East Asian Arch Psychiatry 2016;26:52-9

THEME PAPER

Developing Treatments for Stimulant Abuse: A Brief Overview
C Davidson

Dr Colin Davidson, BSc, PGDip, PhD, St George’s University of London, London, United Kingdom.

Address for correspondence: Dr Colin Davidson, School of Pharmacy and Biomedical Sciences, College of Clinical and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, United Kingdom.
Tel: 44 (0) 1772 893920; Email: cdavidson2@uclan.ac.uk

Submitted: 27 October 2015; Accepted: 4 January 2016


pdf Full Paper in PDF

Abstract

The abuse of stimulants such as cocaine, amphetamine, and methamphetamine is a huge problem in many parts of the world. Abuse of these drugs does not ruin just the user’s life, but also adversely affects those around them. Despite many years of research, there are no approved medications for stimulant dependence, and treatment is focused on psychotherapy and abstinence. Over the last 10 to 20 years, there have been some major changes in approach to medication development for stimulant dependence. These include assessing ligands for non-dopaminergic sites, atypical dopamine transporter ligands, blocking long-term potentiation and / or memory reconsolidation, vaccines against the stimulant, and molecular approaches including pharmacogenomics and gene silencing. Also included in this overview are non-drug treatments such as deep brain stimulation and psychosurgery. This overview highlights recent preclinical and clinical studies of treatment development for stimulant dependence.

Key words: Amphetamine; Cocaine; Drug therapy; Methamphetamine; Therapeutics

Stimulant Misuse Is Widespread and Difficult to Treat

Lots of people take stimulants. In Europe, 5.7 million adults took cocaine and 2.9 million adults took amphetamine in 2015.1 In 2014 there were 0.9 million cocaine-abusing or cocaine-dependent individuals in the US.2 South-East Asia seems to have the biggest problem with amphetamine- type stimulants,3 and Japan has had problems with methamphetamine for over 60 years.4 There are currently no approved pharmacotherapies for stimulant abuse. Behavioural therapies work to a certain extent but are limited.5 Dropout rates of over 50% are common in cocaine dependence treatment programmes6 and it is likely that cocaine withdrawal symptoms greatly contribute to dropout rates.7 The need for new treatments for stimulant dependence is thus critical.

Time Course of Stimulant Dependence

Traditionally, pharmacotherapies have aimed to block the reinforcing effects of stimulants (e.g. blocking the dopamine transporter [DAT]) or alleviate the anxiety or depression associated with stimulant withdrawal. Nonetheless, when the various stages of drug dependence are considered, it becomes clear that there are multiple stages at which therapies can be targeted. In addition, it is likely that different pharmacotherapies will be needed for these different stages. Thus, there are phases of drug-taking that can be targeted (Fig), when stimulant levels and / or extracellular dopamine levels are high. There are also chronic drug withdrawal states where there is dopaminergic hypofunction.8 In general, drugs can be targeted at ‘abstinence initiation’ or ‘relapse prevention’ and these have been reviewed.9 Drug therapies that might be useful in initiating abstinence include modafinil (a mild stimulant currently approved for narcolepsy), propranolol (an anxiolytic), and bupropion (an antidepressant). Relapse prevention pharmacotherapy might include γ-aminobutyric acid (GABA) enhancers such as γ-vinyl GABA, tiagabine, and topiramate to be discussed below.

Nonetheless, if one examines drug dependence more closely we can split the addict’s activities or behaviour into numerous subcategories, each of which could be a target for pharmacotherapy. These subcategories include thinking about taking the drug, drug-taking, euphoria / drug-liking, acute drug withdrawal, compulsion to take the drug, relapse, chronic drug withdrawal, withdrawal-associated depression, withdrawal-associated anxiety, and drug-induced cognitive dysfunction.

Treatments for Stimulant Dependence That Have Thus Far Not Worked

Agonist therapies include drugs that act on the same receptor or transporter as the drug of abuse. In the case of cocaine this is the DAT and dopamine D1-5 receptors. Because cocaine’s acute action is to increase synaptic dopamine, it has indirect agonist effects at the dopamine receptors. The theory behind agonist therapy is that by giving the addict a little bit of what he / she wants, they may not take the primary drug of abuse. As we have seen with heroin, however, the addict can become addicted to the agonist therapy (methadone). Typically the agonist (or substitution) therapy is longer-acting (making it less addictive) and with a better safety profile than the drug of abuse. Amphetamine and methylphenidate have both been used in cocaine or methamphetamine abusers, with only modest effects. Similarly bupropion, a relatively weak DAT inhibitor, has had inconsistent effects in cocaine or methamphetamine dependence. Modafinil is a mild stimulant that also enhances glutamate transmission.10 It has been found to be useful in some small clinical trials, attenuating the euphoric effects of cocaine and reducing cocaine intake.11-15 More recent data have shown that modafinil causes the DAT to prefer its inward (cytosol)-facing state. Thus modafinil appears to react with the DAT more like a substrate (e.g. dopamine) than an uptake blocker like cocaine, but unlike substrates is not moved intracellularly.16 Dopamine transporter ligands with this property have been termed ‘atypical’ DAT inhibitors. More recent long-acting atypical DAT inhibitors include the CTDP series of National Institute on Drug Abuse–sponsored compounds, e.g. CTDP 3134517 and modafinil analogues.18 Thus, the quest for a cocaine agonist therapy continues.19

The opposite treatment of a substitution or maintenance therapy would be an antagonist-based therapy. This type of therapy has worked for opioid abuse, e.g. using the opioid antagonist naltrexone or the partial agonist buprenorphine.20 For cocaine abuse, rather than reducing cocaine intake by providing a low-level dopamine boost, the antagonist therapy blocks dopamine receptors and thus attenuates the rewarding effects of cocaine. Dopamine D3 receptor antagonists have been shown to work in rodent models of drug abuse21 but have not yet been tested in humans. Antagonists at the dopamine D3 receptor may have particular use in the stimulant chronic withdrawal period.22,23 Nonetheless, it is always possible that the addict will just increase their dose of stimulant to overcome the antagonist. As a caveat, one should also be wary of dopamine antagonist–induced extrapyramidal side-effects.

