414
Views
16
CrossRef citations to date
0
Altmetric
Reviews

Novel approaches for the treatment of psychostimulant and opioid abuse – focus on opioid receptor-based therapies

&

Bibliography

  • Termorshuizen F, Krol A, Prins M, et al. Prediction of relapse to frequent heroin use and the role of methadone prescription: an analysis of the Amsterdam Cohort Study among drug users. Drug Alcohol Depend 2005;79:231-40
  • Dennis ML, Scott CK, Funk R, et al. The duration and correlates of addiction and treatment career. J Subst Abuse Treat 2005;28(Suppl 1):S51-62
  • Hughes JR, Gulliver SB, Fenwick KW, et al. Smoking cessation among self-quitters. Health Psychol 1992;11:331-4
  • Stead LF, Lancaster T. Combined pharmacotherapy and behavioural interventions for smoking cessation. Cochrane Database Syst Rev 2012;10:CD008286
  • Castells X, Casas M, Perez-Mana C, et al. Efficacy of psychostimulant drugs for cocaine dependence. Cochrane Database Syst Rev 2010(2):CD007380
  • Mattick RP, Breen C, Kimber J, et al. Methadone maintenance therapy versus no opioid replacement therapy for opioid dependence. Cochrane Database Syst Rev 2009(3):CD002209
  • Minozzi S, Amato L, Vecchi S, et al. Oral naltrexone maintenance treatment for opioid dependence. Cochrane Database Syst Rev 2011(4):CD001333
  • Nutt DJ, King LA, Phillips LD, et al. Drug harms in the UK: a multicriteria decision analysis. Lancet 2010;376:1558-65
  • Samaha AN, Robinson TE. Why does the rapid delivery of drugs to the brain promote addiction? Trends Pharmacol Sci 2005;26:82-7
  • Benowitz NL. Pharmacology of nicotine: addiction, smoking-induced disease, and therapeutics. Annu Rev Pharmacol Toxicol 2009;49:57-71
  • Sanchis-Segura C, Spanagel R. Behavioural assessment of drug reinforcement and addictive features in rodents: an overview. Addict Biol 2006;11:2-38
  • Shalev U, Grimm JW, Shaham Y. Neurobiology of relapse to heroin and cocaine seeking: a review. Pharmacol Rev 2002;54:1-42
  • Tzscehntke TM. Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol 2007;12:227-462
  • Aguilar MA, Rodriguez-Arias M, Minarro J. Neurobiological mechanisms of the reinstatement of drug-conditioned place preference. Brain Res Rev 2009;59:253-77
  • Bossert JM, Marchant NJ, Calu DJ, et al. The reinstatement model of drug relapse: recent neurobiological findings, emerging research topics, and translational research. Psychopharmacology (Berl) 2013;229:453-76
  • Ahmed SH. The science of making drug-addicted animals. Neuroscience 2012;211:107-25
  • Vanderschuren LJ, Ahmed SH, et al. Animal studies of addictive behavior. Cold Spring Harb Perspect Med 2013;3:a011932
  • Marchant NJ, Li X, Shaham Y. Recent developments in animal models of drug relapse. Curr Opin Neurobiol 2013;23:675-83
  • Besancon F. Time to alcohol dependence after abstinence and first drink. Addiction 1993;88:1647-50
  • Goldstein RZ, Volkow ND. Dysfunction of the prefrontal cortex in addiction: neuroimaging findings and clinical implications. Nat Rev Neurosci 2011;12:652-69
  • de Wit H. Priming effects with drugs and other reinforcers. Exp Clin Psychopharmacol 1996;4:5-10
  • Carter BL, Tiffany ST. Meta-analysis of cue-reactivity in addiction research. Addiction 1999;94:327-40
  • Hyman SM, Fox H, Hong KI, et al. Stress and drug-cue-induced craving in opioid-dependent individuals in naltrexone treatment. Exp Clin Psychopharmacol 2007;15:134-43
  • Sinha R. How does stress increase risk of drug abuse and relapse? Psychopharmacology (Berl) 2001;158:343-59
  • Galai N, Safaeian M, Vlahov D, et al. Longitudinal patterns of drug injection behavior in the ALIVE Study cohort. 1988-2000: description and determinants. Am J Epidemiol 2003;158:695-704
