Skip to main content

Long-Term Pharmacotherapy for Opiate (Primarily Heroin) Addiction: Opioid Antagonists and Partial Agonists

  • Chapter
Pharmacological Aspects of Drug Dependence

Part of the book series: Handbook of Experimental Pharmacology ((HEP,volume 118))

Abstract

The action of a specific opioid antagonist is to bind to specific opioid receptors and thus prevent the action of exogenous opioids. Use of such an antagonist will prevent the normal physiological action of the endogenous opioids at their receptors. The first pure specific opioid antagonist which was successfully synthesized, characterized, and patented in 1960 by Dr. J. Fishman with M.J. Lewenstein was naloxone, an alkyl derivative of nor-oxymorphone (BLUMBERG et al. 1961; BLUMBERG and DAYTON 1974). Naloxone was thus the first specific opioid antagonist used clinically and remains extremely useful in the management of accidental or medical opioid overdose. Naloxone has very limited systemic bioavailability, with less than 3% reaching the systemic circulation after oral administration; when it is used for management of narcotic overdose (or when the attempt was made to use it in the more chronic management of opiate dependency) by direct receptor blockade of opiate agonist action in the central nervous system, naloxone must be administered parenterally, and ideally, intravenously (KREEK 1973c; KREEK et al. 1983a,b,c, 1984). Although brief sporadic attempts were made to use naloxone in the maintenance treatment of addiction, it was clear that this use would not be suitable because of the need for a parenteral route of administration. However, studies using naloxone showed that a specific opioid antagonist could block all exogenous opioid effects, and moreover, could reverse the effects of both exogenous and endogenous opioids (KOSTERLITZ and WATT 1968; VEREBEY and MULE 1975; VOLAVKA et al. 1979a,b; MENDELSON et al. 1980; NABER et al. 1981; KREEK and HARTMAN 1982; KREEK et al. 1983a,b,c, 1984; COHEN et al. 1983; KREEK and CULPEPPER-MORGAN 1991.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 109.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  • Albeck H, Woodfield S, Kreek MJ (1989) Quantitative and pharmacokinetic analysis of naloxone in plasma using high performance liquid chromatography with electrochemical detection and solid phase extraction. J Chromatog 488:435–445

    CAS  Google Scholar 

  • Altman JL, Meyer RE, Mirin SM, McNamee HB (1976) Opiate antagonists and the modification of heroin self-administration behavior in man: an experimental study. Int J Add 11(3):485–499

    CAS  Google Scholar 

  • Atkinson RL, Berke LK, Drake CR, Bibbs ML, Williams FL, Kaiser DL (1985) Effects of long-term therapy with naltrexone on body weight in obesity. Clin Pharm Ther 38:419–422

    CAS  Google Scholar 

  • Bickel WK, Stitzer ML, Bigelow GE, Liebson IA, Jasinski DR, Johnson RE (1988a) A clinical trial of buprenorphine: comparison with methadone in the detoxification of heroin addicts. Clin Pharm Ther 43(1):72–78

    CAS  Google Scholar 

  • Bickel WK, Stitzer ML, Bigelow GE, Liebson IA, Jasinski DR, Johnson RE (1988b) Buprenorphine: dose-related blockade of opioid challenge effects in opioid dependent humans. JPET 247:47–53

    CAS  Google Scholar 

  • Bigelow GE, Preston KL (1992) Assessment of buprenorphine in a drug discrimination procedure in humans. NIDA Research Monograph 121:28–37

    PubMed  CAS  Google Scholar 

  • Blachley PH (1975) Naloxone for diagnosis in methadone programs. J Am Med Assoc 244:334–335

    Google Scholar 

  • Blaine JD (1992) Introduction (to buprenorphine: an alternative treatment for opioid dependence). NIDA Research Monograph 121:1–4

    Google Scholar 

  • Blom Y, Bondesson U, Gunne LM (1987) Effects of buprenorphine in heroin addicts. Dr Alc Depend 20:1–7

