Pharmacometric dose optimization of buprenorphine in neonatal opioid withdrawal syndrome

Rena Eudy-Byrne, Nicole Zane, Susan C Adeniyi-Jones, Marc R Gastonguay, Ana Ruiz-Garcia, Gagan Kaushal, Walter K Kraft, Rena Eudy-Byrne, Nicole Zane, Susan C Adeniyi-Jones, Marc R Gastonguay, Ana Ruiz-Garcia, Gagan Kaushal, Walter K Kraft

Abstract

Results from Blinded Buprenorphine OR Neonatal morphine solution (BBORN), a previous phase III trial in infants with neonatal opioid withdrawal syndrome (NOWS), demonstrated that sublingual buprenorphine resulted in a shorter duration of treatment and shorter length of hospital stay than the comparator, oral morphine. Objectives of Buprenorphine Pharmacometric Open Label Research study of Drug Exposure (BPHORE), a new trial with buprenorphine in a similar population, were to (1) optimize initial dose, up-titration to achieve symptom control and weaning steps of pharmacologic treatment and (2) investigate safety of the revised regimen. A pharmacodynamic model linked buprenorphine exposure to NOWS symptom scores. Adaptive dose regimens were simulated using BBORN results to compare dosing regimens for times to stabilization, weaning, and cessation. A clinical trial using model informed doses (BPHORE), was conducted. Simulations indicated benefits in time to stabilization and weaning when up-titration rates increased to 30%. Stabilization time was not greatly impacted by the starting dose. Time to wean and time to cessation were dose dependent. A weaning rate of 25% shortened time to cessation. Ten infants were enrolled in BPHORE using buprenorphine starting dose of 24 µg/kg/day, 33% titration, and 15% wean rate. Five subjects required adjuvant therapy. Half-maximal effective concentration (EC50 ) values indicated maximum buprenorphine doses did not generate maximal effect size, suggesting potential efficacy of a further increased dose if a goal was to reduce the use of adjunct agents. Simulations indicated that further benefits can be gained by increasing starting doses of buprenorphine and increasing wean rates. Use of a model-based analysis to provide focused guidelines for care can be used with goals of reducing treatment time and hospital stays in infants with NOWS.

Conflict of interest statement

The authors declared no competing interests for this work.

© 2021 The Authors. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

FIGURE 1
FIGURE 1
Dosing algorithm used in Blinded Buprenorphine OR Neonatal morphine solution (BBORN) and Buprenorphine Pharmacometric Open Label Research study of Drug Exposure (BPHORE) trials
FIGURE 2
FIGURE 2
Clinical data used in model‐based analysis for BBORN (Blinded Buprenorphine OR Neonatal morphine solution) and BPHORE (Buprenorphine Pharmacometric Open Label Research study of Drug Exposure) studies
FIGURE 3
FIGURE 3
Graphical depiction of endpoints investigated in adaptive dose simulations. Time to stabilization (TTS) is the time from first dose until maximum dose was reached. Time to wean (TTW) is the time from first dose until weaning (weaning was defined as down titration as a result of the sum of the 3 previous MOTHER neonatal abstinence syndrome (NAS) scores being less than 18 and no up‐titrations within 48 h) and can only be reached if stabilization is reached, first. Time to cessation (TTC) is the time from start of weaning until dose was within 10% of initial dose and can only be reached if time to wean ()TTW is reached, first
FIGURE 4
FIGURE 4
Kaplan‐Meir plots for simulated time to event time to stabilization (TTS) are shown at different initial dose levels and at a 25% up‐titration rate. + denotes censoring. The table below the plot summarizes the estimated percentage of patients who have not reached stabilization stratified by starting dose
FIGURE 5
FIGURE 5
Kaplan‐Meir plots for the simulated time to event time to wean (TTW) are shown at different initial dose levels and at a 25% up‐titration rate. “+” denotes censoring. The table below the plot summarizes the estimated percentage of patients who have not reached weaning stratified by starting dose
FIGURE 6
FIGURE 6
Kaplan‐Meir plots for the simulated time to event time to cessation (TTC) are shown at different dose levels and at a 10% weaning rate. “+” denotes censoring. The table below the plot summarizes the estimated percentage of patients who have not reached cessation stratified by starting dose

