Skip to main content

Advertisement

Log in

Can the Chronic Administration of the Combination of Buprenorphine and Naloxone Block Dopaminergic Activity Causing Anti-reward and Relapse Potential?

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Opiate addiction is associated with many adverse health and social harms, fatal overdose, infectious disease transmission, elevated health care costs, public disorder, and crime. Although community-based addiction treatment programs continue to reduce the harms of opiate addiction with narcotic substitution therapy such as methadone maintenance, there remains a need to find a substance that not only blocks opiate-type receptors (mu, delta, etc.) but also provides agonistic activity; hence, the impetus arose for the development of a combination of narcotic antagonism and mu receptor agonist therapy. After three decades of extensive research, the federal Drug Abuse Treatment Act 2000 (DATA) opened a window of opportunity for patients with addiction disorders by providing increased access to options for treatment. DATA allows physicians who complete a brief specialty-training course to become certified to prescribe buprenorphine and buprenorphine/naloxone (Subutex, Suboxone) for treatment of patients with opioid dependence. Clinical studies indicate that buprenorphine maintenance is as effective as methadone maintenance in retaining patients in substance abuse treatment and in reducing illicit opioid use. With that stated, we must consider the long-term benefits or potential toxicity attributed to Subutex or Suboxone. We describe a mechanism whereby chronic blockade of opiate receptors, in spite of only partial opiate agonist action, may ultimately block dopaminergic activity causing anti-reward and relapse potential. While the direct comparison is not as yet available, toxicity to buprenorphine can be found in the scientific literature. In considering our cautionary note in this commentary, we are cognizant that, to date, this is what we have available, and until such a time when the real magic bullet is discovered, we will have to endure. However, more than anything else this commentary should at least encourage the development of thoughtful new strategies to target the specific brain regions responsible for relapse prevention.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Wood E, Li K, Palepu A, Marsh DC, Schechter MT, Hogg RS, Montaner JS, Kerr T (2005) Sociodemographic disparities in access to addiction treatment among a cohort of Vancouver injection drug users. Subst Use Misuse 40:1153–1167

    PubMed  Google Scholar 

  2. Schottenfeld RS, Chawarski MC, Pakes JR, Pantalon MV, Carroll KM, Kosten TR (2005) Methadone versus buprenorphine with contingency management or performance feedback for cocaine and opioid dependence. Am J Psychiatry 162:340–349

    PubMed  Google Scholar 

  3. Dole VP, Nyswander ME (1966) Rehabilitation of heroin addicts after blockade with methadone. N Y State J Med 66:2011–2017

    PubMed  CAS  Google Scholar 

  4. Goldstein A (1991) Heroin addiction: neurobiology, pharmacology, and policy. J Psychoactive Drugs 23:123–133

    PubMed  CAS  Google Scholar 

  5. Butler M, RL Kane, D McAlpine, RG Kathol, SS Fu, H Hagedorn, TJ Wilt (2008) Integration of Mental Health/Substance Abuse and Primary Care No. 173 (Prepared by the Minnesota Evidence-based Practice Center under Contract No. 290-02-0009.) AHRQ Publication No. 09-E003. Rockville, MD. Agency for Healthcare Research and Quality, October

  6. US Department of Health and Human Services and Substance Abuse and Mental Health Services Administration (SAMHSA) (2003) Opioid agonist therapy (OAT) with methadone or LAAM. Available at http://dpt.samhsa.gov/001025optx.htm. Accessed February 26, 2003

  7. Judson BA, Goldstein A (1984) Naltrexone treatment of heroin addiction: one-year follow-up. Drug Alcohol Depend 13:357–365

    PubMed  CAS  Google Scholar 

  8. Blum K, Futterman S, Wallace JE, Schwertner HA (1977) Naloxone-induced inhibition of ethanol dependence in mice. Nature 265:49–51

    PubMed  CAS  Google Scholar 

  9. Arfken CL, Johanson CE, di Menza S, Schuster CR (2010) Expanding treatment capacity for opioid dependence with office-based treatment with buprenorphine: National surveys of physicians. J Subst Abuse Treat 39(2):96–104

    PubMed  Google Scholar 

  10. McNicholas L (2004) SAMHSA clinical guidelines for the use of buprenorphine in the treatment of opioid addiction. Treatment Improvement Protocol, 40. DHHS Publication No. (SMA) 04-3939

  11. Stock C, Shum JH (2004) Bupreorphine:a new pharmacotherapy for opioid addictions treatment. J Pain Palliat Care Pharmacother 18:35–54

