Elsevier

Drug and Alcohol Dependence

Volume 144, 1 November 2014, Pages 1-11
Drug and Alcohol Dependence

Review
Buprenorphine maintenance and mu-opioid receptor availability in the treatment of opioid use disorder: Implications for clinical use and policy

https://doi.org/10.1016/j.drugalcdep.2014.07.035Get rights and content

Highlights

  • We review data on BUP-induced changes in mu-opioid receptor (μOR) availability, pharmacokinetics and clinical efficacy.

  • Opioid withdrawal suppression appears to require ≤50% μOR availability, associated with BUP plasma concentrations ≥1 ng/mL.

  • Blocking opioid reinforcement requires <20% μOR availability, or BUP plasma levels ≥3 ng/mL.

  • Blockade of opioid use for many patients may require total BUP daily doses ≥16 mg, although other factors contribute.

  • Data suggest that fixed limits on BUP doses in clinical care or limits on reimbursement for this care are unwarranted.

Abstract

Background

Sublingual formulations of buprenorphine (BUP) and BUP/naloxone have well-established pharmacokinetic and pharmacodynamic profiles, and are safe and effective for treating opioid use disorder. Since approvals of these formulations, their clinical use has increased. Yet, questions have arisen as to how BUP binding to mu-opioid receptors (μORs), the neurobiological target for this medication, relate to its clinical application. BUP produces dose- and time-related alterations of μOR availability but some clinicians express concern about whether doses higher than those needed to prevent opioid withdrawal symptoms are warranted, and policymakers consider limiting reimbursement for certain BUP dosing regimens.

Methods

We review scientific data concerning BUP-induced changes in μOR availability and their relationship to clinical efficacy.

Results

Withdrawal suppression appears to require ≤50% μOR availability, associated with BUP trough plasma concentrations ≥1 ng/mL; for most patients, this may require single daily BUP doses of 4 mg to defend against trough levels, or lower divided doses. Blockade of the reinforcing and subjective effects of typical doses of abused opioids require <20% μOR availability, associated with BUP trough plasma concentrations ≥3 ng/mL; for most individuals, this may require single daily BUP doses >16 mg, or lower divided doses. For individuals attempting to surmount this blockade with higher-than-usual doses of abused opioids, even larger BUP doses and <10% μOR availability would be required.

Conclusion

For these reasons, and given the complexities of studies on this issue and comorbid problems, we conclude that fixed, arbitrary limits on BUP doses in clinical care or limits on reimbursement for this care are unwarranted.

Introduction

Buprenorphine (BUP), a partial mu-opioid receptor (μOR) agonist, is approved in several countries for treating opioid use disorder. Since its introduction, clinicians and policymakers have made decisions that place limits on treatment dose and duration, which can impact care that patients receive (Clark et al., 2011). These decisions are sometimes implemented inflexibly and defended with certainty, citing anecdotal experience, policy constraints such as cost, and research literature. Such certainty may be unwarranted given the state of science in this field. This review is designed to inform clinicians and policymakers about empirical data underlying BUP dose-related changes in μOR availability and behavior, with emphasis on educating these individuals about evidence-based maintenance dosing practices. We will: (1) examine findings regarding the extent of BUP binding to μORs under varying experimental dosing conditions, (2) describe relationships between μOR availability and plasma concentrations of BUP toward achieving desired clinical effects including withdrawal suppression and blockade of opioid agonist effects, and (3) communicate the complexity of methods and data, and how their interpretation may affect clinical dosing and policy recommendations.

In animal studies, μORs have been implicated in opioid reinforcement (Bertalmio and Woods, 1989, Kreek et al., 2012, Matthes et al., 1996), discriminative stimulus effects (Bertalmio and Woods, 1987, Comer et al., 1993, Dykstra et al., 1988, Walker et al., 1994), and withdrawal effects (France and Woods, 1989, France et al., 1990, Maldonado et al., 1992, Matthes et al., 1996). Administered chronically, BUP antagonizes self-administration of mu-opioid agonists (Mello et al., 1983, Mello and Negus, 1998), which predicts clinical findings. BUP can precipitate withdrawal depending on the level of opioid dependence, agonist on which the subject is dependent, and time since last agonist dose (Kosten and Kleber, 1988, Sell et al., 2003, Walsh et al., 1995a, Woods and Gmerek, 1985, Woods et al., 1992). In humans, BUP precipitates moderate to severe opioid withdrawal in subjects maintained on moderate-dose methadone (60 mg/day), but less withdrawal in participants taking lower doses of methadone (25–30 mg/day) or heroin (Kosten and Kleber, 1988, Kosten et al., 1991, Strain et al., 1992, Walsh et al., 1995a).

