1
|
Davidson PJ, Bowles JM, Faul M, Gaines TL. Spatial proximity and access to buprenorphine or methadone treatment for opioid use disorder in a sample of people misusing opioids in Southern California. J Subst Abuse Treat 2022; 132:108634. [PMID: 34625318 PMCID: PMC10465062 DOI: 10.1016/j.jsat.2021.108634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/15/2021] [Accepted: 09/14/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND In response to the opioid crisis, over the last 10 years substantial strides have been made to increase the availability of evidence-based treatments for opioid use disorder, in particular buprenorphine maintenance, in the United States. Despite these worthwhile efforts, uptake rates of evidence-based treatment remain relatively low. As part of a broader study of opioid misuse, we examined proximity to evidence-based treatment as a potential barrier to treatment access. METHODS In 2017-2018, we surveyed 218 individuals misusing prescription opioids or using street opioids in three Southern Californian counties. The study calculated driving distance from place of residence to the closest treatment provider offering buprenorphine or methadone treatment for opioid use disorders. RESULTS Median distance to providers was 3.8 km (2.4 miles). Seventy one (33%) participants had received some form of treatment in the last 3 months; however, only 26 (40%) of these had received buprenorphine or methadone maintenance treatment. Participants receiving treatment at the time of their interview were traveling an average 16.8 km (10.4 miles) to reach treatment, indicating that as a group this population was both willing and able to seek and engage with treatment. CONCLUSIONS In the suburban and exurban communities in which our study was based, our findings suggest that simple physical proximity to providers of evidence-based treatment for opioid use disorder is no longer a critical barrier. Other barriers to uptake of buprenorphine or methadone maintenance treatment clearly remain and need to be addressed. DISCLAIMER Findings and conclusions in this report are those of the authors and do not necessarily represent the official position of the Centers for Disease Control and Prevention.
Collapse
Affiliation(s)
- P J Davidson
- University of California, San Diego, Department of Medicine, 9500 Gilman Dr, La Jolla, CA 92093-0507, USA.
| | - J M Bowles
- University of California, San Diego, Department of Medicine, 9500 Gilman Dr, La Jolla, CA 92093-0507, USA; Centre on Drug Policy Evaluation, Li Ka Shing Knowledge Institute, St. Michael's Hospital, 209 Victoria St., Toronto, Ontario M5B 3M6, Canada
| | - M Faul
- Health Systems and Trauma Systems Branch, Mailstop F-62, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| | - T L Gaines
- University of California, San Diego, Department of Medicine, 9500 Gilman Dr, La Jolla, CA 92093-0507, USA
| |
Collapse
|
2
|
Abstract
After participating in this activity, learners should be better able to:• Identify the effects of dysregulated opioid signalling in depression• Evaluate the use of opioid compounds and ketamine in patients with depression ABSTRACT: Major depressive disorder (MDD) remains one of the leading causes of disability and functional impairment worldwide. Current antidepressant therapeutics require weeks to months of treatment prior to the onset of clinical efficacy on depressed mood but remain ineffective in treating suicidal ideation and cognitive impairment. Moreover, 30%-40% of individuals fail to respond to currently available antidepressant medications. MDD is a heterogeneous disorder with an unknown etiology; novel strategies must be developed to treat MDD more effectively. Emerging evidence suggests that targeting one or more of the four opioid receptors-mu (MOR), kappa (KOR), delta (DOR), and the nociceptin/orphanin FQ receptor (NOP)-may yield effective therapeutics for stress-related psychiatric disorders. Furthermore, the effects of the rapidly acting antidepressant ketamine may involve opioid receptors. This review highlights dysregulated opioid signaling in depression, evaluates clinical trials with opioid compounds, and considers the role of opioid mechanisms in rapidly acting antidepressants.
Collapse
|
3
|
Rieder TN. Solving the Opioid Crisis Isn't Just a Public Health Challenge-It's a Bioethics Challenge. Hastings Cent Rep 2021; 50:24-32. [PMID: 33448415 DOI: 10.1002/hast.1169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Among those who discuss America's opioid crisis, it is popular to claim that we know what we, as a society, ought to do to solve the problem-we simply don't want it badly enough. We don't lack knowledge; we lack the will to act and to fund the right policies. In fact, I've heard two versions of this. Among those who focus on prescription opioids, it is clear that we ought to stop prescribing so many powerful opioid painkillers. And among my public health colleagues focusing on illicit drug use, it is clear that we ought to expand addiction treatment and harm-reduction services. The problem, however, is that the second claim is not obvious (and, indeed, is denied by many Americans), and the first claim probably isn't even true (at least, not in so crude a form). In short, the opioid crisis presents not only a problem of political will but also one of ethics. It will take work to discover or justify our normative claims in this arena.
Collapse
|
4
|
Zhang CH, Kim K, Jin Z, Zheng F, Zhan CG. Systematic Structure-Based Virtual Screening Approach to Antibody Selection and Design of a Humanized Antibody against Multiple Addictive Opioids without Affecting Treatment Agents Naloxone and Naltrexone. ACS Chem Neurosci 2021; 12:184-194. [PMID: 33356138 DOI: 10.1021/acschemneuro.0c00670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Opioid drug use, especially heroin, is known as a growing national crisis in America. Heroin itself is a prodrug and is converted to the most active metabolite 6-monoacetylmorphine (6-MAM) responsible for the acute toxicity of heroin and then to a relatively less-active metabolite morphine responsible for the long-term toxicity of heroin. Monoclonal antibodies (mAbs) are recognized as a potentially promising therapeutic approach in the treatment of opioid use disorders (OUDs). Due to the intrinsic challenges of discovering an mAb against multiple ligands, here we describe a general, systematic structure-based virtual screening and design approach which has been used to identify a known anti-morphine antibody 9B1 and a humanized antibody h9B1 capable of binding to multiple addictive opioids (including 6-MAM, morphine, heroin, and hydrocodone) without significant binding with currently available OUD treatment agents naloxone, naltrexone, and buprenorphine. The humanized antibody may serve as a promising candidate for the treatment of OUDs. The experimental binding affinities reasonably correlate with the computationally predicted binding free energies. The experimental activity data strongly support the computational predictions, suggesting that the systematic structure-based virtual screening and humanization design protocol is reliable. The general, systematic structure-based virtual screening and design approach will be useful for many other antibody selection and design efforts in the future.
Collapse
Affiliation(s)
- Chun-Hui Zhang
- Molecular Modeling and Biopharmaceutical Center and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536, United States
| | - Kyungbo Kim
- Molecular Modeling and Biopharmaceutical Center and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536, United States
| | - Zhenyu Jin
- Molecular Modeling and Biopharmaceutical Center and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536, United States
| | - Fang Zheng
- Molecular Modeling and Biopharmaceutical Center and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536, United States
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536, United States
| |
Collapse
|
5
|
Calarco CA, Lobo MK. Depression and substance use disorders: Clinical comorbidity and shared neurobiology. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:245-309. [PMID: 33648671 DOI: 10.1016/bs.irn.2020.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mood disorders, including major depressive disorder (MDD), are the most prevalent psychiatric illnesses, and pose an incredible burden to society, both in terms of disability and in terms of costs associated with medical care and lost work time. MDD has extremely high rates of comorbidity with substance use disorders (SUD) as many of the same neurobiological circuits and molecular mechanisms regulate the reward pathways disrupted in both conditions. MDD may induce SUDs, SUD may contribute to MDD development, or underlying vulnerabilities and common life experience may confer risk to developing both conditions. In this chapter we explore theories of MDD and SUD comorbidity, the neurobiological underpinnings of depression, overlapping cellular and molecular pathways for both conditions, and current treatment approaches for these comorbid conditions.
Collapse
Affiliation(s)
- Cali A Calarco
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
6
|
Jacobson ML, Browne CA, Lucki I. Kappa Opioid Receptor Antagonists as Potential Therapeutics for Stress-Related Disorders. Annu Rev Pharmacol Toxicol 2020; 60:615-636. [DOI: 10.1146/annurev-pharmtox-010919-023317] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Exposure to stressful stimuli activates kappa opioid receptor (KOR) signaling, a process known to produce aversion and dysphoria in humans and other species. This endogenous opioid system is dysregulated in stress-related disorders, specifically in major depressive disorder (MDD). These findings serve as the foundation for a growing interest in the therapeutic potential of KOR antagonists as novel antidepressants. In this review, data supporting the hypothesis of dysregulated KOR function in MDD are considered. The clinical data demonstrating the therapeutic efficacy and safety of selective and mixed opioid antagonists are then presented. Finally, the preclinical evidence illustrating the induction of behaviors relevant to the endophenotypes of MDD and KOR antagonist activity in stress-naïve and stress-exposed animals is evaluated. Overall, this review highlights the emergent literature supporting the pursuit of KOR antagonists as novel therapeutics for MDD and other stress-related disorders.