    PubMed  Google Scholar 

  12. Kreek MJ, LaForge KS, Butelman E (2002) Pharmacotherapy of addictions. Nat Rev Drug Discov 1:710–726

    PubMed  CAS  Google Scholar 

  13. O’Brien CP (2005) Anticraving medications for relapse prevention: a possible new class of psychoactive medications. Am J Psychiatry 162:1423–1431

    PubMed  Google Scholar 

  14. Dackis C, O’Brien C (2005) Neurobiology of addiction: treatment and public policy ramifications. Nat Neurosci 8:1431–1436

    PubMed  CAS  Google Scholar 

  15. Heidbreder CA, Hagan JJ (2005) Novel pharmacotherapeutic approaches for the treatment of drug addiction and craving. Curr Opin Pharmacol 5:107–118

    PubMed  CAS  Google Scholar 

  16. Bruijnzeel AW, Marcinkiewcz C, Isaac S, Booth MM, Dennis DM, Gold MS (2007) The effects of buprenorphine on fentanyl withdrawal in rats. Psychopharmacology (Berl) 191:931–941

    CAS  Google Scholar 

  17. Gold MS (1993) Opiate addiction and the locus coeruleus. The clinical utility of clonidine, naltrexone, methadone, and buprenorphine. Psychiatr Clin North Am 16:61–73

    PubMed  CAS  Google Scholar 

  18. Doehring A, Hentig N, Graff J, Salamat S, Schmidt M, Geisslinger G, Harder S, Lötsch J (2009) Genetic variants altering dopamine D2 receptor expression or function modulate the risk of opiate addiction and the dosage requirements of methadone substitution. Pharmacogenet Genomics 19:407–414

    PubMed  CAS  Google Scholar 

  19. Kreek MJ (2000) Methadone-related opioid agonist pharmacotherapy for heroin addiction. History, recent molecular and neurochemical research and future in mainstream medicine. Ann N Y Acad Sci 909:186–216

    PubMed  CAS  Google Scholar 

  20. Yassen A, Olofsen E, Romberg R, Sarton E, Danhof M, Dahan A (2006) Mechanism-based pharmacokinetic-pharmacodynamic modeling of the antinociceptive effect of buprenorphine in healthy volunteers. Anesthesiology 104:1232–1242

    PubMed  CAS  Google Scholar 

  21. Hans G (2007) Buprenorphine—a review of its role in neuropathic pain. J Opioid Manag 3:195–206

    PubMed  Google Scholar 

  22. Chang Y, Moody DE, McCance-Katz EF (2006) Novel metabolites of buprenorphine detected in human liver microsomes and human urine. Drug Metab Dispos 34:440–448

    PubMed  CAS  Google Scholar 

  23. Cowan A (2003) Buprenorphine: new pharmacological aspects. Int J Clin Pract Suppl 133:3–8

    PubMed  CAS  Google Scholar 

  24. Lesscher HM, Bailey A, Burbach JP, Van Ree JM, Kitchen I, Gerrits MA (2003) Receptor-selective changes in mu-, delta- and kappa-opioid receptors after chronic naltrexone treatment in mice. Eur J Neurosci 17:1006–1012

    PubMed  Google Scholar 

  25. Blum K, Chen ALC, Bowirrat A, Downs BW, Waite RL, Reinking J et al (2009) Genes and happiness. Gene Ther Mol Biol 13:91–129

    CAS  Google Scholar 

  26. Gold MS, Dackis CA, Pottash AL, Sternbach HH, Annitto WJ, Martin D, Dackis MP (1982) Naltrexone, opiate addiction, and endorphins. Med Res Rev 2:211–246

    PubMed  CAS  Google Scholar 

  27. Kakko J, Svanborg KD, Kreek MJ, Heilig M (2003) 1-year retention and social function after buprenorphine-assisted relapse prevention treatment for heroin dependence in Sweden: a randomised, placebo-controlled trial Lancet. 22;361(9358):662–668

  28. Blum K, Kozlowski GP (1990) Ethanol and neuromodulator interactions: a cascade model of reward. In: Ollat H, Parvez S, Parvez H (eds) Alcohol and behavior. VSP Press, Utrecht, pp 131–149

    Google Scholar 

  29. Malhotra AK, Lencz T, Correll CU, Kane JM (2007) Genomics and the future of pharmacotherapy in psychiatry. Int Rev Psychiatry 19:523–530

