa S, Bennett MI, Brunelli C, Cherny N, et al. European Palliative Care Research Collaborative (EPCRC), European Association for Palliative Care (EAPC). Use of opioid analgesics in the treatment of cancer pain: evidence‐based recommendations from the EAPC. Lancet Oncology 2012;13:e58‐68.

Clark 2014

Clark K, Currow DC. Methylnaltrexone in palliative care: further research is needed. Journal of Pain and Symptom Management 2014;47:e5‐6.

Cook 2008

Cook SF, Lanza L, Zhou X, Sweeney CT, Goss D, Hollis K, et al. Gastrointestinal side effects in chronic opioid users: results from a population‐based survey. Alimentary Pharmacology & Therapeutics 2008;27:1224‐32.

Coyne 2014

Coyne KS, LoCasale RJ, Datto CJ. Opioid‐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 review. Clinicoeconomic Outcomes Research 2014;6:269‐81.

Deeks 2006

Deeks JJ, Higgins JP, Altman DG, editor(s). Analysing and presenting results. In: Higgins JP, Green S editor(s). Cochrane Handbook for Systematic Reviews of Interventions. 4.2.6. Chichester, UK: Wiley‐Blackwell, 2006.

Diego 2011

Diego L, Atayee R, Helmons P, Hsiao G, von Gunten CF. Novel opioid antagonists for opioid‐induced bowel dysfunction. Expert Opinion on Investigational Drugs 2011;20:1047‐56.

Egger 1997

Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta‐analysis detected by a simple, graphical test. BMJ 1997;315:629‐34.

Ford 2013

Ford AC, Brenner DM, Schoenfeld PS. Efficacy of pharmacological therapies for the treatment of opioid‐induced constipation: systematic review and meta‐analysis. American Journal of Gastroenterology 2013;108:1566‐74.

Guyatt 2013a

Guyatt G, Oxman AD, Sultan S, Brozek J, Glasziou P, Alonso‐Coello P, et al. GRADE guidelines: 11. Making an overall rating of confidence in effect estimates for a single outcome and for all outcomes. Journal of Clinical Epidemiology 2013;66:151‐7.

Guyatt 2013b

Guyatt GH, Oxman AD, Santesso N, Helfand M, Kunz R, Brozek J, et al. GRADE guidelines: 12. Preparing summary of findings tables‐binary outcomes. Journal of Clinical Epidemiology 2013;66:158‐72.

Higgins 2011

Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Jadad 1996

Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ. Assessing the quality of reports of randomized clinical trials: is blinding necessary. Controlled Clinical Trials 1996;17:1‐12.

Janku 2015

Janku F, Moss J, Karp D, Singleton P, Johnson L. Treatment with methylnaltrexone is associated with increased survival in patients with advanced refractory cancers treated for opioid induced constipation. European Journal of Cancer 2015;51:S235.

Ketwaroo 2013

Ketwarro GA, Cheng V, Lembo A. Opioid‐induced bowel dysfunction. Current Gastroenterology Reports 2013;15:344.

Kumar 2012

Kumar NB. Dysfunction of the Bowel/Constipation. Nutritional Management of Cancer Treatment Effects. Berlin/Heidelberg: Springer‐Verlag, 2012.

Leppert 2010

Leppert W. The role of opioid receptor antagonists in the treatment of opioid‐induced constipation: a review. Advances in Therapy 2010;27:714‐30.

Leppert 2015

Leppert W. Emerging therapies for patients with symptoms of opioid‐induced bowel dysfunction. Drug Design, Development and Therapy 2015;9:2215‐31.

Manjiani 2014

Manjiani D, Paul DB, Kunnumpurath S, Kaye AD, Vadivelu N. Availability and utilization of opioids for pain management: global issues. Ochsner Journal 2014;14:208‐15.

Marderstein 2008

Marderstein EL, Delaney CP. Management of postoperative ileus: focus on alvimopan. Therapeutics and Clinical Risk Management 2008;4:965‐73.

Mehta 2016

Mehta N, O'Connell K, Giambrone GP, Baqai A, Diwan S. Efficacy of methylnaltrexone for the treatment of opioid‐induced constipation: a meta‐analysis and systematic review. Postgraduate Medicine 2016;128:282‐9.

Merck 2015

Pharmaceutical Manufacturing Research Services. Highlights of prescribing information, 2015. www.merck.com/product/usa/pi_circulars/e/entereg/entereg_pi.pdf (date accessed 28 March 2017).

NICE 2012

National Institute for Health and Care excellence. Opioids in palliative care: safe and effective prescribing of strong opioids for pain in palliative care of adults, 2012. www.nice.org.uk/guidance/cg140/resources (date accessed 17 November 2016).

NICE 2013

National Institute for Health and Care excellence. Methylnaltrexone for treating opioid‐induced bowel dysfunction in people with advanced illness receiving palliative care (suspended appraisal). NICE technology appraisal guidance (TA277), 2013. www.nice.org.uk/guidance/ta277 (accessed 19 April 2017).

Panchal 2007

Panchal SJ, Muller‐Schwefe P, Wurzelmann JI. Opioid‐induced bowel dysfunction: prevalence, pathophysiology and burden. International Journal of Clinical Practice 2007;61:1181‐7.

Pappagallo 2001

Pappagallo M. Incidence, prevalence, and management of opioid bowel dysfunction. American Journal of Surgery 2001;182 (Suppl):11‐18S.

Pizzi 2012

Pizzi LT, Toner R, Foley K, Thomson E, Chow W, Kim M, et al. Relationship between potential opioid‐related adverse effects and hospital length of stay in patients receiving opioids after orthopedic surgery. Pharmacotherapy 2012;32:502‐14.

Pritchard 2015

Pritchard D, Bharucha A. Management of opioid‐induced constipation for people in palliative care. International Journal of Palliative Nursing 2015;21:272‐80.

Roberts 2015

Roberts I, Ker K. How systematic reviews cause research waste. Lancet 2015;386:1536.

Schulz 2010

Schulz KF, Altman DG, Moher D. CONSORT 2010 statement: updated guidelines for reporting parallel group randomized trials. Annals of Internal Medicine 2010;152:1‐7.

Scottish Palliative Care Guidelines 2014

NHS Scotland. Scottish Palliative Care Guidelines: about the guidelines, 2014. www.palliativecareguidelines.scot.nhs.uk/guidelines (accessed 17 November 2016).

Siemens 2015

Siemens W, Gaertner J, Becker G. Advances in pharmacotherapy for opioid‐induced constipation ‐ a systematic review. Expert Opinion on Pharmacotherapy 2015;16:515‐32.

Sterne 2011

Sterne JAC, Egger M, Moher D. Chapter 10: Addressing reporting biases. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Sykes 1998

Sykes NP. The relationship between opioid use and laxative use in terminally ill cancer patients. Palliative Medicine 1998;12:375‐82.

Wang 2013

Wang C‐Z, Yuan C‐S. Pharmacologic treatment of opioid‐induced constipation. Drugs 2013;22:1225‐7.

WHO 2016

World Health Organization. WHO's cancer pain ladder for adults, 2016. who.int/cancer/palliative/painladder/en/ (accessed 17 November 2016).

Zhang 2013

Zhang Z, Xu X, Ni H. Small studies may overestimate the effect sizes in critical care meta‐analyses: a meta‐epidemiological study. Critical Care 2013;17:R2.

Candy 2011

Candy B, Jones L, Goodman ML, Drake R, Tookman A. Laxatives or methylnaltrexone for the management of constipation in palliative care patients. Cochrane Database of Systematic Reviews 2011, Issue 1. [DOI: 10.1002/14651858.CD003448.pub3]

McNicol 2008

McNicol ED, Boyce D, Schumann R, Carr DB. Mu‐opioid antagonists for opioid‐induced bowel dysfunction. Cochrane Database of Systematic Reviews 2008, Issue 2. [DOI: 10.1002/14651858.CD006332.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Ahmedzai 2012

Methods

Randomised, parallel, controlled, multi‐centre trial. International with sites in Australia, Czech Republic, France, Germany, Hungary, Israel, the Netherlands, Poland, and the UK

Participants

Aim: to investigate whether OXN PR can improve constipation and maintain analgesia compared with OXY PR tablets, in people with cancer.

