Abstract
Background
Cocaine use contines to be a significant public health problem world-wide. However, despite substantial research efforts, no pharmacotherapies are approved for the treatment of cocaine use disorder (CUD).
Argument
Studies have identified positive signals for a range of medications for treating CUD. These include long-acting amphetamine formulations, modafinil, topiramate, doxazosin and combined topiramate and mixed amphetamine salts extended-release (MAS-ER). However, valid conclusions about a medication’s clinical efficacy require nuanced approaches that take into account behavioural phenotypes of the target population (frequency of use, co-abuse of cocaine and other substances, genetic subgroups, psychiatric comorbidity), variables related to the medication (dose, short-/long-acting formulations, titration speed, medication adherence) and other factors that may affect treatment outcomes. Meta-analyses frequently do not account for these co-varying factors, which contributes to a somewhat nihilistic view on pharmacotherapeutic options for CUD. In addition, the predominant focus on abstinence, which is difficult for most patients to achieve, may overshadow more nuanced therapeutic signals.
Conclusion
While there is an emphasis on finding new medications with novel mechanisms of action for treating CUD, currently available medications deserve further investigation based on the existing literature. Evaluating refined metrics of treatment success in well-defined subgroups of patients, and further exploring combination therapies and their synergy with behavioural/psychosocial interventions, are promising avenues to establishing effective therapies for CUD.
Keywords: Cocaine, combination therapies, dopamine agonists, dopamine antagonists, novel mechanisms, pharmacotherapy, positive signals, therapeutic nihilism, treatment
BACKGROUND
In 2017, the United Nations Office on Drugs and Crime (UNODC) reported an all-time high in the estimated global illicit manufacturing of cocaine [1]. In 2018, an estimated 18.1 million people world-wide used cocaine for recreational purposes [2]. Approximately 30% of these cocaine users (12 years or older) resided in the United States, including 874000 ‘new users’ and 977000 individuals diagnosed with cocaine use disorder (CUD) in the past year [3]. Nonetheless, only approximately 19% of people with CUD received treatment for this disorder [3]. In Europe, similar issues have evolved, with an estimated 3.9 million individuals between ages 15 and 64 reporting cocaine use and 73 000 individuals receiving treatment for CUD in 2017 [4].
Chronic cocaine use produces persistent changes in the vasculature that increase the likelihood of myocardial infarction, hypertension, atherosclerosis and stroke [5–9]. It also increases risks for various other health problems, including psychiatric disorders and sexually transmitted infections [10–12]. Psychiatric comorbidities and psychosocial factors, such as poverty, unemployment, homelessness, socio-economic status and legal issues, predict cocaine-related physical and mental health complications [13]. Almost all who seek treatment for CUD receive psychosocial interventions, but most will continue to use cocaine [14]. Pharmacotherapies may augment the effectiveness of psychosocial interventions, but no Food and Drug Administration (FDA)-approved medications for treating CUD are currently available.
Our views on the contribution of cocaine to drug overdoses have undergone a rapid shift. In 2017, a reported 52% of all fatal drug overdoses in the United States involved cocaine (n = 70237) [15]. While adulteration with synthetic opioids, such as fentanyl, may contribute to growing overdose rates [16], recent data indicate that one-quarter of cocaine overdose deaths were without any opioid involvement [15]. In Europe, stimulant overdoses account for a smaller proportion of drug-related deaths, but these rates vary widely by country [4].
PHARMACOTHERAPEUTIC STRATEGIES TO THE TREATMENT OF CUD—WHERE ARE WE?
Substantial efforts have been devoted to identifying medications that could augment the effectiveness of CUD treatments, with several medications having shown some promise. However, a looming sense of nihilism is pervasive—why?
In this paper, we (1) discuss and examine the pharmacological approaches that have thus far been tested for CUD treatment, (2) discuss a number of major issues with prior research constraining our opportunities to find an efficacious medication, (3) highlight potential avenues to pursue and offer critical considerations for future study and (4) encourage our colleagues to persist in their investigative efforts.
PHARMACOLOGY OF COCAINE
Cocaine stimulates the mesolimbic dopamine system—the brain’s reward pathway [17]. Cocaine effects are produced through binding to, and inhibiting the function of, monoamine transporters for dopamine (DA), serotonin (5-HT) and norepinephrine (NE) [18]. These transporters inhibit neuronal communication by facilitating uptake of neurotransmitters from the extracellular space (i.e. the synapse) back into neighbouring neurones. DA accumulates, resulting in stronger and prolonged signalling with the pre- and post-synaptic neurone. This putatively produces the psychomotor stimulant effects [19] and contributes to the pleasurable effects of cocaine (e.g. euphoria). Despite the central role of DA, other neurotransmitter systems (NE, 5-HT) reflect viable targets for modulation with an impact on cocaine sensitization, craving and reinstatement [20].
CURRENT APPROACHES TO TREATING CUD
Pharmacological treatments for CUD to date form four distinct categories: agonists, antagonists/blockers, novel mechanisms and combination pharmacotherapies. Here, we discuss studies showing promising pharmacotherapeutic signals for CUD and examine potential reasons for why some findings have not been replicated. Table 1 depicts characteristics of the clinical trials that are discussed.
Table 1.
Characteristics of clinical trials discussed.
Author, year | Primary disorder | Other SUDs | Other psychiatric conditions | Primary medication used | Other medications | Maximum dose (mg) | Sample size r andomized (n) | Completers (n) | Follow-up period (weeks) |
---|---|---|---|---|---|---|---|---|---|
Dopamine releasers (median sample size randomized: 101; completers: 34) | |||||||||
Grabowski, 2004 [21] | Cocaine | Opioid | None | D-Amp | Methadone | 60 | 120 | 34 | 26 |
Grabowski, 2001 [22] | Cocaine | No | None | D-Amp | None | 60 | 128 | _a | 12 |
Shearer, 2003 [23] | Cocaine | Opioid | None | D-Amp | None | 60 | 30 | 11 | 14 |
Mooney, 2009 [24] | Cocaine | No | None | Meth | None | 30 | 82 | 25 | 8 |
Nuijten, 2016 [25] | Crack cocaine | No | None | D-Amp | Methadone/diacethylmorphine | 60 | 73 | 65 | 12 |
Levin, 2015 [26] | Cocaine | No | ADHD | MAS-ER | None | 60/80 | 123 | 93 | 13 |
Dopamine uptake inhibitors (median sample size randomized: 96; completers: 59) | |||||||||
Anderson, 2009 [27] | Cocaine | No | None | Modafinil | None | 400 | 210 | 125 | 12 |
Dackis, 2012 [28] | Cocaine | No | None | Modafinil | None | 200/400 | 210 | 120 | 8 |
Kampman, 2015 [29] | Cocaine | No | None | Modafinil | None | 300 | 94 | 71 | 8 |
Schmitz, 2012 [30] | Cocaine | No | None | Modafinil/D-AMP | None | 400/60 | 73 | 15 | 16 |
Schmitz, 2014 [31] | Cocaine | No | None | Modafinil | Levodopa/Carbidopa, Naltrexone | 400 | 81 | 40 | 12 |
Levin, 2007 [32] | Cocaine | Opioid | ADHD | Methylphenidate | Methadone | 80 | 98 | 47 | 12 |
Margolin, 1995 [33] | Opioid | Cocaine | None | Bupropion | Methadone | 300 | 149 | 125 | 12 |
Shoptaw, 2008 [34] | Cocaine | No | None | Bupropion | None | 300 | 70 | 12 | 16 |
Cocaine vaccine (median sample size randomized: 212; completers: 110) | |||||||||
Martell, 2009 [35] | Cocaine | Opioid | None | TA-DC | Methadone | 5 vaccinations | 115 | 94 | 24 |
Kosten, 2014 [36] | Cocaine | No | None | TA-DC | None | 5 vaccinations | 310 | 126 | 24 |
GABA modulators (median sample size randomized: 103; completers: 58) | |||||||||
Shoptaw, 2003 [37] | Cocaine | No | None | Baclofen | None | 60 | 70 | 17 | 16 |
Brodie, 2009 [38] | Cocaine | No | None | Vigabatrin | None | 3 | 103 | 52 | 9 |
Kampman, 2004 [39] | Cocaine | No | None | Topiramate | None | 200 | 40 | 34 | 13 |
Johnson, 2013 [40] | Cocaine | Alcohol | None | Topiramate | None | 300 | 142 | 72 | 12 |
Kampman, 2013 [41] | Cocaine | Alcohol | None | Topiramate | None | 300 | 170 | 100 | 13 |
Umbricht, 2014 [42] | Cocaine | Opioid | None | Topiramate | Methadone | 300 | 171 | 113 | 12 |
Nuijten, 2014 [43] | Crackccocaine | Opioid | None | Topiramate | None | 200 | 74 | 58 | 12 |
Noradrenergic agents (median sample size randomized: 98.5; completers: 58) | |||||||||
Shorter, 2013 [44] | Cocaine | No | None | Doxazosin | None | 8 | 35 | 17 | 13 |
Zhang, 2019 [45] | Cocaine | No | None | Doxazosin | None | 8 | 76 | 55 | 12 |
Carroll, 2004 [46] | Cocaine | No | None | Disulfiram | None | 250 | 121 | 53 | 12 |
Schottenfeld, 2014 [47] | Cocaine | Opioid | None | Disulfiram | Buprenorphine | 250 | 177 | 92 | 12 |
Oliveto, 2011 [48] | Cocaine | Opioid | None | Disulfiram | Methadone | 250 | 161 | 110 | 14 |
Kosten, 2013 [49] | Cocaine | Opioid | None | Disulfiram | Methadone | 250 | 74 | 61 | 12 |
Antipsychotics (median sample size randomized: 35; dropout rate: 49.3%)b | |||||||||
Beresford, 2017 [50] | Schizophrenia | Cocaine | None | Aripiprazole | Perphenazine | 30 | 44 | 44 | 8 |
Novel mechanisms (median sample size randomized: 93; completers: 58) | |||||||||
Carroll, 2018 [51] | Cocaine | Opioid | None | Galantamine | Methadone | 8 | 120 | 92 | 12 |
DeVito, 2019 [52] | Cocaine | No | None | Galantamine | None | 16 | 93 | 58 | 13 |
Dakwar, 2019 [53] | Cocaine | No | None | Ketamine | Midazolam | 0.5/kg | 55 | 32 | 5 |
Combination pharmacotherapy (median sample size randomized: 104; completers: 71.5) | |||||||||
Kablinger, 2014 [54] | Cocaine | Alcohol | None | Metyrapone/oxazepam | None | 1500/20 | 45 | 22 | 7 |
Pettinati, 2008 [55] | Cocaine | Alcohol | None | Disulfiram/naltrexone | None | 250/100 | 208 | 95c | 13 |
Mariani, 2012 [56] | Cocaine | No | None | MAS-ER/topiramate | None | 60/150 | 81 | 64 | 12 |
Levin, 2012 [56] | Cocaine | No | None | MAS-ER/topiramate | None | 60/100 | 127 | 79 | 14 |
Medications used to augment behavioural treatments (median sample size randomized: 133; completers: 66) | |||||||||
Kosten, 2003 [57] | Cocaine | Opioid | None | Desipramine | Buprenorphine | 150 | 160 | 78 | 12 |
Schmitz, 2008 [58] | Cocaine | No | None | Levodopa/carbidopa | None | 400/100 | 161 | 66 | 12 |
Poling, 2006 [59] | Opioid | Cocaine | None | Bupropion | Methadone | 300 | 106 | 62 | 25 |
Moeller, 2007 [60] | Cocaine | No | None | Citalopram | None | 20 | 76 | _d | 12 |
D-Amp = dextro-amphetamine; MAS-ER = extended release mixed amphetamine salts; SUD = substance use disorder; TA-DC = anti-cocaine vaccine.
Number of completers not reported (study completion rates for placebo, 15–30 mg and 30–60 mg groups were 22.9, 40.4 and 8.7%, respectively [22]).
The median sample size randomized refers to the 14 studies included in a Cochrane review [61] plus the additional randomized controlled trial (RCT) testing aripiprazole [50]. The dropout rate was derived from studies included in the Cochrane review that reported dropout rate as an outcome.
Number of completers not reported (45.8% of patients who took 80% of their pills while in treatment [55]).
Number of completers not reported (overall, subjects remained in treatment for an average of 4.6 ± 4.4 weeks [60]).
While we do not make any claim to completeness regarding the literature analyzed for this opinion piece, we reviewed all obtainable systematic reviews and meta-analyses on pharmacological treatments for CUD. A recent systematic review of reviews was used to identify those published until November 2019 [62]. A separate systematic search of peer-reviewed articles published between November 2019 and July 2020 was conducted via Pubmed, EMBASE and the Cochrane Database, using the same keyword search terms as those of the aforementioned systematic review of reviews. This search generated one additional systematic review evaluating cannabidiol in the treatment of CUD [63]. Studies of cannabidiol currently remain in the pre-clinical phase, except for one ongoing human trial (NCT02559167; results not yet posted). In addition, we refrained from including a detailed discussion of antidepressants, even though this is the most widely studied drug class for the treatment of CUD. However, findings from three separate systematic reviews, which included 37 (total N = 2891 [64]), 10 (n = 1226 [65]) and 19 (n = 1180 [66]) randomized controlled trials (RCTs), respectively, consistently showed that antidepressants had negligible effects on CUD [62], except for a potential monoamine augmentation of contingency management (CM) treatment [57,60].
An overarching problem with identifying efficacious CUD treatments is that a vast number of different medications have been tested but only a few studies have investigated each individual compound, and fewer still have adequately sized samples (Table 1). Such scarcity of data raises risks for overestimating the therapeutic potential of a medication or prematurely dismissing one with veridical utility. Further, pharmaceutical drug development typically follows a rigorous process of translating pre-clinical studies with animals to human laboratory experiments to human clinical trials. However, a strikingly small percentage of drugs tested in RCTs as treatments for CUD have previously undergone self-administration investigations in both non-human primates and humans [67]. This is somewhat disturbing, given that self-administration models provide the most direct point-to-point correspondence with addictive behaviour in the ‘real world’ [68–70] and offer potentially invaluable insights at low costs. The therapeutic potential of an intervention demonstrated under controlled laboratory models forms the necessary basis from which to pursue future costly clinical trials, specifically those of appropriate scale for detecting efficacious CUD treatments.
Dopamine agonists
Agonist medications share pharmacodynamic mechanisms of action with the illicitly used drug, but typically have distinct pharmacokinetic qualities (e.g. enteral bio-availability, slower onset of action, longer duration of action) [21,71]. Agonist treatment utilizes a substitution approach to replace (or displace) the illicit drug for the purpose of stabilizing functioning. For example, methadone has been a highly effective agonist substitution method for managing opioid use disorder (OUD).
Although stimulants acutely increase available monoamines [DA/noradrenaline (NA)/5HT] [19,72,73], chronic users exhibit blunted monoaminergic functioning (low baseline DA, blunted DA release, low D2/D3 receptor availability). Stimulant-like agonists may help to reduce DA hypoactivity via tonic DA regulation [74] and attenuate the phasic DA responses promoting drug-seeking [75,76]. Moreover, agonist treatments alleviate the intensity of drug cravings and withdrawal symptoms that can contribute to relapse [77,78]. Other benefits of the agonist approach include familiarity with drug effects that may promote medication compliance [79–81].
One concern commonly put forth against agonist approaches is the potential for secondary abuse, given their subjective stimulant-like qualities. However, evidence thus far is relatively weak for abuse liability and cardiovascular risk of agonist medications, including amphetamine in cocaine users [82–85]. While carefully weighing therapeutic benefits against potential risks is undoubtedly important, a lack of (political) willingness to address the necessary regulatory and implementation procedures should not prevent the study of agonist medications or the clinical use of drugs that have proved efficacious. Another concern is the prolonged blunting of DA systems with stimulant maintenance. Evidence for the potential adverse effects associated with long-term stimulant medication use remain unclear, due to a lack of longitudinal studies [86]. However, preliminary evidence from non-human primates suggests that chronic exposure to long-acting stimulant medications does not produce aberrant DA functioning [87].
Dopamine releasers
The most promising signals to date have been obtained with DA releasers, specifically with amphetamine maintenance [88,89]. Dextro-amphetamine (D-amp) yields similar efficacy in decreasing cocaine choice in monkeys as in humans [90]. Under controlled laboratory settings, D-amp maintenance reduced cocaine self-administration in cocaine-dependent participants with comorbid OUD (maintained on buprenorphine) [91] and those without the comorbid disorder [92].
Clinical trials have produced similar results showing that D-amp reduces cocaine use [22,23,82]. One study testing multi-stage dosing showed greatest treatment retention at 15–30 mg, and the lowest total percentage of cocaine-positive urine screens at 30–60 mg [22]. With immediate release (IR) methamphetamine (5 mg, 6×/day) and sustained-release (SR) methamphetamine (30 mg first dose, then 5× placebos), individuals who received the SR formulation had fewer cocaine-positive urine samples and greater reduction in cocaine craving [24]. These effects were attributed to overall higher medication adherence for the first dose of the day (95%), but lower for subsequent capsules. Moreover, cocaine-dependent patients in heroin-assisted treatment who were given 60 mg SR D-Amp exhibited fewer days of cocaine use compared to those treated with placebo [25]. These findings highlight the potential benefits of D-amp in CUD treatment, especially in the context of good medication adherence.
CUD frequently co-occurs with attention deficit hyperactivity disorder (ADHD), and cocaine use may be reflective of attempts to self-medicate [93]. A multi-site clinical trial evaluating the effects of extended-release mixed amphetamine salts (MAS-ER; 60 or 80 mg/day versus placebo) in patients with ADHD and comorbid CUD indicated that the higher dose, combined with cognitive behavioural therapy (CBT), reduced symptoms for both conditions [26].
Dopamine uptake inhibitors
A more recently tested medication with DA-transporter inhibitory properties is modafinil, a cognitive enhancer and wake-promoting agent that binds to the DA transporter, preventing DA re-uptake [94]. Modafinil is well tolerated [95,96] and does not alter acute cardiovascular effects in combination with cocaine or potentiate cocaine-induced euphoria [97]. Human experimental trials showed conflicting results regarding modafinil’s effect on cocaine self-administration [98,99]; however, they differed by the magnitude of presented reinforcers (e.g. choose $1 or 0–20 mg cocaine versus $5 or 0–50 mg cocaine). Reinforcer magnitude has been shown to influence cocaine choice in the context of modafinil maintenance [100], where cocaine choice decreased only when both reward values and response effort demands were high.
In a meta-analysis of 11 RCTs comparing therapeutic outcomes of modafinil versus placebo, seven studies did not provide evidence for superiority of modafinil over placebo in sustaining cocaine abstinence [101]. However, post-hoc analyses from one study revealed that while modafinil maintenance had no effect on cocaine abstinence, those without a history of alcohol use disorder (AUD) exhibited increased percentage of days abstinent by week [27]. In support of these results, individuals with CUD but not AUD who were treated with modafinil were significantly more likely to abstain from cocaine use during the last 3 weeks of the trial than those who received placebo [29].
While these results seem promising, findings of modafinil effects on CUD independent of AUD have not been replicated. While high dropout rates (33–60%) [30,31] and poor medication adherence may have limited conclusions about the effectiveness of modafinil, one study noted some important sex differences [28]. Men treated with the higher modafinil dose (400 mg/day) were more likely to remain abstinent relative to their placebo-treated counterparts, while women treated with placebo had the highest rates of abstinence in the female sample.
Results comparing human laboratory experiments and clinical trials provide a concordance of data supporting the efficacy of methylphenidate (60 mg/day) for treating CUD in individuals with ADHD [32,102]. However, subsequent RCTs investigating methylphenidate as an agonist replacement therapy in participants with CUD, both with and without comorbid ADHD, revealed mainly negative results. It is possible that insufficient dosage strengths may have contributed to the discrepant results [89].
In summary, even though systematic reviews of psychostimulants conclude that there is insufficient evidence to either support or discount their effectiveness for CUD [65,101,103–105], positive signals have been identified in studies testing dopamine agonist treatments, particularly for long-acting amphetamine formulations at sufficiently high doses and modafinil when medication adherence is ensured.
Antagonist/blocker approaches
In general, antagonists are thought to block the euphoric effects of cocaine and facilitate the decrease in cocaine use through extinction. Antagonist or blocker approaches are generally less effective in the treatment of substance use disorders (SUDs) compared to agonists because they require high levels of motivation at treatment initiation and maintenance; nonetheless, their efficacy can be high. A potential safety concern is compensatory drug use to ‘over-ride’ the blockade, especially when it is not a complete blockade. However, in practice these attempts are typically modest, because patients report that they may try the drug of abuse but will not waste their money if the effects are undesirable [106].
Antipsychotics
A Cochrane Review concluded that there is no evidence supporting the use of antipsychotic medications that block DA receptors to treat CUD [61]. However, the results came from only 14 trials with small sample sizes (median sample size of the 14 RCTs included: 33) and moderate to low-quality evidence. In addition, antipsychotics may require an acclimation period to take effect, as exemplified by an RCT testing aripiprazole. In participants with schizophrenia and comorbid CUD, the effect of aripiprazole on craving appeared at week 6 of treatment [50].
Cocaine vaccine
An anti-cocaine vaccine (e.g. TA-CD) is one of several novel approaches utilizing an immunological mechanism of action for the treatment of SUDs. TA-CD, composed of a cocaine hapten conjugated to inactivated cholera toxin B, increases production of antibodies that target the cocaine molecule. The antibodies bind to cocaine in the blood and, because the antigen–antibody complexes are too large to cross the blood–brain barrier, prevent cocaine from entering the brain [107,108].
