Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jan 1.
Published in final edited form as: Curr Pharm Des. 2011;17(14):1351–1358. doi: 10.2174/138161211796150846

Pharmacological Treatment of Cannabis Dependence

AM Weinstein 1,*, David A Gorelick 2
PMCID: PMC3171994  NIHMSID: NIHMS318595  PMID: 21524266

Abstract

Cannabis is the most frequently used illegal psychoactive substance in the world. There is a significant increase in the number of treatment admissions for cannabis use disorders in the past few years, and the majority of cannabis-dependent individuals who enter treatment have difficulty in achieving and maintaining abstinence. Thus, there is increased need for medications that can be used to treat this population. So far, no medication has been shown broadly and consistently effective; none has been approved by any national regulatory authority. Medications studied have included those that alleviate symptoms of cannabis withdrawal (e.g., dysphoric mood, irritability), those that directly affect endogenous cannabinoid receptor function, and those that have shown efficacy in treatment of other drugs of abuse or psychiatric conditions. Buspirone is the only medication to date that has shown efficacy for cannabis dependence in a controlled clinical trial. Results from controlled human laboratory studies and small open-label clinical trials suggest that dronabinol, the COMT inhibitor entacapone, and lithium may warrant further study. Recent pre-clinical studies suggest the potential of fatty acid amide hydrolase (FAAH) inhibitors such as URB597, endocannabinoid-metabolizing enzymes, and nicotinic alpha7 receptor antagonists such as methyllycaconitine (MLA). Controlled clinical trials are needed to evaluate the clinical efficacy of these medications and to validate the laboratory models being used to study candidate medications.

Keywords: Cannabis, withdrawal, dependence, pharmacotherapy, treatment

CANNABIS USE DISORDERS

Cannabis is the most frequently used illegal substance in the world [13]. Cannabis abuse and cannabis dependence are diagnoses recognized in the United States Diagnostic and Statistical Manual of Mental Disorders Fourth Edition (DSM IV) [4] and the WHO International Classification of Diseases, Tenth Revision (ICD-10) [5]. In the United States, the number of individuals with disorders associated with cannabis use is twice that of any other illicit drug [1], with approximately 4 million adults meeting criteria for a life-time diagnosis of cannabis dependence [6]. Relapse rates for cannabis users in treatment are comparable to those found for other drugs of abuse [711].

NEUROPHARMACOLOGICAL MECHANISMS OF CANNABIS DEPENDENCE

Exogenous cannabis (and its primary psychoactive component, Δ-9-tetrahydrocannabinol [THC]) acts on the endogenous cannabinoid (endocannabinoid) system in the brain and other body tissues by binding to two different types of cannabinoid receptors on cell membranes: CB1 and CB2 [12]. CB1 receptors are located primarily in pre-synaptic neurons of the CNS and are responsible for the acute psychological and cardiovascular effects of cannabis. CB2 receptors are located largely in the periphery and modulate immune function and inflammatory response.

Endocannabinoids (endogenous ligands at CB receptors) such as anandamide serve as retrograde neuromodulators of synaptic activity. They are released postsynaptically by a variety of stimuli upon demand, travel across the synaptic cleft, and then activate presynaptic CB receptors. A membrane transporter actively takes anandamide into the cell. Anandamide is then broken down by fatty acid amide hydrolase (FAAH) [1315].

The neuropharmacological mechanism of cannabis dependence may involve interactions of the endocannabinoid system with the dopaminergic and opioid systems. Additionally, CB receptor agonists such as THC act as inhibitors of neurotransmission in acetylcholine, GABA, and glutamatergic pathways. Chronic administration of cannabinoids leads to down-regulation of the CB receptor and receptor function desensitization [16].

THC like other drugs of abuse, releases DA in the mesocortico-limbic regions of animal brains [1719]. PET brain imaging studies in healthy human volunteers provide inconsistent evidence for this action in humans. One study showed modest THC-induced dopamine release in the ventral striatum and dorsal putamen using [11C] raclopride [20]. Another study found no significant effect of THC on [11C] raclopride binding, although THC markedly increased psychosis-like symptoms [21]. A subsequent study using the same methodology found significant decreases in frontal and temporal lobe [11C] raclopride binding after THC challenges, but no changes in the striatum, which is also part of the dopamine reward pathway [22]. Decreased frontal lobe binding significantly correlated with catechol-O-methyltransferase (COMT) status. Therefore, medications that target the brain dopamine reward system may have a role in the treatment of cannabis dependence, as they may for other drugs of abuse.

CANNABIS INTOXICATION

Cannabis intoxication is a syndrome recognized in DSM-IV [4] and ICD-10 [5], with both psychological and behavioral (euphoria, relaxation, increased appetite, impaired memory and concentration), and physical (motor incoordination, tachycardia, orthostatic hypotension), manifestations. Intoxication is usually mild and self-limiting, not requiring pharmacological treatment [23]. The most severe effects (anxiety, panic, psychosis) are best treated symptomatically with a benzodiazepine or second-generation (atypical) anti-psychotic medication. No medication is approved specifically for treatment of cannabis intoxication.

Studies with the selective CB1 receptor antagonist/inverse agonist rimonabant suggest that CB1 receptors mediate many of the acute effects of cannabis in humans. In a double-blind, placebo-controlled study of 63 healthy men with a history of cannabis use, single oral doses of rimonabant produced significant dose-dependent blockade of the subjective intoxication and tachycardia caused by smoking an active (2.64% THC) or placebo (double-blind) cannabis cigarette 2 hours later [24] The 90-mg dose produced about 40% reductions in ratings of "high" "stoned" and "drug effect" (on 100-mm visual-analogue scales) and a 60% reduction in heart rate. Rimonabant alone produced no significant physiological or psychological effects and did not affect peak THC plasma concentration or its time course. This pattern of findings suggests that the observed attenuation of cannabis effects was specifically due to CB1 receptor blockade, and not to reduction in brain THC concentration or counteracting effects of rimonabant.

CB receptor antagonists such as rimonabant might be useful in treating acute cannabis intoxication, in the way that the mu-opioid receptor (mOR) antagonists naloxone and naltrexone are used to treat opiate intoxication. However, such medications are no longer available for clinical use. Rimonant and similar CB1 receptor antagonists were withdrawn from clinical development and use because of psychiatric side-effects associated with their long-term use [25].

THE CANNABIS WITHDRAWAL SYNDROME

Both human laboratory and clinical outpatient studies have established the reliability, validity and time course of the cannabis withdrawal syndrome [26, 27] and the cannabis withdrawal syndrome has been proposed for inclusion in DSM-V [28]. Some US studies suggest that about half of patients in treatment have reported symptoms of the cannabis withdrawal syndrome [23, 2933]. The main symptoms of cannabis withdrawal are anxiety, irritability, depressed mood, restlessness, disturbed sleep, G-I symptoms, and decreased appetite. Most symptoms begin during the first week of abstinence and resolve after a few weeks.

TREATMENT OF CANNABIS WITHDRAWAL SYNDROME

Because symptoms of cannabis withdrawal may serve as negative reinforcement for relapse to cannabis use in individuals trying to abstain [27, 34], pharmacological treatment aimed at alleviating cannabis withdrawal might prevent relapse and reduce dependence.

