Research Articles
Differential Involvement of P-Glycoprotein (ABCB1) in Permeability, Tissue Distribution, and Antinociceptive Activity of Methadone, Buprenorphine, and Diprenorphine: In Vitro and In Vivo Evaluation

https://doi.org/10.1002/jps.21770Get rights and content

Abstract

Conclusions based on either in vitro or in vivo approach to evaluate the P-gp affinity status of opioids may be misleading. For example, in vitro studies indicated that fentanyl is a P-gp inhibitor while in vivo studies indicated that it is a P-gp substrate. Quite the opposite was evident for meperidine. The objective of this study was to evaluate the P-gp affinity status of methadone, buprenorphine and diprenorphine to predict P-gp-mediated drug-drug interactions and to determine a better candidate for management of opioid dependence. Two in vitro (P-gp ATPase and monolayer efflux) assays and two in vivo (tissue distribution and antinociceptive evaluation in mdr1a/b (−/−) mice) assays were used. Methadone stimulated the P-gp ATPase activity only at higher concentrations, while verapamil and GF120918 inhibited its efflux (p < 0.05). The brain distribution and antinociceptive activity of methadone were enhanced (p < 0.05) in P-gp knockout mice. Conversely, buprenorphine and diprenorphine were negative in all assays. P-gp can affect the PK/PD of methadone, but not buprenorphine or diprenorphine. Our report is in favor of buprenorphine over methadone for management of opioid dependence. Buprenorphine most likely is not a P-gp substrate and concerns regarding P-gp-mediated drug-drug interaction are not expected. © 2009 Wiley-Liss, Inc. and the American Pharmacists Association J Pharm Sci 98:4928–4940, 2009

Section snippets

INTRODUCTION

The blood–brain barrier (BBB) acts as a regulator of the flux of xenobiotics between the systemic circulation and the central nervous system (CNS). The permeability of drugs across the BBB has been traditionally considered as a factor of molecular weight, protein binding, H-bonding, degree of ionization and lipid solubility.1 However, a convincing body of evidence suggests that the efflux transporter, P-glycoprotein (P-gp, Abcb1), may also modulate drug disposition into the brain.2, 3, 4, 5, 6,

Drug-Stimulated P-gp ATPase Activity

Drug stimulated P-gp ATPase activity was estimated by Pgp-GIO assay system (Promega, Madison, WI). This method relies on the ATP dependence of the light-generating reaction of firefly luciferase. ATP consumption is detected as a decrease in luminescence. In a 96 well plate, recombinant human P-gp (25 µg) was incubated with P-gp-GIO assay buffer™ (20 µL) (control, n = 4), verapamil (200 µM) (n = 4), sodium orthovanadate (100 µM) (n = 4), methadone (5–100 µM) (n = 3/conc.), buprenorphine (5–100 µM) (n = 

Effect of Modulators on P-gp ATPase Activity

Different concentrations of methadone, buprenorphine, and diprenorphine (Fig. 2) were examined to determine their effects on the P-gp ATPase activity. Each opioid together with a known excess of ATP was incubated with recombinant human P-gp. ATP consumption due to P-gp stimulation by each opioid was detected as a decrease in luminescence, that is, the higher the potency of a compound to stimulate the P-gp ATPase activity, the lower the luminescence signal. The rate of ATP consumption due to

DISCUSSION

The objective of this study was to evaluate the P-gp affinity status of methadone, buprenorphine and diprenorphine to predict P-gp-mediated drug–drug interactions and to determine a better candidate for management of opioid dependence. Classification of opioids or any other compound as a P-gp substrate, nonsubstrate, inducer or inhibitor based on only one assay may be misleading.1,21,23,32 As a result, we used two in vitro (P-gp ATPase and monolayer efflux assays) and two in vivo (opioids'

Acknowledgements

This study was supported in part by University of Maryland Intramural Research grant (A.C.), a Predoctoral Fellowship from the Egyptian Ministry of Higher Education (H.E.H.) and Ralph Shangraw Predoctoral Fellowship from University of Maryland (H.E.H.).

REFERENCES (50)

  • R. Callaghan et al.

    Synthetic and natural opiates interact with P-glycoprotein in multidrug-resistant cells

    J Biol Chem

    (1993)
  • J.E. Polli et al.

    Methods to compare dissolution profiles and a rationale for wide dissolution specifications for metoprolol tartrate tablets

    J Pharm Sci

    (1997)
  • S.Y. Liu et al.

    Simultaneous determination of buprenorphine and its prodrug, buprenorphine propionate, by high-performance liquid chromatography with fluorescence detection: Application to pharmacokinetic studies in rabbits

    J Chromatogr B Analyt Technol Biomed Life Sci

    (2005)
  • F.B. Ross et al.

    Co-administration of sub-antinociceptive doses of oxycodone and morphine produces marked antinociceptive synergy with reduced CNS side-effects in rats

    Pain

    (2000)
  • T. Litman et al.

    Structure-activity relationships of P-glycoprotein interacting drugs: Kinetic characterization of their effects on ATPase activity

    Biochim Biophys Acta

    (1997)
  • M.J. Garrido et al.

    Methadone: A review of its pharmacokinetic/pharmacodynamic properties

    J Pharmacol Toxicol Methods

    (1999)
  • G.D. Eytan et al.

    The role of passive transbilayer drug movement in multidrug resistance and its modulation

    J Biol Chem

    (1996)
  • A.H. Schinkel

    P-Glycoprotein, a gatekeeper in the blood-brain barrier

    Adv Drug Deliv Rev

    (1999)
  • T. Suzuki et al.

    Investigation of transport mechanism of pentazocine across the blood-brain barrier using the in situ rat brain perfusion technique

    J Pharm Sci

    (2002)
  • A. Crowe

    The influence of P-glycoprotein on morphine transport in Caco-2 cells. Comparison with paclitaxel

    Eur J Pharmacol

    (2002)
  • K.M. Mahar Doan et al.

    Passive permeability and P-glycoprotein-mediated efflux differentiate central nervous system (CNS) and non-CNS marketed drugs

    J Pharmacol Exp Ther

    (2002)
  • A.H. Schinkel et al.

    Normal viability and altered pharmacokinetics in mice lacking mdr1-type (drug-transporting) P-glycoproteins

    Proc Natl Acad Sci USA

    (1997)
  • A.H. Schinkel et al.

    Absence of the mdr1a P-Glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin, and cyclosporin A

    J Clin Invest

    (1995)
  • J.H. Lin et al.

    Role of P-glycoprotein in pharmacokinetics: Clinical implications

    Clin Pharmacokinet

    (2003)
  • P.A. Glare et al.

    Dose-ranging study of oxycodone for chronic pain in advanced cancer

    J Clin Oncol

    (1993)
  • Cited by (80)

    • 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

      2022, Pharmacology and Therapeutics
      Citation Excerpt :

      Methadone (Tournier et al., 2010; Tournier et al., 2011), oxycodone (Hassan, Myers, Lee, Coop, & Eddington, 2007; Zwisler et al., 2010), fentanyl (Park et al., 2007), and morphine (Callaghan & Riordan, 1993; Sadhasivam et al., 2015) are substrates for P-gp. Buprenorphine is the only opioid discussed in this review that is not transported by P-gp (Hassan, Myers, Coop, & Eddington, 2009), although its metabolite norbuprenorphine is (Tournier et al., 2010). P-gp is expressed at the apical side of the syncytiotrophoblast (Atkinson, Sibley, Fairbairn, & Greenwood, 2006; Sun et al., 2006) and on the cytotrophoblast, adjacent to the basolateral membrane of the syncytiotrophoblast (Lye et al., 2013), where it promotes transport towards the intervillous space.

    • Pharmacogenomics

      2019, A Practice of Anesthesia for Infants and Children
    • Methadone Metabolism and Drug-Drug Interactions: In Vitro and In Vivo Literature Review

      2018, Journal of Pharmaceutical Sciences
      Citation Excerpt :

      Methadone slightly inhibited P-gp-mediated transport of rhodamine 123 in a concentration-dependent manner in an accumulation assay,28 but inhibited the efflux of rhodamine 123 with a concentration at 50% inhibition value of 2.6 μg/mL.29 Several studies have shown methadone to be a P-gp substrate by comparing plasma and brain levels in wild-type (mdr1+/+) and P-gp-deficient (mdr1−/−) mice.26,30,31 Methadone is not a substrate of the uptake transporters organic cation transporter (OCT) 1, OCT2, OCT3, organic anion transporting protein (OATP) 1A2, OATP1B1, OATP1B3, or OATP2B1.32

    View all citing articles on Scopus
    View full text