Inclusion criteria: people with chronic moderate/severe cancer pain and requiring 24‐h opioid therapy

Exclusion criteria: clinically unstable disease or significant cardiovascular, renal, hepatic, or psychiatric disease; clinically significant gastrointestinal disease or significant structural abnormalities of the gastrointestinal tract; cyclic chemotherapy within 2 weeks before screening visit or planned during the core trial (shown in the past to influence bowel function); radiotherapy that would influence bowel function or pain during the double‐blind phase

Participants: in the intervention arm; mean age 61 years and 48/92 men. In comparison arm; mean age 64 years and 46 men and 46 women. The most common primary cancer sites were breast (19%), lung (13%), and prostate (10%). 26% had bone metastases. At the start of the trial, 183/184 (99.5%) participants had constipation‐induced or worsened by their opioid medication. A similar number were also taking laxatives. All were outpatients.

Interventions

Intervention: OXN PR up to 120 mg/day, n = 92

Comparison: OXY PR up to 120 mg/day, n = 92

Duration: 4 weeks

Outcomes

Primary outcomes: symptoms of constipation as measured by Bowel Function Index, efficacy for management of chronic cancer pain as measured by the Brief Pain Inventory‐Short Form

Secondary outcomes: use of rescue medication, quality of life, and safety

Outcomes measured: at 4 weeks

Notes

Funding: Mundipharma GmbH

Trial registration: NCT00513656/OXN2001

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were assigned to treatments (1:1 allocation ratio) using a pseudo‐random number generator in a computer program."

Allocation concealment (selection bias)

Low risk

Quote: "randomisation schedule prepared by the Clinical Supplies Department of the Sponsor or an associated company."

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Stated double‐blind, no further details provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

133/184 completed the trial. Less than a third in each group dropped out. Similar proportion dropped out in each group

Selective reporting (reporting bias)

Unclear risk

No details provided

Sample size

Unclear risk

50‐199 participants per treatment arm

Bull 2015

Methods

Randomised, controlled, parallel, multi‐centred trial in the USA

Participants

Aim: to determine the efficacy and safety of fixed‐dose subcutaneous methylnaltrexone in people with advanced illness and opioid‐induced constipation in a variety of healthcare situations (inpatient, outpatient, home, hospice, and long‐term care facilities).

Inclusion criteria: participants aged > 18 years with advanced illness and a life expectancy of ≥ 1 month and opioid‐induced constipation (< 3 BM in the last week and no BM in 24 h or 48 h) and who were receiving stable doses of laxatives and opioids

Exclusion criteria: people with a disease process suggestive of gastrointestinal obstruction or clinically significant active diverticular disease, fecal impaction, peritonitis, bowel surgery 10 days before dosing, or fecal ostomy, or with a bodyweight < 38 kg

Participants: 118 men and 112 women. Mean age in intervention arm 65.3 years (SD 12.9) and in placebo arm 65.7 years (SD 13.0). 216/230 of white race. Primary diagnosis cancer in 66% of participants (152/230). The majority (58/78) of the other participants had pulmonary, cardiovascular, or neurological disease

Interventions

Intervention: subcutaneous methylnaltrexone 8 mg (bodyweight of 38 kg to < 62 kg) or 12 mg (bodyweight > 62 kg), n = 116

Comparison: placebo, n = 114

Duration: both were administered every other day over 2 weeks

Outcomes

Primary outcome: percentage of participants with RFBM within 4 h after at the most 2 of the doses in the first week of treatment

Secondary outcomes: % with the first RFBM within 4 h after the first dose, number of BMs within 24 h after dosing per week

Outcomes measured: over 2 weeks

Notes

Funding: technical editorial and medical writing assistance from Salix Pharmaceuticals Limited

Trial registration: NCT00672477

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomly assigned." No other details

Allocation concealment (selection bias)

Unclear risk

Quote: "randomly assigned." No other details

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Stated double blind, no further details provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

27/116 in the intervention group and 20/114 in placebo were lost to follow‐up. Reason for loss were similar in both trial arms.

Selective reporting (reporting bias)

Unclear risk

No details

Sample size

Unclear risk

50‐199 participants per treatment arm

Dupoiron 2017

Methods

Randomised, controlled, parallel trial unclear what country participants were from

Participants

Aim: to evaluate the tolerability and efficacy of OXN PR doses up to oxycodone/naloxone 160 mg/80 mg compared with OXY PR formulation.

Inclusion criteria: adults with cancer and non‐cancer pain requiring opioids on a stable dose of OXY PR for ≥ 4 consecutive days prior to randomisation and have a pain score of ≤ 4 with ≤ 2 doses of OXY PR analgesic rescue medication per day for either the last 3 consecutive days or 4 of the last 7 days. Constipation caused or aggravated by opioids was confirmed by the participant and the investigator and evidenced by a medical need of regular laxatives to have ≥ 3 bowel evacuations per week or by having < 3 bowel evacuations when not taking a laxative

Exclusion criteria: included hypersensitivity to oxycodone, naloxone; active alcohol or drug abuse or history of opioid abuse (or both); unreported illicit drug use (including cannabis); any condition in which opioids were contraindicated or if they had diarrhoea

Participants: 100 men and 143 women randomised, of which a subsample, 46, were people with cancer pain. Mean age in whole sample 57.9 years (SD 11.03) in OXN PR arm and 57.5 years (SD 12.33) in OXY PR arm. Subsample demographics on people with cancer not provided

Interventions

Intervention: starting dose during the double‐blind phase dependent on the effective, stable analgesic dose established in the run‐in period, titration up to maximum daily dose of OXN PR 160 mg/80 mg was permitted after 1 week

Comparison: OXY PR equivalent dosage to participants in the intervention arm

Duration: up to 5 weeks

Outcomes

Primary outcomes: change in mean bowel function scores, pain scores

Secondary outcomes: analgesic and laxative rescue medication, complete SBMs, and quality of life (EuroQol EQ‐5D‐3L)

Outcomes measured: 1, 2, 4, and 5 weeks

Notes

Funding: Mundipharma GmbH

Trial registration: NCT01438567

Study comprised of 3 phases: prerandomisation phase consisting of a screening period and a run‐in period, a double‐blind phase, and an extension phase. In the run‐in phase, OXY PR was titrated to analgesic effect to determine the starting dose to be used after randomisation.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "randomly assigned." No other details

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Double‐blind, participant blinded, no other details on who else was blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

16/120 men and 18/123 women in whole sample dropped out per arm. Number who dropped in the subset of 46 people with cancer not reported

Selective reporting (reporting bias)

Low risk

Bias was unlikely as the trial listed in clinical trial registry reported same primary and secondary outcomes that were presented in the paper

Sample size

High risk

< 50 participants per treatment arm in subsample of people with cancer

Katakami 2017

Methods

Randomised, controlled, parallel, multi‐centred trial in Korea and Japan

Participants

Aim: to evaluate the dose, efficacy, and safety of naldemedine for the treatment of opioid‐induced constipation in people with cancer in Japan and Korea.

Inclusion criteria: adults aged ≥ 18 years with cancer pain, stable regimen of opioid for > 2 weeks, complicated with opioid‐induced constipation despite regular laxative use

Exclusion criteria: constipation potentially attributable to causes other than opioid analgesics

Participants: 134 men and 93 women entered trial. Mean age by trial arm: naldemedine 0.1 mg daily: 65.8 years (SD 11.5), naldemedine 0.2 mg daily: 63.4 years (SD 10.4), naldemedine 0.4 mg daily: 64.2 years (SD 10.7); placebo: 64.2 (SD 9.6). Most participants had lung cancer, other cancers included breast and colorectal. All as graded by the ECOG Performance Status were ambulatory. Care setting not stated

Interventions

Intervention 1: naldemedine 0.1 mg daily, n = 56

Intervention 2: naldemedine 0.2 mg daily, n = 58

Intervention 3: naldemedine 0.4 mg daily, n = 56

Comparison: placebo, n = 57

Duration: all administered daily for 2 weeks

Outcomes

Primary outcome: change from baseline in the frequency of SBM per week

Secondary outcomes: SBM responder rate, change from baseline in frequency of complete SBM, change from baseline in frequency of SBM without straining, adverse events, and opiate withdrawal

Outcomes measured: over 2 weeks

Notes

Funding: Shionogi and Co Ltd

Trial registration: JapicCTI‐111510

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Achieved "using the dynamic allocation procedure of the registration center, where the maximum intergroup difference in the participant number at each study site did not exceed two."

