Skip to main content
Top
Published in: Clinical Pharmacokinetics 6/2009

01-06-2009 | Original Research Article

Morphine Glucuronidation in Preterm Neonates, Infants and Children Younger than 3 Years

Authors: Dr Catherijne A. J. Knibbe, Elke H. J. Krekels, Johannes N. van den Anker, Joost DeJongh, Gijs W. E. Santen, Monique van Dijk, Sinno H. P. Simons, Richard A. van Lingen, Evelyne M. Jacqz-Aigrain, Meindert Danhof, Dick Tibboel

Published in: Clinical Pharmacokinetics | Issue 6/2009

Login to get access

Abstract

Background and objective

A considerable amount of drug use in children is still unlicensed or off-label. In order to derive rational dosing schemes, the influence of aging on glucuronidation capacity in newborns, including preterms, infants and children under the age of 3 years was studied using morphine and its major metabolites as a model drug.

Methods

A population pharmacokinetic model was developed with the nonlinear mixed-effects modelling software NONMEM® V, on the basis of 2159 concentrations of morphine and its glucuronides from 248 infants receiving intravenous morphine ranging in bodyweight from 500 g to 18 kg (median 2.8 kg). The model was internally validated using normalized prediction distribution errors.

Results

Formation clearances of morphine to its glucuronides and elimination clearances of the glucuronides were found to be primarily influenced by bodyweight, which was parameterized using an allometric equation with an estimated exponential scaling factor of 1.44. Additionally, a postnatal age of less than 10 days was identified as a covariate for formation clearance to the glucuronides, independent of birthweight or postmenstrual age. Distribution volumes scaled linearly with bodyweight.

