Skip to main content
  • 2624 Accesses

Abstract

Pharmacotherapeutic interventions for addiction focus on treatment and prevention of dependence. Addiction is a complex biological process and its complete underlying pathophysiology remains elusive. Successful treatment involves both short-term and long-term maintenance pharmacologic approaches linked with various psychosocial support avenues. However, specific agents have been developed for alcohol, nicotine, and opiate dependence. Disulfiram has been used since the 1950s as an aversive therapeutic technique where its metabolites when exposed to alcohol lead to the patient experiencing symptoms of nausea and vomiting, encouraging the patient to abstain from alcohol. Acamprosate is indicated for alcohol abstinence maintenance with the chemical component acetyl-homotaurine measured in pharmacokinetic studies. Acamprosate is primarily renally excreted. Varenicline is a partial agonist of the central nicotinic acetylcholine receptor and approved for the treatment of nicotine dependence. Varenicline is mainly renally excreted where a combined pharmacokinetic-pharmacodynamic model described this compound with an open two-compartment pharmacokinetic model with a linear pharmacodynamic model. Naloxone and naltrexone are opiate antagonists used in a variety of clinical settings. Naloxone is a “rescue” agent for opiate overdose. Naltrexone is used for alcohol dependence and available in an oral and a monthly long-acting depot injection. Naloxone has been combined with pentazocine, buprenorphine, and oxycodone to prevent abuse. Levo-alpha acetyl methadol (LAAM) is a potent derivative of methadone but QTc prolongation occurrences have limited its clinical use.

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 99.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 129.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 199.99
Price excludes VAT (USA)
  • Durable hardcover 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

References

  1. Doering PL, Li RM (2014) Chapter 49. Substance-related disorders II. In: DiPiro JT, Talbert RL, Yee GC et al. (eds) Pharmacotherapy, 9th edn. McGraw Hill, New York. pp. 1003–1018

    Google Scholar 

  2. Wilde MI, Wagstaff AJ (1997) Acamprosate. Drugs 53:1038–1053

    Article  CAS  PubMed  Google Scholar 

  3. Scott LJ, Figgitt DP, Keam SJ, Waugh J (2005) Acamprosate. CNS Drugs 19:445–464

    Article  CAS  PubMed  Google Scholar 

  4. Mason BJ (2001) Treatment of alcohol-dependent outpatients with acamprosate: a clinical review. J Clin Psychiatry 62(Suppl 20):42–48

    CAS  PubMed  Google Scholar 

  5. Saivin S, Hulot T, Chabac S et al (1998) Clinical pharmacokinetics of acamprosate. Clin Pharmacokinet 35:331–345

    Article  CAS  PubMed  Google Scholar 

  6. Peachy JE, Brien JF, Roach CA, Loomis CW (1981) A comparative review of the pharmacological and toxicological properties of disulfiram and calcium carbimide. J Clin Psychopharmacol 1:21–26

    Article  Google Scholar 

  7. Peachy JE, Sellers EM (1981) The disulfiram and calcium carbimide acetaldehyde-mediated ethanol reactions. Pharmacol Ther 15:89–97

    Article  Google Scholar 

  8. Johnansson B (1992) A review of the pharmacokinetics and pharmacodynamics of disulfiram and its metabolites. Acta Psychiatr Scand Suppl 369:15–26

    Article  Google Scholar 

  9. Hudmon KS, Kroon LA, Corelli RL (2012) Chapter 47. Smoking cessation. In: Krinsky DL, Berardi RR, Ferreri SP (eds) Handbook of nonprescription drugs, 17th edn. American Pharmacists Association, Washington DC, pp 885–910

    Google Scholar 

  10. Svensson CK (1987) Clinical pharmacokinetics of nicotine. Clin Pharmacokinet 12:30–40

    Article  CAS  PubMed  Google Scholar 

  11. Benowitz NL, Hukkanen J, Jacob P III (2009) Nicotine chemistry, metabolism, kinetics, and biomarkers. Handb Exp Pharmacol 192:29–60