    PubMed  Google Scholar 

  30. Koob GF, Le Moal M (2008) Addiction and the brain antireward system. Annu Rev Psychol 59:29–53

    PubMed  Google Scholar 

  31. Volkow ND, Fowler JS, Wang GJ, Goldstein RZ (2002) Role of dopamine, the frontal cortex and memory circuits in drug addiction: insight from imaging studies. Neurobiol Learn Mem 78:610–624

    PubMed  CAS  Google Scholar 

  32. Comings DE, Muhleman D, Gysin R (1996) Dopamine D2 receptor (DRD2) gene and susceptibility to posttraumatic stress disorder: a study and replication. Biol Psychiatry 40:368–372

    PubMed  CAS  Google Scholar 

  33. Costa-Mallen P, Costa LG, Checkoway H (2005) Genotype combinations for monoamine oxidase-B intron 13 polymorphism and dopamine D2 receptor TaqIB polymorphism are associated with ever-smoking status among men. Neurosci Lett 385:158–162

    PubMed  CAS  Google Scholar 

  34. da Silva Lobo DS, Vallada HP, Knight J, Martins SS, Tavares H, Gentil V, Kennedy JL (2007) Dopamine genes and pathological gambling in discordant sib-pairs. J Gambl Stud 23:421–433

    PubMed  Google Scholar 

  35. Epstein LH, Temple JL, Neaderhiser BJ, Salis RJ, Erbe RW, Leddy JJ (2007) Food reinforcement, the dopamine D2 receptor genotype, and energy intake in obese and nonobese humans. Behav Neurosci 121:877–889

    PubMed  CAS  Google Scholar 

  36. Kirsch P, Reuter M, Mier D, Lonsdorf T, Stark R, Gallhofer B, Vaitl D, Hennig J (2006) Imaging gene-substance interactions: the effect of the DRD2 TaqIA polymorphism and the dopamine agonist bromocriptine on the brain activation during the anticipation of reward. Neurosci Lett 405:196–201

    PubMed  CAS  Google Scholar 

  37. Ponce G, Jimenez-Arriero MA, Rubio G, Hoenicka J, Ampuero I, Ramos JA, Palomo T (2003) The A1 allele of the DRD2 gene (TaqI A polymorphisms) is associated with antisocial personality in a sample of alcohol-dependent patients. Eur Psychiatry 18:356–360

    PubMed  CAS  Google Scholar 

  38. Radwan GN, Setouhy ME, Mohamed MK, Hamid MA, Israel E, Azem SA, Kamel O, Loffredo CA (2007) DRD2/ANKK1 TaqI polymorphism and smoking behavior of Egyptian male cigarette smokers. Nicotine Tob Res 9:1325–1329

    PubMed  CAS  Google Scholar 

  39. Shahmoradgoli Najafabadi M, Ohadi M, Joghataie MT, Valaie F, Riazalhosseini Y, Mostafavi H, Mohammadbeigi F, Najmabadi H (2005) Association between the DRD2 A1 allele and opium addiction in the Iranian population. Am J Med Genet B Neuropsychiatr Genet 134B:39–41

    PubMed  Google Scholar 

  40. Spangler R, Wittkowski KM, Goddard NL, Avena NM, Hoebel BG, Leibowitz SF (2004) Opiate-like effects of sugar on gene expression in reward areas of the rat brain. Brain Res Mol Brain Res 124:134–142

    PubMed  CAS  Google Scholar 

  41. Spitz MR, Shi H, Yang F, Hudmon KS, Jiang H, Chamberlain RM, Amos CI, Wan Y, Cinciripini P, Hong WK, Wu X (1998) Case–control study of the D2 dopamine receptor gene and smoking status in lung cancer patients. J Natl Cancer Inst 90:358–363

    PubMed  CAS  Google Scholar 

  42. Swan GE, Jack LM, Valdes AM, Ring HZ, Ton CC, Curry SJ, McAfee T (2007) Joint effect of dopaminergic genes on likelihood of smoking following treatment with bupropion SR. Health Psychol 26:361–368

    PubMed  Google Scholar 

  43. Xu K, Lichtermann D, Lipsky RH, Franke P, Liu X, Hu Y, Cao L, Schwab SG, Wildenauer DB, Bau CH, Ferro E, Astor W, Finch T, Terry J, Taubman J, Maier W, Goldman D (2004) Association of specific haplotypes of D2 dopamine receptor gene with vulnerability to heroin dependence in 2 distinct populations. Arch Gen Psychiatry 61:597–606