Conclusions

Model-based simulations show that in newborns, including preterms, infants and children under the age of 3 years, a loading dose in µg/kg and a maintenance dose expressed in µg/kg1.5/h, with a 50% reduction of the maintenance dose in newborns younger than 10 days, results in a narrow range of morphine and metabolite serum concentrations throughout the studied age range. Future pharmacodynamic investigations are needed to reveal target concentrations in this population, after which final dosing recommendations can be made.
Appendix
Available only for authorised users
Literature
1.
go back to reference Conroy S, McIntyre J, Choonara I. Unlicensed and off label drug use in neonates. Arch Dis Child Fetal Neonatal Ed 1999; 80(2): F142–4 Conroy S, McIntyre J, Choonara I. Unlicensed and off label drug use in neonates. Arch Dis Child Fetal Neonatal Ed 1999; 80(2): F142–4
2.
go back to reference Danhof M, de Jongh J, de Lange EC, et al. Mechanism-based pharmacokinetic-pharmacodynamic modeling: biophase distribution, receptor theory, and dynamical systems analysis. Annu Rev Pharmacol Toxicol 2007; 47: 357–400PubMedCrossRef Danhof M, de Jongh J, de Lange EC, et al. Mechanism-based pharmacokinetic-pharmacodynamic modeling: biophase distribution, receptor theory, and dynamical systems analysis. Annu Rev Pharmacol Toxicol 2007; 47: 357–400PubMedCrossRef
3.
go back to reference de Wildt SN, Kearns GL, Leeder JS, et al. Glucuronidation in humans: pharmacogenetic and developmental aspects. Clin Pharmacokinet 1999; 36(6): 439–52PubMedCrossRef de Wildt SN, Kearns GL, Leeder JS, et al. Glucuronidation in humans: pharmacogenetic and developmental aspects. Clin Pharmacokinet 1999; 36(6): 439–52PubMedCrossRef
4.
go back to reference McCarver DG, Hines RN. The ontogeny of human drug-metabolizing enzymes: phase II conjugation enzymes and regulatory mechanisms. J Pharmacol Exp Ther 2002; 300(2): 361–6PubMedCrossRef McCarver DG, Hines RN. The ontogeny of human drug-metabolizing enzymes: phase II conjugation enzymes and regulatory mechanisms. J Pharmacol Exp Ther 2002; 300(2): 361–6PubMedCrossRef
5.
go back to reference Coffman BL, Rios GR, King CD, et al. Human UGT2B7 catalyzes morphine glucuronidation. Drug Metab Dispos 1997; 25(1): 1–4PubMed Coffman BL, Rios GR, King CD, et al. Human UGT2B7 catalyzes morphine glucuronidation. Drug Metab Dispos 1997; 25(1): 1–4PubMed
6.
go back to reference Allegaert K, Rayyan M, de Hoon J, et al. Contribution of glucuronidation to tramadol disposition in early neonatal life. Basic Clin Pharmacol Toxicol 2006; 98(1): 110–2PubMedCrossRef Allegaert K, Rayyan M, de Hoon J, et al. Contribution of glucuronidation to tramadol disposition in early neonatal life. Basic Clin Pharmacol Toxicol 2006; 98(1): 110–2PubMedCrossRef
7.
go back to reference Court MH, Krishnaswamy S, Hao Q, et al. Evaluation of 3′-azido-3′-deoxy-thymidine, morphine, and codeine as probe substrates for UDP-glucur-onosyltransferase 2B7 (UGT2B7) in human liver microsomes: specificity and influence of the UGT2B7*2 polymorphism. Drug Metab Dispos 2003; 31(9): 1125–33PubMedCrossRef Court MH, Krishnaswamy S, Hao Q, et al. Evaluation of 3′-azido-3′-deoxy-thymidine, morphine, and codeine as probe substrates for UDP-glucur-onosyltransferase 2B7 (UGT2B7) in human liver microsomes: specificity and influence of the UGT2B7*2 polymorphism. Drug Metab Dispos 2003; 31(9): 1125–33PubMedCrossRef
8.
go back to reference Barbier O, Turgeon D, Girard C, et al. 3′-azido-3′-deoxythimidine (AZT) is glucuronidated by human UDP-glucuronosyltransferase 2B7 (UGT2B7). Drug Metab Dispos 2000; 28(5): 497–502PubMed Barbier O, Turgeon D, Girard C, et al. 3′-azido-3′-deoxythimidine (AZT) is glucuronidated by human UDP-glucuronosyltransferase 2B7 (UGT2B7). Drug Metab Dispos 2000; 28(5): 497–502PubMed
9.
go back to reference Simons SH, van Dijk M, Van Lingen RA, et al. Routine morphine infusion in preterm newborns who received ventilatory support: a randomized controlled trial. JAMA 2003; 290(18): 2419–27PubMedCrossRef Simons SH, van Dijk M, Van Lingen RA, et al. Routine morphine infusion in preterm newborns who received ventilatory support: a randomized controlled trial. JAMA 2003; 290(18): 2419–27PubMedCrossRef
10.