    Article  CAS  PubMed  Google Scholar 

  12. Faessel HM, Obach RS, Rollema H et al (2010) A review of the clinical pharmacokinetics and pharmacodynamics of varenicline for smoking cessation. Clin Pharmacokinet 49:799–816

    Article  CAS  PubMed  Google Scholar 

  13. Feng B, Obach RS, Burstein AH et al (2008) Effect of human renal cationic transporter inhibition on the pharmacokinetics of varenicline, a new therapy for smoking cessation: an in vitro – in vivo study. Clin Pharmacol Ther 83:567–576

    Article  CAS  PubMed  Google Scholar 

  14. Ravva P, Gastonguay MR, Tensfeldt TG, Faessel HM (2009) Population pharmacokinetic analysis of varenicline in adult smokers. Br J Clin Pharmacol 68:669–681

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Faessel HM, Gibbs MA, Clark DJ et al (2006) Multiple-dose pharmacokinetics of the selective nicotinic receptor partial agonist, varenicline, in healthy smokers. J Clin Pharmacol 46:1439–1448

    Article  CAS  PubMed  Google Scholar 

  16. Kikkawa H, Maruyama N, Fujimoto Y, Hasunuma T (2011) Single and multiple-dose pharmacokinetics of the selective nicotinic receptor partial agonist, varenicline, in healthy Japanese adult smokers. J Clin Pharmacol 51:527–537

    Article  CAS  PubMed  Google Scholar 

  17. Ravva P, Gastonguay MR, Faessel HM, Lee TC, Niaura R (2014) Pharmacokinetic-pharmacodynamic modeling of the effect of varenicline on nicotine craving in adult smokers. Nicotine Tob Res 17(1):106–113. doi:10.1093/intr/ntu154

    Article  PubMed  Google Scholar 

  18. King DP, Paciga S, Pickering E et al (2012) Smoking cessation pharmacogenetics: analysis of varenicline and bupropion in placebo-controlled clinical trials. Neuropsychopharmacol 37:641–650

    Article  CAS  Google Scholar 

  19. Blumberg H, Dayton HB, Wolf PS (1966) Counteraction of narcotic antagonist by the narcotic antagonist Naloxone. Proc Soc Exp Biol Med 123:755–758

    Article  CAS  PubMed  Google Scholar 

  20. Evans LE, Swainson CP, Roscoe P, Prescott LF (1973) Treatment of drug overdose with naloxone, a specific narcotic antagonist. Lancet 1(7601):452–455

    Article  CAS  PubMed  Google Scholar 

  21. Handal KA, Schauben JL, Salamone FR (1983) Naloxone. Ann Emerg Med 12:438–445

    Article  CAS  PubMed  Google Scholar 

  22. Berkowitz BA (1976) The relationship of pharmacokinetics to pharmacologic activity: morphine, methadone, and naloxone. Clin Pharmacokinet 1:219–230

    Article  CAS  PubMed  Google Scholar 

  23. Fishman J, Roffwarg H, Hellman L (1973) Disposition of naloxone in normal narcotic dependent men. J Pharmacol Exp Ther 187:575–580

    CAS  PubMed  Google Scholar 

  24. Weinstein SH, Pfeffer M, Schor JM et al (1971) Metabolites of naloxone in human urine. J Pharm Sci 60:1567–1568

    Article  CAS  PubMed  Google Scholar 

  25. Ngal SH, Berkowitz BA, Yang JC, Hempstead J, Spector S (1976) Pharmacokinetics of naloxone in rats and man. Anesthesiology 44:398–401

    Article  Google Scholar 

  26. Berkowitz BA, Ngal SH, Hempstead J, Spector S (1975) Disposition of naloxone: use of a new radioimmunoassay. J Pharmacol Exp Ther 195:499–504

