Abstract
Marijuana (Cannabis sativa) is the most commonly used illicit drug worldwide as well as in the Unites States. Prolonged use of marijuana or repeated administration of its primary psychoactive constituent, Δ9-tetrahydrocannabinol (THC), can lead to physical dependence in humans and laboratory animals. The changes that occur with repeated cannabis use include alterations in behavioral, physiological, and biochemical responses. A variety of withdrawal responses occur in cannabis-dependent individuals: anger, aggression, irritability, anxiety and nervousness, decreased appetite or weight loss, restlessness, and sleep difficulties with strange dreams. But the long half-life and other pharmacokinetic properties of THC result in delayed expression of withdrawal symptoms, and because of the lack of contiguity between drug cessation and withdrawal responses the latter are not readily recognized as a clinically relevant syndrome. Over the past 30 years, a substantial body of clinical and laboratory animal research has emerged supporting the assertion that chronic exposure to cannabinoids produces physical dependence and may contribute to drug maintenance in cannabis-dependent individuals. However, no medications are approved to treat cannabis dependence and withdrawal. In this review, we describe preclinical and clinical research that supports the existence of a cannabinoid withdrawal syndrome. In addition, we review research evaluating potential pharmacotherapies (e.g., THC, a variety of antidepressant drugs, and lithium) to reduce cannabis withdrawal responses and examine how expanded knowledge about the regulatory mechanisms in the endocannabinoid system may lead to promising new therapeutic targets.
Keywords: Δ9-tetrahydrocannabinol (THC), anandamide, cannabis dependence, CB1 receptor, endogenous cannabinoid (endocannabinoid), fatty acid amide hydrolase (FAAH), marijuana, monoacylglycerol lipase (MAGL), withdrawal
Introduction
For more than 30 years marijuana has been the most-used illicit drug by teenagers and adults in the UnitedStates—42% of 18-year-olds and 82% of 50-year-olds report lifetime use (Johnston et al. 2010). Although the perception persists that marijuana use is innocuous and lacks dependence liability, the first record of cannabis withdrawal was published in the 1940s (Wallace and Cunningham 1944) and the medical community is now beginning to accept the idea that cannabis-related disorders represent a clinically significant public health problem (for review, Weinstein and Gorelick 2011). According to the Drug Abuse Warning Network, marijuana was involved in 374,435 hospital emergency department visits or 37.7% of all such visits involving an illicit drug in 2008, and marijuana users accounted for 18.5% of the 177,879 drug-related emergency department visits that year by patients seeking detoxification or substance abuse treatment services (SAMHSA 2011).
In this review, we discuss data from human surveys, retrospective and clinical studies, and preclinical research characterizing cannabis dependence. The preponderance of evidence suggests that cannabis dependence should be considered an important medical condition that requires clinical intervention. Cannabis-dependent individuals who cease using the drug experience a variety of withdrawal symptoms that are sufficiently severe to contribute to drug maintenance, thus highlighting its addictive properties. We review both clinical and preclinical studies examining a variety of pharmacotherapies to alleviate withdrawal signs.
Almost half a century has passed since the structure of the primary psychoactive constituent of Cannabis sativa, Δ9-tetrahydrocannabinol (THC1), was first elucidated (Gaoni and Mechoulam 1964). Since then, more than 70 other phytocannabinoids have been discovered (Elsohly and Slade 2005) and hundreds, if not thousands, of cannabinoids have been synthesized.
Studies have shown that THC and many other cannabinoids bind to and activate two types of cannabinoid (CB1) receptors that have been cloned: CB1 (Matsuda et al. 1990) and CB2 (Gerard et al. 1991). CB1 receptors, which are heterogeneously expressed throughout the central nervous system (CNS) and periphery (Felder and Glass 1998; Herkenham et al. 1990; Matsuda et al. 1993), are responsible for most of the pharmacological actions of THC, particularly in the CNS. CB2 receptors are associated with immune cells (Klein et al. 2003) and were initially thought to be expressed solely in the periphery, but more recently they were found to be expressed in microglial cells (Cabral and Marciano-Cabral 2005) and in neurons (Van Sickle et al. 2005) in the brain.
A major breakthrough in cannabinoid pharmacology came with the discovery of the endogenous cannabinoids (endocannabinoids) N-arachidonylethanolamine (anandamide) (Devane et al. 1992) and 2-arachidonylglycerol (2-AG1) (Mechoulam et al. 1995; Sugiura et al. 1995). The endocannabinoid system comprises the CB receptors, the endocannabinoids, and the enzymes that regulate endocannabinoid biosynthesis and degradation (Ahn et al. 2008). Blockade of the enzymes fatty acid amide hydrolase (FAAH1) and monoacylglycerol lipase (MAGL1) raises brain levels of anandamide (Kathuria et al. 2003; Lichtman et al. 2004) and 2-AG (Long et al. 2009) respectively. These catabolic enzymes are targets for the development of selective inhibitors to treat cannabis-related disorders as well as pain, inflammation, and anxiety.
We review evidence from laboratory animal and human studies showing that repeated administration of cannabinoids can result in physical dependence. Emerging data indicate that cannabinoid-dependent laboratory animals and humans display physical withdrawal responses upon drug cessation. In addition, we provide an overview of preclinical and clinical research examining pharmacotherapies to treat cannabis dependence. In animal studies, THC administration and inhibition of endocannabinoid catabolic enzymes represent promising approaches to reduce cannabinoid withdrawal responses. We discuss several clinical studies showing that oral THC reduces cannabis withdrawal responses in cannabis-dependent patients.