go back to reference van Dijk M, Bouwmeester NJ, Duivenvoorden HJ, et al. Efficacy of continuous versus intermittent morphine administration after major surgery in 0–3-year-old infants: a double-blind randomized controlled trial. Pain 2002; 98(3): 305–13PubMedCrossRef van Dijk M, Bouwmeester NJ, Duivenvoorden HJ, et al. Efficacy of continuous versus intermittent morphine administration after major surgery in 0–3-year-old infants: a double-blind randomized controlled trial. Pain 2002; 98(3): 305–13PubMedCrossRef
11.
go back to reference van Dijk M, de Boer JB, Koot HM, et al. The reliability and validity of the COMFORT scale as a postoperative pain instrument in 0 to 3-year-old infants. Pain 2000; 84(2–3): 367–77PubMedCrossRef van Dijk M, de Boer JB, Koot HM, et al. The reliability and validity of the COMFORT scale as a postoperative pain instrument in 0 to 3-year-old infants. Pain 2000; 84(2–3): 367–77PubMedCrossRef
12.
go back to reference Van der Marel CD, Peters JW, Bouwmeester NJ, et al. Rectal acetaminophen does not reduce morphine consumption after major surgery in young infants. Br J Anaesth 2007; 98(3): 372–9PubMedCrossRef Van der Marel CD, Peters JW, Bouwmeester NJ, et al. Rectal acetaminophen does not reduce morphine consumption after major surgery in young infants. Br J Anaesth 2007; 98(3): 372–9PubMedCrossRef
13.
go back to reference Brendel K, Comets E, Laffont C, et al. Metrics for external model evaluation with an application to the population pharmacokinetics of gliclazide. Pharm Res 2006; 23(9): 2036–49PubMedCrossRef Brendel K, Comets E, Laffont C, et al. Metrics for external model evaluation with an application to the population pharmacokinetics of gliclazide. Pharm Res 2006; 23(9): 2036–49PubMedCrossRef
14.
go back to reference Comets E, Brendel K, Mentre F. Computing normalised prediction distribution errors to evaluate nonlinear mixed-effect models: the NPDE add-on package for R. Comput Methods Programs Biomed 2008; 90(2): 154–66PubMedCrossRef Comets E, Brendel K, Mentre F. Computing normalised prediction distribution errors to evaluate nonlinear mixed-effect models: the NPDE add-on package for R. Comput Methods Programs Biomed 2008; 90(2): 154–66PubMedCrossRef
15.
go back to reference Brendel K, Dartois C, Comets E, et al. Are population pharmacokinetic and/or pharmacodynamic models adequately evaluated? A survey of the literature from 2002 to 2004. Clin Pharmacokinet 2007; 46(3): 221–34PubMedCrossRef Brendel K, Dartois C, Comets E, et al. Are population pharmacokinetic and/or pharmacodynamic models adequately evaluated? A survey of the literature from 2002 to 2004. Clin Pharmacokinet 2007; 46(3): 221–34PubMedCrossRef
16.
go back to reference Tod M, Jullien V, Pons G. Facilitation of drug evaluation in children by population methods and modelling. Clin Pharmacokinet 2008; 47(4): 231–43PubMedCrossRef Tod M, Jullien V, Pons G. Facilitation of drug evaluation in children by population methods and modelling. Clin Pharmacokinet 2008; 47(4): 231–43PubMedCrossRef
17.
go back to reference Alcorn J, McNamara PJ. Ontogeny of hepatic and renal systemic clearance pathways in infants: part I. Clin Pharmacokinet 2002; 41(12): 959–98PubMedCrossRef Alcorn J, McNamara PJ. Ontogeny of hepatic and renal systemic clearance pathways in infants: part I. Clin Pharmacokinet 2002; 41(12): 959–98PubMedCrossRef
18.
go back to reference Anand KJ, Anderson BJ, Holford NH, et al. Morphine pharmacokinetics and pharmacodynamics in preterm and term neonates: secondary results from the NEOPAIN trial. Br J Anaesth 2008; 101(5): 680–9PubMedCrossRef Anand KJ, Anderson BJ, Holford NH, et al. Morphine pharmacokinetics and pharmacodynamics in preterm and term neonates: secondary results from the NEOPAIN trial. Br J Anaesth 2008; 101(5): 680–9PubMedCrossRef
19.
go back to reference Bouwmeester NJ, Anderson BJ, Tibboel D, et al. Developmental pharmacokinetics of morphine and its metabolites in neonates, infants and young children. Br J Anaesth 2004; 92(2): 208–17PubMedCrossRef Bouwmeester NJ, Anderson BJ, Tibboel D, et al. Developmental pharmacokinetics of morphine and its metabolites in neonates, infants and young children. Br J Anaesth 2004; 92(2): 208–17PubMedCrossRef
20.
go back to reference Saarenmaa E, Neuvonen PJ, Rosenberg P, et al. Morphine clearance and effects in newborn infants in relation to gestational age. Clin Pharmacol Ther 2000; 68(2): 160–6PubMedCrossRef Saarenmaa E, Neuvonen PJ, Rosenberg P, et al. Morphine clearance and effects in newborn infants in relation to gestational age. Clin Pharmacol Ther 2000; 68(2): 160–6PubMedCrossRef
21.