Several studies have tested the effects of medications on cannabis withdrawal [3537]. These medications are either CB receptor agonists that directly suppress the withdrawal syndrome (analogous to using an opiate to suppress heroin withdrawal) or are designed to indirectly alleviate symptoms of cannabis withdrawal (e.g. dysphoric mood, irritability) by influencing the brain circuits that mediate these symptoms. No medication has regulatory approval for the treatment of cannabis withdrawal. The CB receptor agonist THC has shown efficacy in several human laboratory studies and open-label case series. See Table 1 for description of all pharmacological treatment trials for cannabis dependence.

Table 1.

Pharmacological Trials for Cannabis Dependence

1. Anti-Depressants
Study Drug N Dose Design Results
Haney et al., (2001) Bupropion (10) 300mg Randomized Double-blind Placebo-controlled Cross-over Worsened withdrawal
Haney et al., (2003a) Nefazodone (7) 450mg Randomized Double-blind Placebo-controlled Cross-over Improved anxiety
*Cornelius et al., (2005) Fluoxetine (22) 20–40mg Randomized Double-blind Placebo-controlled Cross-over Reduced cannabis use
Haney et al., (2008) Lofexidine+ THC (8) 2.4mg 60mg Placebo-controlled Reduced withdrawal
*Carpenter et al., (2009) Nefazodone Bupropion (106) 300mg 150mg Randomized Double-blind Placebo-controlled Cross-over No effect
Haney et al., (2010) Baclofen Or Mirtazapine (11) 30, 60, 90 mg 30mg Randomized Double-blind Placebo-controlled Cross-over No effect
*Cornelius et al., (2010) Fluoxetine (70) 20 mg Randomized Double-blind Placebo-controlled Cross-over No effect
2. Cannabis Agents

Cannabis Agonists
Study Drug N Dose Design Results
Haney et al., (2004) Oral THC (11) 10mg Randomized Double-blind Placebo-controlled cross-over Reduced withdrawal
Budney et al., (2007) Oral THC (8) 30, 90 mg Randomized Double-blind Placebo-controlled Cross-over Reduced withdrawal
Levin and Kleber (2008) Dronabinol (2) 10–50mg Case studies Mixed results
Cannabis Antagonists
Study Drug N Dose Design Results
Huestis et al., (2001) SR141716 (63) (1, 3,10, 30, 90 mg) Randomized Double-blind Placebo- controlled Cross-over Attenuated effects of cannabis
Huestis et al., (2007) Rimonabant (42) 90mg Double blind parallel groups Attenuated effects of cannabis
3. Other Agents

Opiate Antagonists
Study Drug N Dose Design Results
Wachtel and de Wit (2000) Naltrexone (14) 50mg Double blind Placeb-controlled Cross-over Failed to attenuate dronabinol
Haney et al., (2003b) Naltrexone (9,23) 50mg Double blind Placebo-controlled Cross-over Enhanced subjective effects of THC
Haney et al., (2007) Naltrexone (21) 12mg Double blind Placebo-controlled Cross-over Mixed results
Cooper and Haney, (2010) Naltrexone (29) (12, 25, 50, or 100 mg) Double blind Placebo-controlled Cross-over Enhanced subjective effects of cannabis
Greenwald & Stitzer (2000) Naltrexone (5) (50, or 200 mg) Double-blind Placebo-controlledCross-over No effect
Dopamine Agents
Study Drug N Dose Design Results
Shafa (2009) Entacapone (36) 200mg Open label Reduced craving
Glutamate
Study Drug N Dose Design Results
Gray et al., (2010) N-acetylcysteine (24) 1200mg Open label Reduced self-reported use, but not urine cannabinoid levels
Norepinephrine Reuptake Inhibitor
Study Drug N Dose Design Results
*Tirado et al., (2008) Atomoxetine (13) 25–80mg Open label Reduction in cannabis use but adverse events
*McRae-Clark et al., (2010) Atomoxetine (36) 25–80 mg Double-blind Placebo-controlled No effect
Anxiolytic
Study Drug N Dose Design Results
*McRae et al., (2006) Buspirone (10) Up to 60mg Open label Reduced craving and irritability
*McRae et al., (2009) Buspirone (50) Up to 60 mg Double-blind, Placebo-controlled Reduced cannabis use
Mood Stabilizers
Bowen et al., (2005) Lithium (9) 600–900 mg Open label Reduced withdrawal
Winstock et al., (2009) Lithium (12) 500mg Open label Reduced cannabis use
Haney et al., (2004) Divalproex (7) 1500mg Randomized Double-blind Placebo-controlled Cross-over Worsened withdrawal
*Levin et al., (2004) Divalproex (25) 1500–2000mg Randomized Double-blind Placebo- controlled Cross-over No effect
*

Indicates Clinical Trial

Most published studies have been human laboratory studies of short duration (typically 3–4 days), using an inpatient human laboratory model developed at Columbia University (New York, US) [35]. Participants were non-treatment-seeking volunteers who smoked cannabis many times a day. They smoked cannabis (active or placebo) and received oral medication (active or placebo) each day under double-blind conditions. The protocols used a within-subjects crossover design so that each participant received each active and placebo combination of cannabis and medication [38].

The early laboratory studies evaluated divalproex, an anticonvulsant which is used clinically as a mood stabilizer and to treat epilepsy and migraine headaches [39] buproprion, which is used clinically as an antidepressant and for smoking cessation; and nefazodone, an antidepressant that blocks post-synaptic 5HT-2a receptors and inhibits pre-synaptic 5HT and NE reuptake [40]. Bupropion is thought to exert its clinical effects by inhibiting reuptake of norepinephrine (NE) and dopamine (DA) and possibly by acting as a nicotine receptor antagonist [41]. Single doses of bupropion sustained-release (300 mg/day for 17 days) and divalproex (1500 mg/day for 29 days) actually worsened, rather than improved, some withdrawal symptoms and had no positive effects [42, 43]. A single dose of nefazodone (450 mg/day) decreased some, but not the majority, of cannabis withdrawal symptoms [44].

So far, the only medication successful in suppression of withdrawal symptoms in the laboratory was a single dose of 10mg/day oral synthetic THC (dronabinol) [44]. Oral THC was also more effective than placebo in an outpatient study in which oral THC was given to 8 adult, daily cannabis users who were not seeking treatment in a 40-day, within-subject design study [44]. Participants received daily doses of placebo, 30 mg (10 mg/tid), or 90 mg (30 mg/tid) oral THC during three 5-day periods of abstinence from cannabis use, separated by 7–9-day periods of cannabis smoking as usual. Comparison of measures of withdrawal symptoms across conditions indicated a dose-dependent reduction of withdrawal discomfort by THC. Minimal adverse effects were associated with either THC dose. This demonstration of dose-response effect replicates and extends prior findings of the pharmacological specificity of the cannabis withdrawal syndrome [43].

More recently, the Columbia group has evaluated medications in a more complicated human laboratory design that models both withdrawal and relapse. Regular cannabis users were maintained on each medication condition for 7 inpatient days. Each medication phase was separated by an outpatient washout phase. During the first three inpatient days, placebo cannabis was available for self-administration (withdrawal). For the next 4 days, active cannabis was available for self-administration. Participants paid for self-administered cannabis using study earnings.

The first such study evaluated lofexidine, an agonist at the alpha2-adrenergic receptor that is used to treat opiate withdrawal [46]. Lofexidine was tested both alone and in combination with THC [47]. Eight non-treatment-seeking male regular cannabis users were maintained on each of four medication conditions double-blind: placebo, THC (60 mg/day), lofexidine (2.4 mg/day), and THC (60 mg/day) combined with lofexidine (2.4 mg/day). THC reversed the anorexia and weight loss associated with cannabis withdrawal, and decreased some withdrawal symptoms, but increased sleep onset latency, and did not decrease the resumption of cannabis use when active cannabis was available. Lofexidine, which was sedating, worsened withdrawal-related anorexia and did not robustly attenuate mood symptoms associated with withdrawal, but improved sleep and decreased cannabis relapse. The combination of lofexidine and THC produced the most robust improvements in sleep and decreased cannabis withdrawal, craving, and relapse in daily cannabis smokers relative to either medication alone.

The second such study evaluated baclofen, a GABA B receptor agonist and antispasmodic medication that reduces mood symptoms in heroin withdrawal [48], and mirtazapine, an antidepressant that enhances noradrenergic and serotonergic transmission and decreases withdrawal symptoms in alcohol-dependent patients [49], especially agitation and insomnia [50]. In this study, separate groups received baclofen (60, 90 mg/day) for 16 days (n=10) or mirtazapine (30 mg/day) for 14 days (n=11) [51] Medication administration began when subjects were outpatients prior to each 8-day inpatient phase. On the first inpatient day of each medication condition, participants smoked active cannabis (baclofen group : 3.3% THC; mirtazapine: 6.2% THC). For the next 3 days, participants could self-administer placebo cannabis (withdrawal phase), followed by 4 days in which they could self-administer active cannabis (relapse phase). During active cannabis smoking, baclofen dose-dependently decreased craving for tobacco and cannabis, but had little effect on mood during abstinence and did not decrease relapse. Mirtazapine improved sleep during abstinence, and robustly increased food intake, but had no effect on withdrawal symptoms and did not decrease cannabis relapse. Overall, this human laboratory study did not find evidence to suggest that either baclofen or mirtazapine show promise for the treatment of cannabis withdrawal.

TREATMENT OF CANNABIS DEPENDENCE

3.1. Agonist Approach

One strategy to treat drug dependence is long-term treatment with the same agonist drug or with a cross-tolerant drug to suppress withdrawal and drug craving. This approach is successfully used in the treatment of tobacco (nicotine) dependence (nicotine itself) and opiate dependence (methadone, buprenorphine). It is being studied for treatment of cannabis dependence using synthetic THC which is legally marketed in many countries as an oral medication for appetite stimulation and suppression of nausea and vomiting due to chemotherapy. Questions of medication abuse and diversion must be addressed, as with opiate agonist substitution treatment.

Use of oral synthetic THC in outpatients was reported in a study that showed the potential benefit, as well as questions that arise from the use of this medication in cannabis-abusing populations [52]. Controlled clinical trials of oral THC are currently underway (www.clinicaltrials.gov).

3.2. Antagonist Approach

The antagonist approach uses long-term treatment with a CB1 antagonist to prevent patients from experiencing the pleasurable reinforcing effects of cannabis use, resulting in extinction of drug-seeking and drug-taking behavior. This approach has been used successfully with the mOR antagonist naltrexone in the treatment of opiate dependence [53]. It could be implemented should a CB1 receptor antagonist again become available for human use.

A recent randomized, double blind, parallel group study investigated whether subacute (2-week) treatment with the CB1 receptor antagonist rimonabant (40 mg daily) attenuated the effects of smoked cannabis in 42 healthy men with a history of cannabis use [54]. The repeated daily rimonabant doses attenuated the acute cardiovascular effects of a cannabis cigarette (2.78% THC) to a similar degree as a single 90-mg dose; repeated 40-mg doses attenuated subjective effects after 8 but not 15 days (possibly because of smaller sample size and lower statistical power at day 15). Rimonabant did not significantly affect THC pharmacokinetics, suggesting that the observed effects were due to receptor blockade and not reduced THC levels in the brain.

3.3. Other Approaches

Alternative pharmacotherapy approaches may arise from improved understanding of the neuropharmacology of cannabis use disorders, including the recognition that (i) frequent cannabis use may cause an adaptive down-regulation of brain endocannabinoid signaling, and (ii) genetic traits that favor hyperactivity of the endocannabinoid system in humans may decrease susceptibility to cannabis dependence [55]. These findings suggest that pharmacological agents that elevate brain levels of the endocannabinoid neurotransmitters anandamide and 2-arachidonoylglycerol (2-AG) might alleviate cannabis withdrawal and dependence. One such agent, the FAAH inhibitor URB597, selectively increased anandamide levels in the brain of rodents and primates. Preclinical studies showed that URB597 produced analgesic, anxiolytic-like, and antidepressant-like effects in rodents, which were not accompanied by overt signs of abuse liability. This evidence suggests that FAAH inhibitors such as URB597 might offer a possible therapeutic avenue for the treatment of cannabis withdrawal [55].

3.3.a. Opiate Antagonist Naltrexone

Because animal studies show that mOR antagonists block effects of THC, several human laboratory studies have investigated whether the mOR antagonist naltrexone can reduce the subjective effects of cannabinoids in humans. In cannabis users, pretreatment with high doses of naltrexone (50–200 mg) failed to attenuate or enhanced the subjective effects of THC [56, 57] or smoked cannabis [58]. However, a lower, more mOR-selective dose of naltrexone (12 mg) decreased the intoxicating effects of 20 mg, but not 40 mg, of THC [59]. A recent placebo-controlled study in 29 heavy cannabis smokers found that opioid-receptor blockade by naltrexone (12, 25, 50, or 100 mg daily) enhanced the subjective and cardiovascular effects of cannabis [60]. This pattern of human experimental findings is not completely consistent, but suggests that clinically used doses of naltrexone would not be effective as treatment for cannabis dependence, and might actually increase the abuse liability of cannabis.

3.3.b. Dopamine Agents

Catechol-O-Methyl Transferase (COMT) Inhibitor-- Entacapone

Dopamine (DA) is a major neurotransmitter in the brain’s meso-cortico-limbic reward pathway, believed to be a common pathway involved in drug-seeking for all drugs of abuse [6163] DA deficiency in this reward pathway plays a major role in drug compulsion and craving [64]. Catechol-O-methyl transferase (COMT) is an enzyme that inactivates catecholamine neurotransmitters and plays a pivotal role in regulating homeostatic levels of DA neurotransmitter in the inter-synaptic cleft. COMT inhibitors would increase synaptic DA activity, perhaps counteracting the DA deficiency considered to play a role in drug compulsion and craving. The gene for COMT is located on chromosome 22q11.21 There is some evidence that carriers of the valine158 allele of the COMT gene, who should have increased brain dopamine turnover, are at increased risk for psychotic symptoms and development of schizophrenia if they use cannabis by the age of 18 [65]. However, these findings were not replicated in a later study [66].

As mentioned earlier, THC, like other drugs of abuse, releases DA in the meso-cortico-limbic regions of animal brains. PET brain imaging studies in healthy volunteers so far seems to suggest that THC administration results in modest dopamine release in some human brain regions, but the role of this action in the rewarding effects of THC remains unclear. Therefore, the place in treatment of medications that target the brain dopamine reward system also remains unclear.

Entacapone is a COMT inhibitor approved for the treatment of Parkinson’s disease, in a recent study entacapone (up to 2000 mg/day) was given to 36 patients with cannabis dependence (DSM-IV) in an open-label trial for 12 weeks, continued for 12 months in interested individuals. Entacapone both short-term and long-term significantly decreased craving for cannabis in 52.7% of the patients, but no information was reported on patients’ cannabis use. Entacapone was well tolerated and there was no serious adverse event [67].

3.3.c. Glutamate- N-acetylcysteine (NAC)

The neurotransmitter glutamate has emerged as a potential target in the treatment of addictions, such as cocaine, nicotine, and cannabis dependence. In animal studies, N-acetylcysteine (NAC) reverses drug-induced down-regulation of the cystine-glutamate exchanger, which restores normal regulation of glutamate release, reducing compulsive drug-seeking behaviors [68]. Consistent with this evidence, preliminary studies have demonstrated significant reductions in cocaine craving [69] and cigarette use [70] during NAC treatment.

A recent open-label study gave NAC (1,200 mg) twice daily for 4 weeks to 24 cannabis-dependent males and females who were interested in reducing their cannabis use [71]. Treatment with NAC was well tolerated and associated with significant decreases in self-report measures of cannabis use and craving, but no change in semi-quantitative urine cannabinoid levels.

3.3.d. Norepinephrine Reuptake Inhibitor- Atomoxetine

Cannabis users demonstrate time and dose-dependent impairments in attention, memory, executive function and response inhibition that resemble deficits in patients with attention deficit hyperactivity disorder (ADHD) and share morbidity with this disorder [72]. A recent study [73] evaluated atomoxetine, an ADHD medication with low abuse potential, in an 11-week open-label study of thirteen treatment-seeking, cannabis-dependent patients (25, 40, 80mg/day). For the eight participants who completed the study, there was a trend towards reduction in cannabis use and increase in percent days of abstinence. The majority of patients experienced gastrointestinal adverse events.

A more recent double-blind, placebo-controlled 12-week study of atomoxetine (25–100 mg/day escalating doses) in 38 cannabis-dependent outpatients with concurrent ADHD found no significant change in cannabis use, although there was some improvement in ADHD symptoms [74].

3.3.e. Anxiolytic- Buspirone

Buspirone shares some of the properties of the benzodiazepines and the neuroleptics; it is a 5-HT (1A) receptor agonist [75] and a D2 receptor antagonist [76]. A preliminary 12-week, open-label study in 10 cannabis-dependent men found that buspirone (maximum 60 mg per day) in a 12-week open-label trial significantly reduced frequency and duration of cannabis craving and use and reduced irritability and depression [77]. A following 12-week controlled clinical trial compared buspirone (maximum 60mg/day) vs. placebo, together with motivational interviewing, in 23 cannabis-dependent participants [78]. Among the 24 participants who completed the trial, those randomized to buspirone had a greater percentage of cannabis-negative urine samples (95% CI:7–63%, p<0.05) and a trend towards achieving the first cannabis-negative urine sample sooner than those participants treated with placebo. These findings support the promise of buspirone as a treatment for cannabis dependence.

3.3.f. Mood Stabilizers

Lithium is a mood stabilizer used primarily in the treatment of bipolar disorder (depression and mania), both acutely and chronically. A preclinical study showing that lithium attenuated cannabis withdrawal in rats [79] prompted two small open-label clinical studies. In the first study, lithium (600 to 900 mg/day), administered to 9 adults for 6 days, reduced withdrawal symptoms in 4 of the 9 participants [80]. However, cannabis was admittedly smoked during this period by one of these 4 participants and cannabis abstinence was not verified in the others. In the second study, 20 cannabis-dependent participants received lithium (500 mg 2x/day) for 7 days in an inpatient detoxification facility [81]. Twelve participants completed the 7-day inpatient detoxification. Self-reported cannabis abstinence at post-treatment follow-up sessions was 64% (Day 10), 65% (Day 24), and 41% (Day 90). Participants self-reported cannabis abstinence on 88% of days post-treatment. Five participants reported continuous abstinence that was corroborated with urine toxicology tests on Day 90. These results provide limited support for a double-blind trial of lithium as treatment for cannabis dependence.

A small 6-week controlled clinical trial in 25 cannabis-dependent outpatients also receiving weekly relapse prevention psychotherapy found that divalproex (1500–2000 mg daily, to achieve plasma concentrations of 50–120 ng/mL) did not reduce cannabis use more than placebo and was poorly tolerated by participants [82].

3.3.g. Anti-Depressants

A 13-week controlled clinical trial comparing nefazodone (300mg/day), bupropion-sustained release (150mg/day), or placebo, plus weekly, individual coping skills therapy in 106 cannabis-dependent outpatients found no significant medication effects on cannabis use or cannabis withdrawal symptoms [83]. These results suggest that nefazodone and bupropion-sustained release are not effective in treating cannabis dependence.

TREATMENT OF PATIENTS WITH COMORBID CANNABIS DEPENDENCE AND DEPRESSION

Cannabis users frequently have co-morbid mood symptoms, especially depression [84, 85]. The prevalence of depression in this population suggested that anti-depressant medication might promote abstinence in this population.

Two studies evaluated the selective serotonin reuptake inhibitor (SSRI) anti-depressant medication fluoxetine in this population. A post hoc analysis of 13 cannabis-using patients among a larger sample of alcohol-abusing, depressed adolescents treated with fluoxetine (20–40 mg daily) showed reduction in cannabis and alcohol dependence and depressive symptoms [86]. Five-year follow-up of 10 patients showed that cannabis and alcohol dependence were reduced and academic ability improved, but clinical depression remained problematic. A later 12-week, controlled clinical trial in 70 adolescents and young adults with comorbid major depression and cannabis use disorder found fluoxetine (20 mg daily) no better than placebo in treating either the depressive symptoms or the cannabis- related symptoms [87]. The lack of a significant between-group difference in symptoms may reflect limited medication efficacy, a ceiling effect because of the efficacy of the concurrent psychosocial treatment (cognitive behavioral/motivational enhancement psychotherapy), or low statistical power from small sample size.

4. NEW PRE-CLINICAL DEVELOPMENTS

Several recent studies in animals have used compounds that affect the endocannabinoid system and offer promising leads for future therapeutic agents. First, the benzoflavone moiety from methanol extracts of passiflora incarnate Linneaus reversed the effects of THC in mice [88]. Second, nicotinic alpha7 receptor antagonists such as methyllycaconitine (MLA) antagonized the discriminative effects of cannabinoids at doses that did not produce depressant or toxic effects [89]. Finally, inhibitors of endocannabinoid- metabolizing enzymes reduced rimonabant-induced precipitated withdrawal responses in THC- dependent mice [90]. These results suggest several potential therapeutic agents that warrant further study.

SUMMARY

Results from controlled human laboratory studies suggest that CB1 receptor antagonists, should they again become available for clinical use, might be effective treatment for cannabis intoxication and that oral THC (perhaps combined with an α-adrenergic agonist such as lofexidine) might be effective treatment for cannabis withdrawal. For the treatment of cannabis dependence, there is little data to guide the clinician, as few controlled clinical trials have been conducted. Only buspirone has shown efficacy in such a trial, while atomoxetine, bupropion, divalproex, and nefazadone have not. A few small open-label clinical trials suggest that the COMT inhibitor entacapone, dronabinol, and lithium may warrant further study, although this recommendation is tempered by the weakness of evidence from open-label studies. In contrast, available evidence from human laboratory studies suggests that the mu-opioid receptor antagonist naltrexone may increase the abuse liability of cannabis and therefore should not be used for treatment. Recent pre-clinical studies suggest the potential of FAAH inhibitors such as URB597 for the treatment of cannabis withdrawal and of endocannabinoid-metabolizing enzymes andnicotinic alpha7 receptor antagonists such as methyllycaconitine (MLA) for treatment of cannabis dependence.

In response to the continuing public health problem that they pose, the National Institute on Drug Abuse launched in 2004 a research program to develop medications for treating cannabis use disorders (CUDs) [16] which hopefully will bear fruit in the future.

ACKNOWLEDGEMENT

Dr. Weinstein is supported by the National Institute for Psychobiology in Israel that is funded by the Charles E. Smith Foundation. Dr. Gorelick is supported by the US National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health.

REFERENCES

  • 1.United Nations Office on Drugs and Crime (ONODC) World Drug Report 2010. United Nations Office on Drugs and Crime. http://unp.un.org.
  • 2.Substance Abuse and Mental Health Services Administration. Rockville, MD: 2010. Results from the 2009 National Survey on Drug Use and Health: Volume I. Summary of National Findings. (Office of Applied Studies, NSDUH Series H-38A, HHS Publication No. SMA 10-4856 Findings). [Google Scholar]
  • 3.European Monitoring Centre for Drug and Drug addiction (EMCDDA) Conference proceedings: Identifying Europe's information needs for effective drug policy EM-CDDA, Lisbon, December 2009; Luxembourg: 2009. Publications Office of the European Union ISBN 978-92-9168-409-0 www.emcdda.europa.eu. [Google Scholar]
  • 4.American Psychiatric Association Diagnostic and Statistical Manual of Mental Disorders. 4th ed. Washington, DC: American Psychiatric Association; 1994. [Google Scholar]; Artigas F, Nutt DJ, Shelton R. Mechanism of action of antidepressants. Psychopharmacol Bull. 2002;36 Suppl 2:123–132. [PubMed] [Google Scholar]
  • 5.World Health Organization (WHO) Geneva: WHO; 2007. International Classification of Diseases, Tenth Revision (ICD-10) - Clinical Modification. [Google Scholar]
  • 6.Stinson FS, Ruan WJ, Pickering R, Grant BF. Cannabis use disorders in the USA: prevalence, correlates and co-morbidity. Psycholl Med. 2006;36:1447–1460. doi: 10.1017/S0033291706008361. [DOI] [PubMed] [Google Scholar]
  • 7.Copeland J, Swift W, Roffman R, Stephens RS. A randomized controlled trial of brief cognitive– behavioral interventions for cannabis use disorder. J Subste Abuse Treat. 2001;21:55–64. doi: 10.1016/s0740-5472(01)00179-9. [DOI] [PubMed] [Google Scholar]
  • 8.Kadden RM, Litt MD, Kabela-Cormier E, Petry NM. Abstinence rates following behavioral treatments for marijuana dependence. Addict Behav. 2007;32:1220–1236. doi: 10.1016/j.addbeh.2006.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Stephens RS, Roffman RA, Simpson EE. Treating adult marijuana dependence: a test of the relapse prevention model. J Consult Clin Psychol. 1994;62:92–99. doi: 10.1037//0022-006x.62.1.92. [DOI] [PubMed] [Google Scholar]
  • 10.Stephens RS, Roffman RA, Curtin L. Extended versus brief treatment for marijuana use. J Consult Clin Psychol. 2000;68:898–908. [PubMed] [Google Scholar]
  • 11.Moore BA, Budney AJ. Relapse in outpatient treatment for marijuana dependence. J Subst Abuse Treat. 2003;25:85–89. doi: 10.1016/s0740-5472(03)00083-7. [DOI] [PubMed] [Google Scholar]
  • 12.Pertwee RG. Ligands that target cannabinoid receptors in the brain: from THC to anandamide and beyond. Addict Biol. 2008;13:147–159. doi: 10.1111/j.1369-1600.2008.00108.x. [DOI] [PubMed] [Google Scholar]
  • 13.Mechoulam R. Cannabinoids as Therapeutic Agents. FL: CRC Press; 1986. [PubMed] [Google Scholar]
  • 14.Mechoulam R. Marijuana, Chemistry, Pharmacology, Metabolism and Clinical Effects. New York: Academic Press; 1973. [Google Scholar]
  • 15.Sarel S, Mechoulam R, Agranat I. Trends in Medicinal Chemistry. Oxford, UK: Blackwell Publ.; 1991. [Google Scholar]
  • 16.Montoya ID, Vocci F. Novel medications to treat addictive disorders. Curr Psychiat Rep. 2008;10(5):392–398. doi: 10.1007/s11920-008-0063-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chen JP, Paredes W, Li J, Smith D, Lowinson J, Gardner EL. Delta 9-tetrahydrocannabinol produces naloxone-blockable enhancement of presynaptic basal dopamine efflux in nucleusaccumbens of conscious, freely-moving rats as measured by intracerebral microdialysis. Psychopharmacol (Berl) 1990;102:156–162. doi: 10.1007/BF02245916. [DOI] [PubMed] [Google Scholar]
  • 18.Tanda G, Pontieri FE, Di Chiara G. Cannabinoid and heroin activation of mesolimbic dopamine transmission by a common mu1 opioid receptor mechanism. Science. 1997;276:2048–2050. doi: 10.1126/science.276.5321.2048. [DOI] [PubMed] [Google Scholar]
  • 19.Fadda P, Scherma M, Spano MS, Salis P, Melis V, Fattore L, Fratta W. Cannabinoid self-administration increases dopamine release in the nucleus accumbens. Neuroreport. 2006;17:1629–1632. doi: 10.1097/01.wnr.0000236853.40221.8e. [DOI] [PubMed] [Google Scholar]
  • 20.Bossong MG, van Berckel BNM, Boellaard R, Zuurman L, Schuit RC, Windhurst AD, van Gerven JMA, Ramsey NF, Lammertsma AA, Kah RS. Δ9-Tetrahydrocannabinol induces dopamine release in the human striatum. Neuropsychopharmacol. 2009;34:759–766. doi: 10.1038/npp.2008.138. [DOI] [PubMed] [Google Scholar]
  • 21.Stokes PRA, Mehta MA, Curran HV, Breen G, Grasby PM. Can recreational doses of THC produce significant dopamine release in the humn striatum? Neuroimage. 2009;48:186–190. doi: 10.1016/j.neuroimage.2009.06.029. [DOI] [PubMed] [Google Scholar]
  • 22.Stokes PRA, Egerton A, Watson B, Reid A, Breen G, Lingford-Hughes A, Nutt DJ, Mehta MA. Sinificant decreases in frontal and temporal [11C]- raclopride binding after THC challenge. Neuroimage. 2010;52:1521–1527. doi: 10.1016/j.neuroimage.2010.04.274. [DOI] [PubMed] [Google Scholar]
  • 23.Jaffe JH. Drug addiction and drug abuse. In: Gilman AG, Goodman LS, Murad F, editors. The Pharmacological Basis of Therapeutics. 7th Edition. USA: Macmillan; 1985. [Google Scholar]
  • 24.Huestis MA, Gorelick DA, Heishman SJ, Preston KL, Nelson RA, Moolchan ET, Frank RA. Blockade of effects of smoked marijuana by the CB1-selective cannabinoid receptor antagonist SR141716. Arc Gen Psychiat. 2001;58(4):322–328. doi: 10.1001/archpsyc.58.4.322. [DOI] [PubMed] [Google Scholar]
  • 25.LeFoll B, Gorelick DA, Goldberg S. The future of endocannabinoid-oriented clinical research after CB1 antagonists. Psychopharmacol. 2009;205(1):171–174. doi: 10.1007/s00213-009-1506-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Budney AJ, Hughes JR, Moore BA, Vandrey R. Review of the validity and significance of cannabis withdrawal syndrome. Am J Psychiat. 2004;161(11):1967–1977. doi: 10.1176/appi.ajp.161.11.1967. [DOI] [PubMed] [Google Scholar]
  • 27.Budney AJ, Hughes JR. The cannabis withdrawal syndrome. Curr Opin Psychiat. 2006;19:233–238. doi: 10.1097/01.yco.0000218592.00689.e5. [DOI] [PubMed] [Google Scholar]
  • 28.American Psychiatric Association DSM V development. www.dsm5.org.
  • 29.Budney AJ, Radonovich KJ, Higgins ST, Wong CJ. Adults seeking treatment for marijuana dependence: a comparison with cocaine-dependent treatment seekers. Exp Clin Psychopharmacol. 1998;6(4):419–426. doi: 10.1037//1064-1297.6.4.419. [DOI] [PubMed] [Google Scholar]
  • 30.Budney AJ, Moore BA, Vandrey RA, Hughes JR. Onset, magnitude, and duration of abstinence effects following heavy marijuana use. Drug Alc Dep. 2002;66:S23. [Google Scholar]
  • 31.Budney AJ, Hughes JR, Moore BA, Novy PL. Marijuana abstinence effects in mariujana smokers maintained in thein home environment. Arc Gen Psychiat. 2001;58:917–924. doi: 10.1001/archpsyc.58.10.917. [DOI] [PubMed] [Google Scholar]
  • 32.Stephens RS, Roffman RA, Simpson EE. Treating adult marijuana dependence: a test of the relapse prevention model. J Consult Clin Psychol. 1994;62:92–99. doi: 10.1037//0022-006x.62.1.92. [DOI] [PubMed] [Google Scholar]
  • 33.Stephens RS, Roffman RA, Curtin L. Extended versus brief treatment for marijuana use. J Consult Clin Psychol. 2000;68:898–908. [PubMed] [Google Scholar]
  • 34.Levin KH, Copersino ML, Heishman SJ, Liu F, Kelly DL, Boggs DL, Gorelick DA. Cannabis withdrawal symptoms in non-treatment-seeking adult cannabis smokers. Drug and Alc Dep. 2010;111:120–127. doi: 10.1016/j.drugalcdep.2010.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hart CL. Increasing treatment options for cannabis dependence: a review of potential pharmacotherapies. Drug Alc Dep. 2005;80:147–159. doi: 10.1016/j.drugalcdep.2005.03.027. [DOI] [PubMed] [Google Scholar]
  • 36.Nordstrom BR, Levin FR. Treatment of cannabis use disorders: a review of the literature. Am J Add. 2007;16(5):331–342. doi: 10.1080/10550490701525665. [DOI] [PubMed] [Google Scholar]
  • 37.Benyamina A, Lecacheux M, Blecha L, Reynaud M, Lukasiewcz M. Pharmacotherapy and psychotherapy in cannabis withdrawal and dependence. Expt Rev in Neurotherapy. 2008;8(3):479–491. doi: 10.1586/14737175.8.3.479. [DOI] [PubMed] [Google Scholar]
  • 38.Vandrey R, Haney M. Pharmacotherapy for cannabis dependence: how close are we? CNS Drugs. 2009;23(7):543–553. doi: 10.2165/00023210-200923070-00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Löscher W. Basic pharmacology of valproate: a review after 35 years of clinical use for the treatment of epilepsy. CNS Drugs. 2002;16:669–694. doi: 10.2165/00023210-200216100-00003. [DOI] [PubMed] [Google Scholar]
  • 40.Artigas F, Nutt DJ, Shelton R. Mechanism of action of antidepressants. Psychopharmacol Bull. 2002;36 Suppl 2:123–132. [PubMed] [Google Scholar]
  • 41.Dwoskin LP, Rauhut AS, King-Pospisil KA, Bardo MT. Review of the pharmacology and clinical profile of bupropion, an anti-depressant and tobacco use cessation agent. CNS Drug Rev. 2006;12:178–207. doi: 10.1111/j.1527-3458.2006.00178.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Haney M, Ward AS, Comer SD, Hart CL, Foltin RW, Fischman MW. Bupropion SR worsens mood during marijuana withdrawal in humans. Psychopharmacol. 2001;155:171–179. doi: 10.1007/s002130000657. [DOI] [PubMed] [Google Scholar]
  • 43.Haney M, Hart CL, Vosburg SK, et al. Marijuana withdrawal in humans: effects of oral THC or divalproex. Neuropsychopharmacol. 2004;29:158–170. doi: 10.1038/sj.npp.1300310. [DOI] [PubMed] [Google Scholar]
  • 44.Haney M, Hart CL, Ward AS, Foltin RW. Nefazodone decreased anxiety during marijuana withdrawal in humans. Psychopharmacol (Berl) 2003;165:157–165. doi: 10.1007/s00213-002-1210-3. [DOI] [PubMed] [Google Scholar]
  • 45.Budney AJ, Vandrey RG, Hughes JR, Moore BA, Bahrenburg B. Oral delta-9-tetrahydrocannabinol suppresses cannabis withdrawal symptoms. Drug Alc Dep. 2007;86(1):22–29. doi: 10.1016/j.drugalcdep.2006.04.014. [DOI] [PubMed] [Google Scholar]
  • 46.Levi MS, Borne RF. A review of chemical agents in the pharmacotherapy of addiction. Curr Medic Chem. 2002;9:1807–1818. doi: 10.2174/0929867023368980. [DOI] [PubMed] [Google Scholar]
  • 47.Haney M, Hart CL, Vosburg SK, Comer SD, Reed SC, Foltin RW. Effects of THC and lofexidine in a human laboratory model of marijuana withdrawal and relapse. Psychopharmacol. 2008;197:157–168. doi: 10.1007/s00213-007-1020-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Akhondzadeh S, Ahmadi-Abhari SA, Assadi SM, Shabestari OL, Kashani AR, Farzanehgan ZM. Double-blind randomized controlled trial of baclofen vs. clonidine in the treatment of opiate withdrawal. J Clin Pharmacol Therap. 2000;25:347–353. doi: 10.1046/j.1365-2710.2000.00295.x. [DOI] [PubMed] [Google Scholar]
  • 49.Liappas J, Paparrigopoulos T, Tzavellas E, Rabavilas A. Mirtazapine and venlafaxine in the management of collateral psychopathology during alcohol detoxification. Prog Neuropsychopharmacol. 2005;29:55–60. doi: 10.1016/j.pnpbp.2004.10.005. [DOI] [PubMed] [Google Scholar]
  • 50.Yoon SJ, Pae CU, Kim DJ, Namkoong K, Lee E, Oh DY, Lee YS, Shin DH, Jeong YC, Kim JH, Choi SB, Hwang IB, Shin YC, Cho SN, Lee HK, Lee CT. Mirtazapine for patients with alcohol dependence and comorbid depressive disorders: a multicentre, open label study. Prog Neuropsychopharmacol. 2006;30:1196–1201. doi: 10.1016/j.pnpbp.2006.02.018. [DOI] [PubMed] [Google Scholar]
  • 51.Haney M, Hart CL, Vosburg SK, Comer SD, Collins S, Reed SC, Cooper Z, Foltin RW. Effects of baclofen and mirtazapine on a laboratory model of marijuana withdrawal and relapse. Psychopharmacol. 2010;211:233–244. doi: 10.1007/s00213-010-1888-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Levin FR, Kleber HD. Use of dronabinol for cannabis dependence: two case reports and review. Am J Add. 2008;17:161–164. doi: 10.1080/10550490701861177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Comer SD, Sullivan MA, Yu E, Rothenberg JL, Kleber HD, Kampman K, Dackis C, O’Brien CP. Injectable, Sustained-Release Naltrexone for the Treatment of Opioid Dependence A Randomized, Placebo-Controlled Trial. ArcGen Psychiat. 2006;63:210–218. doi: 10.1001/archpsyc.63.2.210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Huestis MA, Boyd SJ, Heishman SJ, Preston KL, Bonnet D, Le Fur G, Gorelick DA. Single and multiple doses of rimonabant antagonize acute effects of smoked cannabis in male cannabis users. Psychopharmacol (Berl) 2007;194(4):505–515. doi: 10.1007/s00213-007-0861-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Clapper JR, Mangieri RA, Piomelli D. The endocannabinoid system as a target for the treatment of cannabis dependence. Neuropharmacol. 2009;56 Suppl 1:235–243. doi: 10.1016/j.neuropharm.2008.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Wachtel SR, de Wit H. Naltrexone does not block the subjective effects of oral Delta(9)- tetrahydrocannabinol in humans. Drug Alc Dep. 2000;59:251–260. doi: 10.1016/s0376-8716(99)00127-1. [DOI] [PubMed] [Google Scholar]
  • 57.Haney M, Bisaga A, Foltin RW. Interaction between naltrexone and oral THC in heavy marijuana smokers. Psychopharmacol. 2003;166:77–85. doi: 10.1007/s00213-002-1279-8. [DOI] [PubMed] [Google Scholar]
  • 58.Greenwald MK, Stitzer ML. Antinociceptive, subjective and behavioral effects of smoked marijuana in humans. Drug Alc Dep. 2000;59:261–275. doi: 10.1016/s0376-8716(99)00128-3. [DOI] [PubMed] [Google Scholar]
  • 59.Haney M. Opioid antagonism of cannabinoid effects: differences between marijuana smokers and nonmarijuana smokers. Neuropsychopharmacol. 2007;32:1391–1403. doi: 10.1038/sj.npp.1301243. [DOI] [PubMed] [Google Scholar]
  • 60.Cooper ZD, Haney M. Opioid antagonism enhances marijuana's effects in heavy marijuana smokers. Psychopharmacol (Berl) 2010;211(2):141–148. doi: 10.1007/s00213-010-1875-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Koob GF. Drugs of abuse: anatomy, pharmacology and function of reward pathways. Trends Pharmacol Sci. 1992;13:177–184. doi: 10.1016/0165-6147(92)90060-j. [DOI] [PubMed] [Google Scholar]
  • 62.Di Chiara G, North RA. Neurobiology of opiate abuse. Trends Pharmacol Sci. 1992;13:185–193. doi: 10.1016/0165-6147(92)90062-b. [DOI] [PubMed] [Google Scholar]
  • 63.Wise RA. Neurobiology of addiction. Curr op Neurobiol. 1996;6:243–251. doi: 10.1016/s0959-4388(96)80079-1. [DOI] [PubMed] [Google Scholar]
  • 64.Volkow ND, Wang GJ, Fowler JS, Hitzemann R, Angrist B, Gatley SJ, Logan J, Ding YS, Pappas N. Association of Methylphenidate-induced craving with changes in right striatoorbitofrontal metabolism in cocaine abusers: implications in addiction. Am J Psychiat. 1999;156(1):19–26. doi: 10.1176/ajp.156.1.19. [DOI] [PubMed] [Google Scholar]
  • 65.Caspi A, Moffitt TE, Cannon M, McClay J, Murray R, Harrington H, Taylor A, Arseneault L, Williams B, Braithwaite A, Poulton R, Craig IW. Moderation of the effect of adolescent-onset cannabis use on adult psychosis by a functional polymorphism in the catechol-O-methyltransferase gene: longitudinal evidence of a gene X environment interaction. Biol Psychiat. 2005;57(10):1117–1127. doi: 10.1016/j.biopsych.2005.01.026. [DOI] [PubMed] [Google Scholar]
  • 66.Zammit S, Spurlock G, Williams H, Norton N, Williams N, O'Donovan MC, Owen MJ. Genotype effects of CHRNA7, CNR1 and COMT in schizophrenia: interaction with tobacco and cannabis use. Brit J Psychiat. 2007;191:402–407. doi: 10.1192/bjp.bp.107.036129. [DOI] [PubMed] [Google Scholar]
  • 67.Shafa R. COMT- Inhibitors may be a Promising Tool in Treatment of Marijuana Addiction Trials. A J Addict. 2009;18:322. [Google Scholar]
  • 68.Kau KS, Madayag A, Mantsch JR, et al. Blunted cystine-glutamate antiporter function in the nucleus accumbens promotes cocaine induced drug seeking. Neuroscience. 2008;155:530–537. doi: 10.1016/j.neuroscience.2008.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.LaRowe SD, Mardikian P, Malcolm R, Myrick H, Kalivas P, McFarland K, Saladin M, McRae A, Brady K. Safety and tolerability of N-acetylcysteine in cocaine-dependent individuals. Am J Addictn. 2006;15:105–110. doi: 10.1080/10550490500419169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Knackstedt LA, LaRowe S, Mardikian P, et al. The role of cystineglutamate exchange in nicotine dependence in rats and humans. Biol Psychiat. 2009;65:841–845. doi: 10.1016/j.biopsych.2008.10.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Gray KM, Watson NL, Carpenter MJ, Larowe SD. Nacetylcysteine (NAC) in young marijuana users: an open-label pilot study. Am J Add. 2010;19(2):187–189. doi: 10.1111/j.1521-0391.2009.00027.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wilens TE. Attention Deficit Hyperactivity Disorder and Substance Use Disorders. AJ Psychiat. 2006;163:2059–2063. doi: 10.1176/ajp.2006.163.12.2059. [DOI] [PubMed] [Google Scholar]
  • 73.Tirado CF, Goldmanm M, Lynch K, Kampman KM, Obrien CP. Atomoxetine for treatment of marijuana dependence: a report on the efficacy and high incidence of gastrointestinal adverse events in a pilot study. Drug Alc Dep. 2008;94:254–257. doi: 10.1016/j.drugalcdep.2007.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.McRae-Clark AL, Carter RE, Killeen TK, Carpenter MJ, White KG, Brady KT. A Placebo-Controlled Trial of Atomoxetine in Marijuana-Dependent Individuals with Attention Deficit Hyperactivity Disorder. Am J Add. 2010;19(6):481–489. doi: 10.1111/j.1521-0391.2010.00076.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Blier P, Bergeron R, de Montigny C. Selective activation of post-synaptic 5-HT1A receptors induces rapid antidepressant response. Neuropsychopharmacol. 1997;16(5):333–338. doi: 10.1016/S0893-133X(96)00242-4. [DOI] [PubMed] [Google Scholar]
  • 76.Gobert A, Rivet JM, Cistarelli L, Melon C, Millan MJ. Buspirone modulates basal and fluoxetine-stimulated dialysate levels of dopamine, noradrenaline and serotonin in the frontal cortex of freely moving rats: activation of serotonin1A receptors and blockade of alpha2-adrenergic receptors underlie its actions. Neuroscience. 1999;93(4):1251–1262. doi: 10.1016/s0306-4522(99)00211-0. [DOI] [PubMed] [Google Scholar]
  • 77.McRae AL, Brady KT, Carter RE. Buspirone for Treatment of Marijuana Dependence: A Pilot Study. Am J Add. 2006;15:404. doi: 10.1080/10550490600860635. [DOI] [PubMed] [Google Scholar]
  • 78.McRae-Clark AL, Carter RE, Killeen TK, Carpenter MJ, Wahlquist AE, Simpson SA, Brady KT. A placebo-controlled trial of buspirone for the treatment of marijuana dependence. Drug Alc Dep. 2009;105(1–2):132–138. doi: 10.1016/j.drugalcdep.2009.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cui SS, Bowen RC, Gu GB, Hannesson DK, Yu PH, Zhang X. Prevention of cannabinoid withdrawal syndrome by lithium: involvement of oxytocinergic neuronal activation. J Neurosci. 2001;21:9867–9876. doi: 10.1523/JNEUROSCI.21-24-09867.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Bowen R, McIlwrick J, Baetz M, Zhang X. Lithium and marijuana withdrawal. Can J Psychiat. 2005;50:240–241. doi: 10.1177/070674370505000410. [DOI] [PubMed] [Google Scholar]
  • 81.Winstock AR, Lea T, Copeland J. Lithium carbonate in the management of cannabis withdrawal in humans: an open-label study. J Psychopharmacol. 2009;23:84–93. doi: 10.1177/0269881108089584. [DOI] [PubMed] [Google Scholar]
  • 82.Levin FR, McDowell D, Evans SM, Nunes E, Akerele E, Donovan S, Vosburg SK. Pharmacotherapy for marijuana dependence: a double-blind, placebo-controlled pilot study of divalproex sodium. Am J Add. 2004;13:21–32. doi: 10.1080/10550490490265280. [DOI] [PubMed] [Google Scholar]
  • 83.Carpenter KM, McDowell D, Brooks DJ, Cheng WY, Levin FR. A preliminary trial: double-blind comparison of nefazodone, bupropion-SR, and placebo in the treatment of cannabis dependence. Am J Add. 2009;18(1):53–64. doi: 10.1080/10550490802408936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Petronis KR, Samuels JF, Moscicki EK, Anthony JC. An epidemiologic investigation of potential risk factors for suicide attempts. Soc Psychiat Psychiat Epidem. 1990;25:193–199. doi: 10.1007/BF00782961. [DOI] [PubMed] [Google Scholar]
  • 85.Rowe MG, Fleming MF, Barry KL, Manwell LB, Kropp S. Correlates of depression in primary care. Journal of Family Practice. 1995;41:551–558. [PubMed] [Google Scholar]; Sarel S, Mechoulam R, Agranat I. Trends in Medicinal Chemistry. Oxford, UK: Blackwell Publ.; 1991. [Google Scholar]
  • 86.Cornelius JR, Clark DB, Bukstein OG, Bimaher B, Salloum IM, Brown SA. Acute phase and five year follow up study of fluoxetine in adolescents with major depression and comorbid substance abuse disorder: a review. Add Behavs. 2005;30:1824–1833. doi: 10.1016/j.addbeh.2005.07.007. [DOI] [PubMed] [Google Scholar]
  • 87.Cornelius JR, Buksteinb OG, Douaihy AB, Clark DB, Chung TA, Daley DC, Wood DS, Brown SJ. Double-blind fluoxetine trial in comorbid MDD–CUD youth and young adults. Drug Alc Dep. 2010;112:39–45. doi: 10.1016/j.drugalcdep.2010.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Dhawan K, Kumar S, Sharma A. Reversal of cannabinoids (delta9-THC) by the benzoflavone moiety from methanol extract of Passiflora incarnata Linneaus in mice: a possible therapy for cannabinoid addiction. Jf Pharm Pharmacol. 2002;54(6):875–881. doi: 10.1211/0022357021779069. [DOI] [PubMed] [Google Scholar]
  • 89.Solinas M, Scherma M, Fattore L, Stroik J, Wertheim C, Tanda G, Fratta W, Goldberg SR. Nicotinic alpha 7 receptors as a new target for treatment of cannabis abuse. J Neurosci. 2007;27(21):5615–5620. doi: 10.1523/JNEUROSCI.0027-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Schlosburg JE, Carlson BL, Ramesh D, Abdullah RA, Long JZ, Cravatt BF, Lichtman AH. Inhibitors of endocannabinoid-metabolizing enzymes reduce precipitated withdrawal responses in THC-dependent mice. AAPS J. 2009;11(2):342–352. doi: 10.1208/s12248-009-9110-7. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES