Skip to main content
Erschienen in: Clinical Pharmacokinetics 6/2017

29.12.2016 | Review Article

Clinical Pharmacokinetics and Pharmacodynamics of Naloxegol, a Peripherally Acting µ-Opioid Receptor Antagonist

verfasst von: Khanh Bui, Diansong Zhou, Hongmei Xu, Eike Floettmann, Nidal Al-Huniti

Erschienen in: Clinical Pharmacokinetics | Ausgabe 6/2017

Einloggen, um Zugang zu erhalten

Abstract

Naloxegol is a peripherally acting µ-opioid receptor antagonist approved for use as an orally administered tablet (therapeutic doses of 12.5 and 25 mg) for the treatment of opioid-induced constipation. Over a wide dose range (i.e. single supratherapeutic doses up to 1000 mg in healthy volunteers), the pharmacokinetic properties of naloxegol appear to be time- and dose-independent. Naloxegol is rapidly absorbed, with mean time to maximum plasma concentration of <2 h. Following once-daily administration, steady state is achieved within 2–3 days and minimal accumulation is observed. The primary route of naloxegol elimination is via hepatic metabolism, with renal excretion playing a minimal role. In clinical studies, six metabolites were found in feces, urine or plasma, none of which have been identified as unique or disproportionate human metabolites. The major plasma circulating species is naloxegol. There are small effects of mild and moderate renal impairment, age, race, and body mass index on the systemic exposure of naloxegol; however, gender has no effect on the pharmacokinetics of this agent. Naloxegol is a sensitive substrate of cytochrome P450 (CYP) 3A4 and its exposure can be significantly altered by strong or moderate CYP3A modulators. Food increases the bioavailability of naloxegol, and the relative bioavailability of the tablet formulation was not limited by dissolution. Naloxegol in the dose range of 8–125 mg can antagonize morphine-induced peripheral effects without impacting the effect of morphine on the central nervous system, consistent with a peripheral mode of action.
Literatur
2.
Zurück zum Zitat Chou R, Fanciullo GJ, Fine PG, et al. Clinical guidelines for the use of chronic opioid therapy in chronic noncancer pain. J Pain. 2009;10(2):113–30.CrossRefPubMedPubMedCentral Chou R, Fanciullo GJ, Fine PG, et al. Clinical guidelines for the use of chronic opioid therapy in chronic noncancer pain. J Pain. 2009;10(2):113–30.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Pappagallo M. Incidence, prevalence and management of opioid bowel dysfunction. Am J Surg. 2001;182:11s–8s.CrossRefPubMed Pappagallo M. Incidence, prevalence and management of opioid bowel dysfunction. Am J Surg. 2001;182:11s–8s.CrossRefPubMed
4.
Zurück zum Zitat Panchal SJ, Muller-Schwefe P, Wurzelmann JI. Opioid-induced bowel dysfunction: prevalence, pathophysiology and burden. Int J Clin Pract. 2007;61(7):1181–7.CrossRefPubMedPubMedCentral Panchal SJ, Muller-Schwefe P, Wurzelmann JI. Opioid-induced bowel dysfunction: prevalence, pathophysiology and burden. Int J Clin Pract. 2007;61(7):1181–7.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Camilleri M. Opioid-induced constipation: challenges and therapeutic opportunities. Am J Gastroenterol. 2011;106:835–42.CrossRefPubMed Camilleri M. Opioid-induced constipation: challenges and therapeutic opportunities. Am J Gastroenterol. 2011;106:835–42.CrossRefPubMed
6.
Zurück zum Zitat American Society of Pain Educators. Pocket guide: opioid induced constipation. International Guidelines Center; 2010. American Society of Pain Educators. Pocket guide: opioid induced constipation. International Guidelines Center; 2010.
7.
Zurück zum Zitat Sobczak M, Sałaga M, Storr MA, Fichna J. Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract: current concepts and future perspectives. J Gastroenterol. 2014;49(1):24–45.CrossRefPubMed Sobczak M, Sałaga M, Storr MA, Fichna J. Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract: current concepts and future perspectives. J Gastroenterol. 2014;49(1):24–45.CrossRefPubMed
8.
Zurück zum Zitat Bell TJ, Panchal SJ, Miaskowski C, 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.CrossRefPubMed Bell TJ, Panchal SJ, Miaskowski C, 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.CrossRefPubMed
9.
Zurück zum Zitat Drewes AM, Munkholm P, Simrén M, et al. Definition, diagnosis and treatment strategies for opioid-induced bowel dysfunction: recommendations of the Nordic Working Group. Scand J Pain. 2016;11:111–22.CrossRef Drewes AM, Munkholm P, Simrén M, et al. Definition, diagnosis and treatment strategies for opioid-induced bowel dysfunction: recommendations of the Nordic Working Group. Scand J Pain. 2016;11:111–22.CrossRef
