We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Agonist replacement therapy for cocaine dependence: a translational review

    Craig R Rush

    * Author for correspondence

    Department of Behavioral Science, University of Kentucky, College of Medicine, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA.

    Department of Psychiatry, University of Kentucky College of Medicine, 3470 Blazer Parkway, Lexington, KY 40509, USA

    Department of Psychology, University of Kentucky College of Arts and Sciences, Kastle Hall, Lexington, KY 40506, USA

    &
    William W Stoops

    Department of Behavioral Science, University of Kentucky, College of Medicine, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA.

    Department of Psychology, University of Kentucky College of Arts and Sciences, Kastle Hall, Lexington, KY 40506, USA

    Published Online:https://doi.org/10.4155/fmc.11.184

    Cocaine use disorders are prevalent throughout the world. Agonist replacement therapy is among the most effective strategies for managing substance use disorders including nicotine and opioid dependence. This paper reviews the translational literature, including preclinical experiments, human laboratory studies and clinical trials, to determine whether agonist-replacement therapy is a viable strategy for managing cocaine dependence. Discussion is limited to transporter blockers (i.e., methylphenidate) and releasers (i.e., amphetamine analogs) that are available for use in humans in the hope of impacting clinical research and practice more quickly. The translational review suggests that agonist-replacement therapy, especially monoamine releasers, may be effective for managing cocaine dependence. Future directions for medications development are also discussed because the viability of agonist-replacement therapy for cocaine dependence may hinge on identifying novel compounds or formulations that have less abuse and diversion potential.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Substance Abuse and Mental Health Services Administration. Results from the 2008 National Survey on Drug Use and Health: National Findings. Office of Applied Studies, Rockville, MD, USA (2009).
    • Substance Abuse and Mental Health Services Administration. Treatment Episode Data Set (TEDS) Highlights – 2007 National Admissions to Substance Abuse Treatment Services. Office of Applied Studies, Rockville, MD, USA (2009).
    • European Monitoring Centre for Drugs and Drug Addiction. 2009 Annual report on the state of the drugs problem in Europe. Publications Office of the European Union, Luxembourg (2009).
    • Chen K, Scheier LM, Kandel DB. Effects of chronic cocaine use on physical health: a prospective study in a general population sample. Drug Alcohol Depend.43,23–37 (1996).
    • Schwartz BG, Rezkalla S, Kloner RA. Cardiovascular effects of cocaine. Circulation122,2558–2569 (2010).
    • Substance Abuse and Mental Health Services Administration. Results from the 2009 National Survey on Drug Use and Health: National Findings. Office of Applied Studies: Rockville, MD, USA (2010).
    • Rounsaville BJ, Anton SF, Carroll K, Budde D, Prusoff BA, Gawin F. Psychiatric diagnoses of treatment-seeking cocaine abusers. Arch. Gen. Psychiatry48,43–51 (1991).
    • Van Tieu H, Koblin BA. HIV, alcohol and noninjection drug use. Curr. Opin HIV AIDS4,314–318 (2009).
    • Covi L, Hess JM, Schroeder JR, Preston KL. A dose response study of cognitive behavioral therapy in cocaine abusers. J. Subst. Abuse Treat.23,191–197 (2002).
    • 10  McKee SA, Carroll KM, Sinha R et al. Enhancing brief cognitive–behavioral therapy with motivational enhancement techniques in cocaine users. Drug Alcohol Depend.91(1),97–101 (2007).
    • 11  Higgins ST, Heil SH, Lussier JP. Clinical implications of reinforcement as a determinant of substance use disorders. Ann. Rev. Psychol.55,431–461 (2004).
    • 12  Vocci FJ, Montoya ID. Psychological treatments for stimulant misuse, comparing and contrasting those for amphetamine dependence and those for cocaine dependence. Curr. Opin Psychiatry22(3),263–268 (2009).
    • 13  de Lima MS, de Oliveira Soares BG, Reisser AA, Farrell M. Pharmacological treatment of cocaine dependence: a systematic review. Addiction97(8),931–949 (2002).
    • 14  Vocci FJ, Elkashef A. Pharmacotherapy and other treatments for cocaine abuse and dependence. Curr. Opin Psychiatry18,265–270 (2005).
    • 15  Gorelick DA, Gardner EL, Xi ZX. Agents in development for the management of cocaine abuse. Drugs64,1547–1573 (2004).
    • 16  Platt DM, Rowlett JK, Spealman RD. Behavioral effects of cocaine and dopaminergic strategies for preclinical medication development. Psychopharmacology163,265–282 (2002).
    • 17  Cornish JW, Metzger D, Woody GE et al. Naltrexone pharmacotherapy for opioid dependent federal probationers. J. Subst. Abuse Treat.14(6),529–534 (1997).
    • 18  Rea F, Bell JR, Young MR, Mattick RP. A randomized, controlled trial of low dose naltrexone for the treatment of opioid dependence. Drug Alcohol Depend.75,79–88 (2004).
    • 19  Rose JE, Behm FM. Extinguishing the rewarding value of smoke cues: pharmacological and behavioral treatments. Nicotine Tob. Res.6,523–532 (2004).