    CAS  PubMed  Google Scholar 

  27. Cone EF (1976) General procedure for the isolation and identification of a 6-α and 6-β hydroxyl metabolites of narcotic agonists and antagonists with a hydroxymorphine structure. J Chromatogr 129:355–361

    Article  CAS  PubMed  Google Scholar 

  28. Asali LA, Brown KF (1984) Naloxone protein binding in adult and foetal plasma. Eur J Clin Pharmacol 27:459–463

    Article  CAS  PubMed  Google Scholar 

  29. Kanaan M, Daali Y, Dayer P, Desmeules J (2009) P-glycoprotein is not involved in the differential oral potency of naloxone and naltrexone. Fundam Clin Pharmacol 23:543–548

    Article  CAS  PubMed  Google Scholar 

  30. Dowling J, Isbister GK, Kirkpatrick CM, Naidoo D, Graudins A (2008) Population pharmacokinetics of intravenous, intramuscular, and intranasal naloxone in human volunteers. Ther Drug Monit 30:490–496

    CAS  PubMed  Google Scholar 

  31. Yamamoto R, Takasuga S, Yoshida Y et al (2012) In vitro and in vivo transdermal iontophoretic delivery of naloxone, an opioid antagonist. Int J Pharm 422:132–138

    Article  CAS  PubMed  Google Scholar 

  32. Sanaei-Zadeh H (2012) Is transdermal iontophoretic delivery of naloxone sufficient for the management of intoxication in opioid-overdosed patients? Int J Pharm 428:187

    Article  CAS  PubMed  Google Scholar 

  33. Robinson A, Wermeling DP (2014) Intranasal naloxone administration for treatment of opioid overdose. Am J Health Syst Pharm 71:2129–2135

    Article  CAS  PubMed  Google Scholar 

  34. Greenberg MI, Roberts JR, Baskin SI (1980) Endotracheal naloxone reversal of morphine-induced respiratory depression in rabbits. Ann Emerg Med 9:289–292

    Article  CAS  PubMed  Google Scholar 

  35. Goldfrank L, Weisman RS, Errick JK, Lo MW (1986) A dosing nomogram for continuous infusion intravenous naloxone. Ann Emerg Med 15:566–570

    Article  CAS  PubMed  Google Scholar 

  36. Groeger JS, Inturrisi CE (1987) High-dose naloxone: pharmacokinetics in patients in septic shock. Crti Care Med 15:751–756

    Article  CAS  Google Scholar 

  37. Hanes SD, Franklin M, Kuhl DA, Headley AS (1999) Prolonged opioid antagonist with naloxone in chronic renal failure. Pharmacother 19:897–901

    Article  CAS  Google Scholar 

  38. Olofsen E, van Dorp E, Teppema L et al (2010) Naloxone reversal of morphine and morphine-6-glucuronide-induced respiratory depression in healthy volunteers. Anesthesiology 112:1417–1427

    Article  CAS  PubMed  Google Scholar 

  39. Yassan A, Oolofsen E, van Dorp E et al (2007) Mechanism-based pharmacokinetic-pharmacodynamic modeling of the reversal of buprenorphine-induced respiratory depression by naloxone: a study in healthy volunteers. Clin Pharmacokinet 46:965–980

    Article  Google Scholar 

  40. Culpepper-Morgan JA, Inturrisi CE, Portenoy RK et al (1992) Treatment of opioid-induced constipation with oral naloxone: a pilot study. Clin Pharmacol Ther 52:90–95

    Article  CAS  PubMed  Google Scholar 

  41. Gibson CM, Pass SE (2014) Enteral naloxone for the treatment of opioid-induced constipation in the medical intensive care unit. J Crit Care 29:803–807

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  43. Gonzalez JP, Brogden RN (1988) Naltrexone: a review of its pharmacodynamics and pharmacokinetic properties and therapeutic efficacy in the management of opioid-dependence. Drugs 35:192–313