go back to reference Scott CS, Riggs KW, Ling EW, et al. Morphine pharmacokinetics and pain assessment in premature newborns. J Pediatr 1999; 135(4): 423–9PubMedCrossRef Scott CS, Riggs KW, Ling EW, et al. Morphine pharmacokinetics and pain assessment in premature newborns. J Pediatr 1999; 135(4): 423–9PubMedCrossRef
22.
go back to reference Lynn A, Nespeca MK, Bratton SL, et al. Clearance of morphine in postoperative infants during intravenous infusion: the influence of age and surgery. Anesth Anals 1998; 86(5): 958–63 Lynn A, Nespeca MK, Bratton SL, et al. Clearance of morphine in postoperative infants during intravenous infusion: the influence of age and surgery. Anesth Anals 1998; 86(5): 958–63
23.
go back to reference Barrett DA, Barker DP, Rutter N, et al. Morphine, morphine-6-glucuronide and morphine-3-glucuronide pharmacokinetics in newborn infants receiving diamorphine infusions. Br J Clin Pharmacol 1996; 41(6): 531–7PubMedCrossRef Barrett DA, Barker DP, Rutter N, et al. Morphine, morphine-6-glucuronide and morphine-3-glucuronide pharmacokinetics in newborn infants receiving diamorphine infusions. Br J Clin Pharmacol 1996; 41(6): 531–7PubMedCrossRef
24.
go back to reference Mikkelsen S, Feilberg CL, Christensen CB, et al. Morphine pharmacokinetics in premature and mature newborn infants. Acta Paediatr 1994; 83(10): 1025–8PubMedCrossRef Mikkelsen S, Feilberg CL, Christensen CB, et al. Morphine pharmacokinetics in premature and mature newborn infants. Acta Paediatr 1994; 83(10): 1025–8PubMedCrossRef
25.
go back to reference Hartley R, Green M, Quinn M et al. Pharmacokinetics of morphine infusion in premature neonates. Arch Dis Child 1993; 69(1): 55–8PubMedCrossRef Hartley R, Green M, Quinn M et al. Pharmacokinetics of morphine infusion in premature neonates. Arch Dis Child 1993; 69(1): 55–8PubMedCrossRef
26.
go back to reference Pokela ML, Olkkola KT, Seppälä T, et al. Age-related morphine kinetics in infants. Dev Pharmacol Ther 1993; 20(1–2): 26–34PubMed Pokela ML, Olkkola KT, Seppälä T, et al. Age-related morphine kinetics in infants. Dev Pharmacol Ther 1993; 20(1–2): 26–34PubMed
27.
go back to reference McRorie TI, Lynn AM, Nespeca MK, et al. The maturation of morphine clearance and metabolism. Am J Dis Child 1992; 146(8): 972–6PubMed McRorie TI, Lynn AM, Nespeca MK, et al. The maturation of morphine clearance and metabolism. Am J Dis Child 1992; 146(8): 972–6PubMed
28.
go back to reference Choonara I, Lawrence A, Michalkiewicz A, et al. Morphine metabolism in neonates and infants. Br J Clin Pharmacol 1992; 34(5): 434–7PubMedCrossRef Choonara I, Lawrence A, Michalkiewicz A, et al. Morphine metabolism in neonates and infants. Br J Clin Pharmacol 1992; 34(5): 434–7PubMedCrossRef
29.
go back to reference Chay PC, Duffy BJ, Walker JS. Pharmacokinetic-pharmacodynamic relationships of morphine in neonates. Clin Pharmacol Ther 1992; 51(3): 334–42PubMedCrossRef Chay PC, Duffy BJ, Walker JS. Pharmacokinetic-pharmacodynamic relationships of morphine in neonates. Clin Pharmacol Ther 1992; 51(3): 334–42PubMedCrossRef
30.
go back to reference Choonara I, Ekbom Y, Lindstrom B, et al. Morphine sulphation in children. Br J Clin Pharmacol 1990; 30(6): 897–900PubMedCrossRef Choonara I, Ekbom Y, Lindstrom B, et al. Morphine sulphation in children. Br J Clin Pharmacol 1990; 30(6): 897–900PubMedCrossRef
31.
go back to reference Olkkola KT, Maunuksela EL, Korpela R, et al. Kinetics and dynamics of postoperative intravenous morphine in children. Clin Pharmacol Ther 1988; 44(2): 128–36PubMedCrossRef Olkkola KT, Maunuksela EL, Korpela R, et al. Kinetics and dynamics of postoperative intravenous morphine in children. Clin Pharmacol Ther 1988; 44(2): 128–36PubMedCrossRef
32.
33.
go back to reference Anderson BJ, Allegaert K, Holford NH. Population clinical pharmacology of children: modelling covariate effects. Eur J Pediatr 2006; 165(12): 819–29PubMedCrossRef Anderson BJ, Allegaert K, Holford NH. Population clinical pharmacology of children: modelling covariate effects. Eur J Pediatr 2006; 165(12): 819–29PubMedCrossRef
34.
go back to reference Anderson BJ, Holford NH. Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol 2008; 48: 303–32PubMedCrossRef Anderson BJ, Holford NH. Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol 2008; 48: 303–32PubMedCrossRef
35.