Human laboratory studies that incorporate supervised inpatient stays, urine testing, and placebo control enable assessment of the impact of BUP on opioid reinforcing, subjective and physiological effects. Opioid reinforcement is measured using operant drug self-administration procedures whereby participants work on a computer task to earn drugs (e.g., Comer et al., 2001, Greenwald et al., 2013, Mello et al., 1982). Subjective drug-effect assessments include adjective ratings that reflect abuse potential (e.g., “liking”, “good effect”). Multi-item measures of opioid agonist and withdrawal effects are usually assessed. Measures of craving are often included. Physiological indices commonly recorded are pupil diameter, respiratory rate, oxygen saturation, heart rate, and blood pressure.

Sublingual BUP has been shown to dose-dependently decrease the reinforcing effects of heroin (Comer et al., 2001, Comer et al., 2005, Mello and Mendelson, 1980, Mello et al., 1982) and hydromorphone (Greenwald et al., 2002), consistent with reductions of opioid use in outpatient clinical trials (Johnson et al., 1995, Ling et al., 1998, Schottenfeld et al., 1993). Furthermore, many studies have reported BUP dose-dependent attenuation of the subjective effects of opioids (Bickel et al., 1988, Jasinski et al., 1978, Rosen et al., 1994, Schuh et al., 1999, Teoh et al., 1994, Walsh et al., 1995b). BUP also dose-dependently suppress opioid withdrawal symptoms in human laboratory studies (e.g., Greenwald et al., 2003) and outpatient trials (Fudala et al., 1990, Kuhlman et al., 1998), although the latter may be confounded by uncontrolled opioid use.

BUP dosing for opioid dependent patients is informed by FDA recommendations for different formulations and phases of treatment (induction, maintenance, detoxification). At present, the only SL formulations are BUP tablets, BUP/naloxone (NAL) tablets, and BUP/NAL filmstrips. Dosing guidelines are comparable for tablets and film, based on their similar pharmacokinetic and pharmacodynamic properties (Lintzeris et al., 2013). In the context of FDA guidelines, patient-related factors proximally determine clinical practice. Perhaps the most important aspect is that patients with greater levels of illegal/non-medical opioid use (Hillhouse et al., 2011) or comorbid pain severity (Chakrabarti et al., 2010) generally are prescribed higher doses of BUP, which implies there may be an association between greater clinical benefit and higher doses for these patients. Thus, during BUP induction, doses are typically 2–8 mg/day but can be escalated more rapidly depending on safety and need (Amass et al., 2012, Chiang and Hawks, 1994, Whitley et al., 2010). During maintenance, effective doses are typically 8–24 mg/day (Ling et al., 1998, Ling and Smith, 2002) but can be lower or higher (Compton et al., 1996). During detoxification, BUP doses are most often tapered over one to several weeks, usually by halving the dose at each step from the maintenance level (Amass et al., 2004, Ling et al., 2009, Sigmon et al., 2013).

Section snippets

[11C]-carfentanilPositron Emission Tomography (PET) imaging of μORs

The high-affinity, μOR-specific PET radioligand, [11C]-carfentanil (Dannals et al., 1985, Frost et al., 1985, Titeler et al., 1989) offers researchers a reliable and non-invasive means to map brain regional μOR availability. To adhere to the literature, we will refer to changes in receptor availability, which is an indirect measure. Although receptor “occupancy” is an intuitively attractive term, it is misleading. First, “occupancy” suggests an endogenous or exogenous ligand that binds to

Results

A preliminary study (Zubieta et al., 2000) used the SL BUP-mono liquid formulation (tablets have ≈70% bioavailability of liquid formulations; Chawarski et al., 2005, Compton et al., 2006), which was discontinued prior to FDA approval. Three heroin-dependent volunteers were stabilized on daily BUP doses of 2 mg, then 16 mg, then placebo (0 mg) under double-blind conditions. Four days before each PET scan (at each BUP dose), the participant was admitted to an inpatient unit with daily urine testing

Pharmacological issues

BUP maintenance dose is inversely related to μOR availability in opioid-dependent volunteers; however, it is not reasonable to compare receptor availability estimates from Comer et al. (2005) and Greenwald et al. (2003) due to methodological differences. First, these studies differed in measuring receptor availability in vivo (Greenwald et al., 2003) vs. simulation (Comer et al., 2005), which could partly explain disparate estimates for BUP 2-mg (Table 1). Second, estimates from Comer et al.