Allocation concealment (selection bias)

Unclear risk

Probably occurred as allocation provided remotely but not stated specifically

Blinding (performance bias and detection bias)
All outcomes

Low risk

All study team members and participants were blinded to treatment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2 participants, 1/57 in placebo group and 1/56 in naldemedine 0.1 mg were lost to follow‐up.

Selective reporting (reporting bias)

Unclear risk

No details

Sample size

Unclear risk

50‐199 participants per treatment arm

Portenoy 2008

Methods

Randomised, controlled, multi‐centre, parallel‐group trial in the USA

Participants

Aim: to assess the efficacy and safety of subcutaneous methylnaltrexone in a population of people with advanced illness and opioid‐induced constipation, and to clarify whether there was a dose‐response relationship for the purpose of dose selection in further clinical evaluations.

Inclusion criteria: advanced disease (defined as terminal or end‐stage, such as advanced metastatic cancer and AIDS but with a life expectancy of ≥ 4 weeks and stable vital signs) for which they were receiving palliative care and were receiving any opioid drug on a daily basis at a dose that had been stable for ≥ 2 weeks and were expected to remain stable for an additional ≥ 4 weeks, and despite no or conventional laxative therapy they had no BMs for 2 days and reported ongoing constipation, defined as > 2 days with no BM and a score of ≥ 3 on a 5‐point scale assessing constipation‐related distress

Exclusion criteria: fever or otherwise unstable vital signs; liver function test 3 times the upper limit of normal, serum creatinine level 2 times the upper limit, or a platelet count < 50,000/mm3; new regimen or dose change of concurrent gastrointestinal motility‐altering medications during 3 weeks prior to trial enrolment; history of gastrointestinal obstruction or other condition that could compromise drug action; diagnosis of active peritoneal cancer; history of peritoneal catheter placement for chemotherapy or dialysis; known hypersensitivity to methylnaltrexone, naltrexone, or naloxone; or if any investigational drug or experimental product had been administered within the previous 30 days

Participants: 15 men and 18 women. Mean age 61 years (SD 19.0) (range 20‐87 years). 79% were white people. Primary diagnoses at baseline were 28/33 cancer, 3 sickle cell disease, and 2 AIDS. 88% of participants were receiving a laxative at baseline. The mean opioid (morphine equivalent) dose at baseline was 289.9 mg/day (SD 308.0), median 180 mg/day, range 9‐1207 mg/day. Mean number of BMs per week was 1.9. Care setting not stated

Interventions

Intervention 1: subcutaneous methylnaltrexone 1 mg, n = 10

Intervention 2: subcutaneous methylnaltrexone 5 mg, n = 7

Intervention 3: subcutaneous methylnaltrexone 12.5 mg, n = 10

The initial dose range of 1 mg, 5 mg, or 12.5 mg was extended by adding a 20 mg group (n = 6) during the trial while still maintaining the double‐blind.

Duration: 3 doses over 1 week

Outcomes

Primary outcomes: laxative response (BM) within 4 h of the initial dose.

Secondary outcomes: laxation within 4 h of subsequent doses, during the 24‐h period after each dose, time to laxation, use of rescue laxatives, subjective outcomes of constipation‐associated symptoms, pain intensity, symptoms potentially due to opioid withdrawal or adverse events, and participant satisfaction

Outcomes measured: up to 24 h per dose, and 30 days after last dose

Notes

Funding: Progenics Pharmaceuticals

Trial registration: none provided

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "After providing consent, patients were initially randomised in a ratio of 1:1:1 to receive 1 mg, 5 mg, or 12.5 mg of methylnaltrexone."

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Double‐blind, participant blinded, no other details on who was blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

22/33 completed trial. 7 discontinued "at patient request", three from the 12.5 mg arm and one each from the 1 mg and 5 mg arm and two from 20 mg arm. One in the 20mg arm discontinued because of "intolerable" adverse event

Selective reporting (reporting bias)

Unclear risk

No details provided

Sample size

High risk

< 50 participants per treatment arm

Slatkin 2009

Methods

Randomised, controlled, parallel‐group, multi‐centre controlled trial

Participants

Aim: to assess the safety and efficacy of a single subcutaneous injection of methylnaltrexone (0.15 mg/kg or 0.3 mg/kg) versus placebo.

Inclusion criteria: aged > 18 years, advanced illness (such as incurable cancer or end‐stage AIDS and life expectancy 1‐6 months) and opioid‐induced constipation. On a stable opioid regimen for the control of pain/discomfort for ≥ 3 days before randomisation, had a stable scheduled laxative regimen for ≥ 3 days prior to treatment, no clinically significant laxation within 48 h prior to the first trial drug dose, had stable vital signs, and not pregnant and using an effective method of birth control. Baseline laxative regimens taken at time of trial entry could be continued throughout the trial. Rescue laxatives, defined as laxatives administered on an as needed basis were allowed but not within 4 h before or after administration of the double‐blind dose.

Exclusion criteria: previous treatment with methylnaltrexone, naltrexone, or naloxone; recent participation in any other studies involving investigational products; any disease process suggestive of gastrointestinal obstruction; any potential non‐opioid cause of bowel dysfunction; history of current peritoneal catheter for intraperitoneal administration, chemotherapy administration, or dialysis; clinically active diverticular disease; evidence of faecal impaction; surgically acute abdomen; faecal ostomy; pregnancy; or breastfeeding

Participants: 84 American men and 70 American women at 17 hospice and other palliative care settings. Mean age 65.3 years (SD 14.96). Primary diagnosis cancer (125/154), cardiovascular disease (8), HIV/AIDS (1), and other (20). Apart from 8 participants, all had some level of constipation distress. 95% were using a laxative. Oral morphine equivalents, median mg/day 186.5, range 8‐12,2560 mg/day

Interventions

Intervention 1: single subcutaneous injection methylnaltrexone 0.15 mg/kg, n = 47

Intervention 2: single subcutaneous injection methylnaltrexone 0.3 mg/kg, n = 55

Comparison: placebo, n = 52

Duration: 1‐week double‐blind phase, followed by 28‐day open phase

Outcomes

Primary outcome: proportion of participants with rescue‐free laxation (a significant BM) within 4 h after administration of the double‐blind dose. Participants needing rescue laxative or disimpaction within 4 h of dosing were considered non‐responders.

Secondary outcomes: proportion of participants with rescue‐free laxation within 24 h postdosing; improvement in GCIC scale (defined as a rating of slightly better, somewhat better, or much better); improvement in constipation distress (defined as a change by at least 1 category toward none); improvement in stool consistency; changes in baseline pain, symptoms/signs of central opioid withdrawal, and adverse events

Outcomes measured: to 6 days following first dose

Notes

Funding: Progenics Pharmaceuticals

Trial registration: 301/NCT00401362

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...randomly assigned in blocks of three to the three treatment groups in a 1:1:1 ratio. Computer‐generated randomisation scheme performed by a statistician external to the sponsor."

Allocation concealment (selection bias)

Low risk

Quote: "computer‐generated randomisation scheme performed by a statistician external to the sponsor."