Collapse
Affiliation(s)
- Moriah L. Jacobson
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Caroline A. Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| |
Collapse
|
7
|
Zheng Y, Obeng S, Reinecke BA, Chen C, Phansalkar PS, Walentiny DM, Gerk PM, Liu-Chen LY, Selley DE, Beardsley PM, Zhang Y. Pharmacological characterization of 17-cyclopropylmethyl-3,14-dihydroxy-4,5-epoxy-6-[(3'-fluoro-4'-pyridyl)acetamido]morphinan (NFP) as a dual selective MOR/KOR ligand with potential applications in treating opioid use disorder. Eur J Pharmacol 2019; 865:172812. [PMID: 31743739 PMCID: PMC6914219 DOI: 10.1016/j.ejphar.2019.172812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 11/07/2019] [Accepted: 11/14/2019] [Indexed: 01/16/2023]
Abstract
For thousands of years opioids have been the first-line treatment option for pain management. However, the tolerance and addiction potential of opioids limit their applications in clinic. NFP, a MOR/KOR dual-selective opioid antagonist, was identified as a ligand that significantly antagonized the antinociceptive effects of morphine with lesser withdrawal effects than naloxone at similar doses. To validate the potential application of NFP in opioid addiction treatment, a series of in vitro and in vivo assays were conducted to further characterize its pharmacological profile. In calcium mobilization assays and MOR internalization studies, NFP showed the apparent capacity to antagonize DAMGO-induced calcium flux and etorphine-induced MOR internalization. In contrast to the opioid agonists DAMGO and morphine, cells pretreated with NFP did not show apparent desensitization and down regulation of the MOR. Though in vitro bidirectional transport studies showed that NFP might be a P-gp substrate, in warm-water tail-withdrawal assays it was able to antagonize the antinociceptive effects of morphine indicating its potential central nervous system activity. Overall these results suggest that NFP is a promising dual selective opioid antagonist that may have the potential to be used therapeutically in opioid use disorder treatment.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Biological Transport
- CHO Cells
- Caco-2 Cells
- Calcium/metabolism
- Cell Line, Tumor
- Cricetulus
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Humans
- Ligands
- Male
- Mice, Inbred C57BL
- Morphinans/pharmacology
- Narcotic Antagonists/pharmacology
- Opioid-Related Disorders/drug therapy
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Yi Zheng
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, VA, 23298, United States
| | - Samuel Obeng
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, VA, 23298, United States
| | - Bethany A Reinecke
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, VA, 23298, United States
| | - Chongguang Chen
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, United States
| | - Palak S Phansalkar
- Department of Pharmaceutics, Virginia Commonwealth University, 410 North 12th Street, Richmond, VA, 23298, United States
| | - David M Walentiny
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay Street, Richmond, VA, 23298, United States
| | - Phillip M Gerk
- Department of Pharmaceutics, Virginia Commonwealth University, 410 North 12th Street, Richmond, VA, 23298, United States
| | - Lee-Yuan Liu-Chen
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, United States
| | - Dana E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay Street, Richmond, VA, 23298, United States
| | - Patrick M Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay Street, Richmond, VA, 23298, United States; Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, 1112 East Clay Street, Richmond, VA, 23298, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, VA, 23298, United States.
| |
Collapse
|
8
|
Obeng S, Jali A, Zheng Y, Wang H, Schwienteck KL, Chen C, Stevens DL, Akbarali HI, Dewey WL, Banks ML, Liu-Chen LY, Selley DE, Zhang Y. Characterization of 17-Cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(indole-7-carboxamido)morphinan (NAN) as a Novel Opioid Receptor Modulator for Opioid Use Disorder Treatment. ACS Chem Neurosci 2019; 10:2518-2532. [PMID: 30758946 PMCID: PMC6520168 DOI: 10.1021/acschemneuro.9b00038] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The opioid crisis is a significant public health issue with more than 115 people dying from opioid overdose per day in the United States. The aim of the present study was to characterize the in vitro and in vivo pharmacological effects of 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(indole-7-carboxamido)morphinan (NAN), a μ opioid receptor (MOR) ligand that may be a potential candidate for opioid use disorder treatment that produces less withdrawal signs than naltrexone. The efficacy of NAN was compared to varying efficacy ligands at the MOR, and determined at the δ opioid receptor (DOR) and κ opioid receptor (KOR). NAN was identified as a low efficacy partial agonist for G-protein activation at the MOR and DOR, but had relatively high efficacy at the KOR. In contrast to high efficacy MOR agonists, NAN did not induce MOR internalization, downregulation, or desensitization, but it antagonized agonist-induced MOR internalization and stimulation of intracellular Ca2+ release. Opioid withdrawal studies conducted using morphine-pelleted mice demonstrated that NAN precipitated significantly less withdrawal signs than naltrexone at similar doses. Furthermore, NAN failed to produce fentanyl-like discriminative stimulus effects in rats up to doses that produced dose- and time-dependent antagonism of fentanyl. Overall, these results provide converging lines of evidence that NAN functions mainly as a MOR antagonist and support further consideration of NAN as a candidate medication for opioid use disorder treatment.
Collapse
Affiliation(s)
- Samuel Obeng
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Abdulmajeed Jali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Yi Zheng
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Huiqun Wang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Kathryn L. Schwienteck
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Chongguang Chen
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - David L. Stevens
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Hamid I. Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - William L. Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Mathew L. Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Lee-Yuan Liu-Chen
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Dana E. Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| |
Collapse
|
9
|
Blocking drug activation as a therapeutic strategy to attenuate acute toxicity and physiological effects of heroin. Sci Rep 2018; 8:16762. [PMID: 30425345 PMCID: PMC6233155 DOI: 10.1038/s41598-018-35196-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/28/2018] [Indexed: 11/09/2022] Open
Abstract
Heroin is a growing national crisis in America. There is an increasing frequency of heroin overdoses. All of the currently used therapeutic approaches to treatment of heroin abuse and other opioid drugs of abuse focus on antagonizing a brain receptor (particularly µ-opiate receptors). However, it has been known that the therapeutic use of certain µ-opiate receptor antagonist may actually increase heroin overdose. Once overdosed, heroin addicts may continue to get overdosed again and again until fatal. Here we report our design and validation of a novel therapeutic strategy targeting heroin activation based on our analysis of the chemical transformation and functional change of heroin in the body. An effective blocker of heroin activation, such as ethopropazine tested in this study, may be used as a standalone therapy or in combination with a currently available, traditional medications targeting µ-opiate receptors (e.g. naltrexone or its extended-release formulation Vivitrol). The combination therapy would be ideal for heroin abuse treatment as the effects of two therapeutic agents targeting two independent mechanisms are cooperative.
Collapse
|
10
|
Molero Y, Zetterqvist J, Binswanger IA, Hellner C, Larsson H, Fazel S. Medications for Alcohol and Opioid Use Disorders and Risk of Suicidal Behavior, Accidental Overdoses, and Crime. Am J Psychiatry 2018; 175:970-978. [PMID: 30068260 PMCID: PMC6169735 DOI: 10.1176/appi.ajp.2018.17101112] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The authors examined associations between medications for alcohol and opioid use disorders (acamprosate, naltrexone, methadone, and buprenorphine) and suicidal behavior, accidental overdoses, and crime. METHOD In this total population cohort study, 21,281 individuals who received treatment with at least one of the four medications between 2005 and 2013 were identified. Data on medication use and outcomes were collected from Swedish population-based registers. A within-individual design (using stratified Cox proportional hazards regression models) was used to compare rates of suicidal behavior, accidental overdoses, and crime for the same individuals during the period when they were receiving the medication compared with the period when they were not. RESULTS No significant associations with any of the primary outcomes were found for acamprosate. For naltrexone, there was a reduction in the hazard ratio for accidental overdoses during periods when individuals received treatment compared with periods when they did not (hazard ratio=0.82, 95% CI=0.70, 0.96). Buprenorphine was associated with reduced arrest rates for all crime categories (i.e., violent, nonviolent, and substance-related) as well as reduction in accidental overdoses (hazard ratio=0.75, 95% CI=0.60, 0.93). For methadone, there were significant reductions in the rate of suicidal behaviors (hazard ratio=0.60, 95% CI=0.40-0.88) as well as reductions in all crime categories. However, there was an increased risk for accidental overdoses among individuals taking methadone (hazard ratio=1.25, 95% CI=1.13, 1.38). CONCLUSIONS Medications currently used to treat alcohol and opioid use disorders also appear to reduce suicidality and crime during treatment.
Collapse
Affiliation(s)
- Yasmina Molero
- From the Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom; the Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm; the Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Norra Stationsgatan, Stockholm; the Institute for Health Research, Kaiser Permanente Colorado, Denver; the Division of General Internal Medicine, University of Colorado School of Medicine, Aurora; and the School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Johan Zetterqvist
- From the Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom; the Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm; the Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Norra Stationsgatan, Stockholm; the Institute for Health Research, Kaiser Permanente Colorado, Denver; the Division of General Internal Medicine, University of Colorado School of Medicine, Aurora; and the School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Ingrid A Binswanger
- From the Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom; the Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm; the Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Norra Stationsgatan, Stockholm; the Institute for Health Research, Kaiser Permanente Colorado, Denver; the Division of General Internal Medicine, University of Colorado School of Medicine, Aurora; and the School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Clara Hellner
- From the Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom; the Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm; the Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Norra Stationsgatan, Stockholm; the Institute for Health Research, Kaiser Permanente Colorado, Denver; the Division of General Internal Medicine, University of Colorado School of Medicine, Aurora; and the School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Henrik Larsson
- From the Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom; the Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm; the Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Norra Stationsgatan, Stockholm; the Institute for Health Research, Kaiser Permanente Colorado, Denver; the Division of General Internal Medicine, University of Colorado School of Medicine, Aurora; and the School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Seena Fazel
- From the Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom; the Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm; the Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Norra Stationsgatan, Stockholm; the Institute for Health Research, Kaiser Permanente Colorado, Denver; the Division of General Internal Medicine, University of Colorado School of Medicine, Aurora; and the School of Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
11
|
Werle N, Zedillo E. We Can't Go Cold Turkey: Why Suppressing Drug Markets Endangers Society. THE JOURNAL OF LAW, MEDICINE & ETHICS : A JOURNAL OF THE AMERICAN SOCIETY OF LAW, MEDICINE & ETHICS 2018; 46:325-342. [PMID: 30146979 DOI: 10.1177/1073110518782942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This essay argues that policies aimed at suppressing drug use exacerbate the nation's opioid problem. It neither endorses drug use nor advocates legalizing the consumption and sale of all substances in all circumstances. Instead, it contends that trying to suppress drug markets is the wrong goal, and in the midst of an addiction crisis it can be deadly. There is no single, correct drug policy; the right approach depends crucially on the substance at issue, the patterns of use and supply, and the jurisdiction's culture, institutions, and material resources. Decriminalization is no panacea for a nation's drug problems. Nevertheless, either de jure or de facto decriminalization of personal drug possession is a necessary condition for mitigating this crisis.