Cannabis Use, Dependence, and Withdrawal in Humans
Although the current Diagnostic and Statistical Manual of Mental Disorders (4th ed., called the DSM-IV; APA 1994) and International Statistical Classification of Diseases and Related Health Problems (10th ed., called the ICD-10; WHO 2007), the two most common guidebooks used by medical professionals to diagnose substance use disorders, include criteria and symptoms for diagnosing cannabis abuse and dependence, only the latter recognizes a withdrawal syndrome as a component of a cannabis dependence disorder. The DSM-IV reflects the attitude of many in medicine and the general public that cannabis is not a physically addictive substance in which a withdrawal syndrome can produce clinically relevant symptoms of a severity and duration to affect substance-use behavior. This belief is probably due to a multitude of factors, such as the relatively slow onset and unique constellation of the withdrawal syndrome.
Cannabis is the most commonly used illicit drug worldwide. In the United States approximately 56% of young adults (19–28 years old) have at least tried cannabis (Johnston et al. 2010). This high prevalence allows for many people to have personal or anecdotal experience with marijuana without necessarily having personal interactions with dependent users. Although only about 3–4% of individuals who have ever tried cannabis meet the criteria for a cannabis use disorder (compared with 15–25% for cocaine), the total number of Americans classified with such disorders is 4.3 million, more than twice that of cocaine and heroin combined (SAMHSA 2008).
The severity of cannabis withdrawal is not generally associated with symptoms that require hospitalization or are viewed as potentially life threatening. Furthermore, only a subset of regular marijuana users experience a clustering of symptoms upon cessation of use; estimates range from 1 in 6 to half of all such users (Budney et al. 1999; Wiesbeck et al. 1996). Common symptoms observed during cannabis withdrawal include anger, aggression, irritability, anxiety and nervousness, decreased appetite or weight loss, restlessness, and sleep difficulties with strange dreams (Budney and Hughes 2006). Although the immediate physical impact of these symptoms is mild when compared with certain other drugs of abuse, as discussed below the comprehensive impact of the cannabis withdrawal syndrome is becoming better understood.
Controlled Laboratory Studies
Before the cloning of cannabinoid receptors, discovery of the endogenous cannabinoid system, and development of selective cannabinoid agonists and antagonists, early studies of marijuana smokers indicated potential signs of tolerance and withdrawal (Williams et al. 1946). In the 1970s, Jones and colleagues set out to define the physiological and psychoactive effects of cannabis in controlled laboratory settings (Jones and Benowitz 1976; Jones et al. 1981). Human subjects were given varying oral doses of THC in a double-blind fashion, spaced evenly throughout the day to maintain consistent drug levels. THC produced profound tolerance after repeated administration, as assessed by the following: self-reported intoxication, time spent in REM sleep, psychomotor task performance, and numerous autonomic physiological effects. The investigators also identified a subset of behaviors that increased dramatically among subjects during the 4 days after cessation of the drug, including disturbances in sleeping and eating, sweats and chills, tremors and restlessness, and irritability. Most of these symptoms subsided after a resumption of THC intake or marijuana smoking (Jones et al. 1981; Jones and Benowitz 1976). Subsequent studies of marijuana smokers in the laboratory over periods of use and cessation replicated these findings but lacked the controls and precise measurements of the earlier laboratory studies (Georgotas and Zeidenberg 1979; Nowlan and Cohen 1977).
More recently, Haney and colleagues (1999) used data from both laboratory and survey findings to ascertain how heavy users of cannabis respond to use and abstinence in terms of cognitive function, subjective drug effects, and detailed cannabis-specific withdrawal symptoms. Parallel studies using identical methodologies evaluated the effects of oral THC and smoked marijuana. Both types of studies showed increases in ratings of anxiety and irritability and disturbances in food intake, but sleep patterns seemed more sensitive to abstinence from oral THC, and marijuana abstinence impaired performance on a task measuring attention. Other controlled studies reported that chronic marijuana users show deficits associated with complex decision making and cognitive planning (Hermann et al. 2009; Wesley et al. 2011; Whitlow et al. 2004). These studies marked a renewed effort to define the symptoms and impact of cannabis dependence.
Retrospective and Large Population Studies
Although laboratory studies provide for a controlled environment, increased compliance, and around-the-clock data collection, they generally incorporate relatively small sample sizes (on the order of a few dozen) and are conducted on a subset of relatively heavy cannabis users (for a critique of these and other studies discussed in this review, see Smith 2002). In contrast, large datasets are used in retrospective studies in which subjects are asked to recall their own attempts to abstain from marijuana use, providing insight into real-world conditions.
In one such study, thousands of people ranging from cannabis-naïve to daily smokers were interviewed about their cannabis withdrawal symptoms (Wiesbeck et al. 1996). Examining for symptoms similar to those found in laboratory studies, the authors noted that only a small percentage of infrequent and even daily users reported each withdrawal symptom. However, approximately 16% of the regular marijuana smokers recalled distinct clusters of withdrawal symptoms upon cessation, and these subjects were more than twice as likely to meet DSM-III criteria for cannabis and other drug dependencies.
Budney and colleagues (1999) conducted a retrospective study of a group seeking treatment for cannabis use and found that over half the subjects reported at least four moderate withdrawal symptoms during their last attempt to stop. A more recent multisite study established that retrospective reports of specific marijuana withdrawal symptoms were similar in a general population sample to symptoms in treatment-seeking individuals (Mennes et al. 2009).
