Invited Article
A review of pregnancy-induced changes in opioid pharmacokinetics, placental transfer, and fetal exposure: Towards fetomaternal physiologically-based pharmacokinetic modeling to improve the treatment of neonatal opioid withdrawal syndrome

https://doi.org/10.1016/j.pharmthera.2021.108045Get rights and content

Abstract

Physiologically-based pharmacokinetic (PBPK) modeling has emerged as a useful tool to study pharmacokinetics (PK) in special populations, such as pregnant women, fetuses, and newborns, where practical hurdles severely limit the study of drug behavior. PK in pregnant women is variable and everchanging, differing greatly from that in their nonpregnant female and male counterparts typically enrolled in clinical trials. PBPK models can accommodate pregnancy-induced physiological and metabolic changes, thereby providing mechanistic insights into maternal drug disposition and fetal exposure. Fueled by the soaring opioid epidemic in the United States, opioid use during pregnancy continues to rise, leading to an increased incidence of neonatal opioid withdrawal syndrome (NOWS). The severity of NOWS is influenced by a complex interplay of extrinsic and intrinsic factors, and varies substantially between newborns, but the extent of prenatal opioid exposure is likely the primary driver. Fetomaternal PBPK modeling is an attractive approach to predict in utero opioid exposure. To facilitate the development of fetomaternal PBPK models of opioids, this review provides a detailed overview of pregnancy-induced changes affecting the PK of commonly used opioids during gestation. Moreover, the placental transfer of these opioids is described, along with their disposition in the fetus. Lastly, the implementation of these factors into PBPK models is discussed. Fetomaternal PBPK modeling of opioids is expected to provide improved insights in fetal opioid exposure, which allows for prediction of postnatal NOWS severity, thereby opening the way for precision postnatal treatment of these vulnerable infants.

Introduction

Pharmacokinetic (PK) models are mathematical tools that describe drug disposition, which can be used to predict plasma or tissue concentrations at a specific dose (Mould and Upton, 2012, Mould and Upton, 2013). PK models emerged as a cascade of increasing complexity. Conventional population PK models reduce the human body to a central compartment, possibly linked to one or two peripheral compartments, where each compartment does not directly represent a physiological organ. Population PK models are empirically developed (also known as a “top-down” approach), meaning that observed concentration-time data are used to estimate underlying PK parameters. Although still widely used and useful to date, the empirical model's limited anatomical and biological foundation complicates extrapolation to physiological conditions not observed in the study population whose data were used for model development. Hence, physiologically-based pharmacokinetic (PBPK) models materialized (Rowland, Peck, & Tucker, 2011). Although these models are similarly compartmentalized, the compartments reflect actual tissue and organ spaces with corresponding realistic volumes, enabling more comprehensive and mechanism-based modeling.

Through pioneering work by Teorell, the foundation for PBPK modeling was laid as early as 1937 (Teorell, 1937a, Teorell, 1937b). PBPK modeling rose to prominence in the 1960s and 1970s (Himmelstein & Lutz, 1979), and has since been implemented in a variety of clinical areas, such as in supporting dose selection in neonatal and pediatric populations (Strougo et al., 2012; Wagner et al., 2015), evaluating drug-drug interactions (Vieira et al., 2014), and informing clinical trial design (Jones et al., 2015). PBPK models conjoin human body characteristics (e.g., weight, age, blood flow, organ size, enzyme abundance) and drug physicochemical properties (e.g., logP, pKa, blood binding, permeability). In contrast to empirical PK models, PBPK models are developed in a “bottom-up” manner, in which fundamental in vitro data are scaled to conceptualize in vivo PK parameters (also called in vitro-in vivo extrapolation [IVIVE]). In practice, clinical observations are then generally used to validate the PBPK model, optimize physiological or drug-related parameters, or estimate the value of those unknown, in an overall “middle-out” approach (Jamei, Dickinson, & Rostami-Hodjegan, 2009).

Drug disposition in pediatric and neonatal subjects has historically been understudied due to practical concerns in conducting such clinical studies. Still, rapid physiological development and maturation, which inherently affect drug disposition, warrant characterization of pediatric and neonatal PK to identify age-appropriate doses to ensure drug safety and efficacy. Over the past decade, PBPK modeling has emerged as an elegant approach to assess drug behavior in neonates, infants, and children (Verscheijden, Koenderink, Johnson, de Wildt, & Russel, 2020). Another patient group that has historically been excluded from drug research is pregnant women. Although the motivation to do so is evident, this has led to insufficient PK and safety data to guide dosing in pregnant or breastfeeding women for an overwhelming majority of drugs (Carvalho & Wong, 2015; McCormack & Best, 2014). This creates a conundrum for physicians and patients, as it has been shown that in the United States (US) between 34.4 and 39.0% of pregnant women use one or more medications (excluding vitamins and minerals) in the first, second, or third trimester regardless (Daw, Hanley, Greyson, & Morgan, 2011). Pregnant women do not exhibit similar drug disposition as their nonpregnant female and male counterparts typically enrolled in clinical trials (Pariente et al., 2016). Moreover, women carry a vulnerable developing fetus during gestation that needs to be protected from toxic effects of certain xenobiotics. Toxicity is, in part, dependent on placental transfer of the compound, as well as maternal and fetal PK, which rapidly change during gestation. Emerging studies (George et al., 2020; Szeto, Le Merdy, Dupont, Bolger, & Lukacova, 2021) reveal that PBPK models can incorporate pregnancy-related physiological changes and help achieve a better understanding of drug behavior in this special population. In this review, pregnancy-induced changes in opioid PK, placental opioid transfer, and fetal opioid disposition are discussed in the context of neonatal opioid withdrawal syndrome (NOWS). In addition, the implementation of these different elements in PBPK models is described.

Section snippets

Neonatal opioid withdrawal syndrome (NOWS)

The opioid epidemic continues to ravage the US. In 2019, 49,860 people died from opioid overdoses, amounting to an average of one death every ten minutes, which is a six-fold increase since 2000 (Hedegaard, Miniño, & Warner, 2020). The coronavirus disease 2019 (COVID-19) pandemic likely further aggravated the opioid epidemic (Ochalek, Cumpston, Wills, Gal, & Moeller, 2020; Wainwright et al., 2020). Recently, it was found that 6.6% of women self-reported opioid use during pregnancy, of which

Physiological changes affecting opioid disposition during pregnancy

Pregnancy induces a plethora of physiological and anatomical changes in gravid women, including altered drug absorption, and increased volume of distribution, renal blood flow, and hepatic blood flow. These changes and their effect on PK in general have been described previously in great detail (Abduljalil, Furness, Johnson, Rostami-Hodjegan, & Soltani, 2012; Tasnif, Morado, & Hebert, 2016). In this article, the effect of pregnancy-induced physiological changes on specifically the PK of

Anatomy of the placenta

The placenta is the interface between the maternal and fetal circulation and supplies the fetus with oxygen and nutrients, while simultaneously clearing metabolic waste products from the fetal blood. It is a unique organ as it is the only human organ that has a transient existence, arising during early pregnancy and expelling itself following parturition once its purpose has been fulfilled. The placenta acts as a protective barrier for some compounds, but many xenobiotics, driven by the

Towards prediction of fetal opioid disposition and NOWS severity

Thus far, the physiological changes during pregnancy that affect opioid PK and the extent of placental opioid transfer from maternal to fetal circulation have been discussed. Moreover, successful incorporation of these processes in pregnancy PBPK models has been described. Such pregnancy PBPK models can be used to characterize fetal opioid exposure. However, to attain insight into fetal opioid disposition (e.g., metabolism of opioids by the fetus itself and distribution throughout fetal

Conclusion

Pregnancy induces a plethora of physiological and metabolic changes that influence maternal PK. For opioids and their metabolites specifically, promoted activity of CYP and UGT enzymes, increased renal excretion, and, conceivably, reduced biliary secretion are key drivers of altered PK during gestation. These pregnancy-induced changes are not static; rather, conception initiates a cascade of modifications and adaptations that are constantly developing, with some effects possibly

Declaration of Competing Interest

S.L.W. has received a research grant from Chiesi Pharmaceuticals and a consulting fee from Braeburn Pharmaceuticals. All other authors declared no competing interests for this work.

Acknowledgments

M.W.v.H. was supported by the Ritschel Doctoral Fellowship of the University of Cincinnati. T.M. has received a research grant from the Center for Clinical and Translational Science and Training (CCTST); National Center for Advancing Translational Sciences of the National Institutes of Health, under Award Number 2UL1TR001425-05A1.

References (290)

  • J. Cooper et al.

    Placental transfer of fentanyl in early human pregnancy and its detection in fetal brain

    British Journal of Anaesthesia

    (1999)
  • J.S. Dashe et al.

    Relationship between maternal methadone dosage and neonatal withdrawal

    Obstetrics and Gynecology

    (2002)
  • J.M. Davison et al.

    Renal hemodynamics and tubular function normal human pregnancy

    Kidney International

    (1980)
  • R.R. De Alvarez et al.

    Serum protein fractionation in normal pregnancy

    American Journal of Obstetrics and Gynecology

    (1961)
  • R. Desprats et al.

    Maternal and umbilical cord concentrations of fentanyl after epidural analgesia for cesarean section

    European Journal of Obstetrics, Gynecology, and Reproductive Biology

    (1991)
  • T.M. Doberczak et al.

    Neonatal opiate abstinence syndrome in term and preterm infants

    Journal of Pediatrics

    (1991)
  • S. Gil et al.

    P-glycoprotein expression of the human placenta during pregnancy

    Placenta

    (2005)
  • J. Guitton et al.

    Possible involvement of multiple cytochrome P450S in fentanyl and sufentanil metabolism as opposed to alfentanil

    Biochemical Pharmacology

    (1997)
  • J. Hakkola et al.

    Expression of xenobiotic-metabolizing cytochrome P450 forms in human full-term placenta

    Biochemical Pharmacology

    (1996)
  • K. Abduljalil et al.

    Drug dosing during pregnancy-opportunities for physiologically based pharmacokinetic models

    Journal of Pharmacokinetics and Pharmacodynamics

    (2020)
  • K. Abduljalil et al.

    Anatomical, physiological and metabolic changes with gestational age during normal pregnancy: A database for parameters required in physiologically based pharmacokinetic modelling

    Clinical Pharmacokinetics

    (2012)
  • K. Abduljalil et al.

    Fetal physiologically based pharmacokinetic models: systems information on the growth and composition of fetal organs

    Clinical Pharmacokinetics

    (2019)
  • K. Abduljalil et al.

    Fetal physiologically based pharmacokinetic models: systems information on fetal blood components and binding proteins

    Clinical Pharmacokinetics

    (2020)
  • K. Abduljalil et al.

    Fetal physiologically-based pharmacokinetic models: systems information on fetal biometry and gross composition

    Clinical Pharmacokinetics

    (2018)
  • K. Abduljalil et al.

    Fetal physiologically based pharmacokinetic models: systems information on fetal cardiac output and its distribution to different organs during development

    Clinical Pharmacokinetics

    (2021)
  • K. Abduljalil et al.

    Prediction of maternal pharmacokinetics using physiologically based pharmacokinetic models: assessing the impact of the longitudinal changes in the activity of CYP1A2, CYP2D6 and CYP3A4 enzymes during pregnancy

    Journal of Pharmacokinetics and Pharmacodynamics

    (2020)
  • B. Achour et al.

    Expression of hepatic drug-metabolizing cytochrome p450 enzymes and their intercorrelations: a meta-analysis

    Drug Metabolism and Disposition: The Biological Fate of Chemicals

    (2014)
  • B. Achour et al.

    Protein expression of various hepatic uridine 5′-diphosphate glucuronosyltransferase (UGT) enzymes and their inter-correlations: a meta-analysis

    Biopharmaceutics and Drug Disposition

    (2014)
  • M. Andersson et al.

    Detection of morphine-3-sulfate and morphine-6-sulfate in human urine and plasma, and formation in liver cytosol

    Pharmacology Research & Perspectives

    (2014)
  • G.M. Anelli et al.

    Placental ESRRG-CYP19A1 expressions and circulating 17-beta estradiol in IUGR pregnancies

    Frontiers in Pediatrics

    (2019)
  • O. Anoshchenko et al.

    Estimating fetal exposure to the P-gp substrates, corticosteroids, by PBPK modeling to inform prevention of neonatal respiratory distress syndrome

    CPT: Pharmacometrics & Systems Pharmacology

    (2021)
  • O. Anoshchenko et al.

    Gestational age-dependent abundance of human placental transporters as determined by quantitative targeted proteomics

    Drug Metabolism and Disposition: The Biological Fate of Chemicals

    (2020)
  • A. Avdeef et al.

    Octanol-, chloroform-, and propylene glycol dipelargonat-water partitioning of morphine-6-glucuronide and other related opiates

    Journal of Medicinal Chemistry

    (1996)
  • R.E. Azuine et al.

    Prenatal risk factors and perinatal and postnatal outcomes associated with maternal opioid exposure in an urban, low-income, multiethnic US population

    JAMA Network Open

    (2019)
  • B. Bakstad et al.

    Opioid maintenance treatment during pregnancy: occurrence and severity of neonatal abstinence syndrome. A national prospective study

    European Addiction Research

    (2009)
  • E.V. Barker et al.

    Dehydroepiandrosterone sulfotransferase in the developing human fetus: quantitative biochemical and immunological characterization of the hepatic, renal, and adrenal enzymes

    Endocrinology

    (1994)
  • L. de Barros Duarte et al.

    Distribution of fentanyl in the placental intervillous space and in the different maternal and fetal compartments in term pregnant women

    European Journal of Clinical Pharmacology

    (2009)
  • J.R. Bastian et al.

    Dose-adjusted plasma concentrations of sublingual buprenorphine are lower during than after pregnancy

    American Journal of Obstetrics and Gynecology

    (2017)
  • W.T. Beaver et al.

    Comparisons of the analgesic effects of oral and intramuscular oxymorphone and of intramuscular oxymorphone and morphine in patients with cancer

    Journal of Clinical Pharmacology

    (1977)
  • B. Berger et al.

    Cytochrome P450 enzymes involved in metoprolol metabolism and use of metoprolol as a CYP2D6 phenotyping probe drug

    Frontiers in Pharmacology

    (2018)
  • S.R. Bista et al.

    Protein binding of fentanyl and its metabolite nor-fentanyl in human plasma, albumin and α-1 acid glycoprotein

    Xenobiotica

    (2015)
  • C. Blundell et al.

    Placental drug transport-on-a-chip: a microengineered in vitro model of transporter-mediated drug efflux in the human placental barrier

    Advanced Healthcare Materials

    (2018)
  • R. Bouër et al.

    The roles of P-glycoprotein and intracellular metabolism in the intestinal absorption of methadone: in vitro studies using the rat everted intestinal sac

    Fundamental and Clinical Pharmacology

    (1999)
  • P. Bourget et al.

    Models for placental transfer studies of drugs

    Clinical Pharmacokinetics

    (1995)
  • D.Z. Braverman et al.

    Effects of pregnancy and contraceptive steroids on gallbladder function

    New England Journal of Medicine

    (1980)
  • D. Brewster et al.

    Biliary excretion, metabolism and enterohepatic circulation of buprenorphine

    Xenobiotica

    (1981)
  • S.F. Brunk et al.

    Morphine metabolism in man

    Clinical Pharmacology and Therapeutics

    (1974)
  • J. Cao et al.

    Expression of rat hepatic multidrug resistance-associated proteins and organic anion transporters in pregnancy

    American Journal of Physiology: Gastrointestinal and Liver Physiology

    (2002)
  • Y. Chang et al.

    Glucuronidation of buprenorphine and norbuprenorphine by human liver microsomes and UDP-glucuronosyltransferases

    Drug Metabolism Letters

    (2009)
  • N. Chaphekar et al.

    Model-informed dose optimization in pregnancy

    Journal of Clinical Pharmacology

    (2020)
  • Cited by (0)

    View full text