Collapse
Affiliation(s)
- Nick Werle
- Nick Werle, J.D., is a graduate of Yale Law School, a research associate in the International Drug Policy Unit at the London School of Economics and Political Science, and a fellow at Yale's Solomon Center for Health Law and Policy. He received an MSc in economic policy from University College London and an MSc in risk and finance from the London School of Economics and Political Science with support from the U.K.'s Marshall Scholarship. Ernesto Zedillo, Ph.D., is the director of the Yale Center for the Study of Globalization, professor in the field of international economics and politics, and a member of the Global Commission on Drug Policy. He received his M.A. and Ph.D. in economics from Yale University. He was the president of Mexico from 1994-2000
| | - Ernesto Zedillo
- Nick Werle, J.D., is a graduate of Yale Law School, a research associate in the International Drug Policy Unit at the London School of Economics and Political Science, and a fellow at Yale's Solomon Center for Health Law and Policy. He received an MSc in economic policy from University College London and an MSc in risk and finance from the London School of Economics and Political Science with support from the U.K.'s Marshall Scholarship. Ernesto Zedillo, Ph.D., is the director of the Yale Center for the Study of Globalization, professor in the field of international economics and politics, and a member of the Global Commission on Drug Policy. He received his M.A. and Ph.D. in economics from Yale University. He was the president of Mexico from 1994-2000
| |
Collapse
|
12
|
Obeng S, Yuan Y, Jali A, Selley DE, Zhang Y. In vitro and in vivo functional profile characterization of 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3-carboxamido)morphinan (NAQ) as a low efficacy mu opioid receptor modulator. Eur J Pharmacol 2018; 827:32-40. [PMID: 29530590 PMCID: PMC5890425 DOI: 10.1016/j.ejphar.2018.03.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 11/25/2022]
Abstract
Evidence has shown that downstream signaling by mu opioid receptor (MOR) agonists that recruit β-arrestin2 may lead to the development of tolerance. Also, it has been suggested that opioid receptor desensitization and cyclic AMP overshoot contributes to the development of tolerance and occurrence of withdrawal, respectively. Therefore, studies were conducted with 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3-carboxamido)morphinan (NAQ), a MOR selective partial agonist discovered in our laboratory, to characterize its effect on β-arrestin2 recruitment and precipitation of a cyclic AMP overshoot. DAMGO, a MOR full agonist dose-dependently increased β-arrestin2 association with the MOR, whereas NAQ did not. Moreover, NAQ displayed significant, concentration-dependent antagonism of DAMGO-induced β-arrestin2 recruitment. After prolonged morphine treatment of mMOR-CHO cells, there was a significant overshoot of cAMP upon exposure to naloxone, but not NAQ. Moreover, prolonged incubation of mMOR-CHO cells with NAQ did not result in desensitization nor downregulation of the MOR. In functional studies comparing NAQ with nalbuphine in the cAMP inhibition, Ca2+ flux and [35S]GTPγS binding assays, NAQ did not show agonism in the Ca2+ flux assay but showed partial agonism in the cAMP and [35S]GTPγS assays. Also, NAQ significantly antagonized DAMGO-induced intracellular Ca2+ increase. In conclusion, NAQ is a low efficacy MOR modulator that lacks β-arrestin2 recruitment function and does not induce cellular hallmarks of MOR adaptation and fails to precipitate a cellular manifestation of withdrawal in cells pretreated with morphine. These characteristics are desirable if NAQ is pursued for opioid abuse treatment development.
Collapse
Affiliation(s)
- Samuel Obeng
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, P.O. Box 980540, Richmond, VA 23298, United States
| | - Yunyun Yuan
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, P.O. Box 980540, Richmond, VA 23298, United States
| | - Abdulmajeed Jali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay Street, Richmond, VA 23298, United States
| | - Dana E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay Street, Richmond, VA 23298, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, P.O. Box 980540, Richmond, VA 23298, United States.
| |
Collapse
|
13
|
Ma SX, Kwon SH, Seo JY, Hwang JY, Hong SI, Kim HC, Lee SY, Jang CG. Impairment of opiate-mediated behaviors by the selective TRPV1 antagonist SB366791. Addict Biol 2017; 22:1817-1828. [PMID: 27730727 DOI: 10.1111/adb.12460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 12/29/2022]
Abstract
Transient receptor potential vanilloid type 1 (TRPV1), the archetypal member of the vanilloid TRP family, was initially identified as the receptor for capsaicin, the pungent ingredient in hot chili peppers. We previously demonstrated that TRPV1 in the dorsal striatum significantly contributes to morphine reward by using the conditioned place preference paradigm in mice; however, it is unknown whether TRPV1 has the same effect in other reward models. In this study, we investigated the role of TRPV1 in morphine reward by using a self-administration paradigm in rats. We found that treatment with a selective TRPV1 antagonist, SB366791, significantly decreased morphine self-administration on a fixed-ratio 1 schedule or a progressive ratio schedule of reinforcement. In addition, treatment with another selective TRPV1 antagonist, AMG9810, not only significantly prevented morphine self-administration but also prevented morphine-induced c-fos expression in the nucleus accumbens. Furthermore, administration of SB366791 decreased an anxiolytic-like effect during the morphine abstinence period. Moreover, treatment with SB366791 significantly decreased morphine-priming reinstatement. Taken together, our findings suggest that blockade of TRPV1 receptors could provide an approach to limiting morphine addiction.
Collapse
Affiliation(s)
- Shi-Xun Ma
- Department of Pharmacology, School of Pharmacy; Sungkyunkwan University; Korea
| | - Seung-Hwan Kwon
- Department of Pharmacology, School of Pharmacy; Sungkyunkwan University; Korea
| | - Jee-Yeon Seo
- Department of Pharmacology, School of Pharmacy; Sungkyunkwan University; Korea
| | - Ji-Young Hwang
- Department of Pharmacology, School of Pharmacy; Sungkyunkwan University; Korea
| | - Sa-Ik Hong
- Department of Pharmacology, School of Pharmacy; Sungkyunkwan University; Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Neurotoxicology Program, College of Pharmacy; Kangwon National University; Korea
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy; Sungkyunkwan University; Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy; Sungkyunkwan University; Korea
| |
Collapse
|
14
|
Binding mode analyses of NAP derivatives as mu opioid receptor selective ligands through docking studies and molecular dynamics simulation. Bioorg Med Chem 2017; 25:2463-2471. [PMID: 28302509 DOI: 10.1016/j.bmc.2017.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/27/2017] [Accepted: 03/03/2017] [Indexed: 01/02/2023]
Abstract
Mu opioid receptor selective antagonists are highly desirable because of their utility as pharmacological probes for receptor characterization and functional studies. Furthermore, the mu opioid receptors act as an important target in drug abuse and addiction treatment. Previously, we reported NAP as a novel lead compound with high selectivity and affinity towards the mu opioid receptor. Based on NAP, we have synthesized its derivatives and further characterized their binding affinities and selectivity towards the receptor. NMP and NGP were identified as the two most selective MOR ligands among NAP derivatives. In the present study, molecular modeling methods were applied to assess the dual binding modes of NAP derivatives, particularly on NMP and NGP, in three opioid receptors, in order to analyze the effects of structural modifications on the pyridyl ring of NAP on the binding affinity and selectivity. The results indicated that the steric hindrance, electrostatic, and hydrophobic effects caused by the substituents on the pyridyl ring of NAP contributed complimentarily on the binding affinity and selectivity of NAP derivatives to three opioid receptors. Analyses of these contributions provided insights on future design of more potent and selective mu opioid receptor ligands.
Collapse
|
15
|
Digiusto E, Shakeshaft AP, Ritter A, Mattick RP, White J, Lintzeris N, Bell J, Saunders JB. Effects of Pharmacotherapies for Opioid Dependence on Participants' Criminal Behaviour and Expenditure on Illicit Drugs: An Australian National Evaluation (NEPOD). ACTA ACUST UNITED AC 2016. [DOI: 10.1375/acri.39.2.171] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Erol Digiusto
- National Centre in HIV Social Research, University of New South Wales, Australia
- National Drug and Alcohol Research Centre, University of New South Wales, Australia
| | | | - Alison Ritter
- Turning Point Alcohol and Drug Centre Inc., Melbourne,Victoria, Australia
| | - Richard P. Mattick
- National Drug and Alcohol Research Centre, University of New South Wales, Australia
| | - Jason White
- Drug and Alcohol Services Council of South Australia, Parkside, South Australia, Australia
| | - Nicholas Lintzeris
- National Drug and Alcohol Research Centre, University of New South Wales, Australia
- National Addiction Centre, Institute of Psychiatry, Kings College, London, United Kingdom
| | - James Bell
- The Langton Centre, Surry Hills, New South Wales, Australia
| | - John B. Saunders
- Mental Health Centre, Royal Brisbane Hospital, Herston, Queensland,Australia
| | | |
Collapse
|
16
|
Abstract
Medication-assisted treatment of opioid use disorder with physiological dependence at least doubles rates of opioid-abstinence outcomes in randomized, controlled trials comparing psychosocial treatment of opioid use disorder with medication versus with placebo or no medication. This article reviews the current evidence for medication-assisted treatment of opioid use disorder and also presents clinical practice imperatives for preventing opioid overdose and the transmission of infectious disease. The evidence strongly supports the use of agonist therapies to reduce opioid use and to retain patients in treatment, with methadone maintenance remaining the gold standard of care. Combined buprenorphine/naloxone, however, also demonstrates significant efficacy and favorable safety and tolerability in multiple populations, including youth and prescription opioid-dependent individuals, as does buprenorphine monotherapy in pregnant women. The evidence for antagonist therapies is weak. Oral naltrexone demonstrates poor adherence and increased mortality rates, although the early evidence looks more favorable for extended-release naltrexone, which has the advantages that it is not subject to misuse or diversion and that it does not present a risk of overdose on its own. Two perspectives-individualized treatment and population management-are presented for selecting among the three available Food and Drug Administration-approved maintenance therapies for opioid use disorder. The currently unmet challenges in treating opioid use disorder are discussed, as are the directions for future research.