Large-scale data collection, such as data mining of national surveys on drug use, has been a useful approach to investigate cannabis dependence. One study used the Australian National Survey of Mental Health and Wellbeing, in which the criteria of both the DSM-IV and ICD-10 were used to identify cannabis-dependent users. A very high percentage (>85%) of individuals classified as having cannabis dependence disorder by both sets of criteria were identified as using cannabis to abate withdrawal symptoms and failing either to control their use or to abstain (Swift et al. 2001).
A similar large-scale study in the United States used data from the National Epidemiologic Survey on Alcohol and Related Conditions (NESARC). A comparison of frequent (≥3 times per week) use of cannabis alone versus in combination with other drugs showed a similar frequency of reported withdrawal symptoms associated with cannabis, with or without concurrent use of other substances (Hasin et al. 2008). The incidence of symptoms correlated highly with the amount of cannabis used, levels of distress, and subsequent general substance use to relieve symptoms. This association supports the notion that cannabis withdrawal symptoms contribute to the maintenance of cannabis use in dependent individuals.
Outpatient Cohort and Self-Report Studies
The largest volume of published research describes self-report and monitored outpatient studies, which allow for immediate reporting of symptoms and timeline information without constant supervision and monitoring. Measurement of urinary drug metabolites is a common method to verify initial drug use and abstinence from cannabis or other drugs.
Patients identified as heavy cannabis users typically show a moderate set of symptoms, both physical (e.g., weight loss) and psychological (e.g., marijuana craving), which appear within a day of halting cannabis use and usually abate in a week (Budney et al. 2001; Dawes et al. 2006; Kouri and Pope 2000; Preuss et al. 2010). This time course corresponds to rather precipitous drops in THC metabolites in the urine during the first few days of abstinence. Other common symptoms, such as irritability and aggression, can persist for weeks (Budney et al. 2003; Kouri et al. 1999).
Although many of these studies contain sample sizes not much larger than those of laboratory studies, they allow a more thorough examination of how cannabis withdrawal symptoms affect the daily life of users and can be readily performed by numerous treatment clinics to build a converging set of data from multiple sites.
Cannabis Withdrawal Syndrome in Perspective
The exact timeline and symptoms of the cannabis withdrawal syndrome vary across studies, but the growing consensus is that withdrawal symptoms contribute to continued drug use. Cannabis withdrawal is comparable in severity and scope to tobacco withdrawal and contributes to relapse to only a slightly lesser extent (Budney et al. 2008).
Bonn-Miller and Moos (2009) evaluated marijuana use in male inpatients treated for substance use disorders in Department of Veterans Affairs residential substance abuse programs and reported that anxiety symptoms at treatment discharge were associated with a 12-month relapse to marijuana use. Although this finding is consistent with the idea that increases in anxiety after marijuana discontinuation may be predictive of relapse, the study did not report whether these patients were marijuana dependent.
Other studies also show that the occurrence of withdrawal symptoms may predict marijuana users who will relapse soon after a prolonged outpatient abstinence period (Chung et al. 2008; Cornelius et al. 2008) and increases the likelihood that these users will relapse into continued heavy use (Moore and Budney 2003). Withdrawal symptoms do not, however, appear to predict relapse after 2 or more years of abstinence (Arendt et al. 2007).
Studies are beginning to examine the interactions of cannabis use with other drug use and have shown that concurrent cessation of tobacco and cannabis use is associated with temporary increases in withdrawal severity compared with cessation of either alone (Vandrey et al. 2008), and the severity of cannabis withdrawal symptoms may lead to the increased use and craving of alcohol (Peters and Hughes 2010). Cannabis users often differentially use alcohol, tobacco, or cannabis to reduce the severity of specific symptoms (Copersino et al. 2006).
Taken together, the evidence shows that withdrawal from cannabis use produces a distinct syndrome that increases drug craving and use, thus necessitating research into therapeutic treatment.
Preclinical Studies of Cannabinoid Dependence
Preclinical studies in a variety of laboratory animals show that repeated administration of THC or other cannabinoid agonists results in dependence. Animal models for assessing dependence also measure reinforcing and rewarding properties—such as self-administration, conditioned place preference, and intracranial self-stimulation (for a complete review, Panagis et al. 2008)—as well as withdrawal signs, which can include both physiological symptoms and indicators of emotional state.
Characterization of Cannabinoid Withdrawal
The two general approaches used to induce a state of drug withdrawal in preclinical drug dependence studies are spontaneous withdrawal and precipitated withdrawal. Spontaneous withdrawal occurs after abrupt cessation of the drug, which is metabolized and cleared from the body. In precipitated withdrawal, an appropriate selective receptor antagonist is used to displace the agonist from the receptor, resulting in the rapid onset of withdrawal symptoms. The specific withdrawal symptoms, intensity, and duration depend on the pharmacological characteristics of the compound; drugs from the same class generally share similar withdrawal syndromes. The precipitated withdrawal model is used more often than the spontaneous model because of the long half-life of THC and because subtle withdrawal effects in the latter model are difficult to observe and quantify.
The results of preclinical studies using precipitated and spontaneous withdrawal procedures are shown in Table 1.
Table 1.