    PubMed  CAS  Google Scholar 

  44. Xiao C, Zhang J, Krnjeviæ K, Ye JH (2007) Effects of ethanol on midbrain neurons: role of opioid receptors. Alcohol Clin Exp Res 31:1106–1113

    PubMed  CAS  Google Scholar 

  45. Blum K, Braverman ER, Holder JM, Lubar JF, Monastra VJ, Miller D, Lubar JO, Chen TJ, Comings DE (2000) Reward deficiency syndrome: a biogenetic model for the diagnosis and treatment of impulsive, addictive, and compulsive behaviors. J Psychoactive Drugs 32:1–112

    Google Scholar 

  46. Bowirrat A, Oscar-Berman M (2005) Relationship between dopaminergic neurotransmission, alcoholism, and reward deficiency syndrome. Am J Med Genet B Neuropsychiatr Genet 132B:29–37

    PubMed  Google Scholar 

  47. Comings DE, Blum K (2000) Reward deficiency syndrome: genetic aspects of behavioral disorders. Prog Brain Res 126:325–341

    PubMed  CAS  Google Scholar 

  48. Green AI, Zimmet SV, Strous RD, Schildkraut JJ (1999) Clozapine for comorbid substance use disorder and schizophrenia: do patients with schizophrenia have a reward-deficiency syndrome that can be ameliorated by clozapine? Harv Rev Psychiatry 6:287–296

    PubMed  CAS  Google Scholar 

  49. Volkow ND, Wang GJ, Telang F, Fowler JS, Logan J, Jayne M, Ma Y, Pradhan K, Wong C (2007) Profound decreases in dopamine release in striatum in detoxified alcoholics: possible orbitofrontal involvement. J Neurosci 27:12700–12706

    PubMed  CAS  Google Scholar 

  50. Carelli RM (2002) The nucleus accumbens and reward: neurophysiological investigations in behaving animals. Behav Cogn Neurosci Rev 1:281–296

    PubMed  Google Scholar 

  51. Christensen R, Kristensen PK, Bartels EM, Bliddal H, Astrup A (2007) Efficacy and safety of the weight-loss drug rimonabant: a meta-analysis of randomised trials. Lancet 370:1671–1672

    Google Scholar 

  52. Peng XQ, Li X, Li J, Ramachandran PV, Gagare PD, Pratihar D, Ashby CR Jr, Gardner EL, Xi ZX (2008) Effects of gabapentin on cocaine self-administration, cocaine-triggered relapse and cocaine-enhanced nucleus accumbens dopamine in rats. Drug Alcohol Depend 97:207–215

    PubMed  CAS  Google Scholar 

  53. Rothman RB, Blough BE, Baumann MH (2007) Dual dopamine/serotonin releasers as potential medications for stimulant and alcohol addictions. AAPS J 9:E1–E10

    PubMed  Google Scholar 

  54. Suzuki H, Han SD, Lucas LR (2010) Chronic passive exposure to aggression decreases D2 and 5-HT 1B receptor densities. Physiol Behav 99(5):562–570

    PubMed  CAS  Google Scholar 

  55. Chen TJH, Blum K, Chen LCH, Bowirrat A, Downs BW, Madigan MA, Waite RL, Bailey JA, Kerner M, Yelandi S, Giordano J, Morse S, Miller D, Braverman ER (2011) Neurogenetics and clinical evidence for the putative activation of the brain reward circuitry by a neuroadaptagen: proposing an addiction candidate gene panel map. J Psychoactive Drugs 43(2):108–127

    PubMed  Google Scholar 

  56. Blum K, Chen TJ, Meshkin B, Waite RL, Downs BW, Blum SH, Mengucci JF, Arcuri V, Braverman ER, Palomo T (2007) Manipulation of catechol-O-methyl-transferase (COMT) activity to influence the attenuation of substance seeking behavior, a subtype of Reward Deficiency Syndrome (RDS), is dependent upon gene polymorphisms: a hypothesis. Med Hypotheses 69:1054–1060

    PubMed  CAS  Google Scholar 

  57. Blum K, Chen TJ, Downs BW, Bowirrat A, Waite RL, Braverman ER, Madigan M, Oscar-Berman M, DiNubile N, Stice E, Giordano J, Morse S, Gold M (2009) Neurogenetics of dopaminergic receptor supersensitivity in activation of brain reward circuitry and relapse: proposing “deprivation-amplification relapse therapy” (DART). Postgrad Med 121:176–196

    PubMed  Google Scholar 

  58. Chen TJ, Blum K, Payte JT, Schoolfield J, Hopper D, Stanford M, Braverman ER (2004) Narcotic antagonists in drug dependence: pilot study showing enhancement of compliance with SYN-10, amino-acid precursors and enkephalinase inhibition therapy. Med Hypotheses 63:538–548

    PubMed  CAS  Google Scholar 

  59. Mei W, Zhang JX, Xiao Z (2010) Acute effects of sublingual buprenorphine on brain responses to heroin-related cues in early-abstinent heroin addicts: an uncontrolled trial. Neuroscience 170(3):808–815

    PubMed  CAS  Google Scholar 

  60. Yücel M, Lubman DI, Harrison BJ, Fornito A, Allen NB, Wellard RM, Roffel K, Clarke K, Wood SJ, Forman SD, Pantelis C (2007) A combined spectroscopic and functional MRI investigation of the dorsal anterior cingulate region in opiate addiction. Mol Psychiatry 12(611):691–702

    Google Scholar 

  61. De Ridder D, Vanneste S, Kovacs S, Sunaert S, Dom G (2011) Transient alcohol craving suppression by rTMS of dorsal anterior cingulate: an fMRI and LORETA EEG study. Neurosci Lett 496(1):5–10

    PubMed  Google Scholar 

  62. Everitt BJ, Robbins TW (2005) Neural systems of reinforcement for drug addiction: from actions to habits to compulsion. Nat Neurosci 8:1481–1489

    PubMed  CAS  Google Scholar 

  63. Koob GF, Volkow ND (2010) Neurocircuitry of addiction. Neuropsychopharmacology 35(1):217–238

    PubMed  Google Scholar 

  64. Volkow ND, Fowler JS, Wang GJ (2003) The addicted human brain: insights from imaging studies. J Clin Invest 111:1444–1451

    PubMed  CAS  Google Scholar 

  65. Volkow ND, Wang GJ, Ma Y, Fowler JS, Wong C, Jayne M, Telang F, Swanson JM (2006) Effects of expectation on the brain metabolic responses to methylphenidate and to its placebo in non-drugabusing subjects. Neuroimage 32:1782–1792

    PubMed  Google Scholar 

  66. Volkow ND, Wang GJ, Ma Y, Fowler JS, Zhu W, Maynard L, Telang F, Vaska P, Ding YS, Wong C, Swanson JM (2003) Expectation enhances the regional brain metabolic and the reinforcing effects of stimulants in cocaine abusers. J Neurosci 23:11461–11468

    PubMed  CAS  Google Scholar 

  67. Nestler EJ (2009) Reinforcement and addictive disorders. In: Nestler EJ et al (eds) Molecular neuropharmacology: a foundation for clinical neuroscience. McGraw-Hill, New York

    Google Scholar 

  68. Conrad KL, Tseng KY, Uejima JL, Reimers JM, Heng LJ, Shaham Y, Marinelli M, Wolf ME (2008) Formation of accumbens GluR2-lacking AMPA receptors mediates incubation of cocaine craving. Nature 454:118–121

    PubMed  CAS  Google Scholar 

  69. Volkow ND, Fowler JS, Wang GJ, Baler R, Telang F (2009) Imaging dopamine’s role in drug abuse and addiction. Neuropharmacology 56(Suppl 1):3–8

    PubMed  CAS  Google Scholar 

  70. Weiss F (2005) Neurobiology of craving, conditioned reward and relapse. Curr Opin Pharmacol 5:9–19

    PubMed  CAS  Google Scholar 

  71. Wilson SJ, Sayette MA, Fiez JA (2004) Prefrontal responses to drug uses: a neurocognitive analysis. Nat Neurosci 7:211–214

    PubMed  Google Scholar 

  72. Lee TM, Zhou WH, Luo XJ, Yuen KS, Ruan XZ, Weng XC (2005) Neural activity associated with cognitive regulation in heroin users: A fMRI study. Neurosci Lett 15;382(3):211–216

    Google Scholar 

  73. Lawrence NS, Ross TJ, Stein EA (2002) Cognitive mechanisms of nicotine on visual attention. Neuron 24;36(3):539–548

    Google Scholar 

  74. Garavan H, Pankiewicz J, Bloom A, Cho JK, Sperry L, Ross TJ, Salmeron BJ, Risinger R, Kelley D, Stein EA (2000) Cue-induced cocaine craving: neuroanatomical specificity for drug users and drug stimuli. Am J Psychiatry 157:1789–1798