A human laboratory study found that peak plasma antibody levels, which were highly variable between subjects, predicted cocaine’s effects in non-treatment-seeking cocaine-dependent participants [106]. Individuals producing the highest antibody titres had the greatest reductions in positive drug effect ratings and perceived cocaine quality. In addition, self-reported cocaine use showed a trend to decrease as a function of antibody titre.
Results from clinical trials for vaccines are inconsistent. A study of TA-CD administered to methadone-maintained individuals indicated that vaccinated participants who attained high immunoglobulin (Ig)G levels (≥ 43 μg/ml; 38% of vaccinated participants) had more cocaine-free urine samples than those with low IgG levels and those with placebo, during study weeks 9–16 [35]. In addition, the proportion of participants with a 50% reduction in cocaine use was significantly greater in the high IgG level group (53%) than the low IgG level group (23%). These results are promising, but they could not be fully replicated in individuals without comorbid OUD [36]. After week 8, more vaccinated than placebo participants attained abstinence for at least 2 weeks of the trial (24 versus 18%) and the high IgG group (67% vaccinated participants) had the most cocaine-negative urines during the last 2 weeks of treatment [odds ratio (OR) = 3.02]; however, neither result was statistically significant. Notably, almost 3× fewer high than low IgG participants dropped out of the study. The authors recommended for future studies: (1) a more structured setting to increase participant motivation for abstinence, (2) a shift in focus from abstinence initiation to relapse prevention and (3) greater integration of CBT to facilitate sustained abstinence [36].
GABA modulators
Several pre-clinical studies support the potential efficacy of GABAergic medications for the treatment of CUD. GABA is an important modulator of the mesolimbic reward system [109–112], and medications that increase GABAergic activity such as vigabatrin and baclofen have been shown to reduce cocaine self-administration in animal models. Although clinical trials to date have not demonstrated efficacy for baclofen [37], positive outcomes have been found for vigabatrin in a study that ensured high medication adherence [38].
Topiramate, an anticonvulsant with multiple mechanisms of action (Na+ and Ca++ channel blockade; carbonic anhydrase inhibition; GABA potentiation; glutamate antagonism) initially showed compelling evidence for efficacy in CUD treatment. Participants with CUD, both those abstinent [39] or using at baseline [40], showed increased abstinence rates with topiramate treatment relative to placebo. Because rapid dose titration of topiramate can result in uncomfortable central nervous system side effects, a slow titration is required to achieve target dose levels [39]. In individuals with comorbid CUD and AUD, topiramate produced longer periods of cocaine abstinence, even in the absence of differential weekly abstinence rates between those treated with topiramate versus placebo [41]. Among methadone-maintained patients, topiramate did not show superiority over placebo in sustaining cocaine abstinence, regardless of whether or not participants received incentives for drug abstinence [42]. In an open-label trial assessing topiramate as an adjunct to CBT, cocaine smokers reported no reduction in cocaine use, and only 14% of participants took topiramate for 11 of 12 weeks [43]. Negative findings may be attributed to poor medication adherence and discrepancies in dosages used across studies [40,41]. Nonetheless, post-hoc exploratory analyses showed reduced cocaine use in individuals with comorbid CUD and OUD treated with topiramate [43].
Noradrenergic agents
Doxazosin is a long-acting selective α−1 adrenergic antagonist that reduces central noradrenergic activity. Doxazosin has been found to limit the behavioural effects of stimulants, including amphetamine and cocaine [113–115], and attenuate cocaine-induced reinstatement of cocaine-seeking behaviour in rats [116,117]. A promising pilot study revealed that a rapid titration schedule of doxazosin (8 mg/day over 4 weeks) led to more cocaine-negative urines than slower titration (8 mg/day over 8 weeks) or placebo (35, 10 and 14% negative urines, respectively) [44].
In a recent trial, doxazosin-treated (8 mg/day) individuals exhibited a greater reduction in cocaine use relative to those treated with placebo [45]. Genetic subgroup analysis further indicated that the percentage of cocaine-positive urine toxicology screens for doxazosin-treated individuals was lower in the group with lower DA beta-hydroxylase (DβH) levels from T-allele carriers (CT or TT) than the group with higher DβH levels from the DβH CC genotype. This finding suggests that doxazosin may be more effective at blocking NE stimulation when the threshold for NE release is lower (CT/TT group) compared to when it is higher (CC group). A follow-up study by the same group explored pharmacogenetic response to doxazosin treatment based on an alpha-1 adrenoreceptor subtype D (ADRA1D) genetic variant [118]. Given that T-allele carriers with the ADRA1D SNP (T1848A) treated with doxazosin had a greater reduction in cocaine use, this polymorphism constitutes a potential pharmacogenetic marker in pharmacotherapy for CUD [118].
A complex picture of mixed findings is apparent with disulfiram, a copper chelator that acts upon multiple enzymes including DβH, which results in an inhibition of norepinephrine synthesis [119]. For example, disulfiram treatment (versus placebo) in combination with CBT was associated with reduced cocaine use [46]. These effects were most pronounced for participants without AUD at baseline and those who fully abstained from drinking during treatment. Further, buprenorphine-treated participants reported reduced cocaine use following disulfiram treatment [47], with no difference in number of collected cocaine-negative urine samples or consecutive weeks abstinent between disulfiram- and placebo-treated individuals. Another study with methadone-maintained participants compared three disulfiram doses and placebo in combination with CBT. Cocaine-positive urine samples increased over time with the lower two dosages (62.5 or 125 mg/day) but decreased in the 250 mg group [48]. However, there was no difference between the 250 mg disulfiram group and the placebo group.
A pharmacogenomic study found that disulfiram reduced cocaine-positive urines in methadone-stabilized participants by 18% during the course of the 10-week study, and this effect varied as a function of genotype groups [49]. Participants with the normal DβH level genotype showed a reduction by 28% on disulfiram, whereas no effects were found in those with low DβH level genotype. Moreover, men treated with disulfiram sustained a greater number of days abstinent and percentage of drug-free urine samples than those with placebo, whereas women had a moderate outcome regardless of treatment arm [120].
An early systematic review found insufficient evidence to either support or discount the effectiveness of disulfiram for CUD [121], and a recent meta-analysis of seven RCTs found worse retention rates for disulfiram than placebo (relative risk 0.90) [65]. The mechanisms by which disulfiram exerts its effects on cocaine-related behaviours remain unclear. This, and side effects reported in some studies (e.g. hepatotoxicity) [122], may limit its potential use.
Novel mechanisms—tested in humans
Galantamine is a cholinesterase inhibitor that improves concentration and attention. A recent RCT tested galantamine against placebo in the context of standard methadone treatment and computerized CBT as treatments for CUD [51]. Results indicated a reduction in cocaine use frequency over time, with galantamine superior to placebo and CBT superior to methadone alone. There was no evidence of an additive or synergistic effect of combined galantamine and CBT on cocaine use. In a separate RCT that excluded those with comorbid SUDs, neither galantamine dose (8 or 16 mg/day) improved cocaine-use outcomes [52].
Ketamine is a potent N-methyl-D-aspartate receptor (NMDAR) antagonist that is effective in the treatment of severe depression [123]. Ketamine is hypothesized to extend beyond NMDAR modulation to downstream effects on other neurotransmitter systems and pre-frontal synaptogenesis. Subanaesthetic ketamine infusions (0.41 mg/kg) increased motivation to quit cocaine use and reduced cue-induced cocaine craving relative to those who received active control (2 mg lorazepam), and higher dose infusions (0.71 mg/kg) produced even further reductions in cravings [124]. A separate study using the same higher ketamine dose yielded decreased cocaine self-administration 28 hours post-infusion compared to the control condition (0.025 mg/kg midazolam) [125]. Furthermore, ketamine increased self-reported distress tolerance 48 hours post-infusion relative to controls. By creating a reprieve from reactivity to distress, ketamine treatment may help individuals with CUD to access and experience the full benefit of behavioural interventions.
The first clinical trial with ketamine involving individuals with CUD undergoing mindfulness-based relapse prevention showed that a single subanaesthetic ketamine infusion (0.5 mg/kg) was capable of increasing cocaine abstinence relative to an active control (midazolam) [53]. Thirteen ketamine-treated participants remained abstinent during the final 2 weeks of the trial, while only three controls managed to do the same. The study further reported that those in the ketamine group were less likely to drop out of treatment or relapse to cocaine use, and they reported less craving for cocaine.
Combination approaches
Combination pharmacotherapy
Combining pharmacotherapeutic approaches for the treatment of CUD is a viable strategy for a number of reasons: use of lower doses of individual constituents may minimize side effects, additive or synergistic effects may be achieved with combinations and the diverse neurotransmitter systems impacted by stimulant drugs can be targeted [126]. Nonetheless, given that there are no medications with a broad signal for CUD, combining two medications with weak (or no) signals may increase the occurrence of adverse events and thus unfavourably impact the risk: benefit ratio. Another issue of assessing combination pharmacotherapy in clinical trials is that very few individual constituents have been tested against combination approaches. Thus, the superiority of combination over monotherapy remains to be determined.
Thus far, combination approaches that have shown some promise in increasing cocaine abstinence in clinical trials include metyrapone/oxazepam, disulfiram/naltrexone and topiramate/MAS-ER treatment. While there were no treatment group differences in overall analyses for metyrapone/oxazepam [54] and disulfiram/naltrexone treatment [55], subgroup analyses indicated that disulfiram maintenance, both alone and in combination with naltrexone, increased abstinence from cocaine and alcohol, and longer abstinence periods were observed in the high-dose metyrapone/oxazepam group (1500 mg metyrapone/20 mg oxazepam) compared to placebo. The significance of the latter result is attenuated, however, by an overall low retention rate of randomized subjects (22 of 45).
In other studies, combination MAS-ER/topiramate doubled the rate of participants achieving three consecutive weeks of cocaine abstinence (33.3%) relative to placebo (16.7%) [56]. Exploratory post-hoc analyses revealed that MAS-ER/topiramate was more effective for participants who had a greater frequency of cocaine use at baseline. A larger follow-up investigation of MAS-ER/topiramate in heavy, frequent cocaine users showed even greater differences between treatment versus control groups for cocaine abstinence both during the trial (21.9 versus 6%) and ≥ 3 consecutive weeks at the end of the trial (14 versus 0%) [127].
Medications used to augment behavioural treatments
CM is a powerful behavioural technique that uses reinforcers to elicit behaviour change [128]. There is sufficient evidence from reviews to support the effectiveness of CM for CUD [129–133]. The evidence is limited, however, for the efficacy of CBT in the treatment of CUD. CBT offers the benefit of reduced dropout rates, but shows little impact on the maintenance of cocaine abstinence [132].
Several agonists have been tested in conjunction with CM. One RCT compared desipramine (150 mg/day) and placebo in buprenorphine-maintained cocaine users, in addition to either CM or a non-contingent voucher control. Results indicated both independent and additive effects from combined CM/desipramine [57]. Moreover, those who underwent both active treatments returned more drug-free urines (50% of total urines) than all other treatment groups (25–29%). CM and desipramine alone, however, revealed a rapidly increasing drug–urine count across time. Sustained-release levodopa/carbidopa (400/100 mg b.i.d.) versus placebo was also assessed alongside either: (1) clinical management alone (ClinMan), (2) ClinMan and CBT or (3) ClinMan + CBT + CM. Levodopa was found to be superior in increasing cocaine-negative urines and consecutive cocaine abstinence relative to placebo, but only in the context of CM involvement [58]. In methadone-maintained individuals, bupropion, an atypical antidepressant with stimulant properties, combined with CM decreased cocaine-positive urine samples during weeks 3–13 with rates remaining stable in the weeks following [59]. CM alone decreased cocaine use gradually from weeks 14 to 25, while bupropion alone, voucher control and placebo yielded no improvements. Lastly, citalopram (20 mg/day) alongside both CBT and CM produced fewer cocaine-positive urine samples relative to placebo in combination with a similar behavioural regimen [60].
MANY POSITIVE SIGNALS—WHAT HAVE WE BEEN MISSING?
The evidence above suggests that specific pharmacological approaches, either alone or in combination with other interventions, may vary as a function of specific subject-related factors (e.g. biopsychosocial traits and status), the pharmacodynamics of the candidate compound or combination medicine and the interactions of these variables. Thus, future investigation of candidate medications may call for careful consideration of target (sub)populations as well as procedural designs that not only optimize dose response but also medication tolerability and adherence.
Procedural and subject-related factors
Table 2 provides an overview of procedural and subject-related factors that have been shown to impact medication effectiveness. Of note, in the CUD treatment literature, there is a heavy focus upon samples of cocaine users who receive medications for OUD. While the apparent aim is clinically relevant in addressing the high percentage of opioid-dependent individuals who use cocaine, findings are not likely to replicate across other subgroups of cocaine users. This is seen in prior divergent findings with the cocaine vaccine TA-CD [35,36] and galantamine [51,52]. Furthermore, attention to medication adherence is highly inconsistent throughout clinical trials or inadequate (e.g. counting pills [55]). One possible solution to this problem is the employment of biomarkers to track medication exposure. Studies also suffered from poor participant retention rates (see Table 1). The precise reasons for dropout are often unknown or unreported, but this information may prove invaluable to determining better tolerated treatment trial parameters.
Table 2.
Procedural and subject-related factors that may impact treatment effectiveness.
Subject-related factors | |
---|---|
Cocaine use severity | Heavier cocaine users may benefit from MAS-ER/topiramate combination [56,127] |
Comorbid substance use disorders | Comorbid alcohol use disorder may interfere with the benefits of modafinil [27,29] and disulfiram [46] treatment, while topiramate [41] or combination treatment with disulfiram/naltrexone [55] seem well-suited for the treatment of CUD patients who are alcohol-dependent. Topiramate [43] and galantamine [51] seem to be viable options for the treatment of patients with comorbid CUD and OUD |
Co-occurring attention deficit hyperactivity disorder (ADHD) | In patients with co-occurring CUD and ADHD, methylphenidate [32,102] or MAS-ER at robust doses [26] may be the pharmacotherapies of choice to treat symptoms arising from the overlapping neurophysiological deficits |
Sex | Men seem to benefit more from disulfiram [120] and modafinil [28] treatment than women |
Genetic subgroups | Those with normal DβH level genotype may benefit most from disulfiram [49], and those with lower DβH level from doxazosin [45]. In addition, efficacy of doxazosin treatment is highly correlated with the ADRA1D T-allele [118] |
Procedural factors | |
Medication dose | The importance of sufficient medication dose has been highlighted in several trials; e.g. for dextro-amphetamine [22], disulfiram [48], metyrapone [54], topiramate [43] and methylphenidate [32,102] |
Titration schedule | Studies indicate the benefits of slow titration of topiramate to avoid uncomfortable side effects [39] but superiority of fast titration of doxazosin [44] |
Medication adherence | Medication adherence impacts medication effectiveness, which is evident in trials that failed to replicate positive findings for modafinil [28], topiramate [43] or vigabatrin [134] |
Incentive structure | Satisfactory medication adherence (60–70%) [39–41] may be achieved by incentive-based interventions |
Release formulation | ER formulations (taken once a day) show greater adherence compared to IR (dosing multiple times per day) [24], in addition to having lower potential for misuse compared to IR formulations [135–137] |
CUD = cocaine use disorder; OUD = opioid use disorder; DβH = dopamine bseta-hydroxylase; ER = extended release; IR = immediate release; MAS = mixed amphetamine salts.
The pharmacological mechanisms of medications impact treatment outcome differentially across stimulant drugs. Despite many similarities [19,138], promising signals for cocaine do not generalize to other psychostimulants [139]. Studies suggest that DA re-uptake inhibitors (e.g. methylphenidate [140,141]) are more effective for reducing use of DA releasers (e.g. amphetamines), and DA releasers (e.g. amphetamine isomers [22,24,142,143]) are more effective for reducing use of cocaine, a DA re-uptake inhibitor. Thus, pharmacotherapy formulation requires tailoring to the primary drug used as well as to the individualized needs of the patient. To achieve the latter, a combination approach may be beneficial, such as targeting drug withdrawal and cravings and allowing patients to benefit more from behavioural/psychosocial interventions [20].
The issues that may arise from discounting subject-related and procedural factors, when assessing a medication’s clinical utility, are perhaps best depicted in the discrepancy of outcomes between two meta-analyses. A Cochrane Review of 26 RCTs testing nine different psychostimulants as potential CUD interventions (bupropion, D-Amp, lisdexamfetamine, methylphenidate, modafinil, mazindol, methamphetamine, MAS and selegiline) yielded weak evidence to support their viability in facilitating cocaine abstinence [risk ratio (RR) = 1.36), with no overall reduction in cocaine use [103]. However, the number of studies per candidate medication is too limited to provide adequate representation. The pitfall of pooling studies of disparate designs, end-points and sub-populations together, and treating them as though they were homogeneous, is failing to see what the proverbial apple offers that an orange does not. Indeed, a more recent meta-analytical review, specifying only the inclusion of psychostimulants with sufficient dopaminergic potency [modafinil, methylphenidate or prescription amphetamines (MAS, lisdexamphetamine and D-amp)], concluded that the evidence was robust in supporting CUD treatment with psychostimulants. Results were indicative of increased rates of abstinence (RR= 1.45), and prescription amphetamines were efficacious in promoting abstinence (RR = 2.41) [144]. Although the researchers acknowledged that a broad range of factors, including medication dose, ADHD status and concurrent OUD underpins some degree of heterogeneity across trials [144], such sharp contrasts in findings potentially lead to highly divergent paths in both research and clinical development.
Medication expectations: definition and measurement of meaningful end-points
Sustained abstinence, most commonly determined with qualitative urine screens, is the gold standard for determining treatment success in clinical trials evaluating pharmacotherapies for CUD. However, employing urine toxicology results as evidence of treatment success is by no means clear-cut, as a multitude of decisional challenges impact the interpretation of outcome measures (for an overview, see [145]).
A critical issue for interpreting a medication’s effectiveness is the expectation we hold about medication effects themselves. This begs the question: are we having difficulty finding an efficacious medication because of an insistence on abstinence as the only acceptable end-point?
As highlighted by most of the studies referenced here, complete abstinence is difficult to achieve for most individuals with CUD. It is intuitive that other end-points, similar to ‘percent subjects with no heavy drinking days’ as an efficacy end-point for medications used to treat AUD [146,147], may also indicate meaningful change [145,148]. Given the many physical and psychosocial issues that accompany CUD [148], treatment benefits are perhaps better measured through subjective indicators, such as quality of life and daily functioning, or perhaps those on a more macro scale, such as the individual burdens imposed on our health-care resources [145]. Regardless, the constraints of time and resources often preclude the direct observation of such changes. Therefore, reductions in stimulant use that are predictive of clinically relevant improvements in one’s relative functioning and wellbeing may be conceived as useful alternative indicators of treatment success. Nevertheless, the jury is still out on the constituents of meaningful change, and there remains to be established a ‘safe’ level of stimulant use or a standard stimulant drug dose [145].
CONCLUSION—WHERE DO WE GO FROM HERE?
There is an emphasis on finding new medications with novel mechanisms of action for treating CUD (e.g. [63]). In addition, the available results for ketamine are highly promising, notwithstanding the necessity of further investigations involving larger samples and longitudinal designs to establish the long-term behavioural effects and limitations to this intervention. Nonetheless, currently available medications deserve further investigation based on the existing literature; these include long-acting amphetamine formulations, modafinil, topiramate, doxazosin and combined topiramate/MAS-ER treatment.
There is certainly no need for therapeutic nihilism. However, the CUD treatment landscape is pockmarked by the outcomes of under-powered studies with high dropout rates and poor medication adherence. This, and some rather undifferentiated reviews of the literature, may dampen the enthusiasm of seasoned researchers and novitiates alike to pursue further inquiries into medications for CUD. Indeed, this would seem consistent with the recent paucity of primary research examining pharmaceutical interventions for CUD [149]. To find renewed vigour in the search for efficacious treatment models, researchers need persistence matched with adequate financial support and commitment by professional societies and funding agencies. Sorting the manifold issues around rigour in research, pharmacology, medication formulation and characteristics of the patient population is an important first step towards successfully navigating this seemingly serpentine road. Evaluating new and refined metrics of treatment success in well-defined behavioural, genetic and psychiatric patient groups, and further exploring combination therapies as well as their synergy with behavioural/psychosocial interventions, are promising avenues to establishing effective therapies for CUD—perhaps those prescribed by the characteristics of a patient, but tailored individually to their needs.
Acknowledgements
L.B. is funded by an Erwin Schroedinger Fellowship by the Austrian Science Fund (ASF).
Footnotes
Declaration of interests
The authors have no direct financial competing interests on the topic of the article to declare. S.D.C. has received research funding from Alkermes, BioXcel, Braeburn Pharmaceuticals, Cerecor Inc., Corbus, Go Medical, Intracellular Therapies, Janssen and Lyndra. In addition, S.D.C. has consulted for: Alkermes, Charleston Labs, Clinilabs, Collegium, Daiichi Sankyo, Depomed, Egalet, Endo, Epiodyne, Inspirion Delivery Sciences, Janssen, KemPharm, Mallinckrodt, Nektar, Neurolixis, Newron, Opiant, Otsuka, Pfizer and Sun Pharma. She has also received honoraria from the World Health Organization and the US Food and Drug Administration. F.R.L. has received research support from US World Meds and acted as an unpaid consultant for Alkermes, Novartis and US WorldMeds. L.B. and T.C. have no competing interests to report.
References
- 1.United Nations Office on Drugs and Crime. World Drug Report 2019—Executive Summary. 2019. Available at: https://wdr.unodc.org/wdr2019/prelaunch/WDR19_Booklet_1_EXECUTIVE_SUMMARY.pdf (accessed 29 May 2020).