Future Treatments

Drugs That Work via Other Membrane Proteins

The main neurotransmitter involved in drug abuse and reward is undoubtedly dopamine. Many other neurotransmitters are also involved either directly, or as modulators of dopamine levels via action on the dopamine terminals or at the cell body region (the ventral tegmental area). In addition, other transmitter systems are involved in drug withdrawal states and their associated craving, depression, and anxiety. Drugs that block corticotrophin-releasing factor (CRF) and / or noradrenaline might decrease the stimulant-induced anxiety that could lead to relapse.

Stimulants not only increase extracellular dopamine levels, but also those of noradrenaline and 5-HT. Potentiation of 5-HT contributes to cocaine reward24 and transgenic mice devoid of the DAT still exhibit cocaine-seeking behaviour, but not when devoid of the DAT and serotonin transporters.25 It is reasonable to assume then that drugs that act on the 5-HT system may be useful in manipulating stimulant dependence. Another advantage of the 5-HT system is that there are 14 known 5-HT receptors (but only 5 for dopamine) allowing quite specific modulation of only part of the 5-HT system to reduce stimulant dependence,26 but hopefully with fewer side-effects. The use of selective serotonin reuptake inhibitors in cocaine dependence has been disappointing.27 Nonetheless, antagonists of the other actions stimulates the 5-HT2 and 5-HT3 receptors have been tested for stimulant dependence28,29 with small positive effects, and the role of 5-HT2 in cocaine use has been recently reviewed.30

The α1 adrenergic antagonist prazosin can reduce cocaine seeking in rats.31 The α adrenergic agonist guanfacine enhanced inhibitory control and executive function in early abstinent cocaine dependents.32 One might ask how an agonist could reduce noradrenergic function. It is possible that guanfacine preferentially activates the presynaptic α2 receptors, which are inhibitory autoreceptors,33 thus the net effect is lower noradrenaline efflux. Propranolol, a beta-blocker with anxiolytic qualities, has also been found to be useful in patients with severe cocaine withdrawal, increasing the number of patients who stayed in treatment34,35 or remained abstinent.36 The CRF-1 receptor antagonist CP-154,526 can attenuate cocaine or methamphetamine reinstatement (relapse) in rodents37,38 and also reduce cocaine-induced conditioned place preference (cocaine seeking) in rodents.39 Thus far, antagonists at the glutamate N-methyl-D-aspartate (NMDA) receptor, for example, memantine, have been unsuccessful in cocaine dependence.40 Nonetheless N-acetylcysteine, which among other actions stimulates the cysteine-glutamate transporter,41 normalises glutamate levels in the accumbens after cocaine42,43 and may reduce cocaine craving in humans.44-46

Other transmitter systems that might be involved in drug dependence include the orexin, GABAergic and cholinergic systems, as well as pharmacotherapies that act via these systems have been tested. The hypothalamic neuropeptide orexin is released into the accumbens and SB- 334867, an orexin-1 receptor antagonist, inhibits relapse in rodents.47 Tiagabine, used to treat seizures, increases extracellular GABA levels by blocking GABA reuptake.48 It can increase abstinence in cocaine dependence.49 Varenicline, a nicotinic acetylcholine receptor partial agonist, decreased cocaine use50 but had little effect on cocaine abstinence.51 Recently, much research has focused on the cannabinoid system and the CB1 receptor antagonist rimonabant that reversed cocaine sensitisation52 and blocked cue-induced reinstatement of cocaine seeking.53 Regrettably clinical use of rimonabant ceased due to potentially serious side-effects. More recent research has focused on the phytocannabinoid cannabidiol, a weak CB1/2 receptor antagonist. This drug has shown promise in animal models54 and a Canadian trial is currently recruiting for cannabidiol and cocaine dependence (ClinicalTrials.gov identifier: NCT02559167).

Therapeutics That Act via Enzymes

Butyrylcholinesterase is an enzyme that metabolises cocaine. It has been tested in rodents and in people where it may accelerate cocaine metabolism.55 Its use in cocaine dependence has been reviewed.56 Bacterial cocaine esterases (CocE), which also metabolise cocaine, have been isolated from Rhodococcus bacteria that grow in the soil around coca plants. It has had some success in rat models57 but is not stable at physiological temperatures. Mutants of CocE have been developed that are stable at physiological temperatures and can reduce cocaine self-administration in rats. 58 These studies have been reviewed.59 The GABAergic drugs have long been known to be useful in anxiety. Benzodiazepines have been tested in stimulant dependence60 and has led investigators to test drugs that act via GABAergic enzymes: vigabatrin (γ-vinyl GABA), an antiepileptic and an irreversible inhibitor of GABA transaminase (which breaks down GABA), has been shown to increase abstinence in cocaine dependents.61-63

Therapeutics That Block Long-term Potentiation or Affect Memory Reconsolidation

It has been hypothesised that some of the changes seen with long-term stimulant use are similar to those seen in memory formation, including long-term potentiation.64 We can take the memory analogy a step further by considering memory reconsolidation where memories that are reactivated become liable to be modified.65,66 It is thought that by disrupting ‘drug memory’ reconsolidation, when the ‘drug memory’ is particularly labile, we might be able to attenuate drug dependence and this theory has been tested in rodents. Lee et al67-69 used NMDA receptor antagonists to disrupt memory reconsolidation for cocaine, and reduced cocaine- seeking and relapse-like behaviour.

We have previously tried to disrupt cocaine-evoked pharmacological changes by treating rats with the 5-HT3 receptor antagonist ondansetron, not before cocaine injection or self-administration, but by giving ondansetron 3.5 hours after cocaine.70,71 Ondansetron, given after (but not before) cocaine, was able to reduce cocaine self- administration the following day. Giving cocaine as part of a treatment regimen is probably not a great idea and so we tested a cocaine substitute in a similar regimen. We chose pergolide, a clinically used (Parkinson’s disease) dopamine D1/2 receptor agonist as a drug that might evoke a cocaine memory. This pergolide plus ondansetron treatment was able to reverse a previously established cocaine sensitisation72 and attenuated methamphetamine reinstatement (relapse) in rats.73 This work has been reviewed74 and small clinical trials have been completed to establish safety and efficacy for a methylphenidate plus ondansetron treatment regimen (ClinicalTrials.gov identifiers: NCT01290276, NCT01377662). The disruption of drug memory reconsolidation may yet prove to be a fruitful area in clinical trials.

Vaccines for Stimulants

Vaccines for stimulants have been developed so that antibodies to, for example, cocaine are produced, limiting the amount of cocaine that passes from the bloodstream to the brain, reducing its rewarding effects. In the first trial of a cocaine vaccine in humans the vaccine appeared to reduce the euphoric effects of cocaine and increased cocaine abstinence.75 Despite this, results to date have not been as good as hoped due to the relatively long incubation period needed for the required amount of antibodies to be produced. In addition, it is questionable whether enough antibodies could ever be produced to attenuate the effects of the vast amount of cocaine molecules ingested by addicts. A major limitation is that only about 40% of patients treated with the cocaine vaccine actually produced enough antibodies, suggesting that better adjuvants are needed. To this aim, Kosten et al76 have tried to improve the immunogenicity by using keyhole limpet haemocyanin as the carrier protein with improved results and a reduction in cocaine-evoked hyperactivity. The same group has also attempted to improve the cocaine hapten, with succinyl butyl norcocaine proving to be the hapten that produced the most cocaine antibodies.77 To date, however, clinical trials with cocaine vaccines have been disappointing.78 Bacterial proteins have also been tested as both carrier and adjuvant.79 Monoclonal antibodies (produced outside the body) are another mechanism to reduce the effects of cocaine. Research in this area would benefit from behavioural pharmacologists and psychiatrists working more closely with immunologists.

Traditional Herbal Medicines

Traditional medicines from South America, Africa, and Asia all have anecdotal evidence suggesting their use in drug dependence. There are only limited clinical trials but this is certainly an area where more research could be done. In South America, the herbal brew of ayahuasca has been suggested to decrease drug dependence, but research in this area has been hampered by not knowing exactly what is in the brew and concoctions being different in different areas.80,81 In addition, there is a religious element to ayahuasca use that might contribute to any therapeutic effects.

Ibogaine, found in the root of the African shrub Tabernanthe iboga, may be useful in not only cocaine dependence, but also other drugs of abuse.82 Its pharmacology is complicated with affinity at opioid, nicotinic, glutamatergic, and monoamine sites. Levo- tetrahydropalmatine is found in 2 Chinese herbs (Corydalis and Stephania), has a complex pharmacology and has been shown to be useful in rodent models of drug abuse.27

Thunbergia laurifolia, Linn, is a traditional Thai herbal medicine used to treat alcohol and drug addiction and has been tested preclinically and has some mild-stimulant properties that may make it useful as an agonist therapy.83 The use of traditional medicines in the treatment of drug dependence has been reviewed.84 This research area would benefit from researchers in the West working more closely with those from the East.

Looking Further Ahead: Cognitive Enhancers, Deep Brain Stimulation, Molecular Biology, and Psychosurgery?

We might use a personalised medicine approach where we take into account the drug misuser’s gender, age, and so on. We could also target the misuser’s cognitive dysfunction or psychiatric co-morbidity that may contribute to drug misuse and has been reviewed.85 For example, many drugs of abuse can lead to cognitive deficits including response inhibition and memory and attention problems. By treating drug abusers with cognitive enhancers they may be able to better use coping strategies. Galantamine, an acetylcholinesterase inhibitor cognitive enhancer, can reduce cocaine use.86 With the incredible research effort now focused on dementia treatments, it is likely that there will be many new cognitive enhancers becoming available soon, some of which may be useful in stimulant dependence. Similarly, if we could successfully treat the abuser’s co-morbid disorder, we may be able to reduce their drug dependence.87

Deep brain stimulation (DBS) of, for example, basal ganglia circuits, may prove useful in drug dependence. Drug dependence is defined by a compulsion to take the drug and so can be viewed in a similar way to other compulsive disorders such as obsessive-compulsive disorder. Obsessive-compulsive disorder has been treated with DBS and has been reviewed by Pepper et al88 and van Westen et al89 who state that DBS appears to counteract striatal dysfunction through an increase in striatal dopamine and through improvement of reward processing. This sounds very promising for the use of DBS in drug dependence, another condition underpinned by striatal dysfunction.