References

    1. Jones HE, Kraft WK. Analgesia, opioids, and other drug use during pregnancy and neonatal abstinence syndrome. Clin Perinatol. 2019;46:349‐366.
    1. Patrick SW, Barfield WD, Poindexter BB. Neonatal opioid withdrawal syndrome. Pediatrics. 2020;146(5):e2020029074.
    1. Disher T, Gullickson C, Singh B, et al. Pharmacological treatments for neonatal abstinence syndrome: a systematic review and network meta‐analysis. JAMA Pediatr. 2019;173:234‐243.
    1. Kraft WK, Adeniyi‐Jones SC, Chervoneva I, et al. Buprenorphine for the treatment of the neonatal abstinence syndrome. N Engl J Med. 2017;376:2341‐2348.
    1. Hall ES, Rice WR, Folger AT, Wexelblatt SL. Comparison of neonatal abstinence syndrome treatment with sublingual buprenorphine versus conventional opioids. Am J Perinatol. 2018;35(4):405‐412.
    1. Kraft WK, Gibson E, Dysart K, et al. Sublingual buprenorphine for treatment of neonatal abstinence syndrome: a randomized trial. Pediatrics. 2008;122:e601‐e607.
    1. Barrett DA, Barker DP, Rutter N, Pawula M, Shaw PN. Morphine, morphine‐6‐glucuronide and morphine‐3‐glucuronide pharmacokinetics in newborn infants receiving diamorphine infusions. Br J Clin Pharmacol. 1996;41:531‐537.
    1. Kraft WK, Dysart K, Greenspan JS, Gibson E, Kaltenbach K, Ehrlich ME. Revised dose schema of sublingual buprenorphine in the treatment of the neonatal opioid abstinence syndrome. Addiction. 2011;106:574‐580.
    1. Moore JN, Gastonguay MR, Ng CM, et al. The pharmacokinetics and pharmacodynamics of buprenorphine in neonatal abstinence syndrome. Clin Pharmacol Ther. 2018;103:1029‐1037.
    1. Ng CM, Dombrowsky E, Lin H, et al. Population pharmacokinetic model of sublingual buprenorphine in neonatal abstinence syndrome. Pharmacotherapy. 2015;35:670‐680.
    1. R Development Core Team . R: A Language and Environment for Statistical Computing. San Francisco, CA: R Software; 2008.
    1. Moody DE, Slawson MH, Strain EC, Laycock JD, Spanbauer AC, Foltz RL. A liquid chromatographic‐electrospray ionization‐tandem mass spectrometric method for determination of buprenorphine, its metabolite, norbuprenorphine, and a coformulant, naloxone, that is suitable for in vivo and in vitro metabolism studies. Anal Biochem. 2002;306:31‐39.
    1. Elmokadem A, Riggs MM, Baron KT. Quantitative systems pharmacology and physiologically‐based pharmacokinetic modeling with mrgsolve: a hands‐on tutorial. CPT Pharmacometrics Syst Pharmacol. 2019;8:883‐893.
    1. Therneau T. A package for survival analysis in R. 2021.
    1. Liu T, Lewis T, Gauda E, Gobburu J, Ivaturi V. Mechanistic population pharmacokinetics of morphine in neonates with abstinence syndrome after oral administration of diluted tincture of opium. J Clin Pharmacol. 2016;56:1009‐1018.
    1. Wiles JR, Isemann B, Mizuno T, et al. Pharmacokinetics of oral methadone in the treatment of neonatal abstinence syndrome: a pilot study. J Pediatr. 2015;167:1214‐20.e3.
    1. Mizuno T, McPhail BT, Kamatkar S, et al. Physiologic indirect response modeling to describe buprenorphine pharmacodynamics in newborns treated for neonatal opioid withdrawal syndrome. Clin Pharmacokinet. 2021;60:249‐259.
    1. McPhail BT, Emoto C, Butler D, Fukuda T, Akinbi H, Vinks AA. Opioid treatment for neonatal opioid withdrawal syndrome: current challenges and future approaches [published online ahead of print December 31, 2020]. J Clin Pharmacol. 10.1002/jcph.1811.
    1. Shan F, MacVicar S, Allegaert K, et al. Outcome reporting in neonates experiencing withdrawal following opioid exposure in pregnancy: a systematic review. Trials. 2020;21:262–020‐4183‐9.
    1. Greenwald MK, Johanson C‐E, Moody DE, et al. Effects of buprenorphine maintenance dose on mu‐opioid receptor availability, plasma concentrations, and antagonist blockade in heroin‐dependent volunteers. Neuropsychopharmacology. 2003;28:2000‐2009.
    1. Smith J, Lafferty M, Boelig RC, et al. Is maternal methadone dose associated with the severity of neonatal abstinence syndrome? [published online ahead of print December 15, 2020]. Am J Perinatol. 10.1055/s-0040-1721693.
    1. Hayes BD, Klein‐Schwartz W, Doyon S. Toxicity of buprenorphine overdoses in children. Pediatrics. 2008;121:e782‐e786.
    1. Toce MS, Burns MM, O'Donnell KA. Clinical effects of unintentional pediatric buprenorphine exposures: experience at a single tertiary care center. Clin Toxicol (Phila). 2017;55:12‐17.
    1. Marek E, Adeniyi‐Jones SC, Roke L, et al. Ethanol pharmacokinetics in neonates secondary to medication administration, 2015. American Academy of Pediatrics meeting. Available at: .
    1. Kraft WK, Adeniyi‐Jones SC, Ehrlich ME. Buprenorphine for the neonatal abstinence syndrome. N Engl J Med. 2017;377:997‐998.
    1. Czynski AJ, Davis JM, Dansereau LM, et al. Neurodevelopmental outcomes of neonates randomized to morphine or methadone for treatment of neonatal abstinence syndrome. J Pediatr. 2020;219:146‐151.e1.
    1. Ohno M, Motojima K, Okano T, Taniguchi A. Induction of drug‐metabolizing enzymes by phenobarbital in layered co‐culture of a human liver cell line and endothelial cells. Biol Pharm Bull. 2009;32:813‐817.
    1. Wu D, Kraft WK, Ehrlich ME, Barrett JS. Sublingual bioavailability of buprenorphine in newborns with neonatal abstinence syndrome‐‐a case study on physiological and developmental changes using SIMCYP Presented at the American College of Clinical Pharmacology meeting, Philadelphia, PA (2009).
    1. Chang Y, Moody DE, McCance‐Katz EF. Novel metabolites of buprenorphine detected in human liver microsomes and human urine. Drug Metab Dispos. 2006;34:440‐448.
    1. Strougo A, Yassen A, Monnereau C, Danhof M, Freijer J. Predicting the "First dose in children" of CYP3A‐metabolized drugs: evaluation of scaling approaches and insights into the CYP3A7‐CYP3A4 switch at young ages. J Clin Pharmacol. 2014;54:1006‐1015.
    1. Cone EJ, Gorodetzky CW, Yousefnejad D, Buchwald WF, Johnson RE. The metabolism and excretion of buprenorphine in humans. Drug Metab Dispos. 1984;12:577‐581.
    1. Knibbe CA, Krekels EHJ, van den Anker JN, et al. Morphine glucuronidation in preterm neonates, infants and children younger than 3 years. Clin Pharmacokinet. 2009;48:371‐385.
    1. Sanlorenzo LA, Stark AR, Patrick SW. Neonatal abstinence syndrome: an update. Curr Opin Pediatr. 2018;30:182‐186.

Source: PubMed

3
Se inscrever