    PubMed  CAS  Google Scholar 

  75. Blum K, Chen TJ, Morse S, Giordano J, Chen AL, Thompson J, Allen C, Smolen A, Lubar J, Stice E, Downs BW, Waite RL, Madigan MA, Kerner M, Fornari F, Braverman ER (2010) Overcoming qEEG abnormalities and reward gene deficits during protracted abstinence in male psychostimulant and polydrug abusers utilizing putative dopamine D2 agonist therapy: part 2. Postgrad Med 122(6):214–226

    PubMed  Google Scholar 

  76. Lingford-Hughes A (2005) Human brain imaging and substance abuse. Curr Opin Pharmacol 5:42–46

    PubMed  CAS  Google Scholar 

  77. Walsh SL, Gilson SF, Jasinski DR, Stapleton JM, Phillips RL, Dannals RF, Schmidt J, Preston KL, Grayson R, Bigelow GE et al (1994) Buprenorphine reduces cerebral glucose metabolism in polydrug abusers. Neuropharmacology 10:157–170

    CAS  Google Scholar 

  78. London ED, Cascella NG, Wong DF, Phillips RL, Dannals RF, Links JM, Herning R, Grayson R, Jaffe JH, Wagner HN Jr (1990) Cocaine-induced reduction of glucose utilization in human brain. A study using positron emission tomography and [fluorine 18]-fluorodeoxyglucose. Arch Gen Psychiatry 47:567–574

    PubMed  CAS  Google Scholar 

  79. 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(5):723–727

    PubMed  CAS  Google Scholar 

  80. London ED, Broussolle EP, Links JM, Wong DF, Cascella NG, Dannals RF, Sano M, Herning R, Snyder FR, Rippetoe LR et al (1990) Morphine-induced metabolic changes in human brain. Studies with positron emission tomography and [fluorine 18]fluorodeoxyglucose. Arch Gen Psychiatry 47:73–81

    PubMed  CAS  Google Scholar 

  81. Dow-Edwards DL, Milhorat TH, Freed LA, Gintzler AR (1989) Regional brain glucose utilization during and following chronic naltrexone administration: preliminary observations in rat brain. Life Sci 44:571–577

    PubMed  CAS  Google Scholar 

  82. Ritchie T, Noble EP (1996) [3H]naloxone binding in the human brain: alcoholism and the TaqI A D2 dopamine receptor polymorphism. Brain Res 718:193–197

    PubMed  CAS  Google Scholar 

  83. Ritchie T, Noble EP (2003) Association of seven polymorphisms of the D2 dopamine receptor gene with brain receptor-binding characteristics. Neurochem Res 28:73–82

    PubMed  CAS  Google Scholar 

  84. Seeman P (2010) Dopamine D2 receptors as treatment targets in schizophrenia. Clin Schizophr Relat Psychoses 4:56–73

    PubMed  Google Scholar 

  85. Mavridis M, Besson MJ (1999) Dopamine–opiate interaction in the regulation of neostriatal and pallidal neuronal activity as assessed by opioid precursor peptides and glutamate decarboxylase messenger RNA expression. Neuroscience 92:945–966

    PubMed  CAS  Google Scholar 

  86. Chen JF, Aloyo VJ, Qin ZH, Weiss B (1994) Irreversible blockade of D2 dopamine receptors by fluphenazine-N-mustard increases D2 dopamine receptor mRNA and proenkephalin mRNA and decreases D1 dopamine receptor mRNA and mu and delta opioid receptors in rat striatum. Neurochem Int 25:355–366

    PubMed  CAS  Google Scholar 

  87. Yoburn BC, Shah S, Chan K, Duttaroy A, Davis T (1995) Supersensitivity to opioid analgesics following chronic opioid antagonist treatment: relationship to receptor selectivity. Pharmacol Biochem Behav 51:535–539

    PubMed  CAS  Google Scholar 

  88. Poisnel G, Dhilly M, Le Boisselier R, Barre L, Debruyne D (2009) Comparison of five benzodiazepine-receptor agonists on buprenorphine-induced mu-opioid receptor regulation. J Pharmacol Sci 110:36–46

    PubMed  CAS  Google Scholar 

  89. Hervé S, Riachi G, Noblet C, Guillement N, Tanasescu S, Goria O, Thuillez C, Tranvouez JL, Ducrotte P, Lerebours E (2004) Acute hepatitis due to buprenorphine administration. Eur J Gastroenterol Hepatol 16:1033–1037

    PubMed  Google Scholar 

  90. Peyrière H, Tatem L, Bories C, Pageaux GP, Blayac JP, Larrey D (2009) Hepatitis after intravenous injection of sublingual buprenorphine in acute hepatitis C carriers: report of two cases of disappearance of viral replication after acute hepatitis. Ann Pharmacother 43:973–977

    PubMed  Google Scholar 

  91. Jasinski DR, Pevnick JS, Griffith JD (1976) Human pharmacology and abuse potential of the analgesic buprenorphine: a potential agent for treating narcotic addiction. Arch Gen Psychiatry 35:501–516

    Google Scholar 

  92. Gaulier JM, Marquet P, Lacassie E, Dupuy JL, Lachatre G (2000) Fatal intoxication following self-administration of a massive dose of buprenorphine. J Forensic Sci 45:226–228

    PubMed  CAS  Google Scholar 

  93. Kintz P (2001) Deaths involving buprenorphine: a compendium of French cases. Forensic Sci Int 121:65–69

    PubMed  CAS  Google Scholar 

  94. Tracqui A, Tournoud C, Flesch F, Kopferschmitt J, Kintz P, Deveaux M, Ghysel MH, Marquet P, Pépin G, Petit G, Jaeger A, Ludes B (1998) Acute poisoning during substitution therapy based on high-dosage buprenorphine. 29 clinical cases–20 fatal cases. Presse Med 27:557–561

    PubMed  CAS  Google Scholar 

  95. Blum K, Wallace JE, Schwerter HA, Eubanks JD (1976) Morphine suppression of ethanol withdrawal in mice. Experientia 32:79–82

    PubMed  CAS  Google Scholar 

  96. Blum K, Briggs AH, DeLallo L (1983) Clonidine enhancement of ethanol withdrawal in mice. Subst Alcohol Actions Misuse 4:59–63

    PubMed  CAS  Google Scholar 

  97. Hume SP, Lingford-Hughes AR, Nataf V, Hirani E, Ahmad R, Davies AN, Nutt DJ (2007) Low sensitivity of the positron emission tomography ligand [11C]diprenorphine to agonist opiates. J Pharmacol Exp Ther 322:661–667

    PubMed  CAS  Google Scholar 

  98. Feigenbaum J, Yanai J (1984) The role of dopaminergic mechanisms in mediating the central behavioral effects of morphine in rodents. Neuropsychobiology 11(2):98–105

    PubMed  CAS  Google Scholar 

  99. Theile JW, Morikawa H, Gonzales RA, Morrisett RA (2011) GABAergic transmission modulates ethanol excitation of ventral tegmental area dopamine neurons. Neuroscience 13(172):94–103

    Google Scholar 

  100. Galynker I, Schlyer DJ, Dewey SL, Fowler JS, Logan J, Gatley SJ, MacGregor RR, Ferrieri RA, Holland MJ, Brodie J, Simon E, Wolf AP (1996) Opioid receptor imaging and displacement studies with [6-O-[11C] methyl]buprenorphine in baboon brain. Nucl Med Biol 23(3):325–331

    PubMed  CAS  Google Scholar 

  101. Umbricht A, Montoya ID, Hoover DR, Demuth KL, Chiang CT, Preston KL (1999) Naltrexone shortened opioid detoxification with buprenorphine. Drug Alcohol Depend 56:181–190

    PubMed  CAS  Google Scholar 

  102. Bruce RD, Altice FL, Moody DE, Morse GD, Andrews L, Lin SN, Fang WB, Ma Q, Friedland GH (2010) Pharmacokinetic interactions between buprenorphine/naloxone and once-daily lopinavir/ritonavir. J Acquir Immune Defic Syndr 54(5):511–514

    PubMed  CAS  Google Scholar 

  103. Orman JS, Keating GM (2009) Spotlight on buprenorphine/naloxone in the treatment of opioid dependence. CNS Drugs 23:899–902

    PubMed  Google Scholar 

  104. Harris DS, Jones RT, Welm S, Upton RA, Lin E, Mendelson J (2000) Buprenorphine and naloxone co-administration in opiate-dependent patients stabilized on sublingual buprenorphine. Drug Alcohol Depend 61:85–94

    PubMed  CAS  Google Scholar 

  105. Ciraulo DA, Hitzemann RJ, Somoza E, Knapp CM, Rotrosen J, Sarid-Segal O, Ciraulo AM, Greenblatt DJ, Chiang CN (2006) Pharmacokinetics and pharmacodynamics of multiple sublingual buprenorphine tablets in dose-escalation trials. J Clin Pharmacol 46:179–192