Genome-wide association studies have identified numerous genes that contribute to drug abuse including CHRNA3-5 for nicotine dependence and ADH1B and ADH1C genes for alcohol dependence. The DRD2 gene is the dopamine D2 receptor gene and may be involved in numerous central nervous system disorders, including cocaine dependence.90 Other polymorphisms associated with stimulant addiction include DAT91-93 and dopamine receptors,92,94 dopamine beta-hydroxylase,95,96 and brain- derived neurotrophic factor.97 Most recently it has been suggested that numerous genes are involved in drug dependence. Many of these genes were not expected to be involved but have been found to contribute greatly to dependence, for example, genes associated with cell adhesion molecules.98 These studies throw up numerous new targets for developing new treatments for stimulant dependence. Knowing which faulty proteins are involved in drug misuse is the first step in a gene therapy, epigenetic therapy or gene silencing for addictions.99 Finally, from a molecular viewpoint, optogenetics would also be a way to inhibit or activate dysfunctional brain pathways in psychiatric disorders100 including addictions.101-103 Nonetheless, due to ethical reasons, its use in humans would appear unlikely in the near future.

Finally, it is likely that neurosurgery for addiction will be tested to a greater degree in the future. Ablation of the nucleus accumbens core or shell has been examined in rats with respect to heroin self-administration104,105 with the core being shown to be important in heroin-seeking behaviour. Studies in human heroin abusers have had some positive outcomes106,107 but issues have been raised108 including appropriate personality follow-up tests and the possibility of improved DBS treatment being more ethical. Given that the psychostimulants act predominantly via DATs in the accumbens, while μ-opioid receptors are more widely distributed within the brain, one wonders if this surgery might work best in stimulant abusers. Recently, a book has been written on the use of neurosurgery in psychiatric disorders including a chapter on addiction.109 Brain lesions can also happen through strokes and damage to the insula decreases nicotine addiction in humans.110 Lesions of the insula also decrease amphetamine seeking in rats.111 Thus, the insula might be another target, in addition to the accumbens, for DBS or psychosurgery in stimulant addiction.

Concluding Remarks

Some of the pharmacotherapies tested in stimulant dependence as well as some more novel therapies have been highlighted. Some therapies, for example, bupropion, tested for cocaine dependence but with little effect, may nevertheless be useful for methamphetamine dependence.112

We should be aware that a therapeutic drug that fails for one stimulant or drug of abuse may be useful for another, or may be useful in a subpopulation as with a stratified medicine approach. We should also be aware of advances in other areas of the biomedical sciences that might open up new windows of opportunity for drug abuse treatments. Vaccines for cocaine dependence or the use of DBS to attenuate compulsive behaviours are good examples. Advances in molecular biology might lead to gene therapies and optogenetic therapies. Finally, one should always remember that these therapies can or should be used with psychological treatments. It is likely that a combination of drug therapy and psychotherapy will provide the greatest impact.