Blinding (performance bias and detection bias)
All outcomes

Low risk

Quote: "...syringe contents were blinded to patients and staff administering injections." "each syringe had identical volume."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

152/154 completed trial (1 died and 1 was non‐compliant both in trial arm of higher dose of methylnaltrexone)

Analysis on an intention‐to‐treat basis

Selective reporting (reporting bias)

Unclear risk

No details

Sample size

High risk

< 50 participants in 1 of the 2 treatment arms

Although this risk was not relevant to some of our analysis. This is when we combined the trials 2 treatment groups in our exploration of the impact of mu‐opioid antagonists in comparison with placebo.

Sykes 1996

Methods

Randomised, controlled, single‐centre, cross‐over trial

Participants

Aim: to assess in a dose‐ranging trial the use of oral naloxone in opioid‐related constipation in participants with advanced cancer

Inclusion criteria: participants with advanced cancer receiving either morphine or diamorphine analgesia orally. All required laxatives prior to trial and their use was continued during the trial except for lactulose

Exclusion criteria: fecal stomas or history of constipation prior to using opioid analgesia

Participants: 13 men and 14 women patients in a UK hospice. Mean age 64 years, median 65 years, range 44‐88 years. 9 participants had breast cancer; 5 bronchus; 3 prostate; 2 oesophagus, and 1 each of rectum, kidney, bladder, stomach, colon, fallopian tube, malignant melanoma, and fibrosarcoma); 3 participants had liver metastases, 2 had hepatomegaly; no participant had constipation prior to using opioid analgesia

Interventions

Morphine or diamorphine oral (maintenance dose)

Intervention: naloxone oral every 4‐h for total daily dose of 0.5%, 1%, 2%, 5%, 10%, or 20% of total daily dose of morphine. The participants received "one level" (a lower level) of naloxone. Then after 2 participants at 0.5% to 5% had received the drug without slowing bowel transit time the dose was increased. In higher doses, the increase was following no slowing effect in 4 participants, n = 17

Comparison: placebo: chloroform water, n = 17

Duration: 2 days each treatment arm (parallel washout)

Outcomes

Outcomes: small bowel transit time by lactulose/hydrogen breath test; pain by 4‐point scale (0 = no pain, 3 = severe pain)

Notes

Funding: charities, Cancer Relief Macmillan Fund, and the Wolfson Foundation. Naloxone was donated by MacFarlan Smith (pharmaceutical company).

Trial registration: none

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Stated randomised but no further details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Stated double blind but no further details provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data analysis of 12 participants were reported. Of the 5 not included, 1 declined. 4 were withdrawn, 2 because of diarrhoea (1 occurred while on placebo, 1 caused by the lactulose taken as part of the small bowel transit time test), 1 was withdrawn because of general deterioration, and 1 because of nausea which the trialists felt was not related to the intervention).

Selective reporting (reporting bias)

Unclear risk

No details. The study does not declare a primary outcome

Sample size

High risk

< 50 participants per treatment arm

Thomas 2008

Methods

Randomised, controlled, multi‐centre, parallel trial

Participants

Aim: to assess the safety and efficacy of subcutaneous methylnaltrexone for treating opioid‐induced constipation in participants with advanced illness.

Inclusion criteria: participants who had a terminal illness with a life expectancy > 1 month, were receiving stable doses of opioids for analgesia and had opioid‐induced constipation (defined as ≤ 3 laxations in the previous week or no laxation in the previous 48 h) despite having taken laxatives for ≥ 3 days. Participants could continue their baseline laxative regimen throughout the trial and take rescue laxatives as needed, though not within 4 h before or after receiving a dose of the trial drug.

Exclusion criteria: participants whose constipation was not primarily caused by opioids, mechanical gastrointestinal obstruction, an indwelling peritoneal catheter, clinically active diverticular disease, fecal impaction, acute surgical abdomen, and fecal ostomy

Participants: 58 men and 76 women from North America. They were from 27 nursing homes, hospice sites, or other palliative care centres in the USA and Canada (78 with cancer, 15 cardiovascular disease, 14 chronic obstructive pulmonary disease, 8 dementia, and 19 with other diseases). Median age in methylnaltrexone group 70 years (range 34‐93 years) and in the placebo group 72 years (range 39‐98 years). Opioid dose: methylnaltrexone group: mean 417 mg/day, median 150 mg/day, range 9‐4160 mg/day; placebo group: mean 339 mg/day, median 100 mg/day, range 10‐10,160 mg/day. 98% in the methylnaltrexone and 99% in placebo group were using laxatives

Interventions

Intervention: subcutaneous methylnaltrexone 0.15 mg/kg bodyweight, n = 62

Comparison: placebo, n = 71

Dose every other day

Duration of treatment: 2 weeks

Outcomes

Primary outcome: RFBM within 4 h after first dose

Secondary outcomes: laxation within 4 h after ≥ 2 of the first 4 doses. Consistency (from watery to hard) and difficulty of laxation. Adverse events were assessed using the National Cancer Institute's Common Toxicity Criteria (rated on a scale from 'none' to 'very much'). Participants were also assessed on the Modified Himmelsbach Opiate Withdrawal Scale (on 7 symptoms including yawning, lacrimation, rhinorrhoea, perspiration, tremor, piloerection, and restlessness)

Outcomes measured: over 2 weeks

Notes

Funding: Progenics Pharmaceuticals. Trial 302

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation schedule, blocked according to trial centre

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
All outcomes

Low risk

Quote: "The study drugs (40 mg of methylnaltrexone per millilitre or placebo) were provided in identically appearing vials." "Syringe contents were blinded to patients and staff administering injections."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

106/133 completed trial

Selective reporting (reporting bias)

Unclear risk

No details

Sample size

Unclear risk

50‐199 participants per treatment arm

BM: bowel movement; ECOG: Eastern Cooperative Oncology Group; CGIC: Clinical Global Impression of Change; h: hour; n: number of participants; OXN PR: oxycodone/naloxone prolonged release; OXY PR: oxycodone prolonged release; RFBM: rescue‐free bowel movements; SBM: spontaneous bowel movement; SD: standard deviation.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Meissner 2009

Study of people with chronic pain not palliative care or cancer

Mori 2017

Not an RCT

Nadstawek 2008

Study of people with chronic pain not palliative care or cancer

Poelaert 2015

Not an RCT

Vondrackova 2008

Study of people with chronic (low back) pain not palliative care or cancer

RCT: randomised controlled trial.

Characteristics of studies awaiting assessment [ordered by study ID]

Webster 2013

Methods

RCT

Participants

Participants with opioid‐induced constipation. Participants had non‐malignant or cancer‐related pain. No breakdown provided of number with cancer and no subanalysis of effect in group with cancer

Interventions

Naloxegol

Outcomes

Spontaneous bowel movements

Notes

Awaiting responses from authors to clarify population details and further details for analysis

RCT: randomised controlled trial.

Characteristics of ongoing studies [ordered by study ID]

Dimitroulis 2014

Trial name or title

Methylnaltrexone Bromide in the Treatment of Opioid‐Induced Constipation in Lung Cancer Patients

Methods

Single‐centre RCT

Participants

34 participants with a life expectancy of ≥ 3 months receiving treatment for lung cancer. All participants received fentanyl

Interventions

Intervention: methylnaltrexone 12 mg/0.6 mL subcutaneous

Comparison: placebo

Duration: 4 weeks

Drugs administered on alternate days

Outcomes

Laxation

Starting date

Trial completed, no published paper identified

Contact information

Ioannis A Dimitroulis, 6th Pulmonary Department, Sotiria Hospital for Thoracic Diseases, Athens, Greece

Notes

JAPIC‐CTI‐132340

Trial name or title

Phase 3 Study to Evaluate the Efficacy and Safety of Naldemedine for the Treatment of Opioid‐Induced Constipation in Cancer Patients

Methods

RCT

Participants

People with cancer

Interventions

Intervention: naldemedine

Comparison: placebo

Outcomes

Efficacy in improving bowel function and safety

Starting date

Study completed 2016, result presented in abstract only, full paper expected

Contact information

Toshiyuki Harada MD, PhD (harada‐[email protected])