Preclinical studies investigating cannabinoid withdrawal using precipitated and spontaneous withdrawal procedures
Species | Type of withdrawal | Agonist, dosing regimen, route of administration | Dependent variable | Reference |
---|---|---|---|---|
Mouse | Precipitated | THC, 6½ days: 10 mg/kg s.c. (b.i.d.) | Somatic signs | Cook et al. 1998 |
Mouse | Precipitated | THC, 5½ days: 10 20 mg/kg i.p. (b.i.d.) | Somatic signs, adenylyl cyclase overshoot in cerebellum | Hutcheson et al. 1998 |
Mouse | Precipitated | THC, 5½ days: 20 mg/kg i.p. (b.i.d.) | Somatic signs | Valverde et al. 2000 |
Mouse | Precipitated | THC, 6½ days: 10 mg/kg s.c. (b.i.d.) | Somatic signs | Lichtman et al. 2001a |
Mouse | Precipitated | THC, 5½ days: 20 mg/kg i.p. (b.i.d.) | Somatic signs, body weight | Anggadiredja et al. 2003 |
Mouse | Spontaneous | CP, 6½ days: 0.5 mg/kg i.p. (b.i.d.) | Increased locomotor activity, endocrine gene transcription levels | Oliva et al. 2003 |
Mouse | Precipitated | THC, MAR, 5 days: 200 mg; 5 or 10 mg/kg i.v. (s.i.d.) | Somatic signs | Wilson et al. 2006 |
Mouse | Precipitated | THC, 5½ days: 20 mg/kg i.p. (b.i.d.) | Somatic signs | Tourino et al. 2007 |
Mouse | Precipitated | THC, 4½ days: 25 mg/kg s.c. (b.i.d.) | Somatic signs, activity | Huang et al. 2009 |
Mouse | Precipitated | THC, 5½ days: 50 mg/kg s.c. (b.i.d.); 10 mg/kg (s.i.d.) | Somatic signs | Schlosburg et al. 2009 |
Mouse | Precipitated | THC, AEA, 5½ days: 50 mg/kg s.c. (b.i.d.) | Somatic signs | Falenski et al. 2010 |
Mouse | Precipitated | THC, 10 days: 10 mg/kg s.c. (s.i.d.) | Anxiogenic effects (plus maze) | Huang et al. 2010 |
Mouse | Precipitated | JZL 184, THC, 10 days: 40 mg/kg i.p. (s.i.d.) | Somatic signs | Schlosburg et al. 2010 |
Rat | Precipitated | THC, 4 days: 0.5–4; 2.5–20; 12.5–100 mg/kg/hr i.p. | Somatic signs | Aceto et al. 1995 |
Rat | Precipitated | THC, 6½ days: 15 mg/kg i.p. (b.i.d.) | Activity | Tsou et al. 1995 |
Rat | Precipitated, spontaneous | THC, 4 days: 12.5–100; 2.5–20; 0.5–4 mg/kg/24 hr i.p. (cont.) | Somatic signs with precipitated only | Aceto et al. 1996 |
Rat | Precipitated | THC, 6½ days: 15 mg/kg i.p. (b.i.d.) | Somatic signs | Diana et al. 1998 |
Rat | Precipitated | CP, 6½ days: 0.4 mg/kg i.p. (b.i.d.) | Somatic signs, activity | Rubino et al. 1998 |
Rat | Precipitated | THC, 6 days: 10–30; 10–40; 10–50 mg/kg s.c. (b.i.d.) | Operant rate | Beardsley and Martin 2000 |
Rat | Precipitated, spontaneous | WIN, 4 days: 1–8; 2–16; 4–16 mg/kg/24 hr i.p. (cont.) | Somatic signs, body weight | Aceto et al. 2001 |
Rat | Precipitated | THC, 4 days: 12.5–100 mg/kg/24 hr i.p. (cont.) | Somatic signs | Breivogel et al. 2003 |
Rat | Precipitated | HU-210, 5½ days: 100 μg/kg i.p. (b.i.d.) | Somatic signs | Cui et al. 2001 |
Rat | Precipitated | THC, 8 days: 10 mg/kg i.p. (b.i.d.) | Somatic signs, activity | Gonzalez et al. 2004 |
Dog | Precipitated | THC, 10 days: 0.6–1 mg/kg i.v. (b.i.d.) | Somatic signs | Lichtman et al. 1998a |
Monkey | Spontaneous | THC, 10 days: 0.05 mg/kg/hr i.v. | Operant rate suppression | Beardsley et al. 1986 |
Monkey | Precipitated, spontaneous | THC, continuous (drug discrimination study): 1 mg/kg (b.i.d.) | Somatic signs, operant rate suppression | Stewart and McMahon 2010 |
AEA, anandamide; b.i.d., twice a day; HU-210, a synthetic cannabinoid; i.p., intraperitoneally; i.v., intravenously; CP, CP 55940; MAR, marijuana; s.c., subcutaneously; s.i.d, once a day; cont., continuous infusion; THC, Δ9-tetrahydrocannabinol; WIN, WIN 55212-2
Spontaneous Cannabinoid Withdrawal in Laboratory Animals
Abrupt cessation of THC after prolonged administration results in delayed onset and long-duration withdrawal symptoms, so the quantification of abrupt withdrawal signs in laboratory animals is challenging (Huestis 2005) and often yields yields mixed results.