References

  1. European Monitoring Centre for Drugs and Drug Addiction. European drug report 2015. Available from: http://www.emcdda.europa.eu/edr2015. Accessed May 2016.
  2. National Institute on Drug Abuse. DrugFacts: nationwide trends. Available from: https://www.drugabuse.gov/publications/drugfacts/nationwide-trends. Accessed May 2016.
  3. United Nations Office on Drugs and Crime. World drug report 2012. Available from: http://www.unodc.org/unodc/en/data-and-analysis/WDR-2012.html. Accessed May 2016.
  4. Yamamoto J. Recent trends of drug abuse in Japan. Ann N Y Acad Sci 2004;1025:430-8.
  5. Simpson DD, Joe GW, Fletcher BW, Hubbard RL, Anglin MD. A national evaluation of treatment outcomes for cocaine dependence. Arch Gen Psychiatry 1999;56:507-14.
  6. Alterman AI, McKay JR, Mulvaney FD, McLellan AT. Prediction of attrition from day hospital treatment in lower socioeconomic cocaine- dependent men. Drug Alcohol Depend 1996;40:227-33.
  7. Kampman KM, Alterman AI, Volpicelli JR, Maany I, Muller ES, Luce DD, et al. Cocaine withdrawal symptoms and initial urine toxicology results predict treatment attrition in outpatient cocaine dependence treatment. Psychol Addict Behav 2001a;15:52-9.
  8. Kosten TR, George TP, Kosten TA. The potential of dopamine agonists in drug addiction. Expert Opin Invest Drugs 2002;11:491-9.
  9. Kampman KM. The search for medications to treat stimulant dependence. Addict Sci Clin Pract 2008;4:28-35.
  10. Minzenberg MJ, Carter CS. Modafinil: a review of neurochemical actions and effects on cognition. Neuropsychopharmacology 2008;33:1477-502.
  11. Dackis C, O’Brien C. Glutamatergic agents for cocaine dependence. Ann N Y Acad Sci 2003;1003:328-45.
  12. Dackis CA, Kampman KM, Lynch KG, Pettinati HM, O’Brien CP. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology 2005;30:205-11.
  13. Malcolm R, Swayngim K, Donovan JL, DeVane CL, Elkashef A, Chiang N, et al. Modafinil and cocaine interactions. Am J Drug Alcohol Abuse 2006;32:577-87.
  14. Hart CL, Haney M, Vosburg SK, Rubin E, Foltin RW. Smoked cocaine self-administration is decreased by modafinil. Neuropsychopharmacology 2008;33:761-8.
  15. Martinez-Raga J, Knecht C, Cepeda S. Modafinil: a useful medication for cocaine addiction? Review of the evidence from neuropharmacological, experimental and clinical studies. Curr Drug Abuse Rev 2008;1:213-21.
  16. 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.
  17. Peng XQ, Xi ZX, Li X, Spiller K, Li J, Chun L, et al. Is slow- onset long-acting monoamine transport blockade to cocaine as methadone is to heroin? Implications for anti-addiction medications. Neuropsychopharmacology 2010;35:2564-78.
  18. Okunoloa-Bakare OM, Cao J, Kopajtic T, Katz JL, Loland CJ, Shi L, et al. Elucidation of structural elements for selectivity across monoamine transporters: novel 2-[(diphenylmethyl)sulfinyl] acetamide (modafinil) analogues. J Med Chem 2014;57:1000-13.
  19. Reith ME, Blough BE, Hong WC, Jones KT, Schmitt KC, Baumann MH, et al. Behavioral, biological, and chemical perspectives on atypical agents targeting the dopamine transporter. Drug Alcohol Depend 2015;147:1-19.
  20. White JM, Lopatko OV. Opioid maintenance: a comparative review of pharmacological strategies. Expert Opin Pharmacother 2007;8:1- 11.
  21. Heidbreder CA. Selective antagonism at dopamine D3 receptors as a target for drug addiction pharmacotherapy: a review of preclinical evidence. CNS Neurol Disord Drug Targets 2008;7:410-21.
  22. Song R, Bi GH, Zhang HY, Yang RF, Gardner EL, Li J, et al. Blockade of D3 receptors by YQA14 inhibits cocaine’s rewarding effects and relapse to drug-seeking behavior in rats. Neuropharmacology 2014;77:398-405.
  23. Ashby CR Jr, Rice OV, Heidbreder CA, Gardner EL. The selective dopamine D3 receptor antagonist SB-277011A significantly accelerates extinction to environmental cues associated with cocaine- induced place preference in male Sprague-Dawley rats. Synapse 2015;69:512-4.
  24. Pentkowski NS, Acosta JI, Browning JR, Hamilton EC, Neisewander JL. Stimulation of 5-HT(1B) receptors enhances cocaine reinforcement yet reduces cocaine-seeking behavior. Addict Biol 2009;14:419-30.
  25. Sora I, Hall FS, Andrews AM, Itokawa M, Li XF, Wei HB, et al. Molecular mechanisms of cocaine reward: combined dopamine and serotonin transporter knockouts eliminate cocaine place preference. Proc Natl Acad Sci U S A 2001;98:5300-5.
  26. Filip M, Frankowska M, Zaniewska M, Golda A, Przegaliński E. The serotonergic system and its role in cocaine addiction. Pharmacol Rep 2005;57:685-700.
  27. Shorter D, Kosten TR. Novel pharmacotherapeutic treatments for cocaine addiction. BMC Med 2011;9:119.
  28. Johnson BA, Roache JD, Ait-Daoud N, Javors MA, Harrison JM, Elkashef A, et al. A preliminary randomized, double-blind, placebo- controlled study of the safety and efficacy of ondansetron in the treatment of cocaine dependence. Drug Alcohol Depend 2006;84:256- 63.
  29. Johnson BA, Ait-Daoud N, Elkashef AM, Smith EV, Kahn R, Vocci F, et al. A preliminary randomized, double-blind, placebo-controlled study of the safety and efficacy of ondansetron in the treatment of methamphetamine dependence. Int J Neuropsychopharmacol 2008;11:1-14.
  30. Howell LL, Cunningham KA. Serotonin 5-HT2 receptor interactions with dopamine function: implications for therapeutics in cocaine use disorder. Pharmacol Rev 2015;67:176-97.
  31. Zhang XY, Kosten TA. Prazosin, an alpha-1 adrenergic antagonist, reduces cocaine-induced reinstatement of drug-seeking. Biol Psychiatry 2005;57:1202-4.
  32. Fox H, Sofuoglu M, Sinha R. Guanfacine enhances inhibitory control and attentional shifting in early abstinent cocaine-dependent individuals. J Psychopharmacol 2015;29:312-23.
  33. Starke K, Göthert M, Kilbinger H. Modulation of neurotransmitter release by presynaptic autoreceptors. Physiol Rev 1989;69:864-989.
  34. Kampman KM, Rukstalis M, Ehrman R, McGinnis DE, Gariti P, Volpicelli JR, et al. Open trials as a method of prioritizing medications for inclusion in controlled trials for cocaine dependence. Addict Behav 1999;24:287-91.
  35. Kampman KM, Volpiceli JR, Mulvaney F, Alterman AI, Cornish J, Gariti P, et al. Effectiveness of propranolol for cocaine dependence treatment may depend on cocaine withdrawal symptom severity. Drug Alcohol Depend 2001;63:69-78.
  36. Kampman KM, Dackis C, Lynch KG, Pettinati H, Tirado C, Gariti P, et al. A double-blind, placebo-controlled trial of amantadine, propranolol, and their combination for the treatment of cocaine dependence in patients with severe cocaine withdrawal symptoms. Drug Alcohol Depend 2006;85:129-37.
  37. Przegaliński E, Filip M, Frankowska M, Zaniewska M, Papla I. Effects of CP 154,526 a CRF1 receptor antagonist, on behavioral responses to cocaine in rats. Neuropeptides 2005;39:525-33.
  38. Shaham Y, Erb S, Leung S, Buczek Y, Stewart J. CP-154,526, a selective, non-peptide antagonist of the corticotropin-releasing factor1 receptor attenuates stress-induced relapse to drug seeking in cocaine- and heroin-trained rats. Psychopharmacology 1998;137:184- 90.
  39. Moffett MC, Goeders NE. CP-154,526, a CRF type-1 receptor antagonist, attenuates the cue- and methamphetamine-induced reinstatement of extinguished methamphetamine-seeking behavior in rats. Psychopharmacology 2007;190:171-80.
  40. Bisaga A, Aharonovich E, Cheng WY, Levin FR, Mariani JJ, Raby WN, et al. A placebo-controlled trial of memantine for cocaine dependence with high-value voucher incentives during a pre- randomization lead-in period. Drug Alcohol Depend 2010;111:97- 104.
  41. Dean O, Giorlando F, Berk M. N-acetylcysteine in psychiatry: current therapeutic evidence and potential mechanisms of action. J Psychiatry Neurosci 2011;36:78-86.
  42. Madayag A, Lobner D, Kau KS, Mantsch JR, Abdulhameed O, Hearing M, et al. Repeated N-acetylcysteine administration alters plasticity-dependent effects of cocaine. J Neurosci 2007;27:13968- 76.
  43. Moussawi K, Zhou W, Shen H, Reichel CM, See RE, Carr DB, et al. Reversing cocaine-induced synaptic potentiation provides enduring protection from relapse. Proc Natl Acad Sci U S A 2011;108:385-90.
  44. LaRowe SD, Myrick H, Hedden S, Mardikian P, Saladin M, McRae A, et al. Is cocaine desire reduced by N-acetylcysteine? Am J Psychiatry 2007;164:1115-7.
  45. Mardikian PN, LaRowe SD, Hedden S, Kalivas PW, Malcolm RJ. An open-label trial of N-acetylcysteine for the treatment of cocaine dependence: a pilot study. Prog Neuropsychopharmacol Biol Psychiatry 2007;31:389-94.
  46. Amen SL, Placentine LB, Ahmad ME, Li SJ, Mantsch JR, Risinger RC, et al. Repeated N-acetyl cysteine reduces cocaine seeking in rodents and craving in cocaine-dependent humans. Neuropsychopharmacol 2011;36:871-8.
  47. Aston-Jones G, Smith RJ, Moorman DE, Richardson KA. Role of lateral hypothalamic orexin neurons in reward processing and addiction. Neuropharmacology 2009;56:112-21.
  48. Sills GJ. Pre-clinical studies with the GABAergic compounds vigabatrin and tiagabine. Epileptic Disord 2003;5:51-6.
  49. Gonzalez G, Sevarino K, Sofuoglu M, Poling J, Oliveto A, Gonsai K, et al. Tiagabine increases cocaine-free urines in cocaine-dependent methadone-treated patients: results of a randomized pilot study. Addiction 2003;98:1625-32.
  50. Plebani JG, Lynch KG, Yu Q, Pettinati HM, O’Brien CP, Kampman KM. Results of an initial clinical trial of varenicline for the treatment of cocaine dependence. Drug Alcohol Depend 2012;121:163-6.
  51. Poling J, Rounsaville B, Gonsai K, Severino K, Sofuoglu M. The safety and efficacy of varenicline in cocaine using smokers maintained on methadone: a pilot study. Am J Addict 2010;19:401-8.
  52. Gerdeman GL, Schechter JB, French ED. Context-specific reversal of cocaine sensitization by the CB1 cannabinoid receptor antagonist rimonabant. Neuropsychopharmacology 2008;33:2747-59.
  53. Ward SJ, Rosenberg M, Dykstra LA, Walker EA. The CB1 antagonist rimonabant (SR141716) blocks cue-induced reinstatement of cocaine seeking and other context and extinction phenomena predictive of relapse. Drug Alcohol Depend 2009;105:248-55.
  54. Parker LA, Burton P, Sorge RE, Yakiwchuk C, Mechoulam R. Effect of low doses of delta9-tetrahydrocannabinol and cannabidiol on the extinction of cocaine-induced and amphetamine-induced conditioned place preference learning in rats. Psychopharmacology 2004;175:360- 6.
  55. Schindler CW, Goldberg SR. Accelerating cocaine metabolism as an approach to the treatment of cocaine abuse and toxicity. Future Med Chem 2012;4:163-75.
  56. Gorelick DA. Enhancing cocaine metabolism with butyrylcholinesterase as a treatment strategy. Drug Alcohol Depend 1997;48:159-65.
  57. Cooper ZD, Narasimhan D, Sunahara RK, Mierzejewski P, Jutkiewicz EM, Larsen NA, et al. Rapid and robust protection against cocaine- induced lethality in rats by the bacterial cocaine esterase. Mol Pharmacol 2006;70:1885-91.
  58. Collins GT, Brim RL, Narasimhan D, Ko MC, Sunahara RK, Zhan CG, et al. Cocaine esterase prevents cocaine-induced toxicity and the ongoing intravenous self-administration of cocaine in rats. J Pharmacol Exp Ther 2009;331:445-55.
  59. Narasimhan D, Woods JH, Sunahara RK. Bacterial cocaine esterase: a protein-based therapy for cocaine overdose and addiction. Future Med Chem 2012;4:137-50.
  60. Connors NJ, Hoffman RS. Experimental treatments for cocaine toxicity: a difficult transition to the bedside. J Pharmacol Exp Ther 2013;347:251-7.
  61. Brodie JD, Figueroa E, Dewey SL. Treating cocaine addiction: from preclinical to clinical trial experience with gamma-vinyl GABA. Synapse 2003;50:261-5.
  62. Brodie JD, Figueroa E, Laska EM, Dewey SL. Safety and efficacy of gamma-vinyl GABA (GVG) for the treatment of methamphetamine and/or cocaine addiction. Synapse 2005;55:122-5.
  63. Brodie JD, Case BG, Figueroa E, Dewey SL, Robinson JA, Wanderling JA, et al. Randomized, double-blind, placebo-controlled trial of vigabatrin for the treatment of cocaine dependence in Mexican parolees. Am J Psychiatry 2009;166:1269-77.
  64. Gerdeman GL, Partridge JG, Lupica CR, Lovinger DM. It could be habit forming: drugs of abuse and striatal synaptic plasticity. Trends Neurosci 2003;26:184-92.
  65. Lewis DJ. Psychobiology of active and inactive memory. Psychol Bull 1979;86:1054-83.
  66. Nader K. Memory traces unbound. Trends Neurosci 2003;26:65-72.
  67. Lee JL, Di Ciano P, Thomas KL, Everitt BJ. Disrupting reconsolidation of drug memories reduces cocaine-seeking behavior. Neuron 2005;47:795-801.
  68. Lee JL, Milton AL, Everitt BJ. Cue-induced cocaine seeking and relapse are reduced by disruption of drug memory reconsolidation. J Neurosci 2006;26:5881-7.
  69. Milton AL, Lee JL, Everitt BJ. Reconsolidation of appetitive memories for both natural and drug reinforcement is dependent on β-adrenergic receptors. Learn Mem 2008;15:88-92.
  70. Davidson C, Lee TH, Xiong Z, Ellinwood EH. Ondansetron given in the acute withdrawal from a repeated cocaine sensitization dosing regimen reverses the expression of sensitization and inhibits self- administration. Neuropsychopharmacology 2002;27:542-53.
  71. Davidson C, Lazarus C, Lee TH, Ellinwood EH. Ondansetron, given during the acute cocaine withdrawal, attenuates oral cocaine self- administration. Eur J Pharmacol 2004;503:99-102.
  72. Zhang X, Lee TH, Davidson C, Lazarus C, Wetsel WC, Ellinwood EH. Reversal of cocaine-induced behavioral sensitization and associated phosphorylation of the NR2B and GluR1 subunits of the NMDA and AMPA receptors. Neuropsychopharmacology 2007;32:377-87.
  73. Davidson C, Gopalan R, Ahn C, Chen Q, Mannelli P, Patkar AA, et al. Reduction in methamphetamine induced sensitization and reinstatement after combined pergolide plus ondansetron treatment during withdrawal. Eur J Pharmacol 2007;565:113-8.
  74. Lee TH, Szabo ST, Fowler JC, Mannelli P, Mangum OB, Beyer WF, et al. Pharmacologically-mediated reactivation and reconsolidation blockade of the psychostimulant-abuse circuit: a novel treatment strategy. Drug Alcohol Depend 2012;124:11-8.
  75. Martell BA, Mitchell E, Poling J, Gonsai K, Kosten TR. Vaccine pharmacotherapy for the treatment of cocaine dependence. Biol Psychiatry 2005;58:158-64.
  76. Kosten TA, Shen XY, Kinsey BM, Kosten TR, Orson FM. Attenuation of cocaine-induced locomotor activity in male and female mice by active immunization. Am J Addict 2014;23:604-7.
  77. Ramakrishnan M, Kinsey BM, Singh RA, Kosten TR, Orson FM. Hapten optimization for cocaine vaccine with improved cocaine recognition. Chem Biol Drug Des 2014;84:354-63.
  78. Kosten TR, Domingo CB, Shorter D, Orson F, Green C, Somoza E, et al. Vaccine for cocaine dependence: a randomized double-blind placebo-controlled efficacy trial. Drug Alcohol Depend 2014;140:42- 7.
  79. Lockner JW, Eubanks LM, Choi JL, Lively JM, Schlosburg JE, Collins KC, et al. Flagellin as carrier and adjuvant in cocaine vaccine development. Mol Pharm 2015;12:653-62.
  80. McKenna DJ. Clinical investigations of the therapeutic potential of ayahuasca: rationale and regulatory challenges. Pharmacol Ther 2004;102:111-29.
  81. Brierley DI, Davidson C. Developments in harmine pharmacology — implications for ayahuasca use and drug-dependence treatment. Prog Neuropsychopharmacol Biol Psychiatry 2012;39:263-72.
  82. Glick SD, Maisonneuve IM. Development of novel medications for drug addiction: The legacy of an African shrub. Ann N Y Acad Sci 2000;909:88-103.
  83. Thongsaard W, Marsden CA, Morris P, Prior M, Shah YB. Effect of Thunbergia laurifolia, a Thai natural product used to treat drug addiction, on cerebral activity detected by functional magnetic resonance imaging in the rat. Psychopharmacology 2005;180:752- 60.
  84. Lu L, Liu Y, Zhu W, Shi J, Liu Y, Ling W, et al. Traditional medicine in the treatment of drug addiction. Am J Drug Alcohol Abuse 2009;35:1- 11.
  85. Forray A, Sofuoglu M. Future pharmacological treatments for substance use disorder. Br J Clin Pharmacol 2014;77:382-400.
  86. Sofuoglu M, Carroll KM. Effects of galantamine on cocaine use in chronic cocaine users. Am J Addict 2011;20:302-3.
  87. Nunes EV, Levin FR. Treatment of depression in patients with alcohol or other drug dependence: a meta-analysis. JAMA 2004;291:1887-96.
  88. Pepper J, Hariz M, Zrinzo L. Deep brain stimulation versus anterior capsulotomy for obsessive-compulsive disorder: a review of the literature. J Neurosurg 2015;122:1028-37.
  89. van Westen M, Rietveld E, Figee M, Denys D. Clinical outcome and mechanisms of deep brain stimulation for obsessive-compulsive disorder. Curr Behav Neurosci Rep 2015;2:41-8.
  90. Noble EP. The DRD2 gene in psychiatric and neurological disorders and its phenotypes. Pharmacogenomics 2000;1:309-33.
  91. Liu HC, Lin SK, Liu SK, Chen SL, Hu CJ, Chang JG, et al. DAT polymorphism and diverse clinical manifestations in methamphetamine abusers. Psychiatr Genet 2004;14:33-7.
  92. Ballon N, Leroy S, Roy C, Bourdel MC, Olie JP, Charles-Nicolas A, et al. Polymorphisms Taql A of the DRD2, Ball of the DRD3, exon III repeat of the DRD4, and 3’ UTR VNTR of the DAT: association with childhood ADHD in male African-Caribbean cocaine dependents? Am J Med Genet B Neuropsychiatr Genet 2007;144B:1034-41.
  93. Zhou Y, Michelhaugh SK, Schmidt CJ, Liu JS, Bannon MJ, Lin Z. Ventral midbrain correlation between genetic variation and expression of the dopamine transporter gene in cocaine-abusing versus non- abusing subjects. Addict Biol 2014;19:122-31.
  94. Brousse G, Vospan F, Ksouda K, Bloch V, Peoc’h K, Laplanche JL, et al. Could the inter-individual variability in cocaine-induced psychotic effects influence the development of cocaine addiction? Towards a new pharmacogenetic approach to addictions. Med Hypotheses 2010;75:600-4.
  95. Kosten TR, Wu G, Huang W, Harding MJ, Hamon SC, Lappalainen J, et al. Pharmacogenetic randomized trial for cocaine abuse: disulfiram and dopamine β-hydroxylase. Biol Psychiatry 2013;73:219-24.
  96. Kalayasiri R, Verachai V, Gelenter J, Mutirangura A, Malison RT. Clinical features of methamphetamine-induced paranoia and preliminary genetic association with DBH-1021C-T in a Thai treatment cohort. Addiction 2014;109:965-76.
  97. Haerian BS. BDNF rs6265 polymorphism and drug addiction: a systematic review and meta-analysis. Pharmacogenomics 2013;14:2055-65.
  98. Hall FS, Drgonova J, Jain S, Uhl GR. Implications of genome wide association studies for addiction: are our a priori assumptions all wrong? Pharmacol Ther 2013;140:267-79.
  99. Bonoiu AC, Mahajan SD, Ding H, Roy I, Yong KT, Kumar R, et al. Nanotechnology approach for drug addiction therapy: gene silencing using delivery of gold nanorod–siRNA nanoplex in dopaminergic neurons. Proc Natl Acad Sci U S A 2009;106:5546-50.
  100. Touriño C, Eban-Rothschild A, de Lecea L. Optogenetics in psychiatric diseases. Curr Opin Neurobiol 2013;23:430-5.
  101. Cao ZF, Burdakov D, Samyai Z. Optogenetics: potentials for addiction research. Addict Biol 2011;16:519-31.
  102. Stamatakis AM, Stuber GD. Optogenetic strategies to dissect the neural circuits that underlie reward and addiction. Cold Spring Harb Perspect Med 2012;2(11):a011924.
  103. Britt JP, Bonci A. Optogenetic interrogations of the neural circuits underlying addiction. Curr Opin Neurobiol 2013;23:539-45.
  104. Alderson HL, Parkinson JA, Robbins TW, Everitt BJ. The effects of excitotoxic lesions of the nucleus accumbens core or shell regions on intravenous heroin self-administration in rats. Psychopharmacology (Berl) 2001;153:455-63.
  105. Hutcheson DM, Parkinson JA, Robbins TW, Everitt BJ. The effects of nucleus accumbens core and shell lesions on intravenous heroin self- administration and the acquisition of drug-seeking behaviour under a second-order schedule of heroin reinforcement. Psychopharmacology 2001;153:464-72.
  106. Gao G, Wang X, He S, Li W, Wang Q, Liang Q, et al. Clinical study for alleviating opiate drug psychological dependence by a method of ablating the nucleus accumbens with stereotactic surgery. Stereotact Funct Neurosurg 2003;81:96-104.
  107. Yang KJ, Long H, Yuan YW, Qi ST, Xu BT, Wang KW, et al. Stereotactic neurosurgical technique and electrophysiological study in ablating the ventromedial shell of the nucleus accumbens. Stereotact Funct Neurosurg 2014;92:37-43.
  108. Lévêque M, Durand E, Weil AG. Psychosurgery for drug addiction. Stereotact Funct Neurosurg 2014;92:195-6.
  109. Gao G, Wang X. Stereotactic neurosurgery for drug addiction. In: Sun B, Salles AD, editors. Neurosurgical treatments for psychiatric disorders. Netherlands, Springer; 2015:161-73.
  110. Naqvi NH, Rudrauf D, Damasio H, Bechara A. Damage to the insula disrupts addiction to cigarette smoking. Science 2007;315:531-4.
  111. Contreras M, Ceric F, Torrealba F. Inactivation of the interoceptive insula disrupts drug craving and malaise induced by lithium. Science 2007;318:655-8.
  112. Elkashef AM, Rawson RA, Anderson AL, Li SH, Holmes T, Smith EV, et al. Bupropion for the treatment of methamphetamine dependence. Neuropsychopharmacology 2008;33:1162-70.
View My Stats