    • 20  Grabowski J, Rhoades H, Stotts A et al. Agonist-like or antagonist-like treatment for cocaine dependence with methadone for heroin dependence: two double-blind randomized clinical trials. Neuropsychopharmacology29,969–981 (2004).▪ This elegant pair of clinical trials demonstrated that agonist-replacement is likely a more successful strategy for managing cocaine-use disorders than antagonist pharmacotherapy.
    • 21  Karila L, Raynaud M, Aubin HJ et al. Pharmacological treatments for cocaine dependence: is there something new? Curr. Pharm. Des.17(14),1359–1368 (2011).
    • 22  Newton TF, Ling W, Kalechstein AD, Uslaner J, Tervo K. Risperidone pre-treatment reduces the euphoric effects of experimentally administered cocaine. Psychiatry Res.102,227–233 (2001).
    • 23  Grabowski J, Rhoades H, Silverman P et al. Risperidone for the treatment of cocaine dependence: randomized, double-blind trial. J. Clin. Psychopharmacol.20,305–310 (2000).
    • 24  Kampman KM, Pettinati H, Lynch KG, Sparkman T, O’Brien CP. A pilot trial of olanzapine for the treatment of cocaine dependence. Drug Alcohol Depend.70,265–273 (2003).
    • 25  Grabowski J, Shearer J, Merrill J, Negus SS. Agonist-like, replacement pharmacotherapy for stimulant abuse and dependence. Addict. Behav.29,1439–1464 (2004).
    • 26  Mooney ME, Herin DV, Schmitz JM, Moukaddam N, Green CE, Grabowski J. Effects of oral methamphetamine on cocaine use: a randomized, double-blind, placebo-controlled trial. Drug Alcohol Depend.101(1–2),34–41, (2009).▪▪ This clinical trial demonstrated that maintenance on sustained-release methamphetamine substantially decreases cocaine use, which was verified by urine toxicology testing.
    • 27  Henningfield JE. Nicotine medications for smoking cessation. N. Engl. J. Med.333,1196–1203 (1995).
    • 28  Ling W, Rawson RA, Compton MA. Substitution pharmacotherapies for opioid addiction: from methadone to LAAM and buprenorphine. J. Psychoactive Drugs26,119–128 (1994).
    • 29  Silagy C, Lancaster T, Stead L, Mant D, Fowler G. Nicotine replacement therapy for smoking cessation. Cochrane Database Syst. Rev.CD000146 (2004).
    • 30  Sofuoglu M, Kosten TR. Pharmacologic management of relapse prevention in addictive disorders. Psychiatr. Clin. North. Am.27,627–648 (2004).
    • 31  Rush CR, Kollins SH, Pazzaglia PJ. Discriminative-stimulus and participant-rated effects of methylphenidate, bupropion and triazolam in D-amphetamine-trained humans. Exp. Clin. Psychopharmacol.6,32–44 (1998).
    • 32  Stoops WW, Glaser PE, Fillmore MT, Rush CR. Reinforcing, subject-rated, performance and physiological effects of methylphenidate and D-amphetamine in stimulant abusing humans. J. Psychopharmacol.18,534–543 (2004).
    • 33  Negus SS. Choice between heroin and food in nondependent and heroin-dependent rhesus monkeys: effects of naloxone, buprenorphine and methadone. J. Pharmacol. Exp. Ther.317,711–723 (2006).
    • 34  Negus SS, Rice KC. Mechanisms of withdrawal-associated increases in heroin self-administration: pharmacologic modulation of heroin vs food choice in heroin-dependent rhesus monkeys. Neuropsychopharmacology34,899–911 (2009).
    • 35  Czoty PW, Martelle JL, Nader MA. Influence of abstinence and conditions of cocaine access on the reinforcing strength of cocaine in nonhuman primates. Drug Alcohol Depend.85,213–220 (2006).
    • 36  Banks ML, Negus SS. Effects of extended cocaine access and cocaine withdrawal on choice between cocaine and food in rhesus monkeys. Neuropsychopharmacology35,493–504 (2010).
    • 37  Fleckenstein AE, Gibb JW, Hanson GR. Differential effects of stimulants on monoaminergic transporters: pharmacological consequences and implications for neurotoxicity. Eur. J. Pharmacol.406,1–13 (2000).
    • 38  Johanson CE, Fischman MW. The pharmacology of cocaine related to its abuse. Pharmacol. Rev.41,3–52 (1989).
    • 39  Rothman RB, Glowa JR. A review of the effects of dopaminergic agents on humans, animals, and drug-seeking behavior, and its implications for medication development: focus on GBR 12909. Mol. Neurobiol.11,1–19 (1995).
    • 40  Ritz MC, Lamb RJ, Goldberg SR, Kuhar MJ. Cocaine receptors on dopamine transporters are related to self-administration of cocaine. Science237(4819),1219–1223 (1987).▪ This seminal preclinical study definitively demonstrated that the ability of cocaine to block dopamine reuptake is crucial to its abuse-related effects.
    • 41  Rothman RB, Baumann MH, Dersch CM et al. Amphetamine-type central nervous system stimulants release norepinephrine more potently than they release dopamine and serotonin. Synapse39,32–41 (2001).
    • 42  Kuhar MJ, Ritz MC, Boja JW. The dopamine hypothesis of the reinforcing properties of cocaine. Trends Neurosci.14(7),299–302 (1991).
    • 43  Witkin JM. Pharmacotherapy of cocaine abuse: preclinical development. Neurosci. Biobehav. Rev.18,121–142 (1994).
    • 44  Woolverton WL, Johnson KM. Neurobiology of cocaine abuse. Trends Pharmacol. Sci.13,193–200 (1992).
    • 45  Holmes VF. Medical use of psychostimulants: an overview. Int. J. Psychiatry Med.25(1),1–19 (1995).
    • 46  Wilens TE, Biederman J, Spencer TJ, Prince J. Pharmacotherapy of adult attention/deficit hyperactivity disorder: a review. J. Clin. Psychopharmacol.15(4),270–279 (1995).
    • 47  Gatley SJ, Volkow ND, Gifford AN et al. Dopamine-transporter occupancy after intravenous doses of cocaine and methylphenidate in mice and humans. Psychopharmacology146(1),93–100 (1999).
    • 48  Volkow ND, Fowler JS, Gatley JS et al. Comparable changes in synaptic dopamine induced by methylphenidate and cocaine in the baboon brain. Synapse31,59–66 (1999).
    • 49  Volkow ND, Wang GJ, Fowler JS et al. Methylphenidate and cocaine have similar in vivo potency to block the dopamine transporters in the human brain. Life Sci.65,7–12 (1999).
    • 50  Volkow ND, Ding YS, Fowler JS et al. Is methylphenidate like cocaine?. Studies on their pharmacokinetics and distribution in the human brain. Arch. Gen. Psychiatry52,456–463 (1995).
    • 51  Bergman J, Madras BK, Johnson SE, Spealman RD. Effects of cocaine and related drugs in nonhuman primates. III. Self-administration by squirrel monkeys. J. Pharmacol. Exp. Ther.251(1),150–155 (1989).
    • 52  Greenwald MK, Lundahl LH, Steinmiller CL. Sustained release D-amphetamine reduces cocaine but not ‘speedball’-seeking in buprenorphine-maintained volunteers: a test of dual-agonist pharmacotherapy for cocaine/heroin polydrug abusers. Neuropsychopharmacology35(13),2624–2637 (2010).
    • 53  Stoops WW, Glaser PE, Rush CR. Reinforcing, subject-rated, and physiological effects of intranasal methylphenidate in humans: a dose–response analysis. Drug Alcohol Depend.71,179–186 (2003).
    • 54  Rush CR, Baker RW. Behavioral pharmacological similarities between methylphenidate and cocaine in cocaine abusers. Exp. Clin. Psychopharmacol.9(1),59–73 (2001).
    • 55  Wood DM, Emmett-Oglesby MW. Substitution and cross-tolerance profiles of anorectic drugs in rats trained to detect the discriminative stimulus properties of cocaine. Psychopharmacology95,364–368 (1988).
    • 56  Fischman MW, Foltin RW. Cocaine self-administration research: implications for rational pharmacotherapy. In: Cocaine Abuse Research: Pharmacology, Behavior and Clinical Applications. Higgins ST, Katz JL (Eds). Academic Press, San Diego, CA, USA, 181–208 (1998).
    • 57  Bigelow GE, Walsh SL. Evaluation of potential pharmacotherapies: response to cocaine challenge in the human laboratory. In: Cocaine Abuse Research: Pharmacology, Behavior and Clinical Applications. Higgins ST, Katz JL (Eds). Academic Press, San Diego, CA, USA, 209–238 (1998).
    • 58  Glowa JR, Fantegrossi WE. Effects of dopaminergic drugs on food- and cocaine-maintained responding. IV: Continuous cocaine infusions. Drug Alcohol Depend.45,71–79 (1997).
    • 59  Hiranita T, Soto PL, Newman AH, Katz JL. Assessment of reinforcing effects of benztropine analogs and their effects on cocaine self-administration in rats: comparisons with monoamine uptake inhibitors. J. Pharmacol. Exp. Ther.329(2),677–686 (2009).
    • 60  Negus SS. Rapid assessment of choice between cocaine and food in rhesus monkeys: effects of environmental manipulations and treatment with D-amphetamine and flupenthixol. Neuropsychopharmacology28,919–931 (2003).
    • 61  Skjoldager P, Winger G, Woods JH. Effects of GBR 12909 and cocaine on cocaine-maintained behavior in rhesus monkeys. Drug Alcohol Depend.33,31–39 (1993).
    • 62  Collins SL, Levin FR, Foltin RW, Kleber HD, Evans SM. Response to cocaine alone and in combination with methylphenidate in cocaine abusers with ADHD. Drug Alcohol Depend.82,158–167 (2006).
    • 63  Walsh SL, Haberny KA, Bigelow GE. Modulation of intravenous cocaine effects by chronic oral cocaine in humans. Psychopharmacology150,361–373 (2000).
    • 64  Winhusen T, Somoza E, Singal BM et al. Methylphenidate and cocaine: a placebo-controlled drug interaction study. Pharmacol. Biochem. Behav.85,29–38 (2006).
    • 65  Oliveto AH, Rosen MI, Woods SW, Kosten TR. Discriminative stimulus, self-reported and cardiovascular effects of orally administered cocaine in humans. J. Pharmacol. Exp. Ther.272,231–241 (1995).
    • 66  Van Dyke C, Jatlow P, Ungerer J, Barash PG, Byck R. Oral cocaine: plasma concentrations and central effects. Science200,211–213 (1978).
    • 67  Johanson CE, Lundahl LH, Schubiner H. Effects of oral cocaine on intravenous cocaine discrimination in humans. Exp. Clin. Psychopharmacol.15(3),219–227 (2007).
    • 68  Grabowski J, Roache JD, Schmitz JM, Rhoades H, Creson D, Korszun A. Replacement medication for cocaine dependence: methylphenidate. J. Clin. Psychopharmacol.17(6),485–488 (1997).
    • 69  Levin FR, Evans SM, Brooks DJ, Garawi F. Treatment of cocaine dependent treatment seekers with adult ADHD: double-blind comparison of methylphenidate and placebo. Drug Alcohol Depend.87(1),20–29 (2007).
    • 70  Schubiner H, Saules KK, Arfken CL et al. Double-blind placebo-controlled trial of methylphenidate in the treatment of adult ADHD patients with comorbid cocaine dependence. Exp. Clin. Psychopharmacol.10(3),286–294 (2002).
    • 71  Fischman MW, Johanson CE. Ethical and practical issues involved in behavioral pharmacology research that administers drugs of abuse to human volunteers. Behav. Pharmacol.9,479–498 (1998).
    • 72  Perkins KA, Lerman C, Stitzer M et al. Development of procedures for early screening of smoking cessation medications in humans. Clin. Pharmacol. Ther.84,216–221 (2008).
    • 73  Seiden LS, Sabol KE, Ricaurte GA. Amphetamine: effects on catecholamine systems and behavior. Ann. Rev. Pharmacol. Toxicol.33,639–677 (1993).
    • 74  Carroll ME, Lac ST. Acquisition of i.v. amphetamine and cocaine self-administration in rats as a function of dose. Psychopharmacology129,206–214 (1997).
    • 75  Gasior M, Bergman J, Kallman MJ, Paronis CA. Evaluation of the reinforcing effects of monoamine reuptake inhibitors under a concurrent schedule of food and i.v. drug delivery in rhesus monkeys. Neuropsychopharmacology30,758–764 (2005).
    • 76  Richardson NR, Roberts DCS. Progressive ratio schedules in drug self-administration studies in rats: a method to evaluate reinforcing efficacy. J. Neurosci. Methods66,1–11 (1996).
    • 77  Shannon HE, Risner ME. Comparison of behavior maintained by intravenous cocaine and D-amphetamine in dogs. J. Pharmacol. Exp. Ther.229,422–432 (1984).
    • 78  Woolverton WL, Hecht GS, Agoston GE, Katz JL, Newman AH. Further studies of the reinforcing effects of benztropine analogs in rhesus monkeys. Psychopharmacology154,375–382 (2001).
    • 79  Hodos W. Progressive ratio as a measure of reward strength. Science134,943–944 (1961).
    • 80  Johanson CE, Lundahl LH, Lockhart N, Schubiner H. Intravenous cocaine discrimination in humans. Exp. Clin. Psychopharmacol.14(2),99–108 (2006).
    • 81  Kamien JB, Woolverton WL. A pharmacological analysis of the discriminative stimulus properties of D-amphetamine in rhesus monkeys. J. Pharmacol. Exp. Ther.248,938–946 (1989).
    • 82  Li SM, Campbell BL, Katz JL. Interactions of cocaine with dopamine uptake inhibitors or dopamine releasers in rats discriminating cocaine. J. Pharmacol. Exp. Ther.317(3),1088–1096 (2006).
    • 83  Oliveto AH, McCance-Katz E, Singha A, Hameedi F, Kosten TR. Effects of D-amphetamine and caffeine in humans under a cocaine discrimination procedure. Behav. Pharmacol.9,207–217 (1998).
    • 84  Barrett AC, Miller JR, Dohrmann JM, Caine SB. Effects of dopamine indirect agonists and selective D1-like and D2-like agonists and antagonists on cocaine self-administration and food maintained responding in rats. Neuropharmacology47(Suppl. 1),256–273 (2004).
    • 85  Deroche-Gamonet V, Darnaudery M, Bruins-Slot L, Piat F, Le Moal M, Piazza PV. Study of the addictive potential of modafinil in naive and cocaine-experienced rats. Psychopharmacology161(4),387–395 (2002).
    • 86  Chiodo KA, Roberts DC. Decreased reinforcing effects of cocaine following 2 weeks of continuous D-amphetamine treatment in rats. Psychopharmacology206,447–456 (2009).
    • 87  Chiodo KA, Lack CM, Roberts DC. Cocaine self-administration reinforced on a progressive ratio schedule decreases with continuous D-amphetamine treatment in rats. Psychopharmacology200,465–473 (2008).
    • 88  Czoty PW, Martelle JL, Carroll FI, Nader MA. Lower reinforcing strength of the phenyltropane cocaine analogs RTI-336 and RTI-177 compared to cocaine in nonhuman primates. Pharmacol. Biochem. Behav.96(3),274–278 (2010).
    • 89  Czoty PW, Gould RW, Martelle JL, Nader MA. Prolonged attenuation of the reinforcing strength of cocaine by chronic D-amphetamine in rhesus monkeys. Neuropsychopharmacology36(2),539–547 (2011).
    • 90  Negus SS, Mello NK. Effects of chronic D-amphetamine treatment on cocaine- and food-maintained responding under a progressive-ratio schedule in rhesus monkeys. Psychopharmacology167,324–332 (2003a).
    • 91  Negus SS, Mello NK. Effects of chronic D-amphetamine treatment on cocaine- and food-maintained responding under a second-order schedule in rhesus monkeys. Drug Alcohol Depend.70,39–52 (2003b).
    • 92  Negus SS, Mello NK, Blough BE, Baumann MH, Rothman RB. Monoamine releasers with varying selectivity for dopamine/norepinephrine versus serotonin release as candidate ‘agonist’ medications for cocaine dependence: studies in assays of cocaine discrimination and cocaine self-administration in rhesus monkeys. J. Pharmacol. Exp. Ther.320,627–636 (2007).▪▪ This preclinical research demonstrated how potential agonist-replacement medications for cocaine use disorders with varying selectivity for different monoamines produce distinct changes in cocaine and food self-administration.
    • 93  Rush CR, Stoops WW, Sevak RJ, Hays LR. Cocaine choice in humans during D-amphetamine maintenance. J. Clin. Psychopharmacol.30(2),152–159 (2010).▪▪ This study was the first to demonstrate that maintenance on D-amphetamine attenuates the reinforcing effects of cocaine in the human laboratory.
    • 94  Rush CR, Stoops WW, Hays LR. Cocaine effects during D-amphetamine maintenance: a human laboratory analysis of safety, tolerability and efficacy. Drug Alcohol Depend.99,261–271 (2009).
    • 95  Griffiths RR, Troisi JR, Silverman K, Mumford GK. Multiple-choice procedure: an efficient approach for investigating drug reinforcement in humans. Behav. Pharmacol.4(1),3–13 (1993).
    • 96  Griffiths RR, Rush CR, Puhala KA. Validation of the multiple-choice procedure for investigating drug reinforcement in humans. Exp. Clin. Psychopharmacol.4,97–106 (1996).
    • 97  Grabowski J, Rhoades H, Schmitz J et al. Dextroamphetamine for cocaine-dependence treatment: a double-blind randomized clinical trial. J. Clin. Psychopharmacol.21(5),522–526 (2001).
    • 98  Shearer J, Wodak A, van Beek I, Mattick RP, Lewis J. Pilot randomized double blind placebo-controlled study of dexamphetamine for cocaine dependence. Addiction98,1137–1141 (2003).
    • 99  Higgins ST, Delaney DD, Budney AJ et al. A behavioral approach to achieving initial cocaine abstinence. Am. J. Psychiatry148(9),1218–1224 (1991).▪ This clinical study showed that a treatment grounded in operant theory (i.e., abstinence reinforcement) is more effective for reducing cocaine use than more traditional methods (i.e., a 12-step program).
    • 100  Nieuwstraten CE, Dolovich LR. Bupropion versus selective serotonin-reuptake inhibitors for treatment of depression. Ann. Pharmacother.35(12),1608–1613 (2001).
    • 101  Wilkes S. The use of bupropion SR in cigarette smoking cessation. Int. J. Chron. Obstruct. Pulmon. Dis.3,45–53 (2008).
    • 102  Bredeloux P, Dubuc I, Costentin J. Comparisons between bupropion and dexamphetamine in a range of in vivo tests exploring dopaminergic transmission. Br. J. Pharmacol.150,711–719 (2007).
    • 103  Nomikos GG, Damsma G, Wenkstern D, Fibiger HC. Acute effects of bupropion on extracellular dopamine concentrations in rat striatum and nucleus accumbens studied by in vivo microdialysis. Neuropsychopharmacology2(4),273–279 (1989).
    • 104  Kleven MS, Anthony EW, Woolverton WL. Pharmacological characterization of the discriminative stimulus effects of cocaine in rhesus monkeys. J. Pharmacol. Exp. Ther.254(1),312–317 (1990).
    • 105  Griffith JD, Carranza J, Griffith C, Miller LL. Bupropion: clinical assay for amphetamine-like abuse potential. J. Clin. Psychiatry44(5 Pt 2),206–208 (1983).
    • 106  Miller L, Griffith J. A comparison of bupropion, dextroamphetamine and placebo in mixed-substance abusers. Psychopharmacology80(3),199–205 (1983).
    • 107  Peck AW, Bye CE, Clubley M, Henson T, Riddington C. A comparison of bupropion hydrochloride with dexamphetamine and amitriptyline in healthy subjects. Br. J. Clin. Pharmacol.7(5),469–478 (1979).
    • 108  Oliveto A, McCance-Katz FE, Singha A, Petrakis I, Hameedi F, Kosten TR. Effects of cocaine prior to and during bupropion maintenance in cocaine-abusing volunteers. Drug Alcohol Depend.63(2),155–167 (2001).
    • 109  Stoops WW, Lile JA, Hays LR, Glaser PE, Rush CR. Influence of acute bupropion pretreatment on the effects of intranasal cocaine. Addiction (2012) (In press).
    • 110  Levin FR, Evans SM, Brooks DJ, Kalbag AS, Garawi F, Nunes EV. Treatment of methadone-maintained patients with adult ADHD: double-blind comparison of methylphenidate, bupropion and placebo. Drug Alcohol Depend.81(2),137–148 (2006).
    • 111  Margolin A, Kosten T, Petrakis I, Avants SK. Bupropion reduces cocaine abuse in methadone-maintained patients. Arch. Gen. Psychiatry48(1),87 (1991).
    • 112  Margolin A, Kosten TR, Avants SK et al. A multicenter trial of bupropion for cocaine dependence in methadone-maintained patients. Drug Alcohol Depend.40(2),125–131 (1995).
    • 113  Poling J, Oliveto A, Petry N et al. Six-month trial of bupropion with contingency management for cocaine dependence in a methadone-maintained population. Arch. Gen. Psychiatry63(2),219–228 (2006).
    • 114  Shoptaw S, Heinzerling KG, Rotheram-Fuller E et al. Bupropion hydrochloride versus placebo, in combination with cognitive behavioral therapy, for the treatment of cocaine abuse/dependence. J. Addict. Dis.27,13–23 (2008).
    • 115  Broughton RJ, Fleming JA, George CF et al. Randomized, double-blind, placebo-controlled crossover trial of modafinil in the treatment of excessive daytime sleepiness in narcolepsy. Neurology49,444–451 (1997).
    • 116  Miller JL. Modafinil approved for narcolepsy. Am. J. Health Syst. Pharm.56(4),304 (1999).
    • 117  Moldofsky H, Broughton RJ, Hill JD. A randomized trial of the long-term, continued efficacy and safety of modafinil in narcolepsy. Sleep Med.1,109–116 (2000).
    • 118  Teitelman E. Modafinil for narcolepsy. Am. J. Psychiatry158,970–971 (2001).
    • 119  Andersen ML, Kessler E, Murnane KS, McClung JC, Tufik S, Howell LL. Dopamine transporter-related effects of modafinil in rhesus monkeys. Psychopharmacology210(3),439–448 (2010).
    • 120  Madras BK, Xie Z, Lin Z et al. Modafinil occupies dopamine and norepinephrine transporters in vivo and modulates the transporters and trace amine activity in vitro. J. Pharmacol. Exp. Ther.319(2),561–569 (2006).
    • 121  Volkow ND, Fowler JS, Logan J et al. Effects of modafinil on dopamine and dopamine transporters in the male human brain: clinical implications. JAMA301,1148–1154 (2009).
    • 122  Zolkowska D, Jain R, Rothman RB et al. Evidence for the involvement of dopamine transporters in behavioral stimulant effects of modafinil. J. Pharmacol. Exp. Ther.329,738–746 (2009).
    • 123  Dopheide MM, Morgan RE, Rodvelt KR, Schachtman TR, Miller DK. Modafinil evokes striatal [(3)H]dopamine release and alters the subjective properties of stimulants. Eur. J. Pharmacol.568(1–3),112–123 (2007).
    • 124  Gold LH, Balster RL. Evaluation of the cocaine-like discriminative stimulus effects and reinforcing effects of modafinil. Psychopharmacology126(4),286–292 (1996).
    • 125  Newman JL, Negus SS, Lozama A, Prisinzano TE, Mello NK. Behavioral evaluation of modafinil and the abuse-related effects of cocaine in rhesus monkeys. Exp. Clin. Psychopharmacol.18(5),395–408 (2010).
    • 126  Rush CR, Kelly TH, Hays LR, Baker RW, Wooten AF. Acute behavioral and physiological effects of modafinil in drug abusers. Behav. Pharmacol.13(2),105–115 (2002).
    • 127  Rush CR, Kelly TH, Hays LR, Wooten AF. Discriminative-stimulus effects of modafinil in cocaine-trained humans. Drug Alcohol Depend.67(3),311–322 (2002a).
    • 128  Jasinski DR. An evaluation of the abuse potential of modafinil using methylphenidate as a reference. J. Psychopharmacol.14(1),53–60 (2000).
    • 129  Jasinski DR, Kovacevic-Ristanovic R. Evaluation of the abuse liability of modafinil and other drugs for excessive daytime sleepiness associated with narcolepsy. Clin. Neuropharmacol.23(3),149–156 (2000).
    • 130  Malcolm R, Book SW, Moak D, DeVane L, Czepowicz V. Clinical applications of modafinil in stimulant abusers: low abuse potential. Am. J. Addict.11,247–249 (2002).
    • 131  Myrick H, Malcolm R, Taylor B, LaRowe S. Modafinil: preclinical, clinical and post-marketing surveillance – a review of abuse liability issues. Ann. Clin. Psychiatry16(2),101–109 (2004).
    • 132  Vosburg SK, Hart CL, Haney M, Rubin E, Foltin RW. Modafinil does not serve as a reinforcer in cocaine abusers. Drug Alcohol Depend.106,233–236 (2010).
    • 133  Dackis CA, Lynch KG, Yu E et al. Modafinil and cocaine: a double-blind, placebo-controlled drug interaction study. Drug Alcohol Depend.70(1),29–37 (2003).
    • 134  Hart CL, Haney M, Vosburg SK, Rubin E, Foltin RW. Smoked cocaine self-administration is decreased by modafinil. Neuropsychopharmacology33(4),761–768 (2008).
    • 135  Malcolm R, Swayngim K, Donovan JL et al. Modafinil and cocaine interactions. Am. J. Drug Alcohol Abuse32(4),577–587 (2006).
    • 136  Dackis CA, Kampman KM, Lynch KG, Pettinati HM, O’Brien CP. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology30(1),205–211 (2005).
    • 137  Anderson AL, Reid MS, Li SH et al. Modafinil for the treatment of cocaine dependence. Drug Alcohol Depend.104(1–2),133–139 (2009).
    • 138  Carroll FI, Howell LL, Kuhar MJ. Pharmacotherapies for treatment of cocaine abuse: preclinical aspects. J. Med. Chem.42,2721–2736 (1999).
    • 139  Carroll FI, Pawlush N, Kuhar MJ, Pollard GT, Howard JL. Synthesis, monoamine transporter binding properties, and behavioral pharmacology of a series of 3β-(substituted phenyl)-2β-(3´-substituted isoxazol-5-yl)tropanes. J. Med. Chem.47,296–302 (2004).
    • 140  Carroll FI, Runyon SP, Abraham P et al. Monoamine transporter binding, locomotor activity, and drug discrimination properties of 3-(4-substituted-phenyl)tropane-2-carboxylic acid methyl ester isomers. J. Med. Chem.47,6401–6409 (2004).
    • 141  Carroll FI, Howard JL, Howell LL, Fox BS, Kuhar MJ. Development of the dopamine transporter selective RTI-336 as a pharmacotherapy for cocaine abuse. AAPS J.8,E196–E203 (2006).
    • 142  Carroll FI, Fox BS, Kuhar MJ, Howard JL, Pollard GT, Schenk S. Effects of dopamine transporter selective 3-phenyltropane analogs on locomotor activity, drug discrimination, and cocaine self-administration after oral administration. Eur. J. Pharmacol.553,149–156 (2006).
    • 143  Kimmel HL, O’Connor JA, Carroll FI, Howell LL. Faster onset and dopamine transporter selectivity predict stimulant and reinforcing effects of cocaine analogs in squirrel monkeys. Pharmacol. Biochem. Behav.86(1),45–54 (2007).
    • 144  Kimmel HL, Negus SS, Wilcox KM et al. Relationship between rate of drug uptake in brain and behavioral pharmacology of monoamine transporter inhibitors in rhesus monkeys. Pharmacol. Biochem. Behav.90(3),453–462 (2008).
    • 145  Howell LL, Carroll FI, Votaw JR, Goodman MM, Kimmel HL. Effects of combined dopamine and serotonin transporter inhibitors on cocaine self-administration in rhesus monkeys. J. Pharmacol. Exp. Ther.320(2),757–765 (2007).
    • 146  Haile CN, Zhang XY, Carroll FI, Kosten TA. Cocaine self-administration and locomotor activity are altered in Lewis and F344 inbred rats by RTI 336, a 3-phenyltropane analog that binds to the dopamine transporter. Brain Res.1055(1–2),186–195 (2005).
    • 147  Czoty PW, Ginsburg BC, Howell LL. Serotonergic attenuation of the reinforcing and neurochemical effects of cocaine in squirrel monkeys. J. Pharmacol. Exp. Ther.300(3),831–837 (2002).
    • 148  Wee S, Anderson KG, Baumann MH, Rothman RB, Blough BE, Woolverton WL. Relationship between the serotonergic activity and reinforcing effects of a series of amphetamine analogs. J. Pharmacol. Exp. Ther.313,848–854 (2005).
    • 149  Rothman RB, Blough BE, Woolverton WL et al. Development of a rationally designed, low abuse potential, biogenic amine releaser that suppresses cocaine self-administration. J. Pharmacol. Exp. Ther.313,1361–1369 (2005).
    • 150  Negus SS, Baumann MH, Rothman RB, Mello NK, Blough BE. Selective suppression of cocaine- versus food-maintained responding by monoamine releasers in rhesus monkeys: benzylpiperazine, (+)phenmetrazine, and 4-benzylpiperidine. J. Pharmacol. Exp. Ther.329,272–281 (2009).
    • 151  Abreu ME, Bigelow GE, Fleisher L, Walsh SL. Effect of intravenous injection speed on responses to cocaine and hydromorphone in humans. Psychopharmacology154(1),76–84 (2001).
    • 152  Kollins SH, Rush CR, Pazzaglia PJ, Ali JA. Comparison of acute behavioral effects of sustained-release and immediate-release methylphenidate. Exp. Clin. Psychopharmacol.6(4),367–374 (1998).
    • 153  Krishnan SM, Pennick M, Stark JG. Metabolism, distribution and elimination of lisdexamfetamine dimesylate: open-label, single-centre, Phase I study in healthy adult volunteers. Clin. Drug. Investig.28,745–755 (2008).
    • 154  Krishnan SM, Stark JG. Multiple daily-dose pharmacokinetics of lisdexamfetamine dimesylate in healthy adult volunteers. Curr. Med. Res. Opin24,33–40 (2008).
    • 155  Jasinski DR, Krishnan S. Human pharmacology of intravenous lisdexamfetamine dimesylate: abuse liability in adult stimulant abusers. J. Psychopharmacol.23,410–418 (2009).
    • 156  Jasinski DR, Krishnan S. Abuse liability and safety of oral lisdexamfetamine dimesylate in individuals with a history of stimulant abuse. J. Psychopharmacol.23,419–427 (2009).
    • 157  Rothman RB, Dersch CM, Carroll FI, Ananthan S. Studies of the biogenic amine transporters. VIII: identification of a novel partial inhibitor of dopamine uptake and dopamine transporter binding. Synapse43,268–274 (2002).
    • 158  de la Garza R, Johanson CE. Discriminative stimulus properties of intragastrically administered D-amphetamine and pentobarbital in rhesus monkeys. J. Pharmacol. Exp. Ther.243(3),955–962 (1987).
    • 159  Evans SM, Johanson CE. Amphetamine-like effects of anorectics and related compounds in pigeons. J. Pharmacol. Exp. Ther.241,817–825 (1987).
    • 160  Corwin RL, Woolverton WL, Schuster CR, Johanson CE. Anorectics: effects on food intake and self-administration in rhesus monkeys. Alcohol Drug Res.7,351–361 (1987).
    • 161  Griffiths RR, Brady JV, Bradford LD. Predicting the abuse liability of drugs with animal drug self-administration procedures: psychomotor stimulants and hallucinogens. In: Advances in Behavioral Pharmacology, Volume 2. Thompson T, Dews PB (Eds). Academic Press, NY, USA 163–208 (1979).
    • 162  Bramble D. Psychostimulants and psychiatrists: the Trent Adult Psychiatry Psychostimulant Survey. J. Psychopharmacol.14,67–69 (2000).
    • 163  Klein RG. The role of methylphenidate in psychiatry. Arch. Gen. Psychiatry52,429–433 (1995).
    • 164  Goeders NE, McNulty MA, Mirkis S, McAllister KH. Chlordiazepoxide alters intravenous cocaine self-administration in rats. Pharmacol. Biochem. Behav.33,859–866 (1989).
    • 165  Goeders NE, McNulty MA, Guerin GF. Effects of alprazolam on intravenous cocaine self-administration in rats. Pharmacol. Biochem. Behav.44,471–474 (1993).
    • 166  Goeders NE, Guerin GF. Effects of the combination of metyrapone and oxazepam on cocaine and food self-administration in rats. Pharmacol. Biochem. Behav.91,181–189 (2008).
    • 167  Goeders NE, Clampitt DM. Potential role for the hypothalamo-pituitary-adrenal axis in the conditioned reinforcer-induced reinstatement of extinguished cocaine seeking in rats. Psychopharmacology161,222–232 (2002).
    • 168  Keim KL, Sigg EB. Plasma corticosterone and brain catecholamines in stress: effect of psychotropic drugs. Pharmacol. Biochem. Behav.6,79–85 (1977).
    • 169  Yang XM, Gorman AL, Dunn AJ, Goeders, NE. Anxiogenic effects of acute and chronic cocaine administration: neurochemical and behavioral studies. Pharmacol. Biochem. Behav.41,643–650 (1992).
    • 170  Goeders NE, Guerin GF. Effects of surgical and pharmacological adrenalectomy on the initiation and maintenance of intravenous cocaine self-administration in rats. Brain Res.722,145–152 (1996).
    • 171  Lile JA, Stoops WW, Glaser PEA, Hays LR, Rush CR. Subjective and physiological effects of acute intranasal methamphetamine during extended-release alprazolam maintenance. Drug Alcohol Depend.119(3),187–193 (2011).
    • 172  Mumford GK, Evans SM, Fleishaker, JC, Griffiths, RR. Alprazolam absorption kinetics affects abuse liability. Clin. Pharmacol. Ther.57,356–365 (1995).
    • 173  Mumford GK, Rush CR, Griffiths, RR. Abecarnil and alprazolam in humans: behavioral, subjective and reinforcing effects. J. Pharmacol. Exp. Ther.272,570–580 (1995).
    • 174  Rush CR, Griffiths RR. Acute participant-rated and behavioral effects of alprazolam and buspirone, alone and in combination with ethanol, in normal volunteers. Exp. Clin. Psychopharmacol.5,28–38 (1997).
    • 175  Rush CR, Higgins ST, Bickel WK, Hughes JR. Abuse liability of alprazolam relative to other commonly used benzodiazepines: a review. Neurosci. Biobehav. Rev.17,277–285 (1993).
    • 176  Kablinger AS, Lindner MA, Casso S et al. Effects of the combination of metyrapone and oxazepam on cocaine craving and cocaine taking: a double-blind, randomized, placebo-controlled pilot study. J. Psychopharmacol. (2011) (In Press).