  • Leri F, Stewart J, Fischer B, et al. Patterns of opioid and cocaine co-use: a descriptive study in a Canadian sample of untreated opioid-dependent individuals. Exp Clin Psychopharmacol 2005;13:303-10
  • Leri F, Bruneau J, Stewart J. Understanding polydrug use: review of heroin and cocaine co-use. Addiction 2003;98:7-22
  • Matthes HW, Maldonado R, Simonin F, et al. Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the mu-opioid-receptor gene. Nature 1996;383:819-23
  • Bozarth MA, Wise RA. Intracranial self-administration of morphine into the ventral tegmental area in rats. Life Sci 1981;28:551-5
  • Schmitt KC, Rothman RB, Reith ME. Nonclassical pharmacology of the dopamine transporter: atypical inhibitors, allosteric modulators, and partial substrates. J Pharmacol Exp Ther 2013;346:2-10
  • Nieh EH, Kim SY, Namburi P, et al. Optogenetic dissection of neural circuits underlying emotional valence and motivated behaviors. Brain Res 2013;1511:73-92
  • Brauer LH, de Wit H. Subjective responses to d-amphetamine alone and after pimozide pretreatment in normal, healthy volunteers. Biol Psychiatry 1996;39:26-32
  • Ohuoha DC, Maxwell JA, Thomson LE III, et al. Effect of dopamine receptor antagonists on cocaine subjective effects : a naturalistic case study. J Subst Abuse Treat 1997;14:249-58
  • Baik JH. Dopamine signaling in reward-related behaviors. Front Neural Circuits 2013;7:152
  • Volkow ND, Wang GJ, Fowler JS, et al. Addiction circuitry in the human brain. Annu Rev Pharmacol Toxicol 2012;52:321-36
  • Newman AH, Blaylock BL, Nader MA, et al. Medication discovery for addiction: translating the dopamine D3 receptor hypothesis. Biochem Pharmacol 2012;84:882-90
  • Commons KG. Neuronal pathways linking substance P to drug addiction and stress. Brain Res 2010;1314:175-82
  • Yeoh JW, Campbell EJ, James MH, et al. Orexin antagonists for neuropsychiatric disease: progress and potential pitfalls. Front Neurosci 2014;8:36
  • Baimel C, Borgland SL. Hypocretin modulation of drug-induced synaptic plasticity. Prog Brain Res 2012;198:123-31
  • Ogbonmwan YE, Sciolino NR, Groves-Chapman JL, et al. The galanin receptor agonist, galnon, attenuates cocaine-induced reinstatement and dopamine overflow in the frontal cortex. Addict Biol 2014. [Epub ahead of print]
  • Hawes JJ, Brunzell DH, Narasimhaiah R, et al. Galanin protects against behavioral and neurochemical correlates of opiate reward. Neuropsychopharmacology 2008;33:1864-73
  • Piccotto MR, Brabant C, Einstein EB, et al. Effects of galanin on monoaminergic systems and HPA axis: potential mechanisms underlying the effects of galanin on addiction- and stress-related behaviors. Brain Res 2010;1314:206-18
  • Li X, Xi ZX, Markou A. Metabotropic glutamate 7 (mGlu7) receptor: a target for medication development for the treatment of cocaine dependence. Neuropharmacology 2013;66:12-23
  • Fitzgerald PJ. Elevated norepinephrine may be a unifying etiological factor in the abuse of a broad range of substances: alcohol, nicotine, marijuana, heroin, cocaine, and caffeine. Subst Abuse 2013;7:171-83
  • Koob GF, Buck CL, Cohen A, et al. Addiction as a stress surfeit disorder. Neuropharmacology 2014;76(Pt B):370-82
  • Rothman RB, Baumann MH, Dersch CM, et al. Amphetamine-type central nervous system stimulants release norepinephrine more potential than they release dopamine and serotonin. Synapse 2001;226:659-72
  • Ritz MC, Cone EJ, Kuhar MJ. Cocaine inhibition of ligand binding at dopamine, norepinephrine and serotonina transporters: a structure-activity study. Life Sci 1990;46:635-45