    Article  CAS  PubMed  Google Scholar 

  44. Fujimoto JM, Roerig S, Wang RI, Cahtterjie N, Inturrisi CE (1975) Narcotic antagonistic activity of several metabolites of naloxone and naltrexone tested in morphine dependent mice (38558). Proc Soc Exp Biol Med 148:443–448

    Article  CAS  PubMed  Google Scholar 

  45. Cone EJ, Gorodedtzky CW, Yeh S (1974) The urinary excretion profile of naltrexone and metabolites in man. Drug Metab Dispos 2:506–512

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  47. Lee MC, Wagner HN, Tanada S et al (1988) Duration of occupancy of opiate receptors by naltrexone. J Nucl Med 29:1207–1211

    CAS  PubMed  Google Scholar 

  48. Galloway GP, Koch M, Cello R, Smith DE (2005) Pharmacokinetics, safety, and tolerability of a depot formulation naltrexone in alcoholics: an open-label trial. BMC Psychiatry 5:18. doi:10.1186/1471-244X-5-18

    Article  PubMed  PubMed Central  Google Scholar 

  49. Dunbar JL, Truncliff RZ, Hayes SC, Farrell CB (2007) Population pharmacokinetics of extended-release injectable naltrexone (XR-NTX) in patients with alcohol dependence. J Stud Alcohol Drugs 68:862–870

    Article  PubMed  Google Scholar 

  50. Dunbar JL, Turncliff RZ, Dong Q et al (2006) Single and multiple-dose pharmacokinetics of long-acting injectable naltrexone. Alcohol Clin Exp Res 30:480–490

    Article  CAS  PubMed  Google Scholar 

  51. Turncliff RZ, Dunbar JL, Dong Q et al (2005) Pharmacokinetics of long-acting naltrexone in subjects with mild to moderate hepatic impairment. J Clin Pharmacol 45:1259–1267

    Article  CAS  PubMed  Google Scholar 

  52. Bart G (2012) Maintenance medication for opiate addiction: the foundation of recovery. J Addcit Dis 31:207–225

    Article  Google Scholar 

  53. Challoner KR, McCarron MM, Newton EJ (1990) Pentazocine (Talwin) intoxication: report of 57 cases. J Emerg Med 8:67–74

    Article  CAS  PubMed  Google Scholar 

  54. Stahl SM, Kasser IS (1983) Pentazocine overdose. Ann Emerg Med 12:63–65

    Article  Google Scholar 

  55. Legros J, Khalili-Varasteh H, Margetts G (1984) Pharmacologic study of pentazocine-naloxone combination: interest as a potentially nonabusable oral form of pentazocine. Arch Int Pharmacodyn Ther 271:11–21

    CAS  PubMed  Google Scholar 

  56. Swift JQ, Hargreaves KM (1993) Pentazocine analgesia: is there a niche for Talwin Nx? Compendium 14:1048

    CAS  PubMed  Google Scholar 

  57. Lahmeyer HW, Craig RJ (1987) Pentazocine-naloxone: another “addiction-proof” drug of abuse. Int J Addict 22:1163–1166

    Article  CAS  PubMed  Google Scholar 

  58. Baum C, Hsu JP, Nelson RC (1987) The impact of the addition of naloxone on the use and abuse of pentazocine. Public Health Rep 102:426–429

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Reed DA, Schnoll SH (1986) Abuse of pentazocine-naloxone combination. JAMA 256:2562–2564

    Article  CAS  PubMed  Google Scholar 

  60. Levine JD, Gordon CG, Taiwo YO, Coderra TJ (1988) Potentiation of pentazocine analgesia by low-dose naloxone. J Clin Invest 82:1574–1577

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Orman JS, Keating GM (2009) Buprenorphine/naloxone. Drugs 69:577–607