    CAS  Google Scholar 

  • Blumberg H, Dayton HB (1974) Naloxone, naltrexone, and related noroxymorphines. In: Braude MC, Harris LS, May EL, Smith JP, Villarreal JE (eds) Narcotic antagonists. Advances in biochemical psychopharmacology, vol 8. Raven, New York, pp 33–43

    Google Scholar 

  • Blumberg H, Dayton HB, George M, Rapaport DN (1961) N-allylnoroxymorphone: a potent narcotic antagonist. Fed Proc 20:311

    Google Scholar 

  • Borg L, Broe DM, Ho A, Kreek MJ (1995a) Cocaine abuse is decreased with effective methadone maintenance treatment at an urban Department of Veterans Affairs (DVA) Program. In: Harris LS (ed) Problems of Drug Dependence 1994. Proceedings of the 56th Annual Scientific Meeting of the College on Problems of Drug Dependence. NIDA Research Monograph Series, Rockville, MD 153:17

    Google Scholar 

  • Borg L, Ho A, Peters JE, Kreek MJ (1995b) Availability of reliable serum methadone determination for management of symptomatic patients. J Add Dis (in press)

    Google Scholar 

  • Brahen LS, Capone T, Wiechert V, Desiderio D (1977) Naltrexone and cyclazocine: a controlled treatment study. Arch Gen Psych 34:1181–1184

    CAS  Google Scholar 

  • Bullingham RES, McQuay HJ, Moore RA (1983) Clinical pharmacokinetics of narcotic agonist-antagonist drugs. Clin Pharm 8:332–343

    CAS  Google Scholar 

  • Chaisson RE, Bacchetti P, Osmond D, Brodie B, Sande MA, Moss AR (1989) Cocaine use and HIV infection in intravenous drug users in San Francisco. JAMA 261:561–565

    PubMed  CAS  Google Scholar 

  • Chou JZ, Albeck H, Kreek MJ (1993) Determination of nalmefene in plasma by high-performance liquid chromatography with electrochemical detection and its application in pharmacokinetic studies. J Chrom 613:359–364

    CAS  Google Scholar 

  • Cohen MR, Cohen RM, Pickar D, Weingartner H, Murphy DL (1983) High-dose naloxone infusions in normals: dose-dependent behavioral, hormonal, and physiological responses. Arch Gen Psych 40:613–619

    CAS  Google Scholar 

  • Cone EJ (1973) Human metabolite of naltrexone (N-cyclopropylmethylnoroxymorphone) with a novel C-6 isomorphine configuration. Tetrahedron Lett 28:2607–2610

    Google Scholar 

  • Cone EJ, Gorodetzky CW, Yousefnejad D, Buchwald WF, Johnson RE (1984) The metabolism and excretion of buprenorphine in humans. Dr Met Disp 12(5):577–581

    CAS  Google Scholar 

  • Cowan A (1974) Evaluation in nonhuman primates: Evaluation of the physical dependence capacities of oripavine-thebaine partial agonists in patas monkeys. In: Braude MC, Harris LS, May EL, Smith JP, Villarreal JE (eds) Narcotic antagonists. Advances in biochemical psychopharmacology, vol 8. Raven, New York, pp 427 – 438

    Google Scholar 

  • Cowan A, Lewis JW, Macfarlane IR (1971) Analgesic and dependence studies with oripavine partial agonists. Br J Pharmacol 43:461P–462P

    Google Scholar 

  • Cowan A, Lewis JW, Macfarlane IR (1977) Agonist and antagonist properties of buprenorphine, a new antinociceptive agent. Br J Pharmacol 60:537–545

    PubMed  CAS  Google Scholar 

  • Culpepper-Morgan JA, Inturrisi CE, Portenoy RK, Foley K, Houde RW, Marsh F, Kreek MJ (1992) Treatment of opioid induced constipation with oral naloxone: a pilot study. Clin Pharm Ther 23:90–95