  • Torregrossa MM, Taylor JR. Learning to forget: manipulating extinction and reconsolidation processes to treat addiction. Psychopharmacology (Berl) 2013;226:659-72
  • Milton AL, Everitt BJ. The persistence of maladaptive memory: addiction, drug memories and anti-relapse treatments. Neurosci Biobehav Rev 2012;36:1119-39
  • Diergaarde L, Schoffelmeer AN, De Vries TJ. Pharmacological manipulation of memory reconsolidation: towards a novel treatment of pathogenic memories. Eur J Pharmacol 2008;585:453-7
  • Luscher C. Drug-evoked synaptic plasticity causing addictive behavior. J Neurosci 2013;33:17641-6
  • Van den Oever MC, Spijker S, Smit AB. The synaptic pathology of drug addiction. Adv Exp Med Biol 2012;970:469-91
  • Myers KM, Carlezon WA. D-cycloserine effects on extinction of conditioned responses to drug-related cues. Biol Psychiatry 2012;71(11):947-55
  • Lu GY, Wu N, Zhang ZL, et al. Effects of D-cycloserine on extinction and reinstatement of morphine-induced conditioned place preference. Neurosci Lett 2011;503:196-9
  • Tomek SE, Lacrosse AL, Nemirovsky NE, et al. NMDA Receptor Modulators in the Treatment of Drug Addiction. Pharmaceuticals 2013;6:251-68
  • Wiskerke J, Pattij T, Schoffelmeer AN, et al. The role of CB1 receptors in psychostimulant addiction. Addict Biol 2008;13:225-38
  • Bellone C, Luscher C, Mameli M. Mechanisms of synaptic depression triggered by metabotropic glutamate receptors. Cell Mol Life Sci 2008;65:2913-23
  • Nic Dhonnchadha BA, Kantak KM. Cognitive enhancers for facilitating drug cue extinction: insights from animal models. Pharmacol Biochem Behav 2011;99:229-44
  • Niciu MJ, Arias AJ. Targeted opioid receptor antagonists in the treatment of alcohol use disorders. CNS Drugs 2013;27:777-87
  • Levran O, Yuferov V, Kreek MJ. The genetics of the opioid system and specific drug addictions. Hum Genet 2012;131:823-42
  • Lutz PE, Kieffer BL. The multiple facets of opioid receptor function: implications for addiction. Curr Opin Neurobiol 2013;23:473-9
  • Emmerson PJ, Liu MR, Woods JH, et al. Binding affinity and selectivity of opioids at mu, delta and kappa receptors in monkey brain membranes. J Pharmacol Exp Ther 1994;271:1630-7
  • Tang XC, McFarland K, Cagle S, et al. Cocaine-induced reinstatement requires endogenous stimulation of mu-opioid receptors in the ventral pallidum. J Neurosci 2005;25:4512-20
  • Simmons D, Self DW. Role of mu- and delta-opioid receptors in the nucleus accumbens in cocaine-seeking behavior. Neuropsychopharmacology 2009;34:1946-57
  • Perry CJ, McNally GP. A role for the ventral pallidum in context-induced and primed reinstatement of alcohol seeking. Eur J Neurosci 2013;38:2762-73
  • Giuliano C, Robbins TW, Wille DR, et al. Attenuation of cocaine and heroin seeking by μ-opioid receptor antagonism. Psychopharmacology (Berl) 2013;227:137-47
  • Gutierrez-Cuesta J, Burokas A, Mancino S, et al. Effects of genetic deletion of endogenous opioid system components on the reinstatement of cocaine-seeking behavior in mice. Neuropsychopharmacology 2014. [Epub ahead of print]
  • Phillips KA, Epstein DH, Preston KL. Psychostimulant addiction treatment. Neuropharmacology 2014. [Epub ahead of print]
  • Le Merrer J, Becker JA, Befort K, et al. Reward processing by the opioid system in the brain. Physiol Rev 2009;89:1379-412
  • Billa SK, Xia Y, Moron JA. Disruption of morphine-conditioned place preference by a delta2-opioid receptor antagonist: study of mu-opioid and delta-opioid receptor expression at the synapse. Eur J Neurosci 2010;32:625-31