The study of somatic withdrawal signs from repeated administration of THC and other cannabinoids has been examined in different animal species. Although one study in rats reported a variety of abnormal behavior signs, such as tremors, wet-dog shakes, and hyperirritability (Kaymakcalan et al. 1977), other studies failed to find significant abrupt withdrawal signs either in rodents (Aceto et al. 1996; Leite and Carlini 1974) or in pigeons after chronic exposure to THC (McMillan et al. 1973). In contrast, rhesus monkeys chronically infused with intravenously administered THC (0.5 mg/kg of body weight 4x/day for 3 weeks) displayed substantial increases in gross movement, eye contact, and baring of teeth during the first week of abstinence (Fredericks and Benowitz 1980). These behavioral effects reflect “rebound” withdrawal symptoms, as they were initially suppressed by acute administration of THC. In another study, food-reinforced operant responding was decreased in monkeys after abrupt cessation of drug infusions (Beardsley et al. 1986). Spontaneous withdrawal responses also occurred after discontinuation of chronic administration of the full cannabinoid receptor agonists WIN 55212 in rats (Aceto et al. 2001) and CP 55940 in mice (Oliva et al. 2003).
The data from these studies suggest that the ability to observe and quantify spontaneous withdrawal effects in experimental animals depends on many factors including species, cannabinoid selection, duration of drug administration, time point at which withdrawal is assessed, and specific endpoints. Yet, although the spontaneous withdrawal approach presents with considerable challenges and may be prone to false negatives because of the slow elimination of THC and its metabolites (Huestis 2005), it is considered to be more valid than precipitated withdrawal for modeling human cannabis withdrawal.
Precipitated Cannabinoid Withdrawal in Laboratory Animals
The development of CB1 receptor antagonists (e.g., rimonabant; Rinaldi-Carmona et al. 1994) has been highly useful in investigations of precipitated withdrawal in cannabinoid-dependent animals. Rimonabant binds with high affinity to the CB1 receptor and antagonizes the pharmacological effects of many cannabinoid receptor agonist activities in both laboratory animals and humans (Compton et al. 1996; Huestis et al. 2001, 2007; Lichtman et al. 1998b; Rinaldi-Carmona et al. 1994; Winsauer et al. 1999).
Somatic Withdrawal Signs
Soon after the discovery of rimonabant, two independent groups used this antagonist in rats to demonstrate somatic precipitated withdrawal signs—wet-dog shakes, forepaw fluttering, chewing, increased horizontal and vertical activity, retropulsion, and ptosis (Aceto et al. 1995; Tsou et al. 1995). Rimonabant also precipitated a profound withdrawal syndrome in mice that were chronically exposed to either THC or marijuana smoke (Wilson et al. 2006). Withdrawal signs such as paw tremors and wet-dog shakes are observed consistently across all strains (Cook et al. 1998; Huang et al. 2009). Other signs such as mastication, sniffing, and piloerection are of low frequency but are scored in a cannabinoid composite withdrawal index (Hutcheson et al. 1998; Ledent et al. 1999; Lichtman et al. 2001b; Tzavara et al. 2000). In THC-dependent dogs, rimonabant precipitated a withdrawal syndrome that included distinct gastrointestinal signs (e.g., diarrhea, vomiting, excessive salivation), decreases in social behavior, and increases in restless behavior and trembling (Lichtman et al. 1998a).
In naïve animals, rimonabant can also produce pharmacological effects that resemble withdrawal symptoms, although these effects are far less in magnitude than those elicited by rimonabant in cannabinoid-dependent subjects (for review, Lichtman and Martin 2005), including increases in ear scratching, head shakes, and increased grooming behavior (Cook et al. 1998; Darmani and Pandya 2000).
These studies show that it is critical to include appropriate vehicle-treated control groups in studies using a CB1 antagonist to precipitate cannabinoid withdrawal to control for intrinsic effects of the drug at testing. Nonetheless, the observation that rimonabant elicits a far greater magnitude of withdrawal-like behavior (e.g., head shakes and paw tremors) in subjects that experience repeated administration of THC or repeated exposure to marijuana smoke than in control animals (Cook et al. 1998; Wilson et al. 2006) supports the use of the precipitated withdrawal model.
Aversive and Subjective Signs
There are few reports examining aversive or emotional responses in rodents undergoing cannabinoid withdrawal. Rimonabant challenge to THC-dependent mice led to less time in the open-arm component of the elevated plus-maze test (Huang et al. 2010), suggesting an anxiogenic-like effect in subjects undergoing cannabinoid withdrawal, but failed to elicit aversive or dysphoric effects in the conditioned place avoidance test (Hutcheson et al. 1998).
Other studies have focused on subjective signs of cannabinoid withdrawal. Monkeys chronically treated with THC demonstrated robust discrimination of the CB1 antagonist rimonabant (McMahon 2006; McMahon and France 2003; Stewart and McMahon 2010), and THC discontinuation produced a similar discriminative stimulus to rimonabant in THC-treated animals. The data suggest that the interoceptive cues of THC cessation–induced abstinence are mediated by the CB1 receptor.
Precipitated Withdrawal with Other Cannabinoid Agonists
Rimonabant has precipitated withdrawal signs after chronic administration of other cannabinoid agonists such as anandamide, methanandamide, WIN 55212, CP 55940, and HU-210 (Aceto et al. 1998, 2001; Rodriguez de Fonseca et al. 1997; Rubino et al. 1998).
The withdrawal syndrome precipitated after chronic anandamide administration is not as robust as that precipitated with other cannabinoids (Costa et al. 2000; Falenski et al. 2010). Mice with persistently elevated anandamide levels after inhibition of FAAH with the inhibitor URB597 and mice lacking the FAAH enzyme do not show any withdrawal symptoms after rimonabant administration (Falenski et al. 2010; Schlosburg et al. 2009).