Collapse
|
17
|
Li QQ, Sun CY, Luo YX, Xue YX, Meng SQ, Xu LZ, Chen N, Deng JH, Zhai HF, Kosten TR, Shi J, Lu L, Sun HQ. A conjugate vaccine attenuates morphine- and heroin-induced behavior in rats. Int J Neuropsychopharmacol 2015; 18:pyu093. [PMID: 25522425 PMCID: PMC4376548 DOI: 10.1093/ijnp/pyu093] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Currently approved medications for opioid addiction have shown clinical efficacy, but undesired side effects, dependence induced by the medications themselves, and low treatment compliance necessitate the need for novel therapies. METHODS A novel morphine-keyhole limpet hemocyanin conjugate vaccine was synthesized with 6-glutarylmorphine as the hapten and a lengthened linker of 6 carbon atoms. The titer and specificity of the triggered antibody were assessed by enzyme-linked immunosorbent assay. The effects of the vaccine on the morphine-induced elevation of dopamine levels in the nucleus accumbens were determined by high-performance liquid chromatography. The effects of the vaccine on morphine-induced locomotor sensitization and heroin-primed reinstatement of heroin self-administration were also assessed. RESULTS After subcutaneous administration in rats, the vaccine triggered a high antibody titer, with comparable specificity for morphine, 6-acetylmorphine, and heroin, but no interaction with dissimilar therapeutic opioid compounds, including buprenorphine, naloxone, and nalorphine, was observed. The vaccine significantly prevented the elevation of dopamine levels in the nucleus accumbens induced by a single morphine challenge. Moreover, the vaccine prevented the expression of morphine-induced locomotor sensitization and heroin-primed reinstatement of heroin seeking, suggesting its potential for preventing relapse. CONCLUSION These results demonstrate that active immunization with the present vaccine induces a robust morphine/heroin-specific antibody response in rats and attenuates the behavioral effects of morphine and heroin.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Lin Lu
- Peking University Sixth Hospital/Institute of Mental Health (Dr Li, Ms C.-Y. Sun, Dr Luo, Ms Meng, Mr Xu, Ms Deng, Drs Lu, and H.-Q. Sun), and National Institute on Drug Dependence, Peking University, Beijing, China (Drs Dr Li, Ms C.-Y. Sun, Dr Luo, Dr Xue, Ms Meng, Mr Xu, Ms Chen, Ms Deng, Drs Zhai, Shi, and Lu); Institute for Food and Drug Safety Evaluation, National Institutes for Food and Drug Control, Beijing, China (Dr Li); Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China (Drs Lu and H.-Q. Sun); Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas (Dr Kosten)
| | - Hong-Qiang Sun
- Peking University Sixth Hospital/Institute of Mental Health (Dr Li, Ms C.-Y. Sun, Dr Luo, Ms Meng, Mr Xu, Ms Deng, Drs Lu, and H.-Q. Sun), and National Institute on Drug Dependence, Peking University, Beijing, China (Drs Dr Li, Ms C.-Y. Sun, Dr Luo, Dr Xue, Ms Meng, Mr Xu, Ms Chen, Ms Deng, Drs Zhai, Shi, and Lu); Institute for Food and Drug Safety Evaluation, National Institutes for Food and Drug Control, Beijing, China (Dr Li); Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China (Drs Lu and H.-Q. Sun); Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas (Dr Kosten)
| |
Collapse
|
18
|
|
19
|
Zhang Y, Braithwaite A, Yuan Y, Streicher JM, Bilsky EJ. Behavioral and cellular pharmacology characterization of 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3'-carboxamido)morphinan (NAQ) as a mu opioid receptor selective ligand. Eur J Pharmacol 2014; 736:124-30. [PMID: 24815322 PMCID: PMC4073486 DOI: 10.1016/j.ejphar.2014.04.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 04/28/2014] [Indexed: 12/26/2022]
Abstract
Mu opioid receptor (MOR) selective antagonists and partial agonists have been used for the treatment of opioid abuse and addiction. Our recent efforts on the identification of MOR antagonists have provided several novel leads displaying interesting pharmacological profiles. Among them, 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-[(3'-isoquinolyl)acetamido]morphinan (NAQ) showed sub-nanomolar binding affinity to the MOR with significant selectivity over the delta opioid receptor (DOR) and the kappa opioid receptor (KOR). Its central nervous system penetration capacity together with marginal agonism in the MOR-GTPγS binding assay made it a very interesting molecule for developing novel opioid abuse and addiction therapeutic agents. Therefore, further pharmacological characterization was conducted to fully understand its biological profile. At the molecular and cellular level, NAQ not only induced no translocation of β-arrestin2 to the MOR, but also efficaciously antagonized the effect of DAMGO in MOR-βarr2eGFP-U2OS cells in the β-arrestin2 recruitment assay. At the in vivo level, NAQ displayed a potent inhibition of the analgesic effect of morphine in the tail-flick assay (ID50=1.19 mg/kg). NAQ (10 mg/kg) also significantly decreased the hyper-locomotion induced by acute morphine without inducing any vertical jumps. Meanwhile NAQ precipitated lesser withdrawal symptoms in morphine dependent mice than naloxone. In conclusion, NAQ may represent a new chemical entity for opioid abuse and addiction treatment.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, P.O. Box 980540, Richmond, VA 23298, United States.
| | - Amanda Braithwaite
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, 11 Hills Beach Road, Biddeford, ME 04005, United States
| | - Yunyun Yuan
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, P.O. Box 980540, Richmond, VA 23298, United States
| | - John M Streicher
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, 11 Hills Beach Road, Biddeford, ME 04005, United States
| | - Edward J Bilsky
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, 11 Hills Beach Road, Biddeford, ME 04005, United States
| |
Collapse
|
20
|
Mannelli P, Wu LT, Peindl KS, Swartz MS, Woody GE. Extended release naltrexone injection is performed in the majority of opioid dependent patients receiving outpatient induction: a very low dose naltrexone and buprenorphine open label trial. Drug Alcohol Depend 2014; 138:83-8. [PMID: 24602363 PMCID: PMC4017322 DOI: 10.1016/j.drugalcdep.2014.02.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/22/2014] [Accepted: 02/01/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND The approval of extended release injectable naltrexone (XR-NTX; Vivitrol(®)) has introduced a new option for treating opioid addiction, but studies are needed to identify its place within the spectrum of available therapies. The absence of physiological opioid dependence is a necessary and challenging first step for starting XR-NTX. Outpatient detoxification gives poor results and inpatient detoxification is either unavailable or too brief for the physiological effects of opioids to resolve. Here we present findings from an open label study that tested whether the transition from opioid addiction to XR-NTX can be safely and effectively performed in an outpatient setting using very low dose naltrexone and buprenorphine. METHODS Twenty treatment seeking opioid addicted individuals were given increasing doses of naltrexone starting at 0.25mg with decreasing doses of buprenorphine starting at 4 mg during a 7-day outpatient XR-NTX induction procedure. Withdrawal discomfort, craving, drug use, and adverse events were assessed daily until the XR-NTX injection, then weekly over the next month. RESULTS Fourteen of the 20 participants received XR-NTX and 13 completed weekly assessments. Withdrawal, craving, and opioid or other drug use were significantly lower during induction and after XR-NTX administration compared with baseline, and no serious adverse events were recorded. CONCLUSIONS Outpatient transition to XR-NTX combining upward titration of very low dose naltrexone with downward titration of low dose buprenorphine was safe, well tolerated, and completed by most participants. Further studies with larger numbers of subjects are needed to see if this approach is useful for naltrexone induction.
Collapse
Affiliation(s)
- Paolo Mannelli
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States.
| | - Li-Tzy Wu
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| | - Kathleen S Peindl
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| | - Marvin S Swartz
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| | - George E Woody
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
21
|
Matthew-Simmons F, Ritter A. 'Miracle cure' or 'liquid handcuffs': reporting on naltrexone and methadone in the Australian print media. Drug Alcohol Rev 2014; 33:506-14. [PMID: 24635882 DOI: 10.1111/dar.12134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/10/2014] [Indexed: 10/25/2022]
Abstract
INTRODUCTION AND AIMS The news media is an important source of information regarding new developments in medicine and public health interventions. Previous research has indicated that in many cases, reporting on new treatments can be inaccurate or sensationalist. This paper presents analysis of Australian print media reporting on two treatment options for heroin dependence (naltrexone and methadone). The aim of this study was to quantitatively compare the volume and content of Australian print media reporting on these two treatments, one of which had a long history of use in Australia, and the other which was comparatively newer. DESIGN AND METHODS The study constituted a quantitative content analysis of a sample of 859 Australian newspaper articles, published over a 10-year period (1997-2007). Each article paragraph was coded for positive outcomes/benefits of treatment, as well as negative outcomes associated with treatment. RESULTS The analysis revealed that during this period, the Australian print media was significantly more likely to report the potential positive outcomes of naltrexone treatment, compared with the negative outcomes. In contrast, reporting on methadone focused more on the negative outcomes and side effects. DISCUSSION AND CONCLUSIONS The relative frequency by which the benefits of naltrexone were mentioned in this sample of news content is somewhat at odds with the extant efficacy and effectiveness research evidence. The findings suggest that reporting on these treatments in the Australian print media has not been balanced. This type of reporting has potential implications for public attitudes, as well as policy decisions.
Collapse
Affiliation(s)
- Francis Matthew-Simmons
- Drug Policy Modelling Program, National Drug and Alcohol Research Centre, University of New South Wales, Sydney, Australia
| | | |
Collapse
|
22
|
Hulse GK. Improving clinical outcomes for naltrexone as a management of problem alcohol use. Br J Clin Pharmacol 2013; 76:632-41. [PMID: 22946873 PMCID: PMC3853523 DOI: 10.1111/j.1365-2125.2012.04452.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 08/30/2012] [Indexed: 12/13/2022] Open
Abstract
Despite being a relatively effective and safe treatment, the clinical management of alcohol abuse/dependence by oral naltrexone can be compromised due to the patient's non-compliance with daily use of this medication. Over the past decade an increasing body of research has suggested that the use of sustained release depot naltrexone preparations can overcome this issue and deliver improved clinical outcomes. However, at the same time, research findings from diverse areas of pharmacogenetics, neurobiology and behavioural psychology have also been converging to identify variables including genetic markers, patient psychosocial characteristics and drug use history differences, or clusters of these variables that play a major role in mediating the response of alcohol abuse/dependent persons to treatment by naltrexone. While this article does not attempt to review all available data pertaining to an individual alcohol dependent patient's response to treatment by naltrexone, it does identify relevant research areas and highlights the importance of data arising from them. The characterization of clinical markers, to identify those patients who are most likely to benefit from naltrexone and to tailor a more individual naltrexone treatment, will ultimately provide significant benefit to both patients and clinicians by optimizing treatment outcome.
Collapse
Affiliation(s)
- Gary K Hulse
- Addiction Medicine, School of Psychiatry & Clinical Neurosciences (M521), The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
23
|
Health promotion and prevention strategies. Nurs Clin North Am 2013; 48:469-83, vii. [PMID: 23998773 DOI: 10.1016/j.cnur.2013.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opiate dependency is a medical disorder that requires treatment intervention. Primary health care not only entails treatment of illness but also involves disease prevention and health promotion. Based on Pender's revised Health Promotion Model, a descriptive study comparing the health promoting behaviors/practices in abusing and recovering opiate-dependent drug users is analyzed. Using the Health Promoting Lifestyle Profile II, a comparative descriptive, exploratory, nonexperimental design study was conducted to identify key health-promoting behaviors in recovering opiate-dependent drug users. Prevention strategy recommendations are discussed, along with future research recommendations.
Collapse
|
24
|
Binding mode characterization of 6α- and 6β-N-heterocyclic substituted naltrexamine derivatives via docking in opioid receptor crystal structures and site-directed mutagenesis studies: application of the 'message-address' concept in development of mu opioid receptor selective antagonists. Bioorg Med Chem 2013; 21:6405-13. [PMID: 24055076 DOI: 10.1016/j.bmc.2013.08.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/15/2013] [Accepted: 08/23/2013] [Indexed: 01/20/2023]
Abstract
Highly selective opioid receptor antagonists are essential pharmacological probes in opioid receptor structural characterization and opioid agonist functional studies. Currently, there is no highly selective, nonpeptidyl and reversible mu opioid receptor antagonist available. Among a series of naltrexamine derivatives that have been designed and synthesized, two compounds, NAP and NAQ, were previously identified as novel leads for this purpose based on their in vitro and in vivo pharmacological profiles. Both compounds displayed high binding affinity and selectivity to the mu opioid receptor. To further study the interaction of these two ligands with the three opioid receptors, the recently released opioid receptor crystal structures were employed in docking studies to further test our original hypothesis that the ligands recognize a unique 'address' domain in the mu opioid receptor involving Trp318 that facilitates their selectivity. These modeling results were supported by site-directed mutagenesis studies on the mu opioid receptor, where the mutants Y210A and W318A confirmed the role of the latter in binding. Such work not only enriched the 'message-address' concept, also facilitated our next generation ligand design and development.
Collapse
|
25
|
Zhang Y, Elbegdorj O, Yuan Y, Beletskaya IO, Selley DE. Opioid receptor selectivity profile change via isosterism for 14-O-substituted naltrexone derivatives. Bioorg Med Chem Lett 2013; 23:3719-22. [PMID: 23721804 DOI: 10.1016/j.bmcl.2013.05.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 05/01/2013] [Accepted: 05/07/2013] [Indexed: 11/24/2022]
Abstract
Isosterism is commonly used in drug discovery and development to address stability, selectivity, toxicity, pharmacokinetics, and efficacy issues. A series of 14-O-substituted naltrexone derivatives were identified as potent mu opioid receptor (MOR) antagonists with improved selectivity over the kappa opioid receptor (KOR) and the delta opioid receptor (DOR), compared to naltrexone. Since esters are not metabolically very stable under typical physiological conditions, their corresponding amide analogs were thus synthesized and biologically evaluated. Unlike their isosteres, most of these novel ligands seem to be dually selective for the MOR and the KOR over the DOR. The restricted flexibility of the amide bond linkage might be responsible for their altered selectivity profile. However, the majority of the 14-N-substituted naltrexone derivatives produced marginal or no MOR stimulation in the (35)S-GTP[γS] assay, which resembled their ester analogs. The current study thus indicated that the 14-substituted naltrexone isosteres are not bioisosteres since they have distinctive pharmacological profile with the regard to their opioid receptor binding affinity and selectivity.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA.
| | | | | | | | | |
Collapse
|
26
|
Abstract
Heroin addiction, a chronic relapsing disorder characterized by excessive drug taking and seeking, requires constant psychotherapeutic and pharmacotherapeutic interventions to minimize the potential for further abuse. Vaccine strategies against many drugs of abuse are being developed that generate antibodies that bind drug in the bloodstream, preventing entry into the brain and nullifying psychoactivity. However, this strategy is complicated by heroin's rapid metabolism to 6-acetylmorphine and morphine. We recently developed a "dynamic" vaccine that creates antibodies against heroin and its psychoactive metabolites by presenting multihaptenic structures to the immune system that match heroin's metabolism. The current study presents evidence of effective and continuous sequestration of brain-permeable constituents of heroin in the bloodstream following vaccination. The result is efficient blockade of heroin activity in treated rats, preventing various features of drugs of abuse: heroin reward, drug-induced reinstatement of drug seeking, and reescalation of compulsive heroin self-administration following abstinence in dependent rats. The dynamic vaccine shows the capability to significantly devalue the reinforcing and motivating properties of heroin, even in subjects with a history of dependence. In addition, targeting a less brain-permeable downstream metabolite, morphine, is insufficient to prevent heroin-induced activity in these models, suggesting that heroin and 6-acetylmorphine are critical players in heroin's psychoactivity. Because the heroin vaccine does not target opioid receptors or common opioid pharmacotherapeutics, it can be used in conjunction with available treatment options. Thus, our vaccine represents a promising adjunct therapy for heroin addiction, providing continuous heroin antagonism, requiring minimal medical monitoring and patient compliance.
Collapse
|
27
|
Kelty E, Hulse G. Examination of mortality rates in a retrospective cohort of patients treated with oral or implant naltrexone for problematic opiate use. Addiction 2012; 107:1817-24. [PMID: 22487087 DOI: 10.1111/j.1360-0443.2012.03910.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIMS To examine and compare mortality rates in patients treated with oral and implant naltrexone. DESIGN A retrospective cohort study. SETTING A community not-for-profit drug treatment clinic. PARTICIPANTS Patients treated with oral naltrexone (n = 2155, 17 207 patient-years) and implant naltrexone (n = 2389, 11 678 patient-years) for problematic opiate use between August 1997 and December 2009. MEASUREMENTS Crude gender, age, treatment period and cause-specific mortality rates were calculated using data obtained from the National Death Index. FINDINGS Crude mortality rates for patients treated with oral naltrexone [8.78 deaths per 1000 patient-years (ptpy), 95% confidence interval (CI): 7.38-10.17] were significantly different to those treated with implant naltrexone (6.59 ptpy, 95% CI: 5.13-8.06) (P = 0.0339). During the first 4 months following treatment, differences in the two groups were particularly apparent, with a mortality rate of 26.28 ptpy in patients treated with oral naltrexone compared to 7.34 ptpy in patients treated with implant naltrexone (P = 0.0003). Differences in initial mortality rates following treatment were associated predominantly with high rates of opiate overdoses in oral naltrexone patients during the first 4 months following treatment (17.22 ptpy compared with 0.67 ptpy in implant naltrexone patients) (P < 0.0001). CONCLUSIONS The use of implant naltrexone can reduce all-cause mortality and opiate overdose during the first 4 months following treatment compared with patients treated with oral naltrexone.
Collapse
Affiliation(s)
- Erin Kelty
- School of Psychiatry and Clinical Neuroscience, University of Western Australia.
| | | |
Collapse
|
28
|
Viswanath B, Chand P, Benegal V, Murthy P. Agonist treatment in opioid use: advances and controversy. Asian J Psychiatr 2012; 5:125-31. [PMID: 22813654 DOI: 10.1016/j.ajp.2012.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 01/12/2012] [Accepted: 01/19/2012] [Indexed: 10/28/2022]
Abstract
Opioid dependence is a chronic relapsing condition which requires comprehensive care; pharmacological agents form the mainstay of its long term treatment. The two most popular approaches are the harm reduction method using agonists and the complete abstinence method using antagonists. Currently, particularly from the harm minimization perspective and the low feasibility of an abstinence based approach, there is an increasing trend toward agonist treatment. The use of buprenorphine has gained popularity in view of its safety profile and the availability of the buprenorphine-naloxone combination has made it popular as a take-home treatment. This review outlines the pharmacological advances and controversies in this area.
Collapse
Affiliation(s)
- Biju Viswanath
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore 560 029, India.
| | | | | | | |
Collapse
|
29
|
Lobmaier PP, Kunøe N, Gossop M, Waal H. Naltrexone depot formulations for opioid and alcohol dependence: a systematic review. CNS Neurosci Ther 2012; 17:629-36. [PMID: 21554565 DOI: 10.1111/j.1755-5949.2010.00194.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Naltrexone is an opioid receptor antagonist that blocks the reinforcing effects of opioids and reduces alcohol consumption and craving. It has no abuse potential, mild and transient side effects, and thus appears an ideal pharmacotherapy for opioid dependence. Its effectiveness in alcohol dependence is less evident, but compliance with naltrexone combined with psychosocial support has been repeatedly shown to improve drinking outcomes. Limited compliance with oral naltrexone treatment is a known drawback. Several naltrexone implant and injectable depot formulations are being investigated and provide naltrexone release for at least 1 month. Studies among opioid-dependent patients indicate significant reductions in heroin use, but sample sizes are usually small. In alcohol dependence, two large multicenter trials report alcohol and craving reductions for naltrexone and placebo groups, indicating a significant but moderate effect. The pharmacokinetic profile of the injectable formulation indicates reliable naltrexone release over 1 month at therapeutic levels. Implant formulations releasing naltrexone up to 7 months are reported. Findings on safety and tolerability confirm the generally mild adverse effects described for naltrexone tablets. However, further research on therapeutic levels (i.e., opioid blocking) is warranted. The majority of naltrexone implants lacks approval for regular clinical use and larger longitudinal studies are needed. The available naltrexone depot formulations have the potential to significantly improve medication compliance in opioid and alcohol dependence. In certain circumstances, they may constitute a promising new treatment option.
Collapse
|
30
|
Taylor R, Raffa RB, Pergolizzi JV. Naltrexone extended-release injection: an option for the management of opioid abuse. Subst Abuse Rehabil 2011; 2:219-26. [PMID: 24474859 PMCID: PMC3846318 DOI: 10.2147/sar.s17920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The United States Food and Drug Administration (FDA) approved naltrexone, a synthetic competitive antagonist at opioid receptors, in oral form in 1984 for use in the management of opioid abuse and addiction. Because naltrexone and its major metabolite, 6-β-naltrexone, are both competitive antagonists at opioid receptors - and thereby inhibit opioid agonist-induced effects including those desired by abusers - it was hypothesized that once maintained on naltrex-one, opioid-induced desirable effects would be diminished to the point that relapse to illicit use would decline because it was no longer rewarding. However, good medication compliance is a requisite for such a strategy to be effective and a systematic review of oral naltrexone concluded that this method of treatment was not superior for any outcomes measured (ie, retention, abstinence, or side effects) to placebo, psychotherapy, benzodiazepines, or buprenorphine treatment. In addition, the retention rate on oral naltrexone was very low (less than 30%). Recently, the FDA approved an extended-release formulation (intramuscular depot injection) of naltrexone for prevention of relapse to opioid dependence following opioid detoxification and to be used along with counseling and social support. Since it needs to be administered only monthly, as opposed to the daily administration required for the oral formulation, naltrexone injection has the potential for increasing adherence and retention rates. Concerns include liver damage at high doses (oral formulation) and possible opioid overdose if an attempt is made to surmount receptor antagonism by taking higher doses of an opioid agonist or if opioid receptors become "sensitized" under long-term antagonism. The focus of the present review is the current information regarding the safety and efficacy of naltrexone extended-release therapy.
Collapse
Affiliation(s)
| | - Robert B Raffa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Joseph V Pergolizzi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology, Georgetown University School of Medicine, Washington, DC, USA
| |
Collapse
|
31
|
Ling W, Mooney L, Zhao M, Nielsen S, Torrington M, Miotto K. Selective review and commentary on emerging pharmacotherapies for opioid addiction. Subst Abuse Rehabil 2011; 2:181-8. [PMID: 24474855 PMCID: PMC3846315 DOI: 10.2147/sar.s22782] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Pharmacotherapies for opioid addiction under active development in the US include lofexidine (primarily for managing withdrawal symptoms) and Probuphine®, a distinctive mode of delivering buprenorphine for six months, thus relieving patients, clinicians, and regulatory personnel from most concerns about diversion, misuse, and unintended exposure in children. In addition, two recently approved formulations of previously proven medications are in early phases of implementation. The sublingual film form of buprenorphine + naloxone (Suboxone®) provides a less divertible, more quickly administered, more child-proof version than the buprenorphine + naloxone sublingual tablet. The injectable depot form of naltrexone (Vivitrol®) ensures consistent opioid receptor blockade for one month between administrations, removing concerns about medication compliance. The clinical implications of these developments have attracted increasing attention from clinicians and policymakers in the US and around the world, especially given that human immunodeficiency virus/acquired immunodeficiency syndrome and other infectious diseases are recognized as companions to opioid addiction, commanding more efforts to reduce opioid addiction. While research and practice improvement efforts continue, reluctance to adopt new medications and procedures can be expected, especially considerations in the regulatory process and in the course of implementation. Best practices and improved outcomes will ultimately emerge from continued development efforts that reflect input from many quarters.
Collapse
Affiliation(s)
- Walter Ling
- Integrated Substance Abuse Programs, University of California, Los Angeles, CA, USA
| | - Larissa Mooney
- Integrated Substance Abuse Programs, University of California, Los Angeles, CA, USA
| | - Min Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suzanne Nielsen
- Integrated Substance Abuse Programs, University of California, Los Angeles, CA, USA
| | - Matthew Torrington
- Integrated Substance Abuse Programs, University of California, Los Angeles, CA, USA
| | - Karen Miotto
- Integrated Substance Abuse Programs, University of California, Los Angeles, CA, USA
| |
Collapse
|
32
|
Nunes EV, Rothenberg JL, Sullivan MA, Carpenter KM, Kleber HD. Behavioral Therapy to Augment Oral Naltrexone for Opioid Dependence: A Ceiling on Effectiveness? THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2009; 32:503-17. [PMID: 17127538 DOI: 10.1080/00952990600918973] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The effectiveness of antagonist maintenance with oral naltrexone for opioid dependence has been limited by high dropout rates. Behavioral Naltrexone Therapy (BNT) was developed to improve retention on oral naltrexone by integrating voucher incentives, Motivational and Cognitive Behavioral therapies, and a significant other for monitoring medication adherence. In a 6-month, randomized, controlled trial in heroin dependent patients, BNT (N = 36) improved retention in treatment compared to a standard treatment control (Compliance Enhancement (CE); N = 33) (log rank = 4.28; p = .04). Most patients retained beyond 3 months achieved abstinence from opioids, but retention at 6 months was only 22% on BNT and 9% on CE. A systematic review of related controlled trials revealed similar effect sizes in the small to medium range, and substantial dropout. There may be a limit on the extent to which behavioral therapy can overcome poor adherence to oral naltrexone. Future research should consider combinations of behavioral methods with new long-acting injectable or implantable naltrexone formulations.
Collapse
Affiliation(s)
- Edward V Nunes
- New York State Psychiatric Institute, Division on Substance Abuse, New York, New York 10032, USA.
| | | | | | | | | |
Collapse
|
33
|
Wright IV C, Schnoll S, Bernstein D. Risk Evaluation and Mitigation Strategies for Drugs with Abuse Liability. Ann N Y Acad Sci 2008; 1141:284-303. [DOI: 10.1196/annals.1441.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
34
|
Farid W, Dunlop S, Tait R, Hulse G. The effects of maternally administered methadone, buprenorphine and naltrexone on offspring: review of human and animal data. Curr Neuropharmacol 2008; 6:125-50. [PMID: 19305793 PMCID: PMC2647150 DOI: 10.2174/157015908784533842] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Revised: 11/20/2007] [Accepted: 12/11/2007] [Indexed: 11/22/2022] Open
Abstract
Most women using heroin are of reproductive age with major risks for their infants. We review clinical and experimental data on fetal, neonatal and postnatal complications associated with methadone, the current "gold standard", and compare these with more recent, but limited, data on developmental effects of buprenorphine, and naltrexone. Methadone is a micro-opioid receptor agonist and is commonly recommended for treatment of opioid dependence during pregnancy. However, it has undesired outcomes including neonatal abstinence syndrome (NAS). Animal studies also indicate detrimental effects on growth, behaviour, neuroanatomy and biochemistry, and increased perinatal mortality. Buprenorphine is a partial micro-opioid receptor agonist and a kappa-opioid receptor antagonist. Clinical observations suggest that buprenorphine during pregnancy is similar to methadone on developmental measures but is potentially superior in reducing the incidence and prognosis of NAS. However, small animal studies demonstrate that low doses of buprenorphine during pregnancy and lactation lead to changes in offspring behaviour, neuroanatomy and biochemistry. Naltrexone is a non-selective opioid receptor antagonist. Although data are limited, humans treated with oral or sustained-release implantable naltrexone suggest outcomes potentially superior to those with methadone or buprenorphine. However, animal studies using oral or injectable naltrexone have shown developmental changes following exposure during pregnancy and lactation, raising concerns about its use in humans. Animal studies using chronic exposure, equivalent to clinical depot formulations, are required to evaluate safety. While each treatment is likely to have maternal advantages and disadvantages, studies are urgently required to determine which is optimal for offspring in the short and long term.
Collapse
Affiliation(s)
- W.O Farid
- School of Animal Biology, The University of Western Australia, Nedlands, WA 6009, Australia
- School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Nedlands, WA 6009, Australia
| | - S.A Dunlop
- School of Animal Biology, The University of Western Australia, Nedlands, WA 6009, Australia
- Western Australian Institute for Medical Research, The University of Western Australia, Nedlands, WA 6009, Australia
| | - R.J Tait
- School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Nedlands, WA 6009, Australia
| | - G.K Hulse
- School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Nedlands, WA 6009, Australia
| |
Collapse
|
35
|
Leri F. Co-administration of opioid agonists and antagonists in addiction and pain medicine. Expert Opin Pharmacother 2008; 9:1387-96. [DOI: 10.1517/14656566.9.8.1387] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
36
|
Sullivan MA, Garawi F, Bisaga A, Comer SD, Carpenter K, Raby WN, Anen SJ, Brooks AC, Jiang H, Akerele E, Nunes EV. Management of relapse in naltrexone maintenance for heroin dependence. Drug Alcohol Depend 2007; 91:289-92. [PMID: 17681716 PMCID: PMC4153601 DOI: 10.1016/j.drugalcdep.2007.06.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 06/19/2007] [Accepted: 06/20/2007] [Indexed: 11/29/2022]
Abstract
UNLABELLED Opioid dependence is a growing public health problem. Maintenance on the antagonist naltrexone for clinic- or office-based treatment of opioid dependence is plagued by high rates of relapse. This paper identifies critical determinants of lapses to opioid use during naltrexone maintenance. Time retained in treatment was examined as a function of whether lapses to opioid use occurred while adherent to naltrexone (blocked use), or after having missed naltrexone doses (unblocked). METHOD Participants (N=83) met DSM-IV criteria for opioid dependence and identified a significant other willing to participate in their treatment. Following inpatient detoxification, participants were enrolled in a 26-week outpatient course of therapy and naltrexone maintenance. RESULTS Patients with unblocked use had a very high rate of dropout (10% retained at 6 months), dropout usually occurring within 2 weeks after unblocked use. Patients with only blocked use had less dropout (33% retained at 6 months). However, episodes of blocked use were often followed by unblocked use and dropout. CONCLUSIONS During naltrexone maintenance for opioid dependence unblocked opioid use calls for immediate intervention, such as detoxification or switching to the partial agonist buprenorphine. Episodes of blocked use warrant increased clinical attention, such as direct observation of naltrexone ingestion, increased dose, or increased intensity of treatment contact. Maintenance on oral naltrexone is a fragile treatment because it is so easily undermined by episodes of opioid use while non-compliant. New long-acting injectable or implantable formulations of naltrexone may address this limitation and should be investigated for treatment of opioid dependence.
Collapse
Affiliation(s)
- Maria A Sullivan
- College of Physicians and Surgeons of Columbia University, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Davoli M, Bargagli AM, Perucci CA, Schifano P, Belleudi V, Hickman M, Salamina G, Diecidue R, Vigna-Taglianti F, Faggiano F. Risk of fatal overdose during and after specialist drug treatment: the VEdeTTE study, a national multi-site prospective cohort study. Addiction 2007; 102:1954-9. [PMID: 18031430 DOI: 10.1111/j.1360-0443.2007.02025.x] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Specialist drug treatment is critical to overdose prevention; methadone maintenance is effective, but we lack evidence for other modalities. We evaluate the impact of a range of treatments for opiate dependence on overdose mortality. METHODS Prospective cohort study of 10,454 heroin users entering treatment 1998-2001 in Italy followed-up for 10,208 person-years in treatment and 2,914 person-years out of treatment. Standardized overall mortality ratios (SMR) estimate excess mortality risk for heroin users in and out of treatment compared to the general population. Cox models compare the hazard ratio (HR) of overdose between heroin users in treatment and out of treatment. RESULTS There were 41 overdose deaths, 10 during treatment and 31 out of treatment, generating annual mortality rates of 0.1% and 1.1% and SMRs of 3.9 [95% confidence interval (CI) 2.8-5.4] and 21.4 (16.7-27.4), respectively. Retention in any treatment was protective against overdose mortality (HR 0.09 95% CI 0.04-0.19) compared to the risk of mortality out of treatment, independent of treatment type and potential confounders. The risk of a fatal overdose was 2.3% in the month immediately after treatment and 0.77% in the subsequent period; compared to the risk of overdose during treatment the HR was 26.6 (95% CI 11.6-61.1) in the month immediately following treatment and 7.3 (3.3-16.2) in the subsequent period. CONCLUSIONS We demonstrate that a range of treatments for heroin dependence reduces overdose mortality risk. However, the considerable excess mortality risk in the month following treatment indicates the need for greater health education of drug users and implementation of relapse and overdose death prevention programmes. Further investigation is needed to measure and weigh the potential benefits and harms of short-term therapies for opiate use.
Collapse
|
38
|
Gibson AE, Degenhardt LJ. Mortality related to pharmacotherapies for opioid dependence: a comparative analysis of coronial records. Drug Alcohol Rev 2007; 26:405-10. [PMID: 17564876 DOI: 10.1080/09595230701373834] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION AND AIMS The aim of this study was to compare the mortality associated with oral naltrexone, methadone and buprenorphine in opioid dependence treatment, employing a retrospective data analysis using coronial and prescription data. DESIGN AND METHODS The number of deaths were identified through national coronial data and number of treatment recipients were estimated from 2000 to 2003 prescriptions and restricted medications data. Mortality rates were expressed as deaths per number of treatment episodes and per person-years at high and low risk of fatal opioid overdose. RESULTS Thirty-two oral naltrexone, one buprenorphine and 282 methadone-related deaths were identified. Mortality rates in the highest risk period in deaths per 100 person-years were 22.1 (14.6 - 32.2) for oral naltrexone following treatment cessation and 3.0 (2.3 - 3.9) for methadone during treatment induction. Rates in the lowest risk period in deaths per 100 person-years were 1.0 (0.3 - 2.2) during oral naltrexone treatment and 0.34 (0.3 - 0.4) during post-induction methadone treatment. The relative risk of death for oral naltrexone subjects was 7.4 times (high-risk period, p < 0.0001) or 2.8 times (low-risk period, p = 0.055) that of methadone subjects. DISCUSSION AND CONCLUSIONS This is the first comparison of mortality associated with these three pharmacotherapies for opioid dependence. The risk of death related to oral naltrexone appears higher than that related to methadone treatment.
Collapse
Affiliation(s)
- Amy E Gibson
- National Drug and Alcohol Research Centre, University of New South Wales, Australia.
| | | |
Collapse
|
39
|
Tait RJ, Ngo HTT, Hulse GK. Mortality in heroin users 3 years after naltrexone implant or methadone maintenance treatment. J Subst Abuse Treat 2007; 35:116-24. [PMID: 17931824 DOI: 10.1016/j.jsat.2007.08.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Accepted: 08/08/2007] [Indexed: 11/30/2022]
Abstract
Concerns that treatment for heroin dependence using naltrexone may increase suicide rates during treatment and fatal overdoses posttreatment have been expressed. There is also disquiet about mortality during induction onto methadone. We assessed mortality during specific periods following treatment with naltrexone implants or methadone. Data were assembled using the Western Australian Data Linkage System. The methadone cohort comprised all those who started methadone in Western Australia during 2001-2002: The naltrexone cohort comprised all Western Australian heroin-dependent persons who received their first implant in 2001-2002. There were 15 (2.7%) deaths in the methadone cohort (n = 553) and 6 (1.8%) deaths in the naltrexone cohort (n = 341). Mortality rates for the "initial 14-day period," "stable treatment," and "overall" were 94.47, 0.0, and 5.83 deaths/1,000 person-years for the methadone group. In the naltrexone group, the rates "during first treatment (0-6 months)," "post first treatment," and overall were 0.0, 4.21, and 3.76 deaths/1,000 person-years. The age-standardized mortality rate ratio for naltrexone compared to methadone was 0.645 (95% confidence interval = 0.123-1.17). Increased mortality during induction onto methadone was confirmed. Evidence relating naltrexone to either increased suicide or overdose was not found. Overall mortality rates for naltrexone implant were similar to those for methadone, but increased mortality during methadone induction was avoided.
Collapse
Affiliation(s)
- Robert James Tait
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, QE II Medical Center, Nedlands, WA 6009, Australia.
| | | | | |
Collapse
|
40
|
Black A, Khan S, Brown R, Sharp P, Chatfield H, McGuiness C. An evaluation of opioid replacement pharmacotherapy in an urban Aboriginal Health Service. Aust N Z J Public Health 2007; 31:428-32. [DOI: 10.1111/j.1753-6405.2007.00113.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
41
|
Comer SD, Sullivan MA, Hulse GK. Sustained-release naltrexone: novel treatment for opioid dependence. Expert Opin Investig Drugs 2007; 16:1285-94. [PMID: 17685876 DOI: 10.1517/13543784.16.8.1285] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The devastating costs of opioid abuse and dependence underscore the need for effective treatments for these disorders. At present, several different maintenance medications exist for treating opioid dependence, including methadone, buprenorphine and naltrexone. Of these, naltrexone is the only one that possesses no opioid agonist effects. Instead, naltrexone occupies opioid receptors and prevents or reverses the effects produced by opioid agonists. Despite its clear pharmacologic effectiveness, its clinical effectiveness in treating opioid dependence has been disappointing, primarily due to non-compliance with taking the medication. However, the recent availability of sustained-release formulations of naltrexone has renewed interest in this medication. The present paper describes the development of sustained-release naltrexone formulations and discusses the clinical issues associated with their use in treating opioid dependence.
Collapse
Affiliation(s)
- Sandra D Comer
- College of Physicians & Surgeons of Columbia University, New York State Psychiatric Institute, Department of Psychiatry, Unit 120, New York, NY 10032, USA.
| | | | | |
Collapse
|
42
|
Ngo HTT, Tait RJ, Arnold-Reed DE, Hulse GK. Mental health outcomes following naltrexone implant treatment for heroin-dependence. Prog Neuropsychopharmacol Biol Psychiatry 2007; 31:605-12. [PMID: 17229510 DOI: 10.1016/j.pnpbp.2006.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 11/20/2006] [Accepted: 12/05/2006] [Indexed: 11/22/2022]
Abstract
BACKGROUND Oral naltrexone is an approved treatment for opioid dependence. However, the impact of sustained release naltrexone on the mental health of treated opioid users has not been studied. AIMS To assess if naltrexone via implant treatment was associated with any change in (i) risk, (ii) rate, and (iii) duration for hospital morbidity related to several categories of mental disorders among treated heroin users. METHOD A cohort of 359 heroin users treated with sustained release naltrexone via implants in Western Australia was retrospectively followed up for mental health related outcomes via a health record linkage system over an average period of 1.78 years post-treatment. RESULTS Individual patient's risk for hospital mental diagnoses was not altered after naltrexone implant. On a population cohort level, hospital admission rates related to all mental health problems, except mood disorders, declined significantly post-treatment; however, length of hospital stay did not improve. Overall, young, female patients or those with pre-existing mental illness were more likely than other patients to require hospital care for mental health issues following treatment. Longer period of heroin use was associated with poorer mood outcomes. CONCLUSIONS Naltrexone implants were not associated with an increased risk for hospitalisation due to mental illness, and in most cases, were associated with a decrease in mental related hospital admission rate.
Collapse
Affiliation(s)
- Hanh T T Ngo
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, QE II Medical Centre, Nedlands, WA 6009, Australia.
| | | | | | | |
Collapse
|
43
|
|
44
|
Maxwell S, Bigg D, Stanczykiewicz K, Carlberg-Racich S. Prescribing naloxone to actively injecting heroin users: a program to reduce heroin overdose deaths. J Addict Dis 2007; 25:89-96. [PMID: 16956873 DOI: 10.1300/j069v25n03_11] [Citation(s) in RCA: 184] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Heroin overdose deaths have increased alarmingly in Chicago over the past decade. Naloxone, an opioid antagonist with no abuse potential, has been used to reverse opiate overdose in emergency medical settings for decades. We describe here a program to educate opiate users in the prevention of opiate overdose and its reversal with intramuscular naloxone. Participant education and naloxone prescription are accomplished within a large comprehensive harm reduction program network. Since institution of the program in January 2001, more than 3,500 10 ml (0.4 mg/ml) vials of naloxone have been prescribed and 319 reports of peer reversals received. The Medical Examiner of Cook County reported a steady increase in heroin overdose deaths since 1991, with a four-fold increase between 1996 and 2000. This trend reversed in 2001, with a 20% decrease in 2001 and 10% decreases in 2002 and 2003.