go back to reference Coffman BL, King CD, Rios GR, et al. The glucuronidation of opioids, other xenobiotics, and androgens by human UGT2B7Y(268) and UGT2B7H(268). Drug Metab Dispos 1998; 26(1): 73–7PubMed Coffman BL, King CD, Rios GR, et al. The glucuronidation of opioids, other xenobiotics, and androgens by human UGT2B7Y(268) and UGT2B7H(268). Drug Metab Dispos 1998; 26(1): 73–7PubMed
36.
go back to reference Ohno S, Kawana K, Nakajin S. Contribution of UDP-glucuronosyltransferase 1A1 and 1A8 to morphine-6-glucuronidation and its kinetic properties. Drug Metab Dispos 2008; 36(4): 688–94PubMedCrossRef Ohno S, Kawana K, Nakajin S. Contribution of UDP-glucuronosyltransferase 1A1 and 1A8 to morphine-6-glucuronidation and its kinetic properties. Drug Metab Dispos 2008; 36(4): 688–94PubMedCrossRef
37.
go back to reference Stone AN, Mackenzie PI, Galetin A, et al. Isoform selectivity and kinetics of morphine 3- and 6-glucuronidation by human udp-glucuronosyltransferases: evidence for atypical glucuronidation kinetics by UGT2B7. Drug Metab Dispos 2003; 31(9): 1086–9PubMedCrossRef Stone AN, Mackenzie PI, Galetin A, et al. Isoform selectivity and kinetics of morphine 3- and 6-glucuronidation by human udp-glucuronosyltransferases: evidence for atypical glucuronidation kinetics by UGT2B7. Drug Metab Dispos 2003; 31(9): 1086–9PubMedCrossRef
38.
39.
go back to reference Capparelli EV, Englund JA, Connor JD, et al. Population pharmacokinetics and pharmacodynamics of zidovudine in HIV-infected infants and children. J Clin Pharmacol 2003; 43(2): 133–40PubMedCrossRef Capparelli EV, Englund JA, Connor JD, et al. Population pharmacokinetics and pharmacodynamics of zidovudine in HIV-infected infants and children. J Clin Pharmacol 2003; 43(2): 133–40PubMedCrossRef
40.
go back to reference Capparelli EV, Mirochnick M, Dankner WM, et al. Pharmacokinetics and tolerance of zidovudine in preterm infants. J Pediatr 2003; 142(1): 47–52PubMedCrossRef Capparelli EV, Mirochnick M, Dankner WM, et al. Pharmacokinetics and tolerance of zidovudine in preterm infants. J Pediatr 2003; 142(1): 47–52PubMedCrossRef
41.
go back to reference Osborne R, Thompson P, Joel S, et al. The analgesic activity of morphine-6-glucuronide. Br J Clin Pharmacol 1992; 34(2): 130–8PubMedCrossRef Osborne R, Thompson P, Joel S, et al. The analgesic activity of morphine-6-glucuronide. Br J Clin Pharmacol 1992; 34(2): 130–8PubMedCrossRef
42.
go back to reference Faura CC, Collins SL, Moore RA, et al. Systematic review of factors affecting the ratios of morphine and its major metabolites. Pain 1998; 74(1): 43–53PubMedCrossRef Faura CC, Collins SL, Moore RA, et al. Systematic review of factors affecting the ratios of morphine and its major metabolites. Pain 1998; 74(1): 43–53PubMedCrossRef
43.
go back to reference Murthy BR, Pollack GM, Brouwer KL. Contribution of morphine-6-glucuronide to antinociception following intravenous administration of morphine to healthy volunteers. J Clin Pharmacol 2002; 42(5): 569–76PubMedCrossRef Murthy BR, Pollack GM, Brouwer KL. Contribution of morphine-6-glucuronide to antinociception following intravenous administration of morphine to healthy volunteers. J Clin Pharmacol 2002; 42(5): 569–76PubMedCrossRef
44.
go back to reference Smith MT, Watt JA, Cramond T. Morphine-3-glucuronide: a potent antagonist of morphine analgesia. Life Sci 1990; 47(6): 579–85PubMedCrossRef Smith MT, Watt JA, Cramond T. Morphine-3-glucuronide: a potent antagonist of morphine analgesia. Life Sci 1990; 47(6): 579–85PubMedCrossRef
45.
go back to reference Gong QL, Hedner T, Hedner J, et al. Antinociceptive and ventilatory effects of the morphine metabolites: morphine-6-glucuronide and morphine-3-glucuronide. Eur J Pharmacol 1991; 193(1): 47–56PubMedCrossRef Gong QL, Hedner T, Hedner J, et al. Antinociceptive and ventilatory effects of the morphine metabolites: morphine-6-glucuronide and morphine-3-glucuronide. Eur J Pharmacol 1991; 193(1): 47–56PubMedCrossRef
Metadata
Title
Morphine Glucuronidation in Preterm Neonates, Infants and Children Younger than 3 Years
Authors
Dr Catherijne A. J. Knibbe
Elke H. J. Krekels
Johannes N. van den Anker
Joost DeJongh
Gijs W. E. Santen
Monique van Dijk
Sinno H. P. Simons
Richard A. van Lingen
Evelyne M. Jacqz-Aigrain
Meindert Danhof
Dick Tibboel
Publication date
01-06-2009
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 6/2009
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.2165/00003088-200948060-00003

Other articles of this Issue 6/2009

Clinical Pharmacokinetics 6/2009 Go to the issue