Notes

Sponsor Shionogi Limited

NCT00135577

Trial name or title

A Double‐Blind, Placebo‐Controlled, Multi‐Centre Phase IIb Extension Study to Evaluate the Safety and Efficacy of Multiple Alvimopan Dosage Regimens for the Treatment of Opioid‐Induced Bowel Dysfunction in Cancer Pain Subjects

Methods

RCT

Participants

People with cancer

Interventions

Intervention: alvimopan

Comparison: placebo

Outcomes

Laxation

Starting date

2004

Contact information

Sponsor Cubist, collaborator GlaxoSmithKline

Notes

ID NCT00135577

NCT00331045/00101998

Trial name or title

Trial of Alvimopan Drug for Treatment of Constipation due to Prescription Pain Medication in Cancer Patients

Methods

RCT

Participants

People with cancer

Interventions

Intervention: alvimopan

Comparison: placebo

Outcomes

Not stated

Starting date

Start date 2003, completed 2006

Contact information

Funded by Cubist

Notes

IDs NCT00331045 trial terminated early (with 21 participants) "as subject registration did not proceed as expected," NCT00101998 trial completed

NCT01438567

Trial name or title

RCT in Symptoms of Constipation in Subjects with Non‐Malignant or Malignant Pain that Requires Around‐the‐Clock Opioid Therapy taking 50/25‐80/40 mg Twice Daily as Oxycodone/Naloxone Prolonged Release Tablets Compared to Subjects taking 50‐80 mg Twice Daily Oxycodone Prolonged Release Tablets Alone

Methods

RCT

Participants

People with and without cancer pain

Interventions

Intervention: oxycodone/naloxone

Comparison: oxycodone alone

Outcomes

Pain and bowel function

Starting date

Clinical trials website reports trial complete, no publication identified

Contact information

Funded by Mundipharma GmbH, no contact details provided on clinical trials website

Notes

NCT02321397

Trial name or title

To Demonstrate Equivalence in Analgesic Efficacy & Bowel Function Between Oxycodone/naloxone PR Higher Dose & Lower Dose Tablet Strengths in Subjects with Non‐cancer or Cancer Pain

Methods

RCT

Participants

People with and without cancer with pain

Interventions

OXN PR higher‐dose and lower‐dose tablets

Outcomes

Pain and bowel function

Starting date

2014

Contact information

Funded by Mundipharma, no contact details provided on clinical trials website

Notes

NCT02574819

Trial name or title

Trial of Methylnaltrexone in Opioid‐Induced Constipation Patients

Methods

RCT

Participants

People with advanced illness

Interventions

Intervention: Subcutaneous methylnaltrexone

Comparison: placebo

Outcomes

Laxation

Starting date

2015

Contact information

Shiying Yu, [email protected]

Notes

Sponsors: Jiangsu Chia‐tai Tianqing Pharmaceutical Co, Ltd

NCT02745353

Trial name or title

Naloxegol in Cancer Opioid‐Induced Constipation

Methods

Randomised single‐centre trial

Participants

People with cancer

Interventions

Intervention: naloxegol

Comparison: treatment as usual

Outcomes

Laxation, quality of life, and pain

Starting date

May 2016

Contact information

Chelsea Hagmann, [email protected]

Notes

Sponsor: University of California, Collaborator: Astra Zeneca

NCT02839889

Trial name or title

Tolerability, Safety, and Feasibility of Naloxegol in Patients with Cancer and OIC (Opioid Induced Constipation)

Methods

Randomised multi‐centre trial

Participants

People with cancer

Interventions

Intervention: naloxegol

Comparison: placebo

Outcomes

Laxation and pain

Starting date

July 2016

Contact information

Janet Bull, MD, [email protected]

Notes

Sponsor and collaborators: Hospice of Henderson County, Inc and Astra Zeneca

Neefjes 2014

Trial name or title

Clinical Evaluation of the Efficacy of Methylnaltrexone in Resolving Constipation‐Induced by Different Opioid Subtypes Combined with Laboratory Analysis of Immunomodulatory and Antiangiogenic Effects of Methylnaltrexone

Methods

Multi‐centre RCT

Participants

People receiving palliative care with opioid‐induced constipation

Interventions

Intervention: methylnaltrexone

Comparison: unclear

Outcomes

Differences in the efficacy of methylnaltrexone prescribed to resolve opioid‐induced constipation between 3 commonly used opioid subtypes: morphine sulphate, oxycodone, and fentanyl

Starting date

Not stated, protocol published in 2014. Trial ongoing as reported December 2015

Contact information

ECW Neefjes, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, The Netherlands, [email protected]

Notes

ID NCT01955213

Peppin 2013

Trial name or title

Effect of Subcutaneous Methylnaltrexone on Patient‐Reported Outcomes in Advanced Illness Patients with Opioid‐Induced Constipation

Methods

RCT

Participants

People with advanced illness

Interventions

Intervention: methylnaltrexone

Comparison: placebo

Outcomes

Participant‐reported outcomes of constipation distress, bowel movement difficulty, and Global Clinical Impression of Change

Starting date

Not stated, conference abstract with findings published in 2013

Contact information

J Peppin. Progenics Pharmaceuticals Inc, Tarrytown, NY sponsored trial

Notes

Did not include results section so unclear if trial is the same as any identified in a full published paper.

RCT: randomised controlled trial.

Data and analyses

Open in table viewer
Comparison 1. Methylnatrexone versus placebo: rescue‐free laxation:

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Within 24 hours of dose Show forest plot

2

287

Risk Ratio (M‐H, Fixed, 95% CI)

2.77 [1.91, 4.04]

Analysis 1.1

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 1 Within 24 hours of dose.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 1 Within 24 hours of dose.

2 Within 4 hours after 4 of the 7 doses Show forest plot

2

305

Risk Ratio (M‐H, Fixed, 95% CI)

9.98 [4.96, 20.09]

Analysis 1.2

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 2 Within 4 hours after 4 of the 7 doses.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 2 Within 4 hours after 4 of the 7 doses.

3 Within 4 hours of first dose Show forest plot

3

517

Risk Ratio (M‐H, Fixed, 95% CI)

3.87 [2.83, 5.28]

Analysis 1.3

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 3 Within 4 hours of first dose.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 3 Within 4 hours of first dose.

4 Within 4 hours after 1 or 2 doses of the first 4 doses Show forest plot

2

363

Risk Ratio (M‐H, Fixed, 95% CI)

6.89 [4.46, 10.66]

Analysis 1.4

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 4 Within 4 hours after 1 or 2 doses of the first 4 doses.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 4 Within 4 hours after 1 or 2 doses of the first 4 doses.

5 Improvement in constipation distress at day 1 Show forest plot

2

287

Risk Ratio (M‐H, Fixed, 95% CI)

1.87 [1.34, 2.59]

Analysis 1.5

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 5 Improvement in constipation distress at day 1.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 5 Improvement in constipation distress at day 1.

6 Participant global impression of improvement in bowel status at 1 week Show forest plot

2

287

Risk Ratio (M‐H, Fixed, 95% CI)

2.32 [1.64, 3.27]

Analysis 1.6

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 6 Participant global impression of improvement in bowel status at 1 week.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 6 Participant global impression of improvement in bowel status at 1 week.

7 Clinician global impression of improvement in bowel status at 1 week Show forest plot

2

287

Risk Ratio (M‐H, Fixed, 95% CI)

2.37 [1.66, 3.38]

Analysis 1.7

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 7 Clinician global impression of improvement in bowel status at 1 week.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 7 Clinician global impression of improvement in bowel status at 1 week.

Open in table viewer
Comparison 2. Methylnaltrexone versus placebo: serious adverse event

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse event Show forest plot

2

364

Risk Ratio (M‐H, Fixed, 95% CI)

0.59 [0.38, 0.93]

Analysis 2.1

Comparison 2 Methylnaltrexone versus placebo: serious adverse event, Outcome 1 Serious adverse event.

Comparison 2 Methylnaltrexone versus placebo: serious adverse event, Outcome 1 Serious adverse event.

Open in table viewer
Comparison 3. Methylnaltrexone versus placebo: adverse event

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse events Show forest plot

3

518

Risk Ratio (M‐H, Random, 95% CI)

1.17 [0.94, 1.45]

Analysis 3.1

Comparison 3 Methylnaltrexone versus placebo: adverse event, Outcome 1 Adverse events.

Comparison 3 Methylnaltrexone versus placebo: adverse event, Outcome 1 Adverse events.

2 Dropouts due to adverse event Show forest plot

2

363

Risk Ratio (M‐H, Fixed, 95% CI)

1.22 [0.54, 2.76]

Analysis 3.2

Comparison 3 Methylnaltrexone versus placebo: adverse event, Outcome 2 Dropouts due to adverse event.

Comparison 3 Methylnaltrexone versus placebo: adverse event, Outcome 2 Dropouts due to adverse event.

Open in table viewer
Comparison 4. Oxycodone/naloxone prolonged‐release tablets versus oxycodone prolonged‐release: adverse event

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse events Show forest plot

2

234

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.94, 1.24]

Analysis 4.1

Comparison 4 Oxycodone/naloxone prolonged‐release tablets versus oxycodone prolonged‐release: adverse event, Outcome 1 Adverse events.

Comparison 4 Oxycodone/naloxone prolonged‐release tablets versus oxycodone prolonged‐release: adverse event, Outcome 1 Adverse events.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 1 Within 24 hours of dose.
Figuras y tablas -
Analysis 1.1

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 1 Within 24 hours of dose.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 2 Within 4 hours after 4 of the 7 doses.
Figuras y tablas -
Analysis 1.2

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 2 Within 4 hours after 4 of the 7 doses.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 3 Within 4 hours of first dose.
Figuras y tablas -
Analysis 1.3

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 3 Within 4 hours of first dose.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 4 Within 4 hours after 1 or 2 doses of the first 4 doses.
Figuras y tablas -
Analysis 1.4

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 4 Within 4 hours after 1 or 2 doses of the first 4 doses.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 5 Improvement in constipation distress at day 1.
Figuras y tablas -
Analysis 1.5

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 5 Improvement in constipation distress at day 1.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 6 Participant global impression of improvement in bowel status at 1 week.
Figuras y tablas -
Analysis 1.6

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 6 Participant global impression of improvement in bowel status at 1 week.

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 7 Clinician global impression of improvement in bowel status at 1 week.
Figuras y tablas -
Analysis 1.7

Comparison 1 Methylnatrexone versus placebo: rescue‐free laxation:, Outcome 7 Clinician global impression of improvement in bowel status at 1 week.

Comparison 2 Methylnaltrexone versus placebo: serious adverse event, Outcome 1 Serious adverse event.
Figuras y tablas -
Analysis 2.1

Comparison 2 Methylnaltrexone versus placebo: serious adverse event, Outcome 1 Serious adverse event.

Comparison 3 Methylnaltrexone versus placebo: adverse event, Outcome 1 Adverse events.
Figuras y tablas -
Analysis 3.1

Comparison 3 Methylnaltrexone versus placebo: adverse event, Outcome 1 Adverse events.

Comparison 3 Methylnaltrexone versus placebo: adverse event, Outcome 2 Dropouts due to adverse event.
Figuras y tablas -
Analysis 3.2

Comparison 3 Methylnaltrexone versus placebo: adverse event, Outcome 2 Dropouts due to adverse event.

Comparison 4 Oxycodone/naloxone prolonged‐release tablets versus oxycodone prolonged‐release: adverse event, Outcome 1 Adverse events.
Figuras y tablas -
Analysis 4.1

Comparison 4 Oxycodone/naloxone prolonged‐release tablets versus oxycodone prolonged‐release: adverse event, Outcome 1 Adverse events.

Summary of findings for the main comparison. Naldemedine compared to placebo for opioid‐induced bowel dysfunction in cancer and people receiving palliative care

Naldemedine compared to placebo for opioid‐induced bowel dysfunction in cancer and people receiving palliative care

Patient or population: people with cancer and people receiving palliative care with opioid‐induced bowel dysfunction

Settings: cancer care

Intervention: naldemedine

Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo

Naldemedine

Laxation response within 24 hours of dose

Not reported

Laxation response between day 1 and day 14a

375 per 1000

724 per 1000
(510 to 1000)

RR 1.93 (1.36 to 2.74)

NNTB 2.88 (2.04 to 4.92)

225 (1 study)

⊕⊕⊕⊝

Moderateb

Effect on analgesia: opioid withdrawalc

0.1 mg: MD ‐0.13 (‐0.57 to 0.31); 0.2 mg: MD ‐0.40 (‐0.87 to 0.07); 0.4 mg: MD ‐0.02 (‐0.45 to 0.41)

225 (1 study)

⊕⊕⊕⊝

Moderateb

Effect on analgesia: pain intensity

Not reported

Serious adverse eventsa

5 SAEs occurred, all in naldemedine group.

225 (1 study)

⊕⊕⊝⊝
Lowb,d

Adverse eventsa

518 per 1000

704 per 1000 (539 to 927)

RR 1.36 (1.04 to 1.79)

225 (1 study)

⊕⊕⊕⊝

Moderateb

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MD: mean difference; NNTB: number needed to treat for an additional beneficial outcome; RR: risk ratio; SAE: serious adverse events.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

aMeasured by clinician or self‐report and in the case of adverse events using severity grades according to the Common Terminology Criteria for Adverse Events.
bDowngraded by one level for limitations to the study design due to unclear risk of bias (reporting bias).
cMeasured by Clinical Opiate Withdrawal Scale.
dDowngraded by one level for imprecision due to limited number of events.

Figuras y tablas -
Summary of findings for the main comparison. Naldemedine compared to placebo for opioid‐induced bowel dysfunction in cancer and people receiving palliative care
Summary of findings 2. Lower‐dose naldemedine compared to higher‐dose naldemedine for opioid‐induced bowel dysfunction in cancer and people receiving palliative care

Lower‐dose naldemedine compared to higher‐dose naldemedine for opioid‐induced bowel dysfunction in cancer and people receiving palliative care

Patient or population: people with cancer and people receiving palliative care with opioid‐induced bowel dysfunction

Setting: cancer care

Intervention: lower dose naldemedine 0.1 mg daily

Comparison: higher dose naldemedine 0.2 mg or 0.4 mg daily

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Higher dose

0.2 mg/0.4 mg daily

Lower dose

0.1 mg daily

Laxation response within 24 hours of dose

Not reported

Laxation response between day 1 and day 14a

0.1 mg vs 0.2 mg: 776 per 1000

0.1 mg vs 0.4 mg: 821 per 1000

0.1 mg vs 0.2 mg: 564 per 1000

(430 to 739)

0.1 mg vs 0.4 mg: 564 per 1000

(433 to 733)

0.1 mg vs 0.2 mg: RR 0.73 (0.55 to 0.95)

0.1 mg vs 0.4 mg: RR 0.69 (0.53 to 0.89)

226 (1 study)

0.1 mg vs 0.2 mg: n = 113

0.1 mg vs 0.4 mg: n = 111

⊕⊕⊕⊝

Moderateb

Effect on analgesia: opioid withdrawal

Not reported

Effect on analgesia: pain intensity

Not reported

Serious adverse events

Not reported

Adverse events

Not reported

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

aMeasured by self‐report.
bDowngraded by one level for limitations to the study design due to unclear risk of bias (reporting bias).

Figuras y tablas -
Summary of findings 2. Lower‐dose naldemedine compared to higher‐dose naldemedine for opioid‐induced bowel dysfunction in cancer and people receiving palliative care
Summary of findings 3. Naloxone compared with placebo for cancer and people receiving palliative care with opioid‐induced bowel dysfunction

Naloxone compared with placebo for cancer and people receiving palliative care with opioid‐induced bowel dysfunction

Patient or population: people with cancer and people receiving palliative care with opioid‐induced bowel dysfunction

Settings: cancer care

Intervention: naloxone

Comparison: placebo

Outcomes

Illustrative comparative risks*

Relative effect

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo

Naloxone

Laxation response within 24 hours of a dose

Not reported

Laxation response between day 1 and day 14

Not reported

Effect on analgesia: opioid withdrawal

Not reported

Effect on analgesia: pain intensitya

No statistical difference in pain experienced when taking placebo or naloxone. Full data, including pre‐cross‐over results, were not provided.

17 (1 study)

⊕⊝⊝⊝
Very lowb

Serious adverse events

Not reported

Adverse events

Not reported

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

aMeasured using 4‐point scale (0 = no pain, 3 = severe pain).
bDowngraded by three levels due to evidence from one study with a small sample size, which was a cross‐over study with no drug washout between cross‐over, and unclear risk of reporting bias.

Figuras y tablas -
Summary of findings 3. Naloxone compared with placebo for cancer and people receiving palliative care with opioid‐induced bowel dysfunction
Summary of findings 4. Oxycodone/naloxone prolonged release tablets compared with oxycodone prolonged‐released tablets for opioid‐induced bowel dysfunction

Oxycodone/naloxone prolonged release tablets compared with oxycodone prolonged‐released tablets for opioid‐induced bowel dysfunction

Patient or population: people with cancer and people receiving palliative care with opioid‐induced bowel dysfunction

Settings: cancer care

Intervention: oxycodone/naloxone prolonged‐release tablets

Comparison: oxycodone prolonged‐released tablets

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Oxycodone

Oxycodone/naloxone

Laxation response within 24 hours of dose

Not reported

Laxation response between day 1 and day 14

Not reported

Effect on analgesia: opioid withdrawalc

Intervention group: mean 6.64 (SD 5.97) comparison group: mean 7.29 (SD 4.59) at 7 days

184 (1 study)

⊕⊕⊕⊝

Moderateb

Effect on analgesia: pain intensitya

Intervention group: mean 3.50 (SD 1.88) and comparison group: mean 3.52 (SD 1.80) at 4 weeks

184 (1 study)

⊕⊕⊕⊝

Moderateb

Another study, Dupoiron 2017 also found outcome to be similar between trial arms, but did not provide any data.

Serious adverse events

43 per 1000

87 per 1000 (27 to 279)

RR 2.00 (95% CI 0.62 to 6.41)

184 (1 study)

⊕⊕⊝⊝

Lowb,d

Adverse events

754 per 1000

815 per 1000 (709 to 935)

RR 1.08 (95% CI 0.94 to 1.24)

234 (2 studies)

⊕⊕⊕⊝

Moderateb

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; RR: risk ratio; SD: standard deviation.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

aMeasured using the Brief Pain Inventory‐Short Form.
bDowngraded by one level because of study limitations (unclear risk of reporting bias).
cMeasured using the Modified Subjective Opiate Withdrawal Scale.
dDowngraded by one level due to imprecision because of wide confidence intervals.

Figuras y tablas -
Summary of findings 4. Oxycodone/naloxone prolonged release tablets compared with oxycodone prolonged‐released tablets for opioid‐induced bowel dysfunction
Summary of findings 5. Methylnaltrexone compared to placebo for opioid‐induced bowel dysfunction in cancer and people receiving palliative care

Methylnaltrexone compared to placebo for opioid‐induced bowel dysfunction in cancer and people receiving palliative care

Patient or population: people with cancer and people receiving palliative care with opioid‐induced bowel dysfunction

Setting: palliative care

Intervention: methylnaltrexone

Comparison: placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with placebo

Risk with

methylnaltrexone

Laxation response within 24 hours of dosea

195 per 1000

568 per 1000
(431 to 695)

RR 2.77 (1.91 to 4.04)

287
(2 studies)

⊕⊕⊕⊝

Moderateb

Laxation response between day 1 and day 14 (specifically within 4 hours after 4 or more of the 7 doses)a

52 per 1000

517 per 1000
(330 to 699)

RR 9.98 (4.96 to 20.09)

305
(2 studies)

⊕⊕⊕⊝

Moderate

b,c

Effect on analgesia: opioid withdrawald

Study 1: day 1: MD 0.00 (‐0.46 to 0.46); day 14: MD 0.10 (‐0.63 to 0.83)

Study 2: median change to day 2 = 0 in both trials arms

236

(2 studies)

⊕⊕⊕⊝

Moderateb

Effect on analgesia: pain intensitye

Study 1: at 4 hours (methylnaltrexone 0.15 mg/kg: MD ‐0.76 (‐1.47 to 0.05); methylnaltrexone 0.3 mg/kg: MD ‐0.25 (‐0.91 to 0.41)

Study 2: at day 1 and 14 (day 1: MD 0.20 (‐0.62 to 1.02); day 14: MD ‐0.70 (‐1.52 to 0.12)

287

(2 studies)

⊕⊕⊝⊝

Lowb,f

Another study, Bull 2015, found similar pain intensity experienced in trial arms, full data not provided.

Serious adverse events

238 per 1000

142 per 1000
(88 to 219)

RR 0.59 (0.38 to 0.93)

364
(2 studies)

⊕⊕⊕⊝

Moderateb

Adverse events

700 per 1000

815 per 1000
(745 to 869)

RR 1.17

(CI 0.94 to 1.45)

518
(3 studies)

⊕⊕⊝⊝

Lowb,g

Heterogeneity was substantial (74%). It was explained in sensitivity analysis by omitting the trial at a high risk of bias because of small sizes. The effect estimate was reduced. The direction of effect not changed.

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MD: mean difference; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aMeasured by self‐report or clinician report.
bDowngraded once for study limitations because of unclear risk of reporting bias.
cWe did not downgrade for imprecision due to wide confidence intervals because the effect size was large.

dMeasured using the modified Himmelsbach Opioid Withdrawal Scale.
eMeasured by participant‐rated scale 0‐10.
fDowngraded once for inconsistency because of differing estimates of effect.
gDowngraded once for inconsistency because statistical heterogeneity was high across trials.

Figuras y tablas -
Summary of findings 5. Methylnaltrexone compared to placebo for opioid‐induced bowel dysfunction in cancer and people receiving palliative care
Summary of findings 6. Lower‐dose methylnaltrexone compared to higher‐dose methylnaltrexone for opioid‐induced bowel dysfunction in cancer and people receiving palliative care

Lower dose methylnaltrexone compared to higher dose for opioid‐induced bowel dysfunction in cancer and people receiving palliative care

Patient or population: people with cancer and people receiving palliative care with opioid‐induced bowel dysfunction

Setting: palliative care

Intervention 1: lower‐dose methylnaltrexone (study 1: 3 doses, 1 week, 1 mg; study 2: 1 dose, 0.15 mg/kg)

Intervention 2: higher‐dose methylnaltrexone (study 1: 3 doses, 1 week, 5‐12.5 mg; study 2: 1 dose, 0.30 mg/kg)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Higher dose

Lower dose

Laxation response within 24 hours of first dosea

Study 1: 609 per 1000

Study 2: 639 per 1000

Study 1: 499 per 1000 (250 to 100)

Study 2: 681 per 1000 (515 to 904)

Study 1: RR 0.82 (0.41 to 1.66)

Study 2: RR 1.07 (0.81 to 1.42)

135 (2 studies)

Study 1: n = 33

Study 2: n = 102

⊕⊕⊝⊝

Lowb

Unable to combine study data as methylnaltrexone low and higher doses differed per trial

Laxation responsea

At 3 days: 706 per 1000

At 3 days: 332 per 1000
(127 to 882)

At 3 days: RR 0.47 (0.18 to 1.25)

33 participants (1 study)

⊕⊕⊝⊝

Lowb

Unable to combine study data as methylnaltrexone low and higher doses differed per trial

At 5 days: 688 per 1000

At 5 days: 144 per 1000
(21 to 901)

At 3 days: RR 0.21 (0.03 to 1.31)

Effect on analgesia: opioid withdrawalc

MD ‐0.04 (‐0.73 to 0.65)

102 participants

(1 study)

⊕⊕⊝⊝

Lowb

Another study,Portenoy 2008, also found outcome to be similar between trial arms, but did not provide any data

Effect on analgesia: pain intensityd

MD ‐0.51 (‐1.49 to 0.47)

102 participants

(1 study)

⊕⊕⊝⊝

Lowb

Another study, Portenoy 2008, also found outcome to be similar between trial arms, but did not provide any data

Serious adverse event

Not reported

Adverse event

Study 1: 1000 per 1000

Study 2: 800 per 1000

Study 1: 1000 per 1000 (1000 to 1000)

Study 2: 723 per 1000

(580 to 902)

Study 1: RR 1.00 (1.00 to 1.00)

Study 2: RR 0.90 (0.73 to 1.13)

135 (2 studies)

Study 1: n = 33

Study 2: n = 102

⊕⊕⊝⊝

Lowb

Unable to combine study data as methylnaltrexone low and higher doses differed per trial

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MD: mean difference; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

aMeasured by clinician or self‐report.

bDowngraded by two levels for study limitations: one for unclear risk of bias (reporting bias) and one for small sample size (high risk of bias).

cMeasured using the modified Himmelsbach Opioid Withdrawal Scale.

dMeasured by participant‐rated scale 0‐10.

Figuras y tablas -
Summary of findings 6. Lower‐dose methylnaltrexone compared to higher‐dose methylnaltrexone for opioid‐induced bowel dysfunction in cancer and people receiving palliative care
Table 1. Adverse events reported 2% of more participants in the trial of naldemedine

Adverse event

Naldemedine (%)

Placebo (%)

Diarrhoea

67 (39)

14 (25)

Decreased WBC count

9 (5)

3 (5)

Abdominal pain

6 (4)

0 (0)

Vomiting

5 (3)

0 (0)

Bone marrow failure

3 (2)

2 (4)

Decreased appetite

6 (4)

1 (2)

Nasopharyngitis

4 (2)

1 (2)

Nausea

4 (2)

4 (7)

Rash

3 (2)

2 (4)

Decreased platelet count

3 (2)

0 (0)

Decreased total protein

7 (4)

1 (2)

Glucose in urine

4 (2)

1 (2)

Abnormal haematology test

2 (1)

0 (0)

Decreased RBC count

4 (2)

0 (0)

Hypertension

2 (1)

0 (0)

Increased blood alkaline phosphatase

4 (2)

1 (2)

Increased blood lactate dehydrogenase

2 (1)

1 (2)

Increased blood pressure

2 (1)

0 (0)

Increased blood urea

4 (2)

1 (2)

Increased WBC count

1 (2)

2 (4)

Protein present in urine

5 (3)

0 (0)

Upper abdominal pain

3 (2)

1 (2)

RBC: red blood cell; WBC: white blood cell.

All comparisons were not statistically significant.

Figuras y tablas -
Table 1. Adverse events reported 2% of more participants in the trial of naldemedine
Table 2. Sensitivity analyses

Methylnaltrexone vs placeboa

AEs

RR 1.07, 95% CI 0.96 to 1.19

AE of abdominal pain

RR 2.15, 95% CI 1.28 to 3.62

AE of nausea

RR 0.87, 95% CI 0.46 to 1.65

AE of vomiting

RR 0.70, 95% CI 0.33 to 1.47

aomitting trial of high risk of bias.

AE: adverse event; CI: confidence intervals; RR: risk ratio.

Figuras y tablas -
Table 2. Sensitivity analyses
Table 3. Types of adverse event: reported in more than one trial of methylnaltrexone (versus placebo)

Adverse event

RR (95% CI)

I²statistic

on heterogeneity

Abdominal pain

2.39 (1.07 to 5.34)

65%

Diarrhoea

1.02 (0.93 to 1.11)

51%

Dizziness

4.09 (0.99 to 16.83)

0%

Falls

1.02 (0.89 to 1.16)

84%

Flatulence

2.09 (1.07 to 4.08)

0%

Nausea

0.97 (0.89 to 1.06)

63%

Peripheral oedema

1.01 (0.50 to 2.03)

0%

Restlessness

0.83 (0.32 to 2.12)

0%

Somnolence

1.00 (0.93 to 1.08)

73%

Vomiting

0.99 (0.92 to 1.08)

67%

CI: confidence interval; RR: risk ratio.

Figuras y tablas -
Table 3. Types of adverse event: reported in more than one trial of methylnaltrexone (versus placebo)
Table 4. Types of adverse events: reported in only one trial of methylnaltrexone

Adverse event

Methylnaltrexone (%)

Placebo (%)

Abdominal distensiona

1 (2)

6 (8)

Abdominal tendernessa

1 (2)

4 (6)

Astheniaa

4 (6)

4 (6)

Anxietyb

5 (4.9)

0 (0)

Arthralgiab

3 (2.9)

1 (1.9)

Back painc

9 (7.8)

3 (2.9)

Confusional statec

7 (6.0)

9 (7.9)

Dehydrationa

2 (3)

4 (6)

Fatigueb

4 (3.9)

1 (1.9)

Hypotensiona

0 (0)

4 (6)

Increased body temperaturea

5 (8)

2 (3)

Lethergya

4 (6)

4 (6)

Malignant‐neoplasm progressiona

7 (11)

9 (13)

Pain exacerbationb

8 (8)

2 (4)

Rhinorrhoeab

6 (5.9)

1 (1)

Sweating increasedb

8 (7.8)

4 (7.7)

Tachycardiaa

1 (1)

4 (6)

aReported in trial by Thomas 2008.

bReported in trial by Slatkin 2009.

cReported in trial by Bull 2015.

Figuras y tablas -
Table 4. Types of adverse events: reported in only one trial of methylnaltrexone
Comparison 1. Methylnatrexone versus placebo: rescue‐free laxation:

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Within 24 hours of dose Show forest plot

2

287

Risk Ratio (M‐H, Fixed, 95% CI)

2.77 [1.91, 4.04]

2 Within 4 hours after 4 of the 7 doses Show forest plot

2

305

Risk Ratio (M‐H, Fixed, 95% CI)

9.98 [4.96, 20.09]

3 Within 4 hours of first dose Show forest plot

3

517

Risk Ratio (M‐H, Fixed, 95% CI)

3.87 [2.83, 5.28]

4 Within 4 hours after 1 or 2 doses of the first 4 doses Show forest plot

2

363

Risk Ratio (M‐H, Fixed, 95% CI)

6.89 [4.46, 10.66]

5 Improvement in constipation distress at day 1 Show forest plot

2

287

Risk Ratio (M‐H, Fixed, 95% CI)

1.87 [1.34, 2.59]

6 Participant global impression of improvement in bowel status at 1 week Show forest plot

2

287

Risk Ratio (M‐H, Fixed, 95% CI)

2.32 [1.64, 3.27]

7 Clinician global impression of improvement in bowel status at 1 week Show forest plot

2

287

Risk Ratio (M‐H, Fixed, 95% CI)

2.37 [1.66, 3.38]

Figuras y tablas -
Comparison 1. Methylnatrexone versus placebo: rescue‐free laxation:
Comparison 2. Methylnaltrexone versus placebo: serious adverse event

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse event Show forest plot

2

364

Risk Ratio (M‐H, Fixed, 95% CI)

0.59 [0.38, 0.93]

Figuras y tablas -
Comparison 2. Methylnaltrexone versus placebo: serious adverse event
Comparison 3. Methylnaltrexone versus placebo: adverse event

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse events Show forest plot

3

518

Risk Ratio (M‐H, Random, 95% CI)

1.17 [0.94, 1.45]

2 Dropouts due to adverse event Show forest plot

2

363

Risk Ratio (M‐H, Fixed, 95% CI)

1.22 [0.54, 2.76]

Figuras y tablas -
Comparison 3. Methylnaltrexone versus placebo: adverse event
Comparison 4. Oxycodone/naloxone prolonged‐release tablets versus oxycodone prolonged‐release: adverse event

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse events Show forest plot

2

234

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.94, 1.24]

Figuras y tablas -
Comparison 4. Oxycodone/naloxone prolonged‐release tablets versus oxycodone prolonged‐release: adverse event