    Article  CAS  PubMed  Google Scholar 

  62. Middleton LS, Nuzzo PA, Lofwall MR, Moody DE, Walsh SL (2011) The pharmacodynamics and pharmacokinetic profile of intranasal crushed buprenorphine/naloxone tablets in opioid-abusers. Addiction 106:1460–1473

    Article  PubMed  PubMed Central  Google Scholar 

  63. Nath RP, Upton RA, Everhart ET et al (1999) Buprenorphine pharmacokinetics: relative bioavailability of sublingual tablet and liquid formulations. J Clin Pharmacol 39:619–623

    Article  CAS  PubMed  Google Scholar 

  64. Center for Drug Evaluation and Research. Clinical pharmacology and biopharmaceutics review: application number: 20–732, 20–733 (Suboxone®) [online]. Available from URL: http://fda.gov/cder. Accessed 22 Nov 2014

  65. Suboxone commercial web page (2014) Reckitt-Benckiser Inc. Available from: http://www.suboxone.com/. Accessed 22 Nov 2014

  66. Fischer A, Jonsson M, Hjelmstrom P (2015) Pharmaceutical and pharmacokinetic characterization of a novel sublingual buprenorphine/naloxone tablet formulation in healthy volunteers. Drug Dev Ind Pharm 41(1):79–84; early on-line 1–6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Mendelson J, Jones RT (2003) Clinical and pharmacologic evaluation of buprenorphine and naloxone combinations: why the 4:1 ratio for treatment. Drug Alcohol Depend 70:S29–S37

    Article  CAS  PubMed  Google Scholar 

  68. Chaing CN, Hawks RL (2003) Pharmacokinetics of the combination tablet of buprenorphine and naloxone. Drug Alcohol Depend 70:S39–S47

    Article  Google Scholar 

  69. Mintzer MZ, Correia CJ, Strain EC (2004) A dose-effect study of repeated administration of buprenorphine/naloxone on performance in opioid-dependent volunteers. Drug Alcohol Depend 74:205–209

    Article  CAS  PubMed  Google Scholar 

  70. Harris DS, Mendelson JE, Lin ET, Upton RA, Jones RT (2004) Pharmacokinetics and subjective effects of sublingual buprenorphine, alone or in combination with naloxone. Clin Pharmacokinet 43:329–340

    Article  CAS  PubMed  Google Scholar 

  71. Harris DS, Jones RT, Wlem S et al (2000) Buprenorphine and naloxone co-administration in opiate-dependent patients stabilized on sublingual buprenorphine. Drug Alcohol Depend 61:85–94

    Article  CAS  PubMed  Google Scholar 

  72. Parwatikar SD, Knowles RR (1973) Methadone-naloxone in combination for the treatment of heroin addicts. Clin Pharmacol Ther 14:941–948

    Article  CAS  PubMed  Google Scholar 

  73. Nutt JG, Jasinski DR (1974) Methadone-naloxone mixtures for use in methadone maintenance programs. Clin Pharmacol Ther 15:156–166

    Article  CAS  PubMed  Google Scholar 

  74. Loimer N, Presslich O, Grunberger J, Linzmayer L (1991) Combined naloxone-methadone preparations for opiate substitution therapy. J Subst Abuse Treat 8:157–160

    Article  CAS  PubMed  Google Scholar 

  75. Bell J, Shearer J, Ryan A et al (2009) The acceptability, safety, and tolerability of methadone-naloxone in a 50:1 ratio. Exp Clin Psychopharmacol 17:146–153

    Article  CAS  PubMed  Google Scholar 

  76. Mueller-Lissner S (2010) Fixed combination of oxycodone with naloxone: a new way to prevent and treat opioid-induced constipation. Adv Ther 27:581–590

    Article  CAS  PubMed  Google Scholar 

  77. Leppert W (2014) Oxycodone/naloxone in the management of patients with pain and opioid-induced bowel dysfunction. Curr Drug Targets 15:124–135