10.
Zurück zum Zitat Coyne KS, LoCasale RJ, Datto CJ, et al. 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. Clinicoecon Outcomes Res. 2014;6:269–81.CrossRefPubMedPubMedCentral Coyne KS, LoCasale RJ, Datto CJ, et al. 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. Clinicoecon Outcomes Res. 2014;6:269–81.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Christensen HN, Olsson U, From J, Breivik H. Opioid-induced constipation, use of laxatives, and health-related quality of life. Scand J Pain. 2016;11:104–10.CrossRef Christensen HN, Olsson U, From J, Breivik H. Opioid-induced constipation, use of laxatives, and health-related quality of life. Scand J Pain. 2016;11:104–10.CrossRef
12.
Zurück zum Zitat Movantik™ (naloxegol): full prescribing information. Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2015. Movantik™ (naloxegol): full prescribing information. Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2015.
13.
Zurück zum Zitat Moventig® (naloxegol): full prescribing information. Södertälje: AstraZeneca AB; 2014. Moventig® (naloxegol): full prescribing information. Södertälje: AstraZeneca AB; 2014.
14.
Zurück zum Zitat Tack J, Cimen A, Bui K, Sostek M. Naloxegol for opioid-induced constipation: mechanism of action and clinical implications [abstract]. United Eur Gastroenterol J. 2015;3(5 Suppl):A20. Tack J, Cimen A, Bui K, Sostek M. Naloxegol for opioid-induced constipation: mechanism of action and clinical implications [abstract]. United Eur Gastroenterol J. 2015;3(5 Suppl):A20.
15.
Zurück zum Zitat Bui K, Birmingham B, Ulysses Diva U, Berger B. An open-label, randomized, bioavailability study of alternative methods of oral administration of naloxegol in healthy subjects. Clin Pharmacol Drug Dev. 2016 (accepted). Bui K, Birmingham B, Ulysses Diva U, Berger B. An open-label, randomized, bioavailability study of alternative methods of oral administration of naloxegol in healthy subjects. Clin Pharmacol Drug Dev. 2016 (accepted).
16.
Zurück zum Zitat Data on file. Wilmington. DE: AstraZeneca; 2011. Data on file. Wilmington. DE: AstraZeneca; 2011.
17.
Zurück zum Zitat Eldon MA, Song D, Neumann TA, Wolff R, Cheng L, Viegas TX, et al. NKTR-118 (oral PEG-naloxol), a PEGylated derivative of naloxone: demonstration of selective peripheral opioid antagonism after oral administration in preclinical models. American Academy of Pain Management 18thAnnual Clinical Meeting; 27–30 Sep 2007; Las Vegas, NV. Eldon MA, Song D, Neumann TA, Wolff R, Cheng L, Viegas TX, et al. NKTR-118 (oral PEG-naloxol), a PEGylated derivative of naloxone: demonstration of selective peripheral opioid antagonism after oral administration in preclinical models. American Academy of Pain Management 18thAnnual Clinical Meeting; 27–30 Sep 2007; Las Vegas, NV.
18.
Zurück zum Zitat Faassen F, Vogel G, Spanings H, Vromans H. Caco-2 permeability, P-glycoprotein transport ratios and brain penetration of heterocyclic drugs. Int J Pharm. 2003;263:113–22.CrossRefPubMed Faassen F, Vogel G, Spanings H, Vromans H. Caco-2 permeability, P-glycoprotein transport ratios and brain penetration of heterocyclic drugs. Int J Pharm. 2003;263:113–22.CrossRefPubMed
19.
Zurück zum Zitat Al-Huniti N, Chapel S, Xu H, Bui KH, Sostek M. Population pharmacokinetics of naloxegol in a population of 1247 healthy subjects and patients. Br J Clin Pharmacol. 2016;81:89–100.CrossRefPubMed Al-Huniti N, Chapel S, Xu H, Bui KH, Sostek M. Population pharmacokinetics of naloxegol in a population of 1247 healthy subjects and patients. Br J Clin Pharmacol. 2016;81:89–100.CrossRefPubMed
20.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
21.
Zurück zum Zitat Bui K, She F, Zhou D, Butler K, Al-Huniti N, Sostek M. The effect of quinidine, a strong P-glycoprotein inhibitor, on the pharmacokinetics and central nervous system distribution of naloxegol. J Clin Pharmacol. 2015;56(4):497–505.CrossRefPubMed Bui K, She F, Zhou D, Butler K, Al-Huniti N, Sostek M. The effect of quinidine, a strong P-glycoprotein inhibitor, on the pharmacokinetics and central nervous system distribution of naloxegol. J Clin Pharmacol. 2015;56(4):497–505.CrossRefPubMed
22.
Zurück zum Zitat Bui K, She F, Hutchison M, Brunnström Å, Sostek M. Absorption, distribution, metabolism and excretion of [14C]-labeled naloxegol in healthy subjects. Int J Clin Pharmacol Ther. 2015;53:838–46.CrossRefPubMedPubMedCentral Bui K, She F, Hutchison M, Brunnström Å, Sostek M. Absorption, distribution, metabolism and excretion of [14C]-labeled naloxegol in healthy subjects. Int J Clin Pharmacol Ther. 2015;53:838–46.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Eldon MA, Kugler AR, Medve RA, 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.CrossRefPubMed Eldon MA, Kugler AR, Medve RA, 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.CrossRefPubMed
24.
Zurück zum Zitat Eldon MA, Kugler AR, Medve RA, et al. Safety, tolerability, and pharmacokinetics of multiple ascending doses of naloxegol. Clin Pharmacol Drug Dev. 2015;4(6):442–8.CrossRefPubMed Eldon MA, Kugler AR, Medve RA, et al. Safety, tolerability, and pharmacokinetics of multiple ascending doses of naloxegol. Clin Pharmacol Drug Dev. 2015;4(6):442–8.CrossRefPubMed
25.
Zurück zum Zitat 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(9):1542–50.CrossRefPubMed 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(9):1542–50.CrossRefPubMed
26.
Zurück zum Zitat Data on file. Wilmington. DE: AstraZeneca; 2013. Data on file. Wilmington. DE: AstraZeneca; 2013.
27.
Zurück zum Zitat Bui K, She F, Sostek M. The effects of renal impairment on the pharmacokinetics, safety, and tolerability of naloxegol. J Clin Pharmacol. 2014;54:1375–82.CrossRefPubMed Bui K, She F, Sostek M. The effects of renal impairment on the pharmacokinetics, safety, and tolerability of naloxegol. J Clin Pharmacol. 2014;54:1375–82.CrossRefPubMed
28.
Zurück zum Zitat Nolin TD, Naud J, Leblond FA, Pichette V. Emerging evidence of the impact of kidney disease on drug metabolism and transport. Clin Pharmacol Ther. 2008;83:898–903.CrossRefPubMed Nolin TD, Naud J, Leblond FA, Pichette V. Emerging evidence of the impact of kidney disease on drug metabolism and transport. Clin Pharmacol Ther. 2008;83:898–903.CrossRefPubMed
29.
Zurück zum Zitat Bui K, Zhou D, Sostek M, She F, Al-Huniti N. Effects of CYP3A modulators on the pharmacokinetics of naloxegol. J Clin Pharmacol. 2016;56(8):1019–27.CrossRefPubMed Bui K, Zhou D, Sostek M, She F, Al-Huniti N. Effects of CYP3A modulators on the pharmacokinetics of naloxegol. J Clin Pharmacol. 2016;56(8):1019–27.CrossRefPubMed
30.
Zurück zum Zitat Zhou D, Bui K, Sostek M, Al-Huniti N. Simulation and prediction of the drug-drug interaction potential of naloxegol by physiologically based pharmacokinetic modeling. CPT Pharmacomet Syst Pharmacol. 2016;5(5):250–7.CrossRef Zhou D, Bui K, Sostek M, Al-Huniti N. Simulation and prediction of the drug-drug interaction potential of naloxegol by physiologically based pharmacokinetic modeling. CPT Pharmacomet Syst Pharmacol. 2016;5(5):250–7.CrossRef
32.
Zurück zum Zitat Chey WD, Webster L, Sostek M, Lappalainen J, Barker PN, Tack J. Naloxegol for opioid-induced constipation in patients with noncancer pain. N Engl J Med. 2014;370(25):2387–96.CrossRefPubMed Chey WD, Webster L, Sostek M, Lappalainen J, Barker PN, Tack J. Naloxegol for opioid-induced constipation in patients with noncancer pain. N Engl J Med. 2014;370(25):2387–96.CrossRefPubMed
33.
Zurück zum Zitat Al-Huniti N, Nielsen JC, Hutmacher MM, Lappalainen J, Cantagallo K, Sostek M. Population exposure-response modeling of naloxegol in patients with non-cancer-related pain and opioid-induced constipation. CPT Pharmacomet Syst Pharmacol. 2016;5(7):359–66.CrossRef Al-Huniti N, Nielsen JC, Hutmacher MM, Lappalainen J, Cantagallo K, Sostek M. Population exposure-response modeling of naloxegol in patients with non-cancer-related pain and opioid-induced constipation. CPT Pharmacomet Syst Pharmacol. 2016;5(7):359–66.CrossRef
34.
Zurück zum Zitat Gottfridsson C, Carlson G, Lappalainen J, Sostek M. 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.CrossRefPubMed Gottfridsson C, Carlson G, Lappalainen J, Sostek M. 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.CrossRefPubMed
35.
Zurück zum Zitat Webster L, Chey WD, Tack J, Lappalainen J, Diva U, Sostek M. 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.CrossRefPubMed Webster L, Chey WD, Tack J, Lappalainen J, Diva U, Sostek M. 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.CrossRefPubMed
Metadaten
Titel
Clinical Pharmacokinetics and Pharmacodynamics of Naloxegol, a Peripherally Acting µ-Opioid Receptor Antagonist
verfasst von
Khanh Bui
Diansong Zhou
Hongmei Xu
Eike Floettmann
Nidal Al-Huniti
Publikationsdatum
29.12.2016
Verlag
Springer International Publishing
Erschienen in
Clinical Pharmacokinetics / Ausgabe 6/2017
Print ISSN: 0312-5963
Elektronische ISSN: 1179-1926
DOI
https://doi.org/10.1007/s40262-016-0479-z

Weitere Artikel der Ausgabe 6/2017

Clinical Pharmacokinetics 6/2017 Zur Ausgabe