- 2.United Nations Office on Drugs and Crime. World Drug Report 2019: 35 Million People Worldwide Suffer From Drug Use Disorders While Only 1 in 7 People Receive Treatment. 2019. Available at: https://www.unodc.org/unodc/en/frontpage/2019/June/world-drug-report-2019_−35-million-people-worldwide-suffer-from-drug-use-disorders-while-only-1-in-7-people-receive-treatment.html (accessed 29 May 2020).
- 3.Substance Abuse and Mental Health Services Administration (SAHMSA). Key Substance Use and Mental Health Indicators in the United States: Results from the 2018 National Survey on Drug Use and Health. 2019. Available at: https://www.samhsa.gov/data/sites/default/files/cbhsq-reports/NSDUHNationalFindingsReport2018/NSDUHNationalFindingsReport2018.pdf (accessed 29 May 2020).
- 4.European Monitoring Centre for Drugs and Drug Addiction. European Drug Report 2019. 2019. Available at: http://www.emcdda.europa.eu/system/files/publications/11364/20191724_TDAT19001ENN_PDF.pdf (accessed 29 May 2020).
- 5.Brecklin CS, Bauman JL Cardiovascular effects of cocaine: focus on hypertension. J Clin Hypertens 1999; 1: 212–7. [PubMed] [Google Scholar]
- 6.Daras M, Tuchman A, Koppel B, Samkoff L, Weitzner I, Marc J Neurovascular complications of cocaine. Acta Neurol Scand 1994; 90: 124–9. [DOI] [PubMed] [Google Scholar]
- 7.Lange RA, Hillis LD Cardiovascular complications of cocaine use. N Engl J Med 2001; 345: 351–8. Available at: http://www.nejm.org/doi/abs/10.1056/NEJM200108023450507 [DOI] [PubMed] [Google Scholar]
- 8.Lucena J, Blanco M, Jurado C, Rico A, Salguero M, Vazquez R, et al. Cocaine-related sudden death: a prospective investigation in south-West Spain. Eur Heart J 2010; 31: 318–29. Available at: https://academic.oup.com/eurheartj/article-lookup/doi/10.1093/eurheartj/ehp557 [DOI] [PubMed] [Google Scholar]
- 9.Patrizi R, Pasceri V, Sciahbasi A, Summaria F, Rosano GMC, Lioy E Evidence of cocaine-related coronary atherosclerosis in young patients with myocardial infarction. J Am Coll Cardiol 2006; 47: 2120–2. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0735109706004827 [DOI] [PubMed] [Google Scholar]
- 10.Budney AJ, Higgins ST, Hughes JR, Bickel WK Nicotine and caffeine use in cocaine-dependent individuals. J Subst Abuse 1993; 5: 117–30. Available at: https://linkinghub.elsevier.com/retrieve/pii/089932899390056H [DOI] [PubMed] [Google Scholar]
- 11.Rounsaville BJ Psychiatric diagnoses of treatment-seeking cocaine abusers. Arch Gen Psychiatry 1991; 48: 43. Available at: http://archpsyc.jamanetwork.com/article.aspx?doi=10.1001/archpsyc.1991.01810250045005 [DOI] [PubMed] [Google Scholar]
- 12.Van Tieu H, Koblin BA HIV, alcohol, and noninjection drug use. Curr Opin HIV AIDS 2009; 4: 314–8. Available at: http://journals.lww.com/01222929-200907000-00015 [DOI] [PubMed] [Google Scholar]
- 13.Degenhardt L, Hall W Extent of illicit drug use and dependence, and their contribution to the global burden of disease. Lancet 2012; 379: 55–70. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0140673611611380 [DOI] [PubMed] [Google Scholar]
- 14.Knapp WP, Soares B, Farrell M, Silva de Lima M Psychosocial interventions for cocaine and psychostimulant amphetamines related disorders. Cochrane Database Syst Rev 2016; 3: CD003023. http://doi.wiley.com/10.1002/14651858.CD003023.pub2 [Google Scholar]
- 15.Kariisa M, Scholl L, Wilson N, Seth P, Hoots B Drug overdose deaths involving cocaine and psychostimulants with abuse potential—United States, 2003–2017. Morb Mortal Wkly Rep 2019; 68: 388–95. Available at: http://www.cdc.gov/mmwr/volumes/68/wr/mm6817a3.htm?s_cid=mm6817a3_w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Busardò FP, Pichini S, Pacifici R, Karch SB The never-ending public health issue of adulterants in abused drugs. J Anal Toxicol 2016; 40: 561–2. Available at: https://academic.oup.com/jat/article-lookup/doi/10.1093/jat/bkw051 [DOI] [PubMed] [Google Scholar]
- 17.Baik J-H Dopamine signaling in reward-related behaviors. Front Neural Circuits 2013; 7: 152. Available at: http://journal.frontiersin.org/article/10.3389/fncir.2013.00152/abstract [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Rothman RB, Baumann MH, Dersch CM, Romero DV, Rice KC, Carroll FI, et al. Amphetamine-type central nervous system stimulants release norepinephrine more potently than they release dopamine and serotonin. Synapse 2001; 39: 32–41. Available at: http://doi.wiley.com/10.1002/1098-2396%2820010101%2939%3A1%3C32%3A%3AAID-SYN5%3E3.0.CO%3B2-3 [DOI] [PubMed] [Google Scholar]
- 19.Johanson CE, Fischman MW The pharmacology of cocaine related to its abuse. Pharmacol Rev 1989; 41: 3–52. [PubMed] [Google Scholar]
- 20.Shorter D, Domingo CB, Kosten TR Emerging drugs for the treatment of cocaine use disorder: a review of neurobiological targets and pharmacotherapy. Expert Opin Emerg Drugs 2015; 20: 15–29. Available at: http://www.tandfonline.com/doi/full/10.1517/14728214.2015.985203 [DOI] [PubMed] [Google Scholar]
- 21.Grabowski J, Shearer J, Merrill J, Negus SS Agonist-like, replacement pharmacotherapy for stimulant abuse and dependence. Addict Behav 2004; 29: 1439–64. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0306460304002436 [DOI] [PubMed] [Google Scholar]
- 22.Grabowski J, Rhoades H, Schmitz J, Stotts A, Daruzska LA, Creson D, et al. Dextroamphetamine for cocaine-dependence treatment: a double-blind randomized clinical trial. J Clin Psychopharmacol 2001; 21: 522–6. Available at: http://journals.lww.com/00004714-200110000-00010 [DOI] [PubMed] [Google Scholar]
- 23.Shearer J, Wodak A, van Beek I, Mattick RP, Lewis J Pilot randomized double blind placebo-controlled study of dexamphetamine for cocaine dependence. Addiction 2003; 98: 1137–41. Available at: http://doi.wiley.com/10.1046/j.1360-0443.2003.00447.x [DOI] [PubMed] [Google Scholar]
- 24.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 2009; 101: 34–41. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871608003797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Nuijten M, Blanken P, van de Wetering B, Nuijen B, van den Brink W, Hendriks VM Sustained-release dexamfetamine in the treatment of chronic cocaine-dependent patients on heroin-assisted treatment: a randomised, double-blind, placebo-controlled trial. Lancet 2016; 387: 2226–34. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0140673616002051 [DOI] [PubMed] [Google Scholar]
- 26.Levin FR, Mariani JJ, Specker S, Mooney M, Mahony A, Brooks DJ, et al. Extended-release mixed amphetamine salts vs placebo for comorbid adult attention-deficit/hyperactivity disorder and cocaine use disorder. JAMA Psychiatry 2015; 72: 593–602. Available at:; http://archpsyc.jamanetwork.com/article.aspx?doi=10.1001/jamapsychiatry.2015.41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Anderson AL, Reid MS, Li S-H, Holmes T, Shemanski L, Slee A, et al. Modafinil for the treatment of cocaine dependence. Drug Alcohol Depend 2009; 104: 133–9. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871609001562 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Dackis CA, Kampman KM, Lynch KG, Plebani JG, Pettinati HM, Sparkman T, et al. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. J Subst Abuse Treat 2012; 43: 303–12. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0740547212000062 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kampman KM, Lynch KG, Pettinati HM, Spratt K, Wierzbicki MR, Dackis C, et al. A double blind, placebo controlled trial of modafinil for the treatment of cocaine dependence without co-morbid alcohol dependence. Drug Alcohol Depend 2015; 155: 105–10. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871615015963 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Schmitz JM, Rathnayaka N, Green CE, Moeller FG, Dougherty AE, Grabowski J Combination of Modafinil and d-amphetamine for the treatment of cocaine dependence: a preliminary investigation. Front Psychol 2012; 3: 77. Available at: http://journal.frontiersin.org/article/10.3389/fpsyt.2012.00077/abstract [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Schmitz JM, Green CE, Stotts AL, Lindsay JA, Rathnayaka NS, Grabowski J, et al. A two-phased screening paradigm for evaluating candidate medications for cocaine cessation or relapse prevention: modafinil, levodopa–carbidopa, naltrexone. Drug Alcohol Depend 2014; 136: 100–7. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871613005310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.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 2007; 87: 20–9. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871606002778 [DOI] [PubMed] [Google Scholar]
- 33.Margolin A, Kosten TR, Avants SK, Wilkins J, Ling W, Beckson M, et al. A multicenter trial of bupropion for cocaine dependence in methadone-maintained patients. Drug Alcohol Depend 1995; 40: 125–31. Available at: https://linkinghub.elsevier.com/retrieve/pii/0376871695011986 [DOI] [PubMed] [Google Scholar]
- 34.Shoptaw S, Heinzerling KG, Rotheram-Fuller E, Kao UH, Wang P-C, Bholat MA, et al. Bupropion hydrochloride versus placebo, in combination with cognitive behavioral therapy, for the treatment of cocaine abuse/dependence. J Addict Dis 2008; 27: 13–23. Available at: http://www.tandfonline.com/doi/abs/10.1300/J069v27n01_02 [DOI] [PubMed] [Google Scholar]
- 35.Martell BA, Orson FM, Poling J, Mitchell E, Rossen RD, Gardner T, et al. Cocaine vaccine for the treatment of cocaine dependence in methadone-maintained patients. Arch Gen Psychiatry 2009; 66: 1116–23. Available at:; http://archpsyc.jamanetwork.com/article.aspx?doi=10.1001/archgenpsychiatry.2009.128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.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. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871614008291 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Shoptaw S, Yang X, Rotheram-Fuller EJ, Hsieh Y-CM, Kintaudi PC, Charuvastra VC, et al. Randomized placebo-controlled trial of baclofen for cocaine dependence. J Clin Psychiatry 2003; 64: 1440–8. Available at: http://article.psychiatrist.com/?ContentType=START&ID=10000616 [DOI] [PubMed] [Google Scholar]
- 38.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. Available at: http://psychiatryonline.org/doi/abs/10.1176/appi.ajp.2009.08121811 [DOI] [PubMed] [Google Scholar]
- 39.Kampman KM, Pettinati H, Lynch KG, Dackis C, Sparkman T, Weigley C, et al. A pilot trial of topiramate for the treatment of cocaine dependence. Drug Alcohol Depend 2004; 75: 233–40. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871604000882 [DOI] [PubMed] [Google Scholar]