Role of funding source

NIH grants R01 DA015462 (M.K.G.), P50 DA09236 and R01 DA16759 (S.D.C.), and R01 DA025991 and R01 DA035616 (D.A.F.) from the National Institute on Drug Abuse, a research grant (Joe Young Sr./Helene Lycaki funds) from the State of Michigan and the Detroit Wayne Mental Health Authority (M.K.G.), supported the preparation of this manuscript article subtitle

Contributors

M.K.G. wrote the initial draft of the manuscript, and finalized all content, and S.D.C. and D.A.F. contributed equally to the conceptualization and editing of the manuscript. All authors read the manuscript and approved of its submission to the journal, and are indebted to the anonymous reviewers for their extremely helpful critiques.

References (106)

  • R. Gross-Isseroff et al.

    Regionally selective increases in mu opioid receptor density in the brains of suicide victims

    Brain Res.

    (1990)
  • D.S. Harris et al.

    Buprenorphine and naloxone co-administration in opiate-dependent patients stabilized on sublingual buprenorphine

    Drug Alcohol Depend.

    (2000)
  • C. Iribarne et al.

    Involvement of cytochrome P450 3A4 in N-dealkylation of buprenorphine in human liver microsomes

    Life Sci.

    (1997)
  • D.M. Jewett

    A simple synthesis of [11 C]carfentanil

    Nucl. Med. Biol.

    (2001)
  • R.E. Johnson et al.

    A placebo controlled clinical trial of buprenorphine as a treatment for opioid dependence

    Drug Alcohol Depend.

    (1995)
  • T.R. Kosten et al.

    Buprenorphine detoxification from opioid dependence: a pilot study

    Life Sci.

    (1988)
  • P.A. Lester et al.

    Comparison of the in vitro efficacy of mu, delta, kappa and ORL1 receptor agonists and non-selective opioid agonists in dog brain membranes

    Brain Res.

    (2006)
  • J.W. Lewis

    Buprenorphine

    Drug Alcohol Depend.

    (1985)
  • W. Ling et al.

    Buprenorphine: blending practice and research

    J. Subst. Abuse Treat.

    (2002)
  • N. Lintzeris et al.

    A randomized controlled trial of sublingual buprenorphine–naloxone film versus tablets in the management of opioid dependence

    Drug Alcohol Depend.

    (2013)
  • R. Maldonado et al.

    Precipitation of morphine withdrawal syndrome in rats by administration of mu-, delta- and kappa-selective opioid antagonists

    Neuropharmacology

    (1992)
  • L.A. Marsch et al.

    Buprenorphine treatment for opioid dependence: the relative efficacy of daily, twice and thrice weekly dosing

    Drug Alcohol Depend.

    (2005)
  • M.I. Rosen et al.

    Buprenorphine: duration of blockade of effects of intramuscular hydromorphone

    Drug Alcohol Depend.

    (1994)
  • R.S. Schottenfeld et al.

    Buprenorphine: dose-related effects on cocaine and opioid use in cocaine-abusing opioid-dependent humans

    Biol. Psychiatry

    (1993)
  • M. Titeler et al.

    Mu opiate receptors are selectively labeled by [3H]-carfentanil in human and rat brain

    Eur. J. Pharmacol

    (1989)
  • M.E. Wewers et al.

    The effect of chronic administration of nicotine on antinociception, opioid receptor binding and met-enkephalin levels in rats

    Brain Res.

    (1999)
  • S.D. Whitley et al.

    Factors associated with complicated buprenorphine inductions

    J. Subst. Abuse Treat.

    (2010)
  • J.H. Woods et al.

    Substitution and primary dependence studies in animals

    Drug Alcohol Depend.

    (1985)
  • L. Amass et al.

    Bringing buprenorphine–naloxone detoxification to community treatment providers: the NIDA Clinical Trials Network field experience

    Am. J. Addict.

    (2004)
  • L. Amass et al.

    A prospective, randomized, multicenter acceptability and safety study of direct buprenorphine/naloxone induction in heroin-dependent individuals

    Addiction

    (2012)
  • A.J. Bertalmio et al.

    Differentiation between mu and kappa receptor-mediated effects in opioid drug discrimination: apparent pA2 analysis

    J. Pharmacol. Exp. Ther.

    (1987)
  • A.J. Bertalmio et al.

    Reinforcing effect of alfentanil is mediated by mu opioid receptors: apparent pA2 analysis

    J. Pharmacol. Exp. Ther.

    (1989)
  • W.K. Bickel et al.

    Buprenorphine dosing every 1, 2, or 3 days in opioid-dependent patients

    Psychopharmacology (Berl.)

    (1999)
  • W.K. Bickel et al.

    Buprenorphine: dose-related blockade of opioid challenge effects in opioid dependent humans

    J. Pharmacol. Exp. Ther.

    (1988)
  • J.W. Black et al.