In contrast, rimonabant elicited a mild intensity of somatic withdrawal signs in mice treated chronically with the irreversible MAGL inhibitor JZL 184 that produced a tenfold increase in levels of 2-AG (Schlosburg et al. 2010). Thus, increases of the two primary endocannabinoids in the brain result in different consequences of physical dependence. This difference may be related to brain 2-AG levels that are already two orders of magnitude greater than the brain levels of anandamide and to the fact that 2-AG is a full CB1 agonist and anandamide a partial CB1 agonist (Howlett and Mukhopadhyay 2000).
Neurobiological Adaptations during Cannabinoid Dependence
Preclinical studies are now beginning to identify the molecular changes that result from repeated exposure to and cessation of cannabinoid use. Such studies provide indicators of the adaptations that drive withdrawal symptomatology and thus improve strategic targeting of therapeutics.
Chronic administration of cannabinoid agonists results in downregulation of the CB1 receptor in several brain regions as measured by radioligand binding (Breivogel et al. 1999). Similarly, chronic treatment with THC produces a time- and region-dependent desensitization of CB1 activity in the G protein (Breivogel et al. 1999; Romero et al. 1997). Other evidence of dysregulation in the endocannabinoid system includes region-specific alterations in endocannabinoid content after precipitated withdrawal in rats and mice (Gonzalez et al. 2004).
Alterations in numerous other neurotransmitter systems are also associated with withdrawal from THC in rodents. After chronic administration of THC, brain levels of serotonin decrease concurrent with increases in its primary metabolite (Taylor and Fennessy 1978, 1982). The finding that various serotonin uptake inhibitors elicited writhing, backward kicks, jumps, and wet-dog shakes in THC-dependent rats suggests serotonergic involvement in cannabinoid withdrawal-like behavior (Verberne et al. 1980). In addition, histamine levels in the brain decrease during initial exposure to THC and during somatic withdrawal induced by the serotonin reuptake inhibitor clomipramine (Verberne et al. 1985). Several studies have shown evidence of upregulation and release of the stress-related peptide corticotropin-releasing factor upon precipitated withdrawal, a common phenomenon during withdrawal from many drugs of abuse (Gonzalez et al. 2004; Rodriguez de Fonseca et al. 1997).
At the intracellular level, the cyclic adenosine mono-phosphate (cAMP) second messenger signaling system appears to be involved in modulating cannabinoid withdrawal. Rimonabant administered to THC-dependent mice resulted in significant increases in basal and forskolin-stimulated adenylyl cyclase activity in the cerebellum but not in other regions (Hutcheson et al. 1998). Similar results were obtained with calcium-calmodulin-stimulated cyclase activity from the cerebella of THC-dependent rats undergoing precipitated withdrawal (Rubino et al. 2000). Rimonabant-precipitated cannabinoid withdrawal also results in upregulation of protein kinase A (PKA) activity, which is downstream of cAMP, in the cerebella of THC-dependent rats (Tzavara et al. 2000). Infusions of the cAMP blocker Rp-8Br-cAMPs into the cerebellum of rats undergoing precipitated withdrawal attenuated PKA activity and the expression of withdrawal signs. Conversely, infusion of Sp-8Br-cAMPs, a cAMP analogue, into the cerebellum elicited cannabinoid withdrawal somatic signs in drug-naïve mice. These findings provide strong evidence for the functional role of the cAMP cascade, particularly in the cerebellum, in modulating withdrawal from cannabinoids.
Pharmacotherapy for Cannabinoid Withdrawal
Attenuation of Cannabinoid Withdrawal Signs in Laboratory Animals
The development of preclinical cannabinoid withdrawal models has made it possible to evaluate potential therapeutic agents for the treatment of cannabis dependence. Compounds investigated for reducing cannabinoid withdrawal responses include cannabinoid substitutes, such as THC and inhibitors of endocannabinoid catabolic enzymes, and non-cannabinoid drugs, such as lithium and clonidine (Table 2).
Table 2.
Preclinical and clinical studies evaluating pharmacological agents in reducing cannabinoid withdrawal responses
Species | Type of withdrawal | Agonist, dosing regimen | Withdrawal response | Treatment drug | Effect on withdrawal | Reference |
---|---|---|---|---|---|---|
Mouse | Precipitated | THC, 6½ days: 10 mg/kg s.c. (b.i.d.) | Somatic signs | Clonidine | Attenuation | Lichtman et al. 2001a |
Mouse | Precipitated | THC, 5½ days: 20 mg/kg i.p. (b.i.d.) | Somatic signs, body weight | Prostaglandin E2 | Attenuation | Anggadiredja et al. 2003 |
Mouse | Precipitated | THC, MAR, 5 days: 200 mg; 5, 10 mg/kg/i.v. (s.i.d.) | Somatic signs | Marijuana | No effect | Wilson et al. 2006 |
Mouse | Precipitated | THC, 5½ days: 20 mg/kg i.p. (b.i.d.) | Somatic signs | MDMA | Attenuation | Tourino et al. 2007 |
Mouse | Precipitated | THC, 5½ days: 10/50 mg/kg s.c. (s.i.d./b.i.d.) | Somatic signs | URB597, JZL 184 | Attenuation | Schlosburg et al. 2009 |
Rat | Precipitated | HU-210, 5½ days: 100 μg/kg i.p. (b.i.d.) | Somatic signs | Lithium | Attenuation | Cui et al. 2001 |
Monkey | Precipitated, spontaneous | THC, cont., 1 mg/kg (b.i.d.) | Head shaking, withdrawal drug discrimination | THC and CP WIN and clonidine; diazepam and cocaine | Full attenuation Partial attenuation of drug discrimination No attenuation |
Stewart and McMahon 2010 |
Human | Spontaneous | MAR, 6 ± 1 days/wk, 6–7 joints/day, inhalation | Subjective ratings | Bupropion | Exacerbation | Haney et al. 2001 |
Human | Spontaneous | MAR, ad libitum/5 joints/day, inhalation | Subjective ratings | Nefazodone | Partial attenuation | Haney et al. 2003 |
Human | Spontaneous | MAR, 1 joint/session, inhalation | Craving, subjective ratings | THC, divalproex | Attenuation; decreased craving, but exacerbated other symptoms | Haney et al. 2004 |
Human | Spontaneous | MAR, ad libitum, inhalation | Subjective ratings | Divalproex | No effect | Levin et al. 2004 |
Human | Spontaneous | MAR, ad libitum | MAR use | Quetiapine (open-label, pilot study) | Reduction in use | Potvin et al. 2004 |
Human | Spontaneous | MAR, 1–15 joints daily, inhalation | Somatic, MCQ | Lithium (pilot study) | Inconclusive | Bowen et al. 2005 |
Human | Spontaneous | MAR, ad libitum, inhalation | Subjective ratings | Buspirone (pilot study) | Attenuation | McRae et al. 2006 |
Human | Spontaneous | MAR, 28½ days/month, 2.6 times/day average, inhalation | Various addiction mood inventories | THC | Attenuation | Budney et al. 2007 |
Human | Spontaneous | MAR, ad libitum | Subjective ratings | THC Lofexidine |
Reversed anorexia weight loss, decreased some withdrawal symptoms, but increased sleep onset latency, did not decrease marijuana relapse; Worsened abstinence- related anorexia, did not attenuate withdrawal, but improved sleep decreased relapse | Haney et al. 2008 |
Human | Spontaneous | MAR, ad libitum | Subjective ratings | Atomoxetine | No effect (adverse gastrointestinal effects) | Tirado et al. 2008 |
Human | Spontaneous | MAR, ad libitum | Subjective ratings | Buspirone | No significant effects | McRae-Clark et al. 2009 |
Human | Spontaneous | MAR, ad libitum | Subjective ratings | Lithium (open-label study) | Inconclusive results | Winstock et al. 2009 |
Abbreviations: b.i.d., twice a day; CP, CP 55940; HU-210, a synthetic cannabinoid; i.p., intraperitoneally; i.v., intravenously; MAR, marijuana; MDMA, 3,4-methylenedioxymethamphetamine; MCQ, Marijuana Craving Questionnaire; s.c., subcutaneously; s.i.d., once a day; THC, Δ9-tetrahydrocannabinol; WIN, WIN 55212-2; cont., continuous infusion
Early studies reported that reintroduction of THC (1) alleviated decreases in operant responding in nonhuman primates undergoing spontaneous withdrawal (Beardsley et al. 1986), (2) decreased clomipramine-precipitated THC withdrawal symptoms in rats (Verberne et al. 1981), and, more recently, (3) reversed rimonabant-precipitated withdrawal signs in THC-dependent mice (Lichtman et al. 2001a). In addition, the α2-adrenergic agonist clonidine dose-dependently attenuated the intensity of precipitated somatic withdrawal signs (Lichtman et al. 2001a) and reduced rimonabant-induced head shaking and rimonabant discriminative stimulus in THC-dependent monkeys (Stewart and McMahon 2010).
In contrast, THC dose-dependently attenuated the intensity of rimonabant-precipitated paw tremors in mice rendered dependent on marijuana smoke, but marijuana itself failed to reverse the precipitated withdrawal effect (Wilson et al. 2006). The lack of effect of marijuana itself was attributed to the lower THC brain levels after exposure to marijuana smoke (203 ng of THC per g of brain tissue) compared with intravenous injection of THC (1862 ng of THC per g of brain tissue).
Morphine also reduced the intensity of rimonabant-precipitated withdrawal signs in THC-dependent mice (Lichtman et al. 2001b), and lithium, a mood stabilizer, prevented the expression of withdrawal signs precipitated by the CB1 antagonist AM 251 in rats treated repeatedly with the potent synthetic cannabinoid HU-210 (Cui et al. 2001).
With the discovery of endocannabinoids, there has been interest in targeting the degradative enzymes of these naturally occurring ligands (Clapper et al. 2009; Piomelli 2004). Schlosburg and colleagues (2009) showed that the FAAH inhibitor URB597 attenuated the intensity of paw tremors and head shakes in THC-dependent mice challenged with rimonabant and that the selective MAGL inhibitor JZL 184 also alleviated the intensity of withdrawal signs.
If targeting endocannabinoid catabolic enzymes is indeed a viable approach to treat cannabis withdrawal, it is important to know whether these inhibitors would themselves have abuse or dependence liability. FAAH inhibitors have been extensively investigated in a variety of such paradigms. Importantly, it has been demonstrated that URB597 does not produce rewarding effects in the rat conditioned place preference paradigm, does not substitute for THC in the drug discrimination paradigm, and is not self-administered by nonhuman primates (Gobbi et al. 2005; Justinova et al. 2008). In addition, rimonabant does not precipitate any apparent withdrawal symptoms in mice treated subchronically with URB597 (Schlosburg et al. 2009) or the FAAH inhibitor PF-3845 (Schlosburg et al. 2010).