- 40.Johnson BA, Ait-Daoud N, Wang X-Q, Penberthy JK, Javors MA, Seneviratne C, et al. Topiramate for the treatment of cocaine addiction. JAMA Psychiatry 2013; 70: 1338–46. Available at:; http://archpsyc.jamanetwork.com/article.aspx?doi=10.1001/jamapsychiatry.2013.2295 [DOI] [PubMed] [Google Scholar]
- 41.Kampman KM, Pettinati HM, Lynch KG, Spratt K, Wierzbicki MR, O’Brien CP A double-blind, placebo-controlled trial of topiramate for the treatment of comorbid cocaine and alcohol dependence. Drug Alcohol Depend 2013; 133: 94–9. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871613002019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Umbricht A, DeFulio A, Winstanley EL, Tompkins DA, Peirce J, Mintzer MZ, et al. Topiramate for cocaine dependence during methadone maintenance treatment: a randomized controlled trial. Drug Alcohol Depend 2014; 140: 92–100. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871614008254 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Nuijten M, Blanken P, van den Brink W, Hendriks V Treatment of crack-cocaine dependence with topiramate: a randomized controlled feasibility trial in the Netherlands. Drug Alcohol Depend 2014; 138: 177–84. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871614000799 [DOI] [PubMed] [Google Scholar]
- 44.Shorter D, Lindsay JA, Kosten TR The alpha-1 adrenergic antagonist doxazosin for treatment of cocaine dependence: a pilot study. Drug Alcohol Depend 2013; 131: 66–70. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871612004620 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhang X, Nielsen DA, Domingo CB, Shorter DI, Nielsen EM, Kosten TR Pharmacogenetics of dopamine β- hydroxylase in cocaine dependence therapy with doxazosin. Addict Biol 2019; 24: 531–8. Available at: https://onlinelibrary.wiley.com/doi/abs/10.1111/adb.12611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Carroll KM, Fenton LR, Ball SA, Nich C, Frankforter TL, Shi J, et al. Efficacy of disulfiram and cognitive behavior therapy in cocaine-dependent outpatients. Arch Gen Psychiatry 2004; 61: 264–72. Available at:; http://archpsyc.jamanetwork.com/article.aspx?doi=10.1001/archpsyc.61.3.264 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Schottenfeld RS, Chawarski MC, Cubells JF, George TP, Lappalainen J, Kosten TR Randomized clinical trial of disulfiram for cocaine dependence or abuse during buprenorphine treatment. Drug Alcohol Depend 2014; 136: 36–42. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871613005231 [DOI] [PubMed] [Google Scholar]
- 48.Oliveto A, Poling J, Mancino MJ, Feldman Z, Cubells JF, Pruzinsky R, et al. Randomized, double blind, placebo-controlled trial of disulfiram for the treatment of cocaine dependence in methadone-stabilized patients. Drug Alcohol Depend 2011; 113: 184–91. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871610002681 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.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. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0006322312006233 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Beresford T, Buchanan J, Thumm EB, Emrick C, Weitzenkamp D, Ronan PJ Late reduction of cocaine cravings in a randomized, double-blind trial of aripiprazole vs Perphenazine in schizophrenia and comorbid cocaine dependence. J Clin Psychopharmacol 2017; 37: 657–63. Available at: http://journals.lww.com/00004714-201712000-00005 [DOI] [PubMed] [Google Scholar]
- 51.Carroll KM, Nich C, DeVito EE, Shi JM, Sofuoglu M Galantamine and computerized cognitive behavioral therapy for cocaine dependence. J Clin Psychiatry 2018; 79: 17m11669. Available at: http://www.psychiatrist.com/JCP/article/Pages/2018/v79n01/17m11669.aspx [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.DeVito EE, Carroll KM, Babuscio T, Nich C, Sofuoglu M Randomized placebo-controlled trial of galantamine in individuals with cocaine use disorder. J Subst Abuse Treat 2019; 107: 29–37. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0740547219303009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Dakwar E, Nunes EV, Hart CL, Foltin RW, Mathew SJ, Carpenter KM, et al. A single ketamine infusion combined with mindfulness-based behavioral modification to treat cocaine dependence: a randomized clinical trial. Am J Psychiatry 2019; 176: 923–30. Available at: http://ajp.psychiatryonline.org/doi/10.1176/appi.ajp.2019.18101123 [DOI] [PubMed] [Google Scholar]
- 54.Kablinger AS, Lindner MA, Casso S, Hefti F, DeMuth G, Fox BS, 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 2012; 26: 973–81. Available at: http://journals.sagepub.com/doi/10.1177/0269881111430745 [DOI] [PubMed] [Google Scholar]
- 55.Pettinati HM, Kampman KM, Lynch KG, Xie H, Dackis C, Rabinowitz AR, et al. A double blind, placebo-controlled trial that combines disulfiram and naltrexone for treating co-occurring cocaine and alcohol dependence. Addict Behav 2008; 33: 651–67. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0306460307003139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mariani JJ, Pavlicova M, Bisaga A, Nunes EV, Brooks DJ, Levin FR Extended-release mixed amphetamine salts and Topiramate for cocaine dependence: a randomized controlled trial. Biol Psychiatry 2012; 72: 950–6. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0006322312005379 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kosten T, Oliveto A, Feingold A, Poling J, Sevarino K, McCance-Katz E, et al. Desipramine and contingency management for cocaine and opiate dependence in buprenorphine maintained patients. Drug Alcohol Depend 2003; 70: 315–25. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871603000322 [DOI] [PubMed] [Google Scholar]
- 58.Schmitz JM, Mooney ME, Moeller FG, Stotts AL, Green C, Grabowski J Levodopa pharmacotherapy for cocaine dependence: choosing the optimal behavioral therapy platform. Drug Alcohol Depend 2008; 94: 142–50. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871607004358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Poling J, Oliveto A, Petry N, Sofuoglu M, Gonsai K, Gonzalez G, et al. Six-month trial of bupropion with contingency management for cocaine dependence in a methadone-maintained population. Arch Gen Psychiatry 2006; 63: 219–28. Available at:; http://archpsyc.jamanetwork.com/article.aspx?doi=10.1001/archpsyc.63.2.219 [DOI] [PubMed] [Google Scholar]
- 60.Moeller FG, Schmitz JM, Steinberg JL, Green CM, Reist C, Lai LY, et al. Citalopram combined with behavioral therapy reduces cocaine use: a double-blind, placebo-controlled trial. Am J Drug Alcohol Abuse 2007; 33: 367–78. Available at: http://www.tandfonline.com/doi/full/10.1080/00952990701313686 [DOI] [PubMed] [Google Scholar]
- 61.Indave BI, Minozzi S, Pani PP, Amato L Antipsychotic medications for cocaine dependence. Cochrane Database Syst Rev 2016; 3: CD006306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ronsley C, Nolan S, Knight R, Hayashi K, Klimas J, Walley A, et al. Treatment of stimulant use disorder: a systematic review of reviews. Hashimoto K, editor. PLOS ONE 2020; 15: e0234809. Available at: https://dx.plos.org/10.1371/journal.pone.0234809 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Rodrigues LA, Caroba MES, Taba FK, Filev R, Gallassi AD Evaluation of the potential use of cannabidiol in the treatment of cocaine use disorder: a systematic review. Pharmacol Biochem Behav 2020; 196: 172982. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0091305720300307 [DOI] [PubMed] [Google Scholar]
- 64.Pani PP, Trogu E, Vecchi S, Amato L Antidepressants for cocaine dependence and problematic cocaine use. Cochrane Database Syst Rev 2011; 12: CD002950. [DOI] [PubMed] [Google Scholar]
- 65.Chan B, Kondo K, Freeman M, Ayers C, Montgomery J, Kansagara D Pharmacotherapy for cocaine use disorder—a systematic review and meta-analysis. J Gen Intern Med 2019; 34: 2858–73. Available at: http://link.springer.com/10.1007/s11606-019-05074-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Torrens M, Fonseca F, Mateu G, Farré M Efficacy of antidepressants in substance use disorders with and without comorbid depression: a systematic review and meta-analysis. Drug Alcohol Depend 2005; 78: 1–22. [DOI] [PubMed] [Google Scholar]
- 67.Czoty PW, Stoops WW, Rush CR Evaluation of the ‘pipeline’ for development of medications for cocaine use disorder: a review of translational preclinical, human laboratory, and clinical trial research. Pharmacol Rev 2016; 68: 533–62. Available at: http://pharmrev.aspetjournals.org/lookup/doi/10.1124/pr.115.011668 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Panlilio LV, Goldberg SR Self-administration of drugs in animals and humans as a model and an investigative tool. Addiction 2007; 102: 1863–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Haney M, Spealman R Controversies in translational research: drug self-administration. Psychopharmacology 2008; 199: 403–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Comer SD, Ashworth JB, Foltin RW, Johanson CE, Zacny JP, Walsh SL The role of human drug self-administration procedures in the development of medications. Drug Alcohol Depend 2008; 96: 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Rothman RB, Blough BE, Baumann MH Appetite suppressants as agonist substitution therapies for stimulant dependence. Ann NY Acad Sci 2006; 965: 109–26. Available at: http://doi.wiley.com/10.1111/j.1749-6632.2002.tb04155.x [DOI] [PubMed] [Google Scholar]
- 72.Koob GF Neural mechanisms of drug reinforcement. Ann NYAcad Sci 1992; 654: 171–91. Available at: http://doi.wiley.com/10.1111/j.1749-6632.1992.tb25966.x [DOI] [PubMed] [Google Scholar]
- 73.Ritz M, Lamb R, Goldberg SR, Kuhar M Cocaine receptors on dopamine transporters are related to self-administration of cocaine. Science 1987; 237: 1219–23. Available at: https://www.sciencemag.org/lookup/doi/10.1126/science.2820058 [DOI] [PubMed] [Google Scholar]
- 74.Grace AA Phasic versus tonic dopamine release and the modulation of dopamine system responsivity: a hypothesis for the etiology of schizophrenia. Neuroscience 1991; 41: 1–24. Available at: https://linkinghub.elsevier.com/retrieve/pii/030645229190196U [DOI] [PubMed] [Google Scholar]