    PubMed  CAS  Google Scholar 

  106. Elkader A, Sproule B (2005) Buprenorphine: clinical pharmacokinetics in the treatment of opioid dependence. Clin Pharmacokinet 44:661–680

    PubMed  CAS  Google Scholar 

  107. Chiang CN, Hawks RL (2003) Pharmacokinetics of the combination tablet of buprenorphine and naloxone. Drug Alcohol Depend 70(2 Suppl):S39–S47

    PubMed  CAS  Google Scholar 

  108. Fang WB, Chang Y, McCance-Katz EF, Moody DE (2009) Determination of naloxone and nornaloxone (noroxymorphone) by high-performance liquid chromatography-electrospray ionization- tandem mass spectrometry. J Anal Toxicol 33:409–417

    PubMed  CAS  Google Scholar 

  109. Kosarac B, Fox AA, Collard CD (2009) Effect of genetic factors on opioid action. Curr Opin Anaesthesiol 22:476–482

    PubMed  Google Scholar 

  110. Zhou SF, Liu JP, Chowbay B (2009) Polymorphism of human cytochrome P450 enzymes and its clinical impact. Drug Metab Rev 41:89–295

    PubMed  CAS  Google Scholar 

  111. Van den Oever MC, Lubbers BR, Goriounova NA, Li KW, Van der Schors RC, Loos M et al (2010) Extracellular matrix plasticity and GABAergic inhibition of prefrontal cortex pyramidal cells facilitates relapse to heroin seeking. Neuropsychopharmacology 35:2120–2133

    PubMed  Google Scholar 

  112. Walley AY, Alperen JK, Cheng DM, Botticelli M, Castro-Donlan C, Samet JH, Alford DP (2008) Office-based management of opioid dependence with buprenorphine: clinical practices and barriers. J Gen Intern Med 23:1393–1398

    PubMed  Google Scholar 

  113. Miller DK, Bowirrat A, Manka M, Miller M, Stokes S, Manka D, Allen C, Gant C, Downs BW, Smolen A, Stevens E, Yeldandi S, Blum K (2010) Acute intravenous synaptamine complex variant KB220™ “normalizes” neurological dysregulation in patients during protracted abstinence from alcohol and opiates as observed using quantitative electroencephalographic and genetic analysis for reward polymorphisms: part 1, pilot study with 2 case reports. Postgrad Med 122:188–213

    PubMed  Google Scholar 

  114. Blum K, Mark S, Gold MS (2011) Neuro-chemical activation of brain reward meso-limbic circuitry is associated with relapse prevention and drug hunger: a commentary. Med Hypotheses 76(4):576–584

    Google Scholar 

  115. Chen AL, Chen TJ, Waite RL, Reinking J, Tung HL, Rhoades P, Downs BW, Braverman E, Braverman D, Kerner M, Blum SH, DiNubile N, Smith D, Oscar-Berman M, Prihoda TJ, Floyd JB, O’Brien D, Liu HH, Blum K (2009) Hypothesizing that brain reward circuitry genes are genetic antecedents of pain sensitivity and critical diagnostic and pharmacogenomic treatment targets for chronic pain conditions. Med Hypotheses 72:14–22

    PubMed  Google Scholar 

Download references

Acknowledgments

We thank the staff of PATH Foundation NY. The writing of this article was funded in part by LifeGen, Inc., San Diego, CA, USA. Other support came from NIAAA grants R01-AA07112 and K05-AA00219 and the Medical Research Service of the US Department of Veterans Affairs (to MO-B).

Authors’ Contributions

KB wrote the first draft of the paper. The figures were developed by Margret Madigan, an AB. The entire final article was edited by Margaret Madigan. JF, JG, JB, SS, TS, MO-B, UD, MK, SM, FF, BWD, THJC, ALCC, DB, and ERB reviewed and edited the manuscript. UD was responsible for literature search and appropriate reference style. All the authors read and approved the final manuscript.

Competing Interests

The patented KB220Z therapy has been exclusively licensed to LifeGen, Inc., whereby KB and BWD owns stock. JG, and FF are partners with LifeGen, Inc. All other authors declare that they have no competing interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kenneth Blum.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Blum, K., Chen, T.J.H., Bailey, J. et al. Can the Chronic Administration of the Combination of Buprenorphine and Naloxone Block Dopaminergic Activity Causing Anti-reward and Relapse Potential?. Mol Neurobiol 44, 250–268 (2011). https://doi.org/10.1007/s12035-011-8206-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-011-8206-0

Keywords

Navigation