  • Le Merrer J, Faget L, Matidas A, et al. Cues predicting drug or food reward restore morphine-induced place conditioning in mice lacking delta opioid receptors. Psychopharmacology (Berl) 2012;223:99-106
  • Charbogne P, Kieffer BL, Befort K. 15 years of genetic approaches in vivo for addiction research: Opioid receptor and peptide gene knockout in mouse models of drug abuse. Neuropharmacology 2014;76(Pt B):204-17
  • Klenowski P, Morgan M, Bartlett SE. The role of δ-opioid receptors in learning and memory underlying the development of addiction. Br J Pharmacol 2014. [Epub ahead of print]
  • Dikshtein Y, Barnea R, Kronfeld N, et al. Beta-endorphin via the delta opioid receptor is a major factor in the incubation of cocaine craving. Neuropsychopharmacology 2013;38:2508-14
  • van Rijn RM, Defriel JN, Whistler JL. Pharmacological traits of delta opioid receptors: pitfalls or opportunities? Psychopharmacology (Berl) 2013;228:1-18
  • Ciccocioppo R, Martin-Fardon R, Weiss F. Effect of selective blockade of mu(1) or delta opioid receptors on reinstatement of alcohol-seeking behavior by drug-associated stimuli in rats. Neuropsychopharmacology 2002;27:391-9
  • Nielsen CK, Simms JA, Bito-Onon JJ, et al. The delta opioid receptor antagonist, SoRI-9409, decreases yohimbine stress-induced reinstatement of ethanol-seeking. Addict Biol 2012;17:224-34
  • van Rijn RM, Brissett DI, Whistler JL. Dual efficacy of delta opioid receptor-selective ligands for ethanol drinking and anxiety. J Pharmacol Exp Ther 2010;335:133-9
  • McLaughlin JP, Marton-Popovici M, Chavkin C. Kappa opioid receptor antagonism and prodynorphin gene disruption block stress-induced behavioral responses. J Neurosci 2003;23:5674-83
  • Beardsley PM, Howard JL, Shelton KL, et al. Differential effects of the novel kappa opioid receptor antagonist, JDTic, on reinstatement of cocaine-seeking induced by footshock stressors vs cocaine primes and its antidepressant-like effects in rats. Psychopharmacology (Berl) 2005;183:118-26
  • Redila VA, Chavkin C. Stress-induced reinstatement of cocaine seeking is mediated by the kappa opioid system. Psychopharmacology (Berl) 2008;200:59-70
  • Bruchas MR, Land BB, Chavkin C. The dynorphin/kappa opioid system as a modulator of stress-induced and pro-addictive behaviors. Brain Res 2010;1314:44-55
  • Schenk S, Partridge B, Shippenberg TS. Reinstatement of extinguished drug-taking behavior in rats: effect of the kappa-opioid receptor agonist, U69593. Psychopharmacology 2000;151:85-90
  • Schenk S, Partridge B. Effect of the kappa-opioid receptor agonist, U69593, on reinstatement of extinguished amphetamine self-administration behavior. Pharmacol Biochem Behav 2001;68:629-34
  • McLaughlin JP, Land BB, Li S, et al. Prior activation of kappa opioid receptors by U50,488 mimics repeated forced swim stress to potentiate cocaine place preference conditioning. Neuropsychopharmacology 2006;31:787-94
  • McLaughlin JP, Li S, Valdez J, et al. Social defeat stress-induced behavioral responses are mediated by the endogenous kappa opioid system. Neuropsychopharmacology 2006;31:1241-8
  • Carey AM, Borozny K, Aldrich JV, et al. Reinstatement of cocaine place-conditioning prevented by the peptide kappa-opioid receptor antagonist arodyn. Eur J Pharmacol 2007;569:84-9
  • Aldrich JV, Patkar KA, McLaughlin JP. Zyklophin, a systemically active selective kappa opioid receptor peptide antagonist with short duration of action. Proc Natl Acad Sci USA 2009;106:18396-401
  • Sedki F, Eigenmann K, Gelinas J, et al. A role for kappa-, but not mu-opioid, receptor activation in acute food deprivation-induced reinstatement of heroin seeking in rats. Addict Biol 2014. [Epub ahead of print]