- 75.Aragona BJ, Cleaveland NA, Stuber GD, Day JJ, Carelli RM, Wightman RM Preferential enhancement of dopamine transmission within the nucleus accumbens shell by cocaine is attributable to a direct increase in phasic dopamine release events. J Neurosci 2008; 28: 8821–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Wanat MJ, Willuhn I, Clark JJ, Phillips PEM Phasic dopamine release in appetitive behaviors and drug addiction. Curr Drug Abuse Rev 2009; 2: 195–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Negus SS, Banks ML Medications development for opioid abuse. Cold Spring Harb Perspect Med 2013; 3: a012104–a012104. Available at: http://perspectivesinmedicine.cshlp.org/lookup/doi/10.1101/cshperspect.a012104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Koob GF Brain stress systems in the amygdala and addiction. Brain Res 2009; 1293: 61–75. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0006899309006027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Preston KL, Umbricht A, Epstein DH Methadone dose increase and abstinence reinforcement for treatment of continued heroin use during methadone maintenance. Arch Gen Psychiatry 2000; 57: 395. Available at: http://archpsyc.jamanetwork.com/article.aspx?doi=10.1001/archpsyc.57.4.395 [DOI] [PubMed] [Google Scholar]
- 80.Rush CR, Essman WD, Simpson CA, Baker RW Reinforcing and subject-rated effects of methylphenidate and d-amphetamine in non-drug-abusing humans. J Clin Psychopharmacol 2001; 21: 273–86. Available at: http://journals.lww.com/00004714-200106000-00005 [DOI] [PubMed] [Google Scholar]
- 81.Stoops WW, Glaser PEA, Fillmore MT, Rush CR Reinforcing, subject-rated, performance and physiological effects of methylphenidate and d-amphetamine in stimulant abusing humans. J Psychopharmacol 2004; 18: 534–43. Available at: http://journals.sagepub.com/doi/10.1177/0269881104047281 [DOI] [PubMed] [Google Scholar]
- 82.Grabowski J, Rhoades H, Stotts A, Cowan K, Kopecky C, Dougherty A, et al. Agonist-like or antagonist-like treatment for cocaine dependence with methadone for heroin dependence: two double-blind randomized clinical trials. Neuropsychopharmacology 2004; 29: 969–81. Available at: http://www.nature.com/articles/1300392 [DOI] [PubMed] [Google Scholar]
- 83.Comer SD, Mogali S, Saccone PA, Askalsky P, Martinez D, Walker EA, et al. Effects of acute Oral naltrexone on the subjective and physiological effects of Oral D-amphetamine and smoked cocaine in cocaine abusers. Neuropsychopharmacology 2013; 38: 2427–38. Available at: http://www.nature.com/articles/npp2013143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lane SD Comparison of caffeine and d-amphetamine in cocaine-dependent subjects: differential outcomes on subjective and cardiovascular effects, reward learning, and salivary Paraxanthine. J Addict Res Ther 2014; 5: 176. Available at: https://www.omicsonline.org/open-access/comparison-of-caffeine-and-damphetamine-in-cocainedependent-subjects-differential-outcomes-on-subjective-and-cardiovascular-effects-reward-learning-2155-6105.1000176.php?aid=25611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Rush CR, Stoops WW, Hays LR Cocaine effects during d-amphetamine maintenance: a human laboratory analysis of safety, tolerability and efficacy. Drug Alcohol Depend 2009; 99: 261–71. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871608003128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Wang GJ, Volkow ND, Wigal T, Kollins SH, Newcorn JH, Telang F, et al. Long-term stimulant treatment affects brain dopamine transporter level in patients with attention deficit hyperactive disorder. PLOS ONE 2013; 8: e63023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Gill KE, Pierre PJ, Daunais J, Bennett AJ, Martelle S, Gage HD, et al. Chronic treatment with extended release methylphenidate does not alter dopamine systems or increase vulnerability for cocaine self-administration: a study in nonhuman primates. Neuropsychopharmacology 2012; 37: 2555–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Negus SS, Henningfield J Agonist medications for the treatment of cocaine use disorder. Neuropsychopharmacology 2015; 40: 1815–25. Available at: http://www.nature.com/articles/npp2014322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Stoops W, Rush C Agonist replacement for stimulant dependence: a review of clinical research. Curr Pharm Des 2013; 19: 7026–35. Available at: http://www.eurekaselect.com/openurl/content.php?genre=article&issn=1381-6128&volume=19&issue=40&spage=7026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Lile JA, Johnson AR, Banks ML, Hatton KW, Hays LR, Nicholson KL, et al. Pharmacological validation of a translational model of cocaine use disorder: effects of d-amphetamine maintenance on choice between intravenous cocaine and a nondrug alternative in humans and rhesus monkeys. Exp Clin Psychopharmacol 2020; 28: 169–80. Available from: http://doi.apa.org/getdoi.cfm?doi=10.1037/pha0000302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.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. Neuropsychopharmacology 2010; 35: 2624–37. Available at: http://www.nature.com/articles/npp2010175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Rush CR, Stoops WW, Sevak RJ, Hays LR Cocaine choice in humans during d-amphetamine maintenance. J Clin Psychopharmacol 2010; 30: 152–9. Available at: http://journals.lww.com/00004714-201004000-00008 [DOI] [PubMed] [Google Scholar]
- 93.Mariani JJ, Khantzian EJ, Levin FR The self-medication hypothesis and psychostimulant treatment of cocaine dependence: an update. Am J Addict 2014; 23: 189–93. Available at: http://doi.wiley.com/10.1111/j.1521-0391.2013.12086.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Volkow ND, Fowler JS, Logan J, Alexoff D, Zhu W, Telang F, et al. Effects of modafinil on dopamine and dopamine transporters in the male human brain. JAMA 2009; 301: 1148–54. Available at:; http://jama.jamanetwork.com/article.aspx?doi=10.1001/jama.2009.351 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wong YN, Wang L, Hartman L, Simcoe D, Chen Y, Laughton W, et al. Comparison of the single-dose pharmacokinetics and tolerability of modafinil and dextroamphetamine administered alone or in combination in healthy male volunteers. J Clin Pharmacol 1998; 38: 971–8. Available at: http://doi.wiley.com/10.1002/j.1552-4604.1998.tb04395.x [DOI] [PubMed] [Google Scholar]
- 96.Hellriegel ET, Arora S, Nelson M, Robertson P Steady-state pharmacokinetics and tolerability of Modafinil given alone or in combination with methylphenidate in healthy volunteers. J Clin Pharmacol 2001; 41: 895–904. Available at: http://doi.wiley.com/10.1177/00912700122010690 [DOI] [PubMed] [Google Scholar]
- 97.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. Available at: http://www.tandfonline.com/doi/full/10.1080/00952990600920425 [DOI] [PubMed] [Google Scholar]
- 98.Hart CL, Haney M, Vosburg SK, Rubin E, Foltin RW Smoked cocaine self-administration is decreased by Modafinil. Neuropsychopharmacology 2008; 33: 761–8. Available at: http://www.nature.com/articles/1301472 [DOI] [PubMed] [Google Scholar]
- 99.Verrico CD, Haile CN, Mahoney JJ III, Thompson-Lake DGY, Newton TF, De La Garza R II Treatment with modafinil and escitalopram, alone and in combination, on cocaine-induced effects: a randomized, double blind, placebo-controlled human laboratory study. Drug Alcohol Depend 2014; 141: 72–8. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871614008771 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Foltin RW, Haney M, Bedi G, Evans SM Modafinil decreases cocaine choice in human cocaine smokers only when the response requirement and the alternative reinforcer magnitude are large. Pharmacol Biochem Behav 2016; 150–151: 8–13. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0091305716301514 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sangroula D, Motiwala F, Wagle B, Shah VC, Hagi K, Lippmann S Modafinil treatment of cocaine dependence: a systematic review and meta-analysis. Subst Use Misuse 2017; 52: 1292–306. Available at: https://www.tandfonline.com/doi/full/10.1080/10826084.2016.1276597 [DOI] [PubMed] [Google Scholar]
- 102.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 2006; 82: 158–67. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871605002826 [DOI] [PubMed] [Google Scholar]
- 103.Castells X, Cunill R, Pérez-Mañá C, Vidal X, Capellà D Psychostimulant drugs for cocaine dependence. Cochrane Database Syst Rev 2016; 9: CD007380 Available from: http://doi.wiley.com/10.1002/14651858.CD007380.pub4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Pérez-Mañá C, Castells X, Vidal X, Casas M, Capellà D Efficacy of indirect dopamine agonists for psychostimulant dependence: a systematic review and meta-analysis of randomized controlled trials. J Subst Abuse Treat 2011; 40: 109–22. [DOI] [PubMed] [Google Scholar]
- 105.Dürsteler K, Berger E-M, Strasser H, Caflisch C, Mutschler J, Herdener M, et al. Clinical potential of methylphenidate in the treatment of cocaine addiction: a review of the current evidence. Subst Abuse Rehabil 2015; 6: 61–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Haney M, Gunderson EW, Jiang H, Collins ED, Foltin RW Cocaine-specific antibodies blunt the subjective effects of smoked cocaine in humans. Biol Psychiatry 2010; 67: 59–65. Available at: https://linkinghub.elsevier.com/retrieve/pii/S000632230901052X [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Fox BS Development of a therapeutic vaccine for the treatment of cocaine addiction. Drug Alcohol Depend 1997; 48: 153–8. Available at: https://linkinghub.elsevier.com/retrieve/pii/S037687169700121X [DOI] [PubMed] [Google Scholar]
- 108.Orson FM, Kinsey BM, Singh RAK, Wu Y, Kosten TR Vaccines for cocaine abuse. Hum Vaccin 2009; 5: 194–9. Available at: http://www.tandfonline.com/doi/abs/10.4161/hv.5.4.7457 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Dewey S, Smith G, Logan J, Brodie J, Yu D, Ferrieri R, et al. GABAergic inhibition of endogenous dopamine release measured in vivo with 11C-raclopride and positron emission tomography. J Neurosci 1992; 12: 3773–80. Available at: http://www.jneurosci.org/lookup/doi/10.1523/JNEUROSCI.12-10-03773.1992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kushner SA, Dewey SL, Kornetsky C Gamma-vinyl GABA attenuates cocaine-induced lowering of brain stimulation reward thresholds. Psychopharmacology 1997; 133: 383–8. Available at: http://link.springer.com/10.1007/s002130050418 [DOI] [PubMed] [Google Scholar]
- 111.Schiffer W, Gerasimov M, Bermel R, Brodie J, Dewey S Stereoselective inhibition of dopaminergic activity by gamma vinyl-GABA following a nicotine or cocaine challenge: a pet/microdialysis study. Life Sci 2000; 66: PL169–PL173. Available at: https://linkinghub.elsevier.com/retrieve/pii/S002432050000432X [DOI] [PubMed] [Google Scholar]
- 112.Schiffer W Gamma vinyl-GABA differentially modulates NMDA antagonist-induced increases in mesocortical versus mesolimbic DA transmission. Neuropsychopharmacology 2001; 25: 704–12. Available at: http://www.nature.com/doifinder/10.1016/S0893-133X(01)00268-8 [DOI] [PubMed] [Google Scholar]