    Google Scholar 

  • Des Jarlais DC, Wenston J, Friedman SR, Sotheran JL, Maslansky R, Marmor M (1992) Crack cocaine use in a cohort of methadone maintenance patients. J Sub Ab Tr 9:319–325

    Google Scholar 

  • Dixon R, Hsiao J, Taaffe W, Hahn E, Tuttle R (1984) Nalmefene: radioimmunoassay for a new opioid antagonist. J Pharm Sci 73(11):1645–1646

    PubMed  CAS  Google Scholar 

  • Dixon R, Howes J, Gentile J, Hsu H-B, Hsiao J, Garg D, Weidler D, Meyer M, Tuttle R (1986) Nalmefene: intravenous safety and kinetics of a new opioid antagonist 39:49–53

    CAS  Google Scholar 

  • Dixon R, Gentile J, Hsu H-B, Hsiao J, Howes J, Garg D, Weidler D (1987) Nalmefene: safety and kinetics after single and multiple oral doses of a new opioid antagonist. J Clin Pharm 27:233–239

    CAS  Google Scholar 

  • Dole VP, Nyswander ME, Kreek MJ (1966a) Narcotic blockade. Arch Intern Med 118:304–309

    CAS  Google Scholar 

  • Dole VP, Nyswander ME, Kreek MJ (1966b) Narcotic blockade: a medical technique for stopping heroin use by addicts. Trans Assoc Am Physicians 79:122–136

    CAS  Google Scholar 

  • Fudala PJ, Jaffe JH, Dax EM, Johnson RE (1990) Use of buprenorphine in the treatment of opioid addiction. II. Physiologic and behavioral effects of daily and alternate-day administration and abrupt withdrawal. Clin Pharm Ther 47:525–534

    CAS  Google Scholar 

  • Goldstein A (1976) Heroin addiction: sequential treatment employing pharmacologic supports. Arch Gen Psych 33:353–358

    CAS  Google Scholar 

  • Gonzalez JP, Brogden RN (1988) Naltrexone: a review of its pharmacodynamic and pharmacokinetic properties and therapeutic efficacy in the management of opioid dependence. Drugs 35:192–213

    PubMed  CAS  Google Scholar 

  • Greenstein RA, Arndt IC, McLellan AT, OBrien CP, Evans B (1984) Naltrexone: a clinical perspective. J Clin Psych 45[9(2)]:25–28

    CAS  Google Scholar 

  • Hanbury R, Sturiano V, Cohen M, Stimmel B, Aguillaume C (1986) Cocaine use in persons on methadone maintenance. Adv Alcohol Subst Abuse 6:97–106

    PubMed  CAS  Google Scholar 

  • Hahn EF, Lahita R, Kreek MJ, Duma C, Inturrisi CE (1983) Naloxone radioimmunoassay: an improved antiserum. J Pharm Pharmacol 35:833–836

    PubMed  CAS  Google Scholar 

  • Heel RC, Brogden RN, Speight TM, Avery GS (1979) Buprenorphine: a review of its pharmacological properties and therapeutic efficacy. Drugs 17:81–110

    PubMed  CAS  Google Scholar 

  • Hollister LE, Schwin RL, Kasper P (1977) Naltrexone treatment of opiate-dependent persons. Dr Alc Dep 2:203–209

    CAS  Google Scholar 

  • Holman BL, Mendelson J, Garada B, Teoh SK, Hallgring E, Johnson KA, Mello NK (1993) Regional cerebral blood flow improves with treatment in chronic cocaine polydrug users. J Nucl Med 34:723–727

    PubMed  CAS  Google Scholar 

  • Hussain MA, Shefter E (1988) Naltrexone-3-salicylate (a prodrug of naltrexone): synthesis and pharmacokinetics in dogs. Pharm Res 5(2):113–115

    PubMed  CAS  Google Scholar 

  • Inturrisi CE (1976) Disposition of narcotics and narcotic antagonists. Ann NY Acad Sci 273–287