    Operational models of pharmacological agonism

    Proc. R. Soc. Lond. B.

    (1983)
  • C.N. Chiang et al.

    Development of a buprenorphine–naloxone combination drug for the treatment of drug addiction

  • R.E. Clark et al.

    The evidence doesn’t justify steps by state Medicaid programs to restrict opioid addiction treatment with buprenorphine

    Health Aff.

    (2011)
  • S.D. Comer et al.

    Buprenorphine sublingual tablets: effects on IV heroin self-administration by humans

    Psychopharmacology (Berl.)

    (2001)
  • S.D. Comer et al.

    Discriminative stimulus effects of BW373U86: a nonpeptide ligand with selectivity for delta opioid receptors

    J. Pharmacol. Exp. Ther.

    (1993)
  • S.D. Comer et al.

    Buprenorphine/naloxone reduces the reinforcing and subjective effects of heroin in heroin-dependent volunteers

    Psychopharmacology

    (2005)
  • P. Compton et al.

    Buprenorphine as a pharmacotherapy for opiate addiction

    Am. J. Addict.

    (1996)
  • M. Connock et al.

    Methadone and buprenorphine for the management of opioid dependence: a systematic review and economic evaluation

    Health Technol. Assess.

    (2007)
  • M.F. DosSantos et al.

    Reduced basal ganglia μ-opioid receptor availability in trigeminal neuropathic pain: a pilot study

    Mol. Pain

    (2012)
  • L.A. Dykstra et al.

    Discriminative and analgesic effects of mu and kappa opioids: in vivo pA2 analysis

    Psychopharmacol. Ser.

    (1988)
  • A. Elkader et al.

    Buprenorphine: clinical pharmacokinetics in the treatment of opioid dependence

    Clin. Pharmacokinet.

    (2005)
  • A. Fareed et al.

    Treatment outcome for flexible dosing buprenorphine maintenance treatment

    Am. J. Drug Alcohol Abuse

    (2012)
  • C.P. France et al.

    Apparent affinity of opioid antagonists in morphine-treated rhesus monkeys discriminating between saline and naltrexone

    J. Pharmacol. Exp. Ther.

    (1990)
  • C.P. France et al.

    Discriminative stimulus effects of naltrexone in morphine-treated rhesus monkeys

    J. Pharmacol. Exp. Ther.

    (1989)
  • J.J. Frost et al.

    Imaging opiate receptors in the human brain by positron tomography

    J. Comp. Assist. Tomogr.

    (1985)
  • P.J. Fudala et al.

    Buprenorphine/Naloxone Collaborative Study Group, 2003. Office-based treatment of opiate addiction with sublingual-tablet formulation of buprenorphine and naloxone

    N. Engl. J. Med.

    (2003)
  • Cited by (123)

    • A Neuropharmacological Model to Explain Buprenorphine Induction Challenges

      2022, Annals of Emergency Medicine
      Citation Excerpt :

      In this situation, buprenorphine microdosing might promote mu-opioid receptor resensitization while not triggering withdrawal symptoms; conversely, high-dose buprenorphine could be used to maximize agonist signaling and resensitization. In contrast, the ease of buprenorphine induction is not expected to differ between various opioids when there is significant preinduction withdrawal (in our clinical experience, more so when there are physiological withdrawal signs) because buprenorphine uniformly suppresses withdrawal regardless of the preceding agonist.78,79 Thus, microdosing is counterproductive (insufficient agonist signaling due to low buprenorphine dosing) when the patient is experiencing preinduction withdrawal symptoms, regardless of how the withdrawal symptoms developed—from abstinence, naloxone, or prior doses of buprenorphine.

    • A novel long-acting formulation of oral buprenorphine/naloxone produces prolonged decreases in fentanyl self-administration by rhesus monkeys

      2022, Drug and Alcohol Dependence
      Citation Excerpt :

      Part of the reason for the high relapse rates may be that sublingual immediate-release formulations require daily administration, which can reduce adherence. Moreover, immediate-release forms of buprenorphine are associated with high peak blood levels, which can induce euphoria and lead to abuse and diversion, and low trough levels, which can impact efficacy (Greenwald et al., 2014). Long-acting implants and injectable formulations of buprenorphine are administered less frequently and can reduce peak and trough effects, and recent data suggest that they may be comparable to or more effective than sublingual buprenorphine (Lofwall et al., 2018; Rosenthal et al., 2016).

    • Buprenorphine and Cardiac Surgery: Navigating the Challenges of Pain Management

      2022, Journal of Cardiothoracic and Vascular Anesthesia
    View all citing articles on Scopus
    View full text