  • Schlosburg JE, Whitfield TW, Park PE, et al. Long-term antagonism of κ opioid receptors prevents escalation of and increased motivation for heroin intake. J Neurosci 2013;33:19384-92
  • Smith JS, Schidler AG, Martinelli E, et al. Stress-induced activation of the dynorphin/κ-opioid receptor system in the amygdala potentiates nicotine conditioned place preference. J Neurosci 2012;32:1488-95
  • Grella SL, Funk D, Coen K, et al. Role of the kappa-opioid receptor system in stress-induced reinstatement of nicotine seeking in rats. Behav Brain Res 2014;265:188-97
  • Van’t Veer A, Carlezon WA. Role of kappa-opioid receptors in stress and anxiety-related behavior. Psychopharmacology (Berl) 2013;229:435-52
  • Walker BM, Koob GF. Pharmacological evidence for a motivational role of kappa-opioid systems in ethanol dependence. Neuropsychopharmacology 2008;33:643-52
  • Kovacs KM, Szakall I, O’Brien D, et al. Decreased oral self-administration of alcohol in kappa-opioid receptor knock-out mice. Alcohol Clin Exp Res 2005;29:730-8
  • Le AD, Poulos CX, Harding S, et al. Effects of naltrexone and fluoxetine on alcohol self-administration and reinstatement of alcohol seeking induced by priming injections of alcohol and exposure to stress. Neuropsychopharmacology 1999;21:435-44
  • Schank JR, Goldstein AL, Rowe KE, et al. The kappa opioid receptor antagonist JDTic attenuates alcohol seeking and withdrawal anxiety. Addict Biol 2012;17:634-47
  • Huang P, Kehner GB, Cowan A, et al. Comparison of pharmacological activities of buprenorphine and norbuprenorphine: norbuprenorphine is a potent opioid agonist. J Pharmacol Exp Ther 2001;297:688-95
  • Giordano AL, Nock B, Cicero TJ. Antagonist-induced up-regulation of the putative epsilon opioid receptor in rat brain: comparison with kappa, mu and delta opioid receptors. J Pharmacol Exp Ther 1990;255:536-40
  • Rothman RB, Gorelick DA, Heishman SJ, et al. An open-label study of a functional opioid kappa antagonist in the treatment of opioid dependence. J Subst Abuse Treat 2000;18:277-81
  • Gerra G, Fantoma A, Zaimovic A. Naltrexone and buprenorphine combination in the treatment of opioid dependence. J Psychopharmacol 2006;20:806-14
  • McCann DJ. Potential of buprenorphine/naltrexone in treating polydrug addiction and co-occurring psychiatric disorders. Clin Pharmacol Ther 2008;83:627-30
  • Greenwald M, Johanson CE, Bueller J, et al. Buprenorphine duration of action: mu-opioid receptor availability and pharmacokinetic and behavioral indices. Biol Psychiatry 2007;61:101-10
  • Dum JE, Herz A. In vivo receptor-binding of the opiate partial agonist, buprenorphine, correlated with its agonistic and antagonistic actions. Br J Pharmacology 1981;74:627-33
  • McAleer SD, Mills RJ, Polack T, et al. Pharmacokinetics of high-dose buprenorphine following single administration of sublingual tablet formulations in opioid naive healthy male volunteers under a naltrexone block. Drug Alcohol Depend 2003;72:75-83
  • Kirchmayer U, Davoli M, Verster AD, et al. A systematic review on the efficacy of naltrexone maintenance treatment in opioid dependence. Addiction 2002;97:1241-9
  • Mucha RF, Millan MJ, Herz A. Aversive properties of naloxone in non-dependent (naïve) rats may involve blockade of central beta-endorphin. Psychopharmacology (Berl) 1985;86:281-5
  • Parker LA, Rennie M. Naltrexone-induced aversions - assessment by place conditioning, taste reactivity, and taste avoidance paradigms. Pharmacol Biochem Behav 1991;41:559-65
  • Suzuki T, Shiozaki Y, Masukawa Y, et al. The role of mu-opioid and kappa-opioid receptors in cocaine-induced conditioned place preference. Jpn J Pharmacol 1992;58:435-42
  • Le Pen G, Wichmann J, Moreau JL, et al. The orphanin receptor agonist RO 64-6198 does not induce place conditioning in rats. Neuroreport 2002;13:451-4
  • Kotlinska J, Wichmann J, Legowska A, et al. Orphanin FQ/nociceptin but not Ro 65-6570 inhibits the expression of cocaine-induced conditioned place preference. Behav Pharmacol 2002;13:229-35
  • Sakoori K, Murphy NP. Central administration of nociceptin/orphanin FQ blocks the acquisition of conditioned place preference to morphine and cocaine, but not conditioned place aversion to naloxone in mice. Psychopharmacology (Berl) 2004;172:129-36
  • Rutten K, De Vry J, Bruckmann W, et al. Effects of the NOP receptor agonist Ro65-6570 on the acquisition of opiate- and psychostimulant-induced conditioned place preference in rats. Eur J Pharmacol 2010;645:119-26
  • Shoblock JR, Wichmann J, Maidment NT. The effect of a systemically active ORL-1 agonist, Ro 64-6198, on the acquisition, expression, extinction, and reinstatement of morphine conditioned place preference. Neuropharmacology 2005;49:439-46
  • Martin-Fardon R, Ciccocioppo R, Massi M, et al. Nociceptin prevents stress-induced ethanol- but not cocaine-seeking behavior in rats. Neuroreport 2000;11:1939-43
  • Wee S, Vendruscolo LF, Misra KK, et al. A combination of buprenorphine and naltrexone blocks compulsive cocaine intake in rodents without producing dependence. Sci Transl Med 2012;4:146ra110
  • Cordery SF, Taverner A, Ridzwan IE, et al. A non-rewarding, non-aversive buprenorphine/naltrexone combination attenuates drug-primed reinstatement to cocaine and morphine in rats in a conditioned place preference paradigm. Addict Biol 2014;19:575-86
  • Mooney LJ, Nielsen S, Saxon A, et al. Cocaine use reduction with buprenorphine (CURB): rationale, design, and methodology. Contemp Clin Trials 2013;34:196-204
  • Kumar V, Ridzwan IE, Grivas K, et al. Selectively promiscuous opioid ligands: discovery of high affinity/low efficacy opioid ligands with substantial NOP receptor affinity. J Med Chem 2014;57:4049-57
  • Brunet A, Orr SP, Tremblay J, et al. Effect of post-retrieval propranolol on psychophysiologic responding during subsequent script-driven traumatic imagery in post-traumatic stress disorder. J Psychiatr Res 2008;42:503-6
  • Schiller D, Monfils MH, Raio CM, et al. Preventing the return of fear in humans using reconsolidation update mechanisms. Nature 2010;463(7277):49-53
  • Kroes MC, Strange BA, Dolan RJ. Beta-adrenergic blockade during memory retrieval in humans evokes a sustained reduction of declarative emotional memory enhancement. J Neurosci 2010;30:3959-63
  • Xue YX, Luo YX, Wu P, et al. A memory retrieval-extinction procedure to prevent drug craving and relapse. Science 2012;336(6078):241-5
  • Badiani A, Belin D, Epstein D, et al. Opiate versus psychostimulant addiction: the differences do matter. Nat Rev Neurosci 2011;12:685-700
  • Peters J, Pattij T, De Vries TJ. Targeting cocaine versus heroin memories: divergent roles within ventromedial prefrontal cortex. Trends Pharmacol Sci 2013;34:689-95
  • Villiger JW. Binding of buprenorphine to opiate receptors. Regulation by guanyl nucleotides and metal ions. Neuropharmacology 1984;23:373-5
  • Selley DE, Sim LJ, Xiao R, et al. mu-Opioid receptor-stimulated guanosine-5’-O-(gamma-thio)-triphosphate binding in rat thalamus and cultured cell lines: signal transduction mechanisms underlying agonist efficacy. Mol Pharmacol 1997;51:87-96

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.