    Article  CAS  PubMed  Google Scholar 

  78. Meissner W, Leyendecker P, Mueller-Lissner S et al (2009) A randomized controlled trial with prolonged-release oral oxycodone and naloxone to prevent and reverse opioid-induced constipation. Eur J Pain 13:56–64

    Article  CAS  PubMed  Google Scholar 

  79. Vondrackova D, Leyendecker P, Meissner W et al (2008) Analgesic effect and safety of oxycodone in combination with naloxone as prolonged release tablets in patients with moderate to severe chronic pain. J Pain 9:1144–1154

    Article  CAS  PubMed  Google Scholar 

  80. Smith K, Hopp M, Mundin G et al (2008) Single- and multiple dose pharmacokinetic evaluation of oxycodone and naloxone in an opioid agonist/antagonist prolonged-release combination in healthy adult volunteers. Clin Ther 30:2051–2068

    Article  CAS  PubMed  Google Scholar 

  81. Smith K, Hopp M, Mundin G et al (2011) Naloxone as part of a prolonged release oxycodone/naloxone combination reduces oxycodone-induced slowing of gastrointestinal transit in healthy volunteers. Expert Opin Investig Drugs 20:427–439

    Article  CAS  PubMed  Google Scholar 

  82. Mercadante S, Ferrera P, Adile C (2011) High doses of oxycodone-naloxone combination may provide poor analgesia. Support Care Cancer 19:1471–1472

    Article  PubMed  Google Scholar 

  83. Duggan ST, Scott LJ (2010) Morphine/Naltrexone. CNS Drugs 24:527–538

    Article  CAS  PubMed  Google Scholar 

  84. Johnson F, Setnik B (2011) Morphine sulfate and naltrexone hydrochloride extended release capsules: naltrexone release, pharmacodynamics and tolerability. Pain Physician 14:391–406

    PubMed  Google Scholar 

  85. Johnson F, Ciric S, Boudriau S, Swearingen D, Stauffer J (2010) Food effects on the pharmacokinetics of morphine sulfate and naltrexone hydrochloride extended release capsules. Adv Ther 27:846–858

    Article  CAS  PubMed  Google Scholar 

  86. Johnson FK, Stark JG, Bieberdorf FA, Stauffer J (2010) Relative oral bioavailability of morphine and naltrexone derived from crushed morphine sulfate and naltrexone hydrochloride extended-release capsules versus intact product and versus naltrexone solution: a single-dose, randomized-sequence, open-label, three-way crossover trial in healthy volunteers. Clin Ther 32:1149–1164

    Article  CAS  PubMed  Google Scholar 

  87. Katz N, Sun S, Jonhson F, Stauffer J (2010) ALO-01 (morphine sulfate and naltrexone hydrochloride) extended release capsules in the treatment of chronic pain of osteoarthritis of the hip or knee: pharmacokinetics, efficacy, and safety. J Pain 11:303–311

    Article  CAS  PubMed  Google Scholar 

  88. King Pharmaceuticals Inc. Embeda® (morphine sulfate and naltrexone hydrochloride) extended release capsules for oral use [online]. Available from URL: http://www.accessdata.fda.gov/drugsatfda_docs/label/2009/

  89. Stauffer J, Setnik B, Sokolowski M (2009) Subjective effects and safety of whole and tampered morphine sulfate and naltrexone hydrochloride (ALO-01) extended-release capsules versus morphine solution and placebo in experienced non-dependent opioid users: a randomized, double-blind, placebo-controlled, crossover study. Clin Drug Investig 29:777–790

    Article  CAS  PubMed  Google Scholar 

  90. Finn P, Wolcock K (1997) Levo-alpha acetyl methadol (LAAM). J Subst Abuse Treat 14:559–564

    Article  CAS  PubMed  Google Scholar 

  91. Center for Substance Abuse Treatment (1995). Treatment Improvement Protocol (TIP) Series, No 22. LAAM in the treatment of opiate addiction. Rockville, MD: Department of Health and Human Services

    Google Scholar 

  92. Weineke H, Conrads H, Wolstein J et al (2009) Levo-α-acetylmethadol (LAAM) induced QTc prolongation – results from a controlled clinical trial. Eur J Med Res 14:7–12

    Article  Google Scholar 

  93. US Food and Drug Administration: Product Discontinuation Notice (2003) ORLAAM® (Levomethadyl hydrochloride) Oral Solution, 10 mg/mL, CII. Available at http://www.fda.gov

  94. Kharasch ED, Whittington D, Hoffer C et al (2005) Paradoxical role of cytochrome P450 3A in the bioactivation and clinical effects of levo-α-acetylmethadol. Clin Pharmacokinet 44:731–751

    Article  CAS  PubMed  Google Scholar 

  95. Henderson GL, Wilson BK, Lau DH (1977) Plasma l-alpha-acetylmethadol (LAAM) after acute and chronic administration. Clin Pharmacol Ther 21:16–25

    Article  CAS  PubMed  Google Scholar 

  96. Crettol S, Digon P, Golay KP, Brawand M, Eap CB (2007) In vitro P-glycoprotein-mediated transport of (R), (S), (R, S)-methadone, LAAM and their main metabolites. Pharmacology 80:304–311

    Article  CAS  PubMed  Google Scholar 

  97. Moody DE, Walsh SL, Rollins DE, Neff JA, Huang W (2004) Ketoconazole, a cytochrome P450 3A4 inhibitor, markedly increases concentrations of levo-acetyl-alpha-methadol in opioid-naïve individuals. Clin Pharmacol Ther 76:154–166

    Article  CAS  PubMed  Google Scholar 

  98. Huang W, Bemis PA, Slawson MH, Moody DE (2003) Determination of l-α-acetylmethadol (LAAM), norLAAL, and dinorLAAM in clinical and in vitro samples using liquid chromatography with electrospray ionization and tandem mass spectrometry. J Pharm Sci 92:10–20

    Article  CAS  PubMed  Google Scholar 

  99. Eissenberg T, Stitzer ML, Bigelow GE, Buchhalter AR, Walsh SL (1999) Relative potency of levo-α-acetylmethadol and methadone in humans under acute dosing conditions. J Pharmacol Exp Ther 289:936–945

    CAS  PubMed  Google Scholar 

  100. Henderson GL, North-Root H, Kuttab SH (1977) Metabolism and disposition of l-α-acetylmethadol in the rat. Drug Metab Dispos 5:321–328

    CAS  PubMed  Google Scholar 

  101. Walsh SL, Johnson RE, Cone EJ, Begelow GE (1998) Intravenous and oral l-α-acetylmethadol: pharmacodynamics and pharmacokinetics in humans. J Pharmacol Exp Ther 285:71–82

    CAS  PubMed  Google Scholar 

  102. Newcombe DA, Bochner F, White JM, Somogyi AA (2004) Evaluation of levo-alpha-acetylmethadol (LAAM) as an alternative treatment for methadone maintenance patients who regularly experience withdrawal: a pharmacokinetic and pharmacodynamic analysis. Drug Alcohol Depend 76:63–72

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michael W. Jann PharmD .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2016 Springer International Publishing Switzerland

About this chapter

Cite this chapter

Jann, M.W. (2016). Anti-addiction Agents. In: Jann, M., Penzak, S., Cohen, L. (eds) Applied Clinical Pharmacokinetics and Pharmacodynamics of Psychopharmacological Agents. Adis, Cham. https://doi.org/10.1007/978-3-319-27883-4_14

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-27883-4_14

  • Published:

  • Publisher Name: Adis, Cham

  • Print ISBN: 978-3-319-27881-0

  • Online ISBN: 978-3-319-27883-4

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics