Skip to main content

Advertisement

Log in

Insights into the Use of Peripherally Acting μ-Opioid Receptor Antagonists (PAMORAs) in Oncologic Patients: from Scientific Evidence to Real Clinical Practice

  • Palliative and Supportive Care (MP Davis, Section Editor)
  • Published:
Current Treatment Options in Oncology Aims and scope Submit manuscript

Opinion statement

Management of chronic pain is crucial to improve the quality of life of cancer and palliative care patients. Opioid-based treatments used to control pain can be prolonged over time. Unfortunately, constipation is one of the most disturbing adverse effects of long-term use of opioids. Opioid-induced constipation (OIC) occurs when opioids bind to the specific receptors present in the gastrointestinal (GI) tract, and can affect any patients receiving chronic opioid therapy, including cancer patients. The limited efficacy of laxatives to treat OIC symptoms prompted the search for new therapeutic strategies. Peripherally acting μ-opioid receptor antagonists (PAMORAs) have recently emerged as new effective drugs for OIC management due to their specific binding to enteric μ-receptors. Little information is available on the use of PAMORAs in real-life practice for OIC treatment in cancer patients. In this paper, a panel of experts specializing in cancer and palliative care pools their clinical experience with PAMORAs in cancer patients presenting OIC and highlights the importance of timing and choice of therapy in achieving prompt OIC management and benefitting patients.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Hughes PA, et al. Opioidergic effects on enteric and sensory nerves in the lower GI tract: basic mechanisms and clinical implications. Am J Physiol Gastrointest Liver Physiol. 2016;311(3):G501–13.

    Article  PubMed  Google Scholar 

  2. Azizi Z, Javid Anbardan S, Ebrahimi Daryani N. A review of the clinical manifestations, pathophysiology and management of opioid bowel dysfunction and narcotic bowel syndrome. Middle East J Dig Dis. 2014;6(1):5–12.

    PubMed  PubMed Central  Google Scholar 

  3. •• Larkin PJ, et al. Diagnosis, assessment and management of constipation in advanced cancer: ESMO Clinical Practice Guidelines. Ann Oncol. 2018;29(Suppl 4):iv111–25 ESMO Clinical Practice Guidelines that highlights the impact of constipation and discuss pharmacological and non-pharmacological options for management, as well as the challenges of treatments in older cancer patients at enhanced risk of constipation.

    Article  CAS  PubMed  Google Scholar 

  4. •• Farmer AD, et al. Pathophysiology and management of opioid-induced constipation: European expert consensus statement. United European Gastroenterol J. 2019;7(1):7–20 An European expert panel evaluates current evidence in the literature regarding pathophysiology and clinical evaluation of OIC, the importance of early OIC recognition and the need of combined therapies as well as provides a management algorithm for day-to-day clinical practice.

    Article  CAS  PubMed  Google Scholar 

  5. Bell TJ, et al. The prevalence, severity, and impact of opioid-induced bowel dysfunction: results of a US and European Patient Survey (PROBE 1). Pain Med. 2009;10(1):35–42.

    Article  PubMed  Google Scholar 

  6. Drossman DA. Functional Gastrointestinal Disorders: History, Pathophysiology, Clinical Features and Rome IV. Gastroenterology. 2016;S0016-5085(16):00223–7. https://doi.org/10.1053/j.gastro.2016.02.032.

  7. Drossman DA, Hasler WL. Rome IV-functional GI disorders: disorders of gut-brain interaction. Gastroenterology. 2016;150(6):1257–61.

    Article  PubMed  Google Scholar 

  8. Mearin F, et al. Bowel Disorders. Gastroenterology. 2016;S0016-5085(16):00222–5. https://doi.org/10.1053/j.gastro.2016.02.031.

  9. Crockett S, Greer KB, Sultan S. Opioid-induced constipation (OIC) Guideline. Gastroenterology. 2019;156(1):228.

    Article  PubMed  Google Scholar 

  10. • Davies A, et al. MASCC recommendations on the management of constipation in patients with advanced cancer. Support Care Cancer. 2020;28(1):23–33 The Palliative Care Study Group of the MASCC provide 15 recommendations with varying guideline categories for assessment, treatment and re-assessment of constipation in advanced cancer, with varying levels of evidence.

    Article  PubMed  Google Scholar 

  11. Christensen HN, et al. Opioid-induced constipation, use of laxatives, and health-related quality of life. Scand J Pain. 2016;11:104–10.

    Article  PubMed  Google Scholar 

  12. Emmanuel A, et al. Laxatives do not improve symptoms of opioid-induced constipation: results of a patient survey. Pain Med. 2017;18(10):1932–40.

    PubMed  Google Scholar 

  13. LoCasale RJ, et al. The impact of opioid-induced constipation among chronic pain patients with sufficient laxative use. Int J Clin Pract. 2015;69(12):1448–56.

    Article  CAS  PubMed  Google Scholar 

  14. Sandhu A, et al. Cardiovascular disorders associated with naloxone monotherapy and in fixed-dose combination with opioids: Data from international safety surveillance. Int J Cardiol. 2016;212:360–3.

    Article  PubMed  Google Scholar 

  15. Ahmedzai SH, et al. Long-term safety and efficacy of oxycodone/naloxone prolonged-release tablets in patients with moderate-to-severe chronic cancer pain. Support Care Cancer. 2015;23(3):823–30.

    Article  PubMed  Google Scholar 

  16. Smith K, et al. Naloxone as part of a prolonged release oxycodone/naloxone combination reduces oxycodone-induced slowing of gastrointestinal transit in healthy volunteers. Expert Opin Investig Drugs. 2011;20(4):427–39.

    Article  CAS  PubMed  Google Scholar 

  17. Kumar L, Barker C, Emmanuel A. Opioid-induced constipation: pathophysiology, clinical consequences, and management. Gastroenterol Res Pract. 2014;2014:141737.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Luthra P, et al. Efficacy of pharmacological therapies for the treatment of opioid-induced constipation: systematic review and network meta-analysis. Gut. 2019;68(3):434–44. https://doi.org/10.1136/gutjnl-2018-316001.

  19. •• Nishie K, et al. Peripherally acting mu-opioid antagonist for the treatment of opioid-induced constipation: Systematic review and meta-analysis. J Gastroenterol Hepatol. 2019;34(5):818–29 Systematic review of the efficacy of PAMORAs in patients with opioid-induced constipation.

    Article  CAS  PubMed  Google Scholar 

  20. Chey WD, et al. Naloxegol for opioid-induced constipation in patients with noncancer pain. N Engl J Med. 2014;370(25):2387–96.

    Article  PubMed  Google Scholar 

  21. Libran Oriol A, et al. Peripheral acting Mu opioid receptor antagonists in the treatment of the opioid-induced constipation: review. Rev Soc Esp Dolor. 2020;27(1):37–49.

    Google Scholar 

  22. Erowele GI. Alvimopan (Entereg), a Peripherally acting mu-opioid receptor antagonist for postoperative Ileus. P T. 2008;33(10):574–83.

    PubMed  PubMed Central  Google Scholar 

  23. Schmidt WK. Alvimopan* (ADL 8-2698) is a novel peripheral opioid antagonist. Am J Surg. 2001;182(5A Suppl):27S–38S.

    Article  CAS  PubMed  Google Scholar 

  24. Webster L, et al. Alvimopan, a peripherally acting mu-opioid receptor (PAM-OR) antagonist for the treatment of opioid-induced bowel dysfunction: results from a randomized, double-blind, placebo-controlled, dose-finding study in subjects taking opioids for chronic non-cancer pain. Pain. 2008;137(2):428–40.

    Article  CAS  PubMed  Google Scholar 

  25. • van Malderen K, Halawi H, Camilleri M. Insights on efficacious doses of PAMORAs for patients on chronic opioid therapy or opioid-naive patients. Neurogastroenterol Motil. 2018;30(5):e13250 This review evaluates the efficacious doses of PAMORAs for opioid-naïve or chronic opioid users.

    Article  PubMed  Google Scholar 

  26. Katakami N, et al. Randomized phase III and extension studies of naldemedine in patients with opioid-induced constipation and cancer. J Clin Oncol. 2017;35(34):3859–66.

    Article  CAS  PubMed  Google Scholar 

  27. Katakami N, et al. Phase IIb, randomized, double-blind, placebo-controlled study of naldemedine for the treatment of opioid-induced constipation in patients with cancer. J Clin Oncol. 2017;35(17):1921–8.

    Article  CAS  PubMed  Google Scholar 

  28. Baker DE. Formulary drug review: naldemedine. Hosp Pharm. 2017;52(7):464–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Markham A. Naldemedine: first global approval. Drugs. 2017;77(8):923–7.

    Article  PubMed  Google Scholar 

  30. Kanemasa T, et al. Profile of Naldemedine, a Peripherally Acting μ-Opioid Receptor Antagonist: Comparison with Naloxone and Naloxegol. J Pharmacol Exp Ther. 2020;373(3):438–44. https://doi.org/10.1124/jpet.119.264515.

  31. Webster L, et al. Randomised clinical trial: the long-term safety and tolerability of naloxegol in patients with pain and opioid-induced constipation. Aliment Pharmacol Ther. 2014;40(7):771–9.

    Article  CAS  PubMed  Google Scholar 

  32. Tack J, et al. Efficacy and safety of naloxegol in patients with opioid-induced constipation and laxative-inadequate response. United European Gastroenterol J. 2015;3(5):471–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Webster L, et al. A 12-week extension study to assess the safety and tolerability of naloxegol in patients with noncancer pain and opioid-induced constipation. J Opioid Manag. 2016;12(6):405–19.

    Article  PubMed  Google Scholar 

  34. Garnock-Jones KP. Naloxegol: a review of its use in patients with opioid-induced constipation. Drugs. 2015;75(4):419–25.

    Article  CAS  PubMed  Google Scholar 

  35. •• Dols MC, et al. Analysis of the efficacy and safety of naloxegol administered to patients with cancer and opioid-induced constipation with laxative-inadequate response: a real-world 12 months of follow-up study. J Clin Oncol. 2020;38(15_suppl):e24155–5 This real-world-data study confirms long-term efficacy, safety and tolerability of naloxegol in cancer patients with OIC maintaining pain control.

  36. •• Davies A, et al. Naloxegol for patients with cancer pain diagnosed with opioid induced constipation (OIC): NACASY observational study. J Clin Oncol. 2020;38:e19350–0 This real-world study evaluates safety and efficacy of naloxegol in patients with cancer pain diagnosed with OIC.

  37. Gottfridsson C, et al. Evaluation of the effect of Naloxegol on cardiac repolarization: a randomized, placebo- and positive-controlled crossover thorough QT/QTc study in healthy volunteers. Clin Ther. 2013;35(12):1876–83.

    Article  CAS  PubMed  Google Scholar 

  38. Eldon MA, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamic effects of naloxegol at peripheral and central nervous system receptors in healthy male subjects: a single ascending-dose study. Clin Pharmacol Drug Dev. 2015;4(6):434–41.

    Article  CAS  PubMed  Google Scholar 

  39. White WB, et al. Cardiovascular safety of the selective mu-opioid receptor antagonist naloxegol: a novel therapy for opioid-induced constipation. J Cardiovasc Pharmacol Ther. 2018;23(4):309–17.

    Article  CAS  PubMed  Google Scholar 

  40. Al-Huniti N, et al. Population pharmacokinetics of naloxegol in a population of 1247 healthy subjects and patients. Br J Clin Pharmacol. 2016;81(1):89–100.

    Article  CAS  PubMed  Google Scholar 

  41. •• Raffa RB, Taylor R Jr, Pergolizzi JV Jr. Treating opioid-induced constipation in patients taking other medications: Avoiding CYP450 drug interactions. J Clin Pharm Ther. 2019;44(3):361–71 This article reviews the potential for metabolic drug interactions of PAMORAs with other medications in patients treated for OIC and other comorbidities.

    Article  CAS  PubMed  Google Scholar 

  42. Moventing®(naloxegol), Full prescribing information. 2019 (last update), Bloemlaan 2, 2132NP Hoofddorp, The Netherlands: Kyowa Kirin Holdings B.V. [Last access January 28]: https://www.ema.europa.eu/en/documents/productinformation/moventig-epar-product-information_en.pdf.

  43. Rizmoic®(naldemedine), Full prescribing information. 2020 (last update), Kingsfordweg 151, 1043GR Amsterdan, The Netherlands: Shionogi B.V. [Last access January 28]: https://www.ema.europa.eu/en/documents/productinformation/rizmoic-epar-product-information_en.pdf

  44. •• Fallon M, et al. Management of cancer pain in adult patients: ESMO Clinical Practice Guidelines. Ann Oncol. 2018;29(Suppl 4):iv166–91 ESMO Clinical Practice Guidelines that recommends the use of laxatives for prevention and treatment of opioid-induced intestinal dysfunctions in oncological patients.

    Article  CAS  PubMed  Google Scholar 

  45. Relistor®(methylnaltrexone), Full prescribing information. 2019 (last update), Jankovcova 1569/2c,Lighthouse 17,000 Prague 7, Czech Republic: PharmaSwiss Ceska Republika s.r.o. [Last access January 28]: https://www.ema.europa.eu/en/documents/productinformation/relistor-epar-product-information_en.pdf.

  46. Schmulson MJ, Drossman DA. What Is New in Rome IV. J Neurogastroenterol Motil. 2017;23(2):151–63.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Szigethy E, Schwartz M, Drossman D. Narcotic bowel syndrome and opioid-induced constipation. Curr Gastroenterol Rep. 2014;16(10):410.

    Article  PubMed  Google Scholar 

  48. Muller-Lissner S, et al. Opioid-Induced Constipation and Bowel Dysfunction: A Clinical Guideline. Pain Med. 2017;18(10):1837–63.

    PubMed  Google Scholar 

  49. Gibson PR, Morrison G. Effects of methylnaltrexone in patients with narcotic bowel syndrome: a pilot observational study. Intern Med J. 2012;42(8):907–12.

    Article  CAS  PubMed  Google Scholar 

  50. Kurlander JE, Drossman DA. diagnosis and treatment of narcotic bowel syndrome. Nat Rev. Gastroenterol Hepatol. 2014;11(7):410–8.

    CAS  Google Scholar 

  51. Bui K, She F, Sostek M. The effects of mild or moderate hepatic impairment on the pharmacokinetics, safety, and tolerability of naloxegol. J Clin Pharmacol. 2014;54(12):1368–74.

    Article  CAS  PubMed  Google Scholar 

  52. Rotshteyn Y, Boyd TA, Yuan CS. Methylnaltrexone bromide: research update of pharmacokinetics following parenteral administration. Expert Opin Drug Metab Toxicol. 2011;7(2):227–35.

    Article  PubMed  Google Scholar 

  53. Fukumura K, et al. The Influence of Renal or Hepatic Impairment on the Pharmacokinetics, Safety, and Tolerability of Naldemedine. Clin Pharmacol Drug Dev. 2020;9(2):162–74. https://doi.org/10.1002/cpdd.690.

  54. Webster LR, et al. A renal impairment subgroup analysis of the safety and efficacy of naldemedine for the treatment of opioid-induced constipation in patients with chronic non-cancer pain receiving opioid therapy. J Pain Res. 2020;13:605–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Streicher JM, Bilsky EJ. Peripherally acting micro-opioid receptor antagonists for the treatment of opioid-related side effects: mechanism of action and clinical implications. J Pharm Pract. 2017:897190017732263.

  56. Osaka I, et al. Safety and efficacy of naldemedine in cancer patients with opioid-induced constipation: a pooled, subgroup analysis of two randomised controlled studies. ESMO Open. 2019;4(4):e000527. https://doi.org/10.1136/esmoopen-2019-000527.

  57. Bull J, et al. Fixed-dose subcutaneous methylnaltrexone in patients with advanced illness and opioid-induced constipation: results of a randomized, placebo-controlled study and open-label extension. J Palliat Med. 2015;18(7):593–600.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Thomas J, et al. Methylnaltrexone for opioid-induced constipation in advanced illness. N Engl J Med. 2008;358(22):2332–43.

    Article  CAS  PubMed  Google Scholar 

  59. Cobo Dols M, et al. Analysis of the efficacy of naloxegol in a real-world 12 weeks of follow-up study, in patients with cancer and opioid-induced constipation with laxativeinadequate response. Poster presented at: EFIC Congress 2019. Pain in Europe XI; September 4, 2019; Valencia, Spain. https://doi.org/10.26226/morressier.5d402fa18f2158d25ec13161.

  60. Webster LR, et al. Long-term use of naldemedine in the treatment of opioid-induced constipation in patients with chronic noncancer pain: a randomized, double-blind, placebocontrolled phase 3 study. Pain. 2018;159(5):987–94. https://doi.org/10.1097/j.pain.0000000000001174.

  61. Rao SS, Go JT. Update on the management of constipation in the elderly: new treatment options. Clin Interv Aging. 2010;5:163–71.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Lynch T. Management of drug-drug interactions: considerations for special populations--focus on opioid use in the elderly and long term care. Am J Manag Care. 2011;17(Suppl 11):S293–8.

    PubMed  Google Scholar 

  63. Martin CM, Forrester CS. Anticipating and managing opioid side effects in the elderly. Consult Pharm. 2013;28(3):150–9.

    Article  PubMed  Google Scholar 

  64. • Bui K, et al. Clinical pharmacokinetics and pharmacodynamics of naloxegol, a peripherally acting micro-opioid receptor antagonist. Clin Pharmacokinet. 2017;56(6):573–82 This article describes the pharmacokinetic/dynamic properties of naloxegol.

    Article  CAS  PubMed  Google Scholar 

  65. Wild J, et al. Safety and Efficacy of Naldemedine for the Treatment of Opioid-Induced Constipation in Patients with Chronic Non-Cancer Pain Receiving Opioid Therapy: A Subgroup Analysis of Patients ≥ 65 Years of Age. Drugs Aging. 2020;37(4):271–9. https://doi.org/10.1007/s40266-020-00753-2.

Download references

Acknowledgments

The authors thank Dr. Susana Cañón, (Medical Statistics Consulting, S.L., Valencia, Spain) for providing medical writing support in accordance with Good Publication Practice (GPP3) guidelines, which was funded by Kyowa Kirin Farmacéutica S.L.U. (Madrid, Spain).

Funding

The authors disclose receipt of the following financial support for the research, authorship, and publication of this article: Kyowa Kirin Farmacéutica S.L.U. supported this work but had no role in the preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ana Fernández-Montes MD.

Ethics declarations

Conflict of Interest

All authors have received compensation from Kyowa Kirin Farmacéutica S.L.U. for service as consultants. Ana Fernández-Montes has received grants from Kyowa Kirin outside the submitted work. José Luis-Fírvida has received compensation from Astra Zeneca, Bristol-Myers Squibb, Lilly, Roche, and Takeda for service as consultant, not related to the submitted work. Guillermo de Velasco has received grants/ personal fees from Pfizer, Roche, Ipsen, Astellas, Bayer, Bristol-Myers Squibb, Novartis, Janssen, Merck Sharp & Dohme, and Merck, outside the submitted work. Santiago Aguín has received personal personal fees from Merck, Merck Sharp & Dohme, and Bristol-Myers Squibb, outside the submitted work. José Luis Fírvida has received compensation from Astra Zeneca, Bristol Myers Squibb, Lilly, Merck Sharp & Dohme, Roche, and Takeda for service as consultant, not related to the submitted work. María Guirado-Risueño has received personal fees and non-financial support from Kyowa Kirin and Roche and personal fees from Merck Sharp & Dohme, Bristol-Myers Squibb, and Astra Zeneca, outside the submitted work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This Article is part of Topical Collection on Palliative and Supportive Care

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fernández-Montes, A., de Velasco, G., Aguín, S. et al. Insights into the Use of Peripherally Acting μ-Opioid Receptor Antagonists (PAMORAs) in Oncologic Patients: from Scientific Evidence to Real Clinical Practice. Curr. Treat. Options in Oncol. 22, 26 (2021). https://doi.org/10.1007/s11864-021-00816-5

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11864-021-00816-5

Keywords

Navigation