- 113.Drouin C, Darracq L, Trovero F, Blanc G, Glowinski J, Cotecchia S, et al. α1b-adrenergic receptors control locomotor and rewarding effects of psychostimulants and opiates. J Neurosci 2002; 22: 2873–84. Available at: http://www.jneurosci.org/lookup/doi/10.1523/JNEUROSCI.22-07-02873.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Blanc G, Trovero F, Vezina P, Hervé D, Godeheu A-M, Glowinski J, et al. Blockade of Prefronto-cortical α1-adrenergic receptors prevents locomotor hyperactivity induced by subcortical D-amphetamine injection. Eur J Neurosci 1994; 6: 293–8. Available at: http://doi.wiley.com/10.1111/j.1460-9568.1994.tb00272.x [DOI] [PubMed] [Google Scholar]
- 115.Darracq L, Blanc G, Glowinski J, Tassin J-P Importance of the noradrenaline–dopamine coupling in the locomotor activating effects of d-amphetamine. J Neurosci 1998; 18: 2729–39. Available at: http://www.jneurosci.org/lookup/doi/10.1523/JNEUROSCI.18-07-02729.1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zhang XY, Kosten TA Prazosin, an α−1 adrenergic antagonist, reduces cocaine-induced reinstatement of drug-seeking. Biol Psychiatry 2005; 57: 1202–4. Available at: https://linkinghub.elsevier.com/retrieve/pii/S000632230500168X [DOI] [PubMed] [Google Scholar]
- 117.Zhang X-Y, Kosten TA Previous exposure to cocaine enhances cocaine self-administration in an alpha 1-adrenergic receptor dependent manner. Neuropsychopharmacology 2007; 32: 638–45. Available at: http://www.nature.com/articles/1301120 [DOI] [PubMed] [Google Scholar]
- 118.Shorter DI, Zhang X, Domingo CB, Nielsen EM, Kosten TR, Nielsen DA Doxazosin treatment in cocaine use disorder: pharmacogenetic response based on an alpha-1 adrenoreceptor subtype D genetic variant. Am J Drug Alcohol Abuse 2020; 46: 184–93. Available at: https://www.tandfonline.com/doi/full/10.1080/00952990.2019.1674864 [DOI] [PubMed] [Google Scholar]
- 119.Gaval-Cruz M, Weinshenker D Mechanisms of disulfiram-induced cocaine abstinence: Antabuse and cocaine relapse. Mol Interv 2009; 9: 175–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Nich C, McCance-Katz EF, Petrakis IL, Cubells JF, Rounsaville BJ, Carroll KM Sex differences in cocaine-dependent individuals’ response to disulfiram treatment. Addict Behav 2004; 29: 1123–8. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0306460304000784 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Pani PP, Trogu E, Vacca R, Amato L, Vecchi S, Davoli M Disulfiram for the treatment of cocaine dependence. Cochrane Database Syst Rev 2010; 1: CD007024. [DOI] [PubMed] [Google Scholar]
- 122.Weinshenker D Cocaine sobers up. Nat Med 2010; 16: 969–70. Available at: http://www.nature.com/articles/nm0910-969 [DOI] [PubMed] [Google Scholar]
- 123.Jones JL, Mateus CF, Malcolm RJ, Brady KT, Back SE Efficacy of ketamine in the treatment of substance use disorders: a systematic review. Front Psychol 2018; 9: 277. Available at: https://www.frontiersin.org/article/10.3389/fpsyt.2018.00277/full [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Dakwar E, Levin F, Foltin RW, Nunes EV, Hart CL The effects of subanesthetic ketamine infusions on motivation to quit and cue-induced craving in cocaine-dependent research volunteers. Biol Psychiatry 2014; 76: 40–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Dakwar E, Hart CL, Levin FR, Nunes EV, Foltin RW Cocaine self-administration disrupted by the N-methyl-D-aspartate receptor antagonist ketamine: a randomized, crossover trial. Mol Psychiatry 2017; 22: 76–81. Available at: http://www.nature.com/articles/mp201639 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Stoops WW, Rush CR Combination pharmacotherapies for stimulant use disorder: a review of clinical findings and recommendations for future research. Exp Rev Clin Pharmacol 2014; 7: 363–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Levin FR, Mariani JJ, Pavlicova M, Choi CJ, Mahony AL, Brooks DJ, et al. Extended release mixed amphetamine salts and topiramate for cocaine dependence: a randomized clinical replication trial with frequent users. Drug Alcohol Depend 2020; 206: 107700. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871619304776 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Petry NM Contingency management: what it is and why psychiatrists should want to use it. Psychiatrist 2011; 35: 161–3. Available at: https://www.cambridge.org/core/product/identifier/S1758320900006909/type/journal_article [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Schierenberg A, van Amsterdam J, van den Brink WE, Goudriaan A Efficacy of contingency management for cocaine dependence treatment: a review of the evidence. Curr Drug Abuse Rev 2012; 5: 320–31. Available at: http://www.eurekaselect.com/openurl/content.php?genre=article&issn=1874-4737&volume=5&issue=4&spage=320 [DOI] [PubMed] [Google Scholar]
- 130.Roozen HG, Boulogne JJ, Van Tulder MW, Van Den Brink W, De Jong CAJ, Kerkhof AJFM A systematic review of the effectiveness of the community reinforcement approach in alcohol, cocaine and opioid addiction. Drug Alcohol Depend 2004; 74: 1–13. [DOI] [PubMed] [Google Scholar]
- 131.Schumacher JE, Milby JB, Wallace D, Meehan D-C, Kertesz S, Vuchinich R, et al. Meta-analysis of day treatment and contingency-management dismantling research: Birmingham homeless cocaine studies (1990–2006). J Consult Clin Psychol 2007; 75: 823–8. Available at: http://doi.apa.org/getdoi.cfm?doi=10.1037/0022-006X.75.5.823 [DOI] [PubMed] [Google Scholar]
- 132.De Crescenzo F, Ciabattini M, D’Alò GL, De Giorgi R, Del Giovane C, Cassar C, et al. Comparative efficacy and acceptability of psychosocial interventions for individuals with cocaine and amphetamine addiction: a systematic review and network meta-analysis. PLOS Med 2018; 15: e1002715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Farronato NS, Dürsteler-Macfarland KM, Wiesbeck GA, Petitjean SA A systematic review comparing cognitive-behavioral therapy and contingency management for cocaine dependence. J Addict Dis 2013; 32: 274–87. [DOI] [PubMed] [Google Scholar]
- 134.Somoza EC, Winship D, Gorodetzky CW, Lewis D, Ciraulo DA, Galloway GP, et al. A multisite, double-blind, placebo-controlled clinical trial to evaluate the safety and efficacy of Vigabatrin for treating cocaine dependence. JAMA Psychiatry 2013; 70: 630–7. Available at: http://archpsyc.jamanetwork.com/article.aspx?doi=10.1001/jamapsychiatry.2013.872 [DOI] [PubMed] [Google Scholar]
- 135.Bright GM Abuse of medications employed for the treatment of ADHD: results from a large-scale community survey. Medscape Gen Med 2008; 10: 111. [PMC free article] [PubMed] [Google Scholar]
- 136.Kollins SH, Rush CR, Pazzaglia PJ, Ali JA Comparison of acute behavioral effects of sustained-release and immediate-release methylphenidate. Exp Clin Psychopharmacol 1998; 6: 367–74. Available at: http://doi.apa.org/getdoi.cfm?doi=10.1037/1064-1297.6.4.367 [DOI] [PubMed] [Google Scholar]
- 137.Parasrampuria DA, Schoedel KA, Schuller R, Silber SA, Ciccone PE, Gu J, et al. Do formulation differences Alter abuse liability of methylphenidate? J Clin Psychopharmacol 2007; 27: 459–67. Available at: http://journals.lww.com/00004714-200710000-00008 [DOI] [PubMed] [Google Scholar]
- 138.Seiden L Amphetamine: effects on catecholamine systems and behavior. Annu Rev Pharmacol Toxicol 1993; 33: 639–77. Available at: http://pharmtox.annualreviews.org/cgi/doi/10.1146/annurev.pharmtox.33.1.639 [DOI] [PubMed] [Google Scholar]
- 139.Siefried KJ, Acheson LS, Lintzeris N, Ezard N Pharmacological treatment of methamphetamine/amphetamine dependence: a systematic review. CNS Drugs 2020; 34: 337–65. Available at: http://link.springer.com/10.1007/s40263-020-00711-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Tiihonen J, Kuoppasalmi K, Föhr J, Tuomola P, Kuikanmäki O, Vorma H, et al. A comparison of aripiprazole, methylphenidate, and placebo for amphetamine dependence. Am J Psychiatry 2007; 164: 160–2. Available at: http://psychiatryonline.org/doi/abs/10.1176/ajp.2007.164.1.160 [DOI] [PubMed] [Google Scholar]
- 141.Grabowski J, Roache JD, Schmitz JM, Rhoades H, Creson D, Korszun A Replacement medication for cocaine dependence. J Clin Psychopharmacol 1997; 17: 485–8. Available at: http://journals.lww.com/00004714-199712000-00008 [DOI] [PubMed] [Google Scholar]
- 142.Galloway GP, Buscemi R, Coyle JR, Flower K, Siegrist JD, Fiske LA, et al. A randomized, placebo-controlled trial of sustained-release dextroamphetamine for treatment of methamphetamine addiction. Clin Pharmacol Ther 2011; 89: 276–82. Available at: http://doi.wiley.com/10.1038/clpt.2010.307 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Longo M, Wickes W, Smout M, Harrison S, Cahill S, White JM Randomized controlled trial of dexamphetamine maintenance for the treatment of methamphetamine dependence. Addiction 2010; 105: 146–54. Available at: http://doi.wiley.com/10.1111/j.1360-0443.2009.02717.x [DOI] [PubMed] [Google Scholar]
- 144.Tardelli VS, Bisaga A, Arcadepani FB, Gerra G, Levin FR, Fidalgo TM Prescription psychostimulants for the treatment of stimulant use disorder: a systematic review and meta-analysis. Psychopharmacology 2020; 237: 2233–55. Available at: http://link.springer.com/10.1007/s00213-020-05563-3 [DOI] [PubMed] [Google Scholar]
- 145.Kiluk BD, Carroll KM, Duhig A, Falk DE, Kampman K, Lai S, et al. Measures of outcome for stimulant trials: ACTTION recommendations and research agenda. Drug Alcohol Depend 2016; 158: 1–7. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0376871615017342 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Falk D, Wang XQ, Liu L, Fertig J, Mattson M, Ryan M, et al. Percentage of subjects with no heavy drinking days: evaluation as an efficacy endpoint for alcohol clinical trials. Alcohol Clin Exp Res 2010; 34: 2022–34. Available at: http://doi.wiley.com/10.1111/j.1530-0277.2010.01290.x [DOI] [PubMed] [Google Scholar]
- 147.Food and Drug Administration (FDA). Food and Drug Administration Draft Guidance. Alcoholism: Developing Drugs for Treatment, Guidance for Industry. Silver Spring, MD: FDA; 2015. [Google Scholar]
- 148.Winchell C, Rappaport BA, Roca R, Rosebraugh CJ Re-analysis of methamphetamine dependence treatment trial. CNS Neurosci Ther 2012; 18: 367–8. Available at: https://onlinelibrary.wiley.com/doi/abs/10.1111/j.1755-5949.2011.00288.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Buchholz J, Saxon AJ Medications to treat cocaine use disorders. Curr Opin Psychiatry 2019; 32: 275–81. Available at: https://journals.lww.com/00001504-201907000-00004 [DOI] [PubMed] [Google Scholar]