    Google Scholar 

  • Jasinski DR, Pevnick JS, Griffith JD (1978) Human pharmacology and abuse potential of the analgesic buprenorphine. Arch Gen Psych 35:501–516

    CAS  Google Scholar 

  • Johnson RE, Fudala PJ (1992) Development of buprenorphine for the treatment of opioid dependence. NIDA Research Monography 121:120–141

    CAS  Google Scholar 

  • Johnson RE, Cone EJ, Henningfield JE, Fudala PJ (1989) Use of buprenorphine in the treatment of opiate addiction. I. Physiologic and behavioral effects during a rapid dose induction. Clin Pharm Ther 46:335–343

    CAS  Google Scholar 

  • Johnson RE, Jaffe JH, Fudala PJ (1992) A controlled trial of buprenorphine treatment for opioid dependence. JAMA 267:2750–2755

    PubMed  CAS  Google Scholar 

  • Judson BA, Himmelberger DU, Goldstein A (1980) The naloxone test for opiate dependence. Clin Pharm Ther 27(4):492–501

    CAS  Google Scholar 

  • Judson BA, Carney TM, Goldstein A (1981) Naltrexone treatment of heroin addiction: efficacy and safety in a double-blind dosage comparison. Dr Alc Dep 7(4):325–346

    CAS  Google Scholar 

  • Kleber HD, Kosten TR, Gaspari J, Topazian M (1985) Nontolerance to the opioid antagonism of naltrexone. Bio Psych 20:66–72

    CAS  Google Scholar 

  • Kogan MJ, Verebey K, Mule SJ (1977) Estimation of the systemic availability and other pharmacokinetic parameters in altrexone in man after acute and chronic oral administration. Res Comm Chem Path Pharm 18(1):29–34

    CAS  Google Scholar 

  • Kosten TR, Kleber HD (1991) Buprenorphine detoxification from opioid dependence: a pilot study. Life Sci 42:635–641

    Google Scholar 

  • Kosten TR, Kreek MJ, Raghunath J, Kleber HD (1986a) A preliminary study of beta endorphin during chronic naltrexone maintenance treatment in ex-opiate addicts. Life Sci 39:55–59

    CAS  Google Scholar 

  • Kosten TR, Kreek MJ, Raghunath J, Kleber HD (1986b) Cortisol levels during chronic naltrexone maintenance treatment in ex-opiate addicts. Bio Psych 21:217–220

    CAS  Google Scholar 

  • Kosten TR, Kleber HD, Morgan C (1989a) Role of opioid antagonists in treating intravenous cocaine abuse. Life Sci 44:887–892

    CAS  Google Scholar 

  • Kosten TR, Kleber HD, Morgan C (1989b) Treatment of cocaine abuse with buprenorphine. Bio Psych 26:170–172

    Google Scholar 

  • Kosten TR, Morgan CJ, Kleber HD (1989c) Buprenorphine treatment of cocaine abuse. NIDA Res Mono 95:461

    CAS  Google Scholar 

  • Kosten TR, Morgan C, Kleber HD (1992a) Phase II clinical trials of buprenorphine: detoxification and induction onto naltrexone. NIDA Research Monograph 121:101–119

    CAS  Google Scholar 

  • Kosten TR, Morgan C, Kreek MJ (1992b) Beta-endorphin levels during heroin, methadone, buprenorphine and naloxone challenges: Preliminary findings. Biolog Psych 32:523–528

    CAS  Google Scholar 

  • Kosten TR, Schottenfeld R, Ziedonis D, Falcioni J (1993) Buprenorphine versus methadone maintenance for opioid dependence. J New Merit Dis 181(6):358–364

    CAS  Google Scholar 

  • Kosterlitz HW, Watt AJ (1968) Kinetic parameters of narcotic agonists and anta-gonists. In: Committee on Problems of Drug Dependence, Proceedings of the 30th meeting in Indianapolis, IN

    Google Scholar 

  • Kreek MJ (1972) Medical safety, side effects and toxicity of methadone. In: Proceedings of the Fourth National Conference on Methadone Treatment NAPANNIMH, pp 171–174

    Google Scholar 

  • Kreek MJ (1973a) Medical safety and side effects of methadone in tolerant individuals. JAMA 223:665–668

    CAS  Google Scholar 

  • Kreek MJ (1973b) Physiological implications of methadone treatment. In: Proceedings, Fifth National Conference on Methadone Treatment. National Association for the Prevention of Addiction to Narcotics 2:85–91

    Google Scholar 

  • Kreek MJ (1973c) Plasma and urine levels of methadone. NYS J Med 73(23):2773–2777

    CAS  Google Scholar 

  • Kreek MJ (1992a) The addict as a patient. In: Lowinson JH, Ruiz P, Millman RB, Langrod JG (eds) Substance abuse: a comprehensive textbook. Williams and Wilkins, Baltimore, pp 997–1009

    Google Scholar 

  • Kreek MJ (1992b) Epilogue: Medical maintenance treatment for heroin addiction, from a retrospective and prospective viewpoint. In: State methadone maintenance treatment guidelines. Office for Treatment Improvement, Division for State Assistance 255–272

    Google Scholar 

  • Kreek MJ (1992c) Rationale for maintenance pharmacotherapy of opiate dependence. Res Pub Assoc Res Nerv Ment Dis 70:205–230

    CAS  Google Scholar 

  • Kreek MJ, Culpepper-Morgan J (1991) Neuroendocrine (HPA) and gastrointestinal effects of opiate antagonists: possible therapeutic application. In: Harris LS (ed) Problems of Drug Dependence 1990. Proceedings of the 52nd Annual Scientific Meeting of the Committee on Problems of Drug Dependence. NIDA Research Monograph Series, Rockville, MD, DHHS Pub#(ADM)91–1753, 105:168–174

    Google Scholar 

  • Kreek MJ, Hartman N (1982) Chronic use of opioids and antipsychotic drugs: side effects, effects on endogenous opioids, and toxicity. Annals NY Acad Sci 151–172

    Google Scholar 

  • Kreek MJ, Schaefer RA, Hahn EF, Fishman J (1983a) Naloxone, a specific opioid antagonist, reverses chronic idiopathic constipation. Lancet 2/5:261–262

    Google Scholar 

  • Kreek MJ, Schaefer RA, Hahn EF, Fishman J (1983b) Naloxone in chronic constipation. Letters to the editors. Lancet 758b

    Google Scholar 

  • Kreek MJ, Wardlaw SL, Hartman N, Raghunath J, Friedman J, Schneider B, Frantz AG (1983c) Circadian rhythms and levels of beta-endorphin, ACTH, and cortisol during chronic methadone maintenance treatment in humans. Life Sci Supp I, 33:409–411

    Google Scholar 

  • Kreek MJ, Schneider BS, Raghunath J, Plevy S (1984) Prolonged (24-hour) infusion of the opioid antagonist naloxone does not significantly alter plasma levels of cortisol and ACTH in humans. Abstracts of the Seventh International Congress of Endocrinology, International Congress Series 652. Excerpta Medica, Amsterdam, p 845

    Google Scholar 

  • Lehmann KA, Reichling U, Wirtz R (1988) Influence of naloxone on the postoperative analgesic and respiratory effects of buprenorphine. Eur J Clin Pharm 34:343–352

    CAS  Google Scholar 

  • Lewis JW (1974) Ring C-bridged derivatives of thebaine and oripavine. In: Braude MC, Harris LS, May EL, Smith JP, Villarreal JE (eds) Narcotic antagonists. Advances in biochemical psychopharmacology, vol 8. Raven, New York, pp 123–136

    Google Scholar 

  • Lewis JW (1985) Buprenorphine. Dr Alc Dep 14:363–372

    CAS  Google Scholar 

  • Lewis JW, Readhead MJ (1970) Novel analgetics and molecular rearrangements in morphine-thebaine group XVIII 3-deoxy-6,14-endo-etheno-6,7,8,14-tetrahydrooripavines. J Med Chem 13:525–527

    PubMed  CAS  Google Scholar 

  • Lewis JW, Walter D (1992) Buprenorphine: background to its development as a treatment for opiate dependence. NIDA Research Monograph 121:5–11

    PubMed  CAS  Google Scholar 

  • Ling W, Wesson DR (1984) Naltrexone treatment for addicted health-care professionals: A collaborative private practice experience. J Clin Psych 45[9(2)]:46–48

    CAS  Google Scholar 

  • Martin WR, Jasinski DR, Mansky PA (1973) Naltrexone, an antagonist for the treatment of heroin dependence. Arch Gen Psych 28:784–791

    CAS  Google Scholar 

  • McQuay HJ, Bullingham RES, Paterson GMC, Moore RA (1980) Clinical effects of buprenorphine during and after operation. Br J Anaesth 52: 1013 – 1019

    PubMed  CAS  Google Scholar 

  • Mello NK, Mendelson JH (1980) Buprenorphine suppresses heroin use by heroin addicts. Science 207:657–659

    PubMed  CAS  Google Scholar 

  • Mello NK, Mendelson JH (1992) Primate studies of the behavioral pharmacology of buprenorphine. NIDA Research Monograph 121:61–100

    PubMed  CAS  Google Scholar 

  • Mello NK, Mendelson JM, Kuehnle JC (1982) Buprenorphine effects on human heroin self-administration: an operate analysis. JPET 223:30–39

    CAS  Google Scholar 

  • Mello NK, Lukas SE, Mendelson JH (1985) Buprenorphine effects on cigarette smoking. Psychopharmacology 86:417–425

    PubMed  CAS  Google Scholar 

  • Mendelson JH, Mello NK (1992) Human laboratory studies of buprenorphine. NIDA Research Monograph 121:38–60

    PubMed  CAS  Google Scholar 

  • Mendelson JH, Ellingboe J, Keuhnle JC, Mello NK (1979) Effects of naltrexone on mood and neuroendocrine function in normal adult males. Psychoneuroendoerinology 3:231–236

    Google Scholar 

  • Mendelson JH, Ellingboe J, Kuehnle JC, and Mello NK (1980) Heroin and naltrexone effects on pituitary-gonadal hormones in man: Interaction of steroid feedback effects, tolerance, and supersensitivity. JPET 214(3):503–506

    CAS  Google Scholar 

  • Mendelson JH, Ellingboe J, Mello NK, Kuehnle J (1982) Buprenorphine effects on plasma luteinizing hormone and prolactin in male heroin addicts. JPET 220:252–255

    CAS  Google Scholar 

  • Mendelson JH, Mello NK, Teoh SK, Lloyd-Jones JG, Clifford JM (1989) Naloxone suppresses buprenorphine stimulation of plasma prolactin. J Clin Psychopharm 9(2):105–109

    CAS  Google Scholar 

  • Meyer MC, Straughn AB, Lo M-W, Schary WL, Whitney CC (1984) Bioequivalence, dose-proportionality, and pharmacokinetics of naltrexone after oral administration. J Clin Psych 45[9(2)]:15–19

    CAS  Google Scholar 

  • Michel ME, Bolger G, Weissman B-A (1985) Binding of a new opiate antagonist, nalmefene, to rat brain membranes. Meth Find Exp Clin Pharm 7(4):175–177

    CAS  Google Scholar 

  • Mirin SM, Mendelson JM, Ellingboe J, Meyer RE (1976) Acute effect of heroin and naltrexone on testosterone and gonadotropin secretion: a pilot study. Psychoneuroend 1:359–369

    Google Scholar 

  • Naber D, Pickar D, Davis GC, Cohen RM, Jimerson DC, Elchisak MA, Defraites EG, Kalin NH, Risch SC, Buchsbaum MS (1981) Naloxone effects onβendorphin, cortisol, prolactin, growth hormone, HVA and MHPG in plasma of normal volunteers. Psychopharmacology 74:125–128

    PubMed  CAS  Google Scholar 

  • National Research Council Committee on Clinical Evaluation of Narcotic Antagonists (1978) Clinical evaluation of naltrexone treatment of opiate-dependent individuals. Arch Gen Psych 35:335–340

    Google Scholar 

  • O’Brien CP, Greenstein RA, Mintz J, Woody GE (1975) Clinical experience with naltrexone. Am J Dr Alc Ab 2: 365 – 377

    Google Scholar 

  • Olley JE, Tiong GKL (1988) Letters to the editor: Plasma levels of opioid material in man following sublingual and intravenous administration of buprenorphine: exogenous/endogenous opioid interaction? J Pharm Pharmacol 40:666–667

    PubMed  CAS  Google Scholar 

  • Pickworth WB, Johnson RE, Holicky BA, Cone EJ (1993) Subjective and physiologic effects of intravenous buprenorphine in humans 53:570–576

    CAS  Google Scholar 

  • The Possilpark Group (1993) Drug injectors in Glasgow: a community at risk? A report from a multidisciplinary group. Health Bulletin 51(6):418–429

    Google Scholar 

  • Quigley AJ, Bredemeyer DE, Seow SS (1984) A case of buprenorphine abuse. Med J Aust 142:425–426

    Google Scholar 

  • Reisinger M (1985) Buprenorphine as new treatment for heroin dependence. Dr Alc Depend 16:257–262

    CAS  Google Scholar 

  • Resnick R, Fink M, Freedman AM (1974) High-dose cyclazocine therapy of opiate dependence. Am J Psych 131(5):595–597

    CAS  Google Scholar 

  • Resnick RB, Resnick E, Galanter M (1991) Buprenorphine responders: a diagnostic subgroup of heroin addicts? Prog Neuro-Psychopharmacol Biol Psychiat 15:531–538

    CAS  Google Scholar 

  • Resnick RB, Galanter M, Pycha C, Cohen A, Grandison P, Flood N (1992) Buprenorphine: an alternative to methadone for heroin dependence treatment. Psychopharm Bull 28(1):109–113

    CAS  Google Scholar 

  • Rosen MI, Pearsall HR, McDougle CJ, Price LH, Woods SW, Kosten TR (1993) Effects of acute buprenorphine on responses to intranasal cocaine: a pilot study. Am J Dr Alc Ab 19(4):451–464

    CAS  Google Scholar 

  • Saarialho-Kere U, Mattila MJ, Paloheimo M, Seppala T (1987) Psychomotor, respiratory, and neuroendocrinological effects of buprenorphine and amitriptyline in healthy volunteers 139–146

    Google Scholar 

  • Seow SSW, Quigley AJ, Ilett KF, Dusci LJ, Swensen G, Harrison-Stewart A, Rappaport L (1986) Buprenorphine: a new maintenance opiate? Med J Austr 144:407–411

    CAS  Google Scholar 

  • Strain EC, Preston KL, Liebson IA, Bigelow GE (1992) Acute effects of buprenorphine, hydromorphone, and naloxone in methadone-maintained volunteers. JPET 261:985–993

    CAS  Google Scholar 

  • Tempel A, Zukin RS, Gardner EL (1982) Supersensitivity of brain opiate receptor subtypes after chronic naltrexone treatment. Life Sci 31:1401–1404

    PubMed  CAS  Google Scholar 

  • Tennant FS, Rawson RA, Cohen AJ, Mann A (1984) Clinical experience with naltrexone in suburban opioid addicts. J Clin Psych 45[9(2)]:42–45

    Google Scholar 

  • Teoh SK, Mendelson JH, Mello NK, Kuehnle J, Sintavanarong P, Rhoades EM (1993) Acute interactions of buprenorphine with intravenous cocaine and morphine: An investigational new drug phase I safety evaluation. J Clin Psychopharm 13 (2): 87 – 99

    CAS  Google Scholar 

  • U.S. patent #3,332,950, July 25, 1967

    Google Scholar 

  • Verebey K, Mule SJ (1975) Naltrexone pharmacology, pharmacokinetics, and metabolism: current status. Am J Dr Alc Ab 2(3–4):357–363

    CAS  Google Scholar 

  • Verebey K, Volavka J, Mule SJ, Resnick RB (1976) Naltrexone: disposition, metabolism, and effects after acute and chronic dosing. Clin Pharm Ther 20:315–328

    CAS  Google Scholar 

  • Volavka J, Resnick RB, Kestenbaum RS, Freedman AM (1976) Short-term effects of naltrexone in 155 heroin ex-addicts. Bio Psych 11(6):679–685

    CAS  Google Scholar 

  • Volavka J, Cho D, Mallya A, Bauman J (1979a) Naloxone increases ACTH and cortisol levels in man. NE J Med 300(18):1056–1057

    CAS  Google Scholar 

  • Volavka J, Mallya A, Bauman J, Pevnick J, Cho D, Reker D, James B, Dornbush R (1979b) Hormonal and other effects of naltrexone in normal men. Adv Exp Med Bio 116:291–305

    CAS  Google Scholar 

  • Volavka J, Bauman J, Pevnick J, Reker D, James B, Cho D (1980) Short–term hormonal effects of naloxone in man. Psychoneuroend 5:225–234

    CAS  Google Scholar 

  • Wall ME, Brine DR, Perez-Reyes M (1981) Metabolism and disposition of naltrexone in man after oral and intravenous administration. Dr Met Disp 9(4):369–375

    CAS  Google Scholar 

  • Wall ME, Perez-Reyes M, Brine DR, Cook CE (1984) Naltrexone disposition in man after subcutaneous administration. Dr Met Disp 12(6):677–682

    CAS  Google Scholar 

  • Weinberg DS, Inturrisi CE, Reidenberg B, Moulin DE, Nip TJ, Wallenstein S, Houde RW, Foley KM (1988) Sublingual absorption of selected opioid analgesics. Clin Pharm Ther 44:335–342

    CAS  Google Scholar 

  • Woods JH, France CP, Winger GD (1992) Behavioral pharmacology of buprenorphine: issues relevant to its potential in treating drug abuse. NIDA Research Monograph 121:12–27

    PubMed  CAS  Google Scholar 

  • Zaks A, Jones T, Fink M, Freedman AM (1971) Naloxone treatment of opiate dependence. JAMA 215(13):2108–2110

    PubMed  CAS  Google Scholar 

  • Zukin RS, Sugarman JR, Fitz-Syage ML, Gardner EL, Zukin SR, Gintzler AR (1982) Naltrexone-induced opiate receptor supersensitivity. Brain Res 245:288–292

    Google Scholar 

Download references

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 1996 Springer-Verlag Berlin Heidelberg

About this chapter

Cite this chapter

Kreek, M.J. (1996). Long-Term Pharmacotherapy for Opiate (Primarily Heroin) Addiction: Opioid Antagonists and Partial Agonists. In: Schuster, C.R., Kuhar, M.J. (eds) Pharmacological Aspects of Drug Dependence. Handbook of Experimental Pharmacology, vol 118. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-642-60963-3_15

Download citation

  • DOI: https://doi.org/10.1007/978-3-642-60963-3_15

  • Publisher Name: Springer, Berlin, Heidelberg

  • Print ISBN: 978-3-642-64631-7

  • Online ISBN: 978-3-642-60963-3

  • eBook Packages: Springer Book Archive

Publish with us

Policies and ethics