Collapse
|
45
|
Van den Brink W, Haasen C. Evidenced-based treatment of opioid-dependent patients. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2006; 51:635-46. [PMID: 17052031 DOI: 10.1177/070674370605101003] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To provide an overview of treatment options for opioid-dependent patients. METHOD We screened all published studies on the treatment of opioid dependence, with a special focus on systematic literature reviews, formal metaanalyses, and recent trials. RESULTS Both clinical experience and neurobiological evidence indicate that opioid dependence is a chronic relapsing disorder. Treatment objectives depend on the pursued goals: crisis intervention, abstinence-oriented treatment (detoxification and relapse prevention), or agonist maintenance treatment. The high quality of solid evidence in the literature demonstrates that there are numerous effective interventions available for the treatment of opioid dependence. Crisis intervention, frequently necessary owing to the high overdose rate, can be effectively handled with naloxone. Abstinence-oriented interventions are effective for only a few motivated patients with stable living conditions and adequate social support. Agonist maintenance treatment is considered the first line of treatment for opioid dependence. Numerous studies have shown efficacy for methadone and buprenorphine treatment, while maintenance with other agonists is also becoming available to a greater extent. Maintenance treatment with diamorphine should be made available for the small group of treatment-resistant, severely dependent addicts. Other harm-reduction measures can serve to engage individuals with opioid addiction who are not in treatment. CONCLUSION Opioid dependence is a chronic relapsing disease that is difficult to cure, but effective treatments are available to stabilize patients and reduce harm, thereby increasing life expectancy and quality of life.
Collapse
Affiliation(s)
- Wim Van den Brink
- Academic Medical Center, University of Amsterdam, Department of Psychiatry, The Netherlands.
| | | |
Collapse
|
46
|
Roozen HG, de Waart R, van der Windt DAWM, van den Brink W, de Jong CAJ, Kerkhof AJFM. A systematic review of the effectiveness of naltrexone in the maintenance treatment of opioid and alcohol dependence. Eur Neuropsychopharmacol 2006; 16:311-23. [PMID: 16361086 DOI: 10.1016/j.euroneuro.2005.11.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 10/28/2005] [Accepted: 11/04/2005] [Indexed: 12/30/2022]
Abstract
This systematic review summarises evidence of the effectiveness of naltrexone (NTX) and the added value of psychosocial treatment in the maintenance treatment of opioid and alcohol dependence. Studies were selected through a literature search conducted in March 2004. Seven opioid and seventeen alcohol studies were identified. When possible, meta-(regression) analyses were performed. There is lack of evidence about the effectiveness of NTX in the maintenance treatment of opioid dependence. There is evidence for the effectiveness and applicability of NTX in the management of alcohol dependence. The opioid studies combined NTX with a variety of psychosocial interventions, which plagued the evaluation of their value. Concomitant psychosocial interventions used in the alcohol studies were mainly cognitive behavioural, which seems to be more effective than NTX combined with supportive therapy. Available data do not allow firm conclusions regarding the added effect of psychosocial interventions. However, the data suggest that a combination of naltrexone with cognitive behavioural relapse prevention therapy is beneficial in alcohol dependent patients.
Collapse
Affiliation(s)
- Hendrik G Roozen
- Novadic-Kentron, Network for Addiction Treatment Services, Roosendaal/Sint-Oedenrode, The Netherlands.
| | | | | | | | | | | |
Collapse
|
47
|
Comer SD, Sullivan MA, Yu E, Rothenberg JL, Kleber HD, Kampman K, Dackis C, O'Brien CP. Injectable, sustained-release naltrexone for the treatment of opioid dependence: a randomized, placebo-controlled trial. ARCHIVES OF GENERAL PSYCHIATRY 2006; 63:210-8. [PMID: 16461865 PMCID: PMC4200530 DOI: 10.1001/archpsyc.63.2.210] [Citation(s) in RCA: 243] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
CONTEXT Oral naltrexone can completely antagonize the effects produced by opioid agonists. However, poor compliance with naltrexone has been a major obstacle to the effective treatment of opioid dependence. OBJECTIVE To evaluate the safety and efficacy of a sustained-release depot formulation of naltrexone in treating opioid dependence. DESIGN AND SETTING Randomized, double-blind, placebo-controlled, 8-week trial conducted at 2 medical centers. PARTICIPANTS Sixty heroin-dependent adults. INTERVENTIONS Participants were stratified by sex and years of heroin use (> or = 5 vs < 5) and then were randomized to receive placebo or 192 or 384 mg of depot naltrexone. Doses were administered at the beginning of weeks 1 and 5. All participants received twice-weekly relapse prevention therapy, provided observed urine samples, and completed other assessments at each visit. MAIN OUTCOME MEASURES Retention in treatment and percentage of opioid-negative urine samples. RESULTS Retention in treatment was dose related, with 39%, 60%, and 68% of patients in the placebo, 192 mg of naltrexone, and 384 mg of naltrexone groups, respectively, remaining in treatment at the end of 2 months. Time to dropout had a significant main effect of dose, with mean time to dropout of 27, 36, and 48 days for the placebo, 192 mg of naltrexone, and 384 mg of naltrexone groups, respectively. The percentage of urine samples negative for opioids, methadone, cocaine, benzodiazepines, and amphetamine varied significantly as a function of dose. When the data were recalculated without the assumption that missing urine samples were positive, a main effect of group was not found for any drugs tested except cocaine, where the percentage of cocaine-negative urine samples was lower in the placebo group. Adverse events were minimal and generally mild. This formulation of naltrexone was well tolerated and produced a robust, dose-related increase in treatment retention. CONCLUSION These data provide new evidence of the feasibility, efficacy, and tolerability of long-lasting antagonist treatments for opioid dependence.
Collapse
Affiliation(s)
- Sandra D Comer
- Division on Substance Abuse, New York State Psychiatric Institute, and Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York 10032, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Waal H, Frogopsahl G, Olsen L, Christophersen AS, Mørland J. Naltrexone implants -- duration, tolerability and clinical usefulness. A pilot study. Eur Addict Res 2006; 12:138-44. [PMID: 16778434 DOI: 10.1159/000092115] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Naltrexone blocks opioid effects effectively, but poor compliance limits the clinical usefulness in the treatment of opioid dependence. Long-acting implanted formulations might increase the clinical feasibility. Several implants have been produced, but few clinical reports have been published. This paper describes an open trial with an Australian implant. This implant is claimed to have duration of up to six months with double implants and acceptable levels of side effects. This was explored in the present pilot study with 13 opioid-dependent patients. By single implant of 1.8 g naltrexone the duration judged by naltrexone plasma levels above 1 ng/ml naltrexone was between 2 and 4 months. Double implants maintained such plasma levels for 5-6.5 months. Clinically, the implants appeared promising. Side effects were minimal. During the period with adequate plasma levels of naltrexone, use of opioids was absent and use of other psychoactive drugs reduced. At 1-year follow-up, the patients rated the implants highly positively.
Collapse
Affiliation(s)
- Helge Waal
- Unit for Addiction Medicine, Institute of Psychiatry, University of Oslo, Oslo, Norway.
| | | | | | | | | |
Collapse
|
49
|
Dodge R, Sindelar J, Sinha R. The role of depression symptoms in predicting drug abstinence in outpatient substance abuse treatment. J Subst Abuse Treat 2005; 28:189-96. [PMID: 15780549 DOI: 10.1016/j.jsat.2004.12.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Revised: 10/28/2004] [Accepted: 12/01/2004] [Indexed: 10/25/2022]
Abstract
OBJECTIVE This study examined the role of depressive symptoms in the context of specific demographic and individual treatment characteristics in predicting drug abstinence at discharge from outpatient substance abuse treatment. METHODS Data from 827 clients entering a large public funded outpatient substance abuse treatment program were analyzed using logistic regression to assess the effects of depressive symptoms on drug abstinence status at discharge. Analyses on the effects of gender, race, age, education level, frequency of drug use, insurance status, referral source, and length of stay in treatment on drug abstinence status at discharge were also conducted. RESULTS Higher depressive symptom scores significantly predicted a decreased likelihood of clients' abstinence at discharge even after accounting for other significant demographic and treatment variables such as insurance status, race, age, primary drug of choice, frequency of drug use at admission and length of stay in treatment. CONCLUSION The findings suggest that depression symptoms are an important factor affecting successful substance abuse treatment outcomes. Treatment approaches that address depressive symptoms are likely to enhance substance abuse treatment outcomes in real world clinical settings.
Collapse
Affiliation(s)
- Rebecca Dodge
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA.
| | | | | |
Collapse
|
50
|
Digiusto E, Shakeshaft A, Ritter A, O'Brien S, Mattick RP. Serious adverse events in the Australian National Evaluation of Pharmacotherapies for Opioid Dependence (NEPOD). Addiction 2004; 99:450-60. [PMID: 15049745 DOI: 10.1111/j.1360-0443.2004.00654.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIMS The study estimated serious adverse event (SAE) rates among entrants to pharmacotherapies for opioid dependence, during treatment and after leaving treatment. DESIGN A longitudinal study based on data from 12 trials included in the Australian National Evaluation of Pharmacotherapies for Opioid Dependence (NEPOD). PARTICIPANTS AND SETTINGS A total of 1244 heroin users and methadone patients treated in hospital, community and GP settings. Intervention Six trials included detoxification; all included treatment with methadone, buprenorphine, levo-alpha-acetyl-methadol (LAAM) or naltrexone. FINDINGS During 394 person-years of observation, 79 SAEs of 28 types were recorded. Naltrexone participants experienced 39 overdoses per 100 person-years after leaving treatment (44% occurred within 2 weeks after stopping naltrexone). This was eight times the rate recorded among participants who left agonist treatment. Rates of all other SAEs were similar during treatment versus out of treatment, for both naltrexone-treated and agonist-treated participants. Five deaths occurred, all among participants who had left treatment, at a rate of six per 100 person-years. Total SAE rates during naltrexone and agonist treatments were similar (20, 14 per 100 person-years, respectively). Total SAE and death rates observed among participants who had left treatment were three and 19 times the corresponding rates during treatment. CONCLUSIONS Individuals who leave pharmacotherapies for opioid dependence experience higher overdose and death rates compared with those in treatment. This may be due partly to a participant self-selection effect rather than entirely to pharmacotherapy being protective. Clinicians should alert naltrexone treatment patients in particular about heroin overdose risks. Duty of care may extend beyond cessation of dosing.
Collapse
Affiliation(s)
- E Digiusto
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, Australia.
| | | | | | | | | |
Collapse
|