There is also supporting biochemical evidence that chronic treatment with FAAH inhibitors does not lead to long-term neural adaptations. Once-daily dosing with URB597 for 5 weeks exerted its pharmacological effects without altering CB1 messenger RNA levels (Bortolato et al. 2007). Likewise, repeated dosing of PF 3845 did not lead to desensitization or downregulation of CB1 receptors and did not alter CB1 receptor–mediated synaptic plasticity of hippocampal neurons. In contrast, repeated treatment with a high dose of the MAGL inhibitor JZL184 led to mild cannabinoid withdrawal signs after rimonabant administration, as well as regionally dependent changes in CB1 receptor downregulation and desensitization in the brain and impaired endocannabinoid-mediated synaptic plasticity (Schlosburg et al. 2010).
The differential actions of prolonged MAGL and FAAH inhibition are not fully understood and may be partly attributed to (1) higher concentrations of 2-AG compared with anandamide after sustained blockade of the respective degradative enzyme of each endocannabinoid and/or (2) differences in the efficacy of these endocannabinoids at the CB1 receptor. It remains to be established whether repeated administration of lower doses of JZL184 leads to cannabinoid dependence or changes in CB1 receptor function. Nonetheless, these studies provide proof of principle that bolstering endogenous anandamide or 2-AG through the inhibition of their respective catabolic enzymes may be viable approaches to reduce cannabis withdrawal symptoms.
Attenuation of Cannabis Withdrawal Symptoms in Humans
In recent years efforts to identify treatments for cannabis dependence disorders have increased considerably. Most of the current research is limited to small-scale laboratory models and small open-label trials. Medications investigated in the clinical laboratory setting include cannabinoid substitutes (e.g., THC) and a select group of noncannabinoid agents (e.g., divalproex, buspirone, bupropion, lithium, nefazodone, and lofexidine; see Table 2). Many of these compounds were selected because of their effectiveness in treating specific symptom clusters or their overall clinical evidence in treating opiate or tobacco-use disorders. For a recent review of pharmacological treatment of cannabis dependence, see Weinstein and Gorelick (2011).
Cannabinoid Substitution
Dronabinol, or oral synthetic THC, has been reliably reported to reduce withdrawal symptoms in cannabis-dependent individuals. In the first inpatient study, dronabinol (10 mg, 5x/day for 6 days) significantly decreased drug cravings, anxiety, chills, misery, and troubled sleep and reversed decreases in food intake (Haney et al. 2004). The findings of this study were extended to an outpatient setting that showed that dronabinol (10 or 30 mg, 3x/day for 15 days) attenuated cannabis withdrawal symptoms such as aggression, craving, troubled sleep, and irritability (Budney et al. 2007). In a subsequent study, dronabinol administered at 20 mg, three times a day for 8 days, decreased symptoms of cannabis withdrawal such as restlessness, anorexia, and chills (Haney et al. 2008). The most recent study reported that dronabinol (200 mg, 2x/day for 8–10 weeks) improved treatment retention and alleviated withdrawal symptoms in marijuana-dependent subjects (Levin et al. 2011).
Haney and colleagues (2008) also showed that a combination of dronabinol and lofexidine, an α2-adrenergic receptor agonist, produced the most robust improvements in sleep and decreased marijuana withdrawal symptoms and cravings relative to each medication by itself. Yet, despite the pre-clinical efficacy of another α2-adrenergic receptor agonist, clonidine, lofexidine by itself had a sedating effect, worsened abstinence-related anorexia, and did not robustly attenuate withdrawal, although it improved sleep and decreased the likelihood of marijuana relapse.
Differences in the effects of α2-adrenergic receptor agonists between preclinical and clinical studies may point to a possible limitation of cannabinoid withdrawal models. However, clonidine has not been evaluated for efficacy in reducing withdrawal in cannabis-dependent patients, and lofexidine has yet to be examined in preclinical models of cannabinoid withdrawal. In addition, the use of pharmacotherapy for cannabis withdrawal needs to be weighed against side effects, as clonidine and, to a lesser extent, lofexidine produce orthostatic hypotension.
In all of the studies described above, dronabinol itself did not produce any adverse effects, was well tolerated, and lower doses were not significantly distinguishable from placebo treatment.
Noncannabinoid Agents
Various other medications have been evaluated in inpatient studies for the treatment of cannabis withdrawal, largely with mixed results.
The antidepressant nefazodone (450 mg/day) decreased anxiety and muscle pain but did not alleviate irritability, misery, or troubled sleep during marijuana withdrawal (Haney et al. 2003). However, hepatotoxicity with this drug resulted in its removal from the market.
Bupropion (a drug used commonly in the cessation of tobacco use and for the attenuation of associated withdrawal symptoms) administered at 300 mg/day worsened ratings of irritability, restlessness, depression, and troubled sleep compared with placebo maintenance during marijuana withdrawal (Haney et al. 2001). The failure of bupropion to treat marijuana withdrawal underscores the need to treat cannabinoid withdrawal as a unique syndrome and not simply as another form of smoking cessation.
The clinical utility of atomoxetine (a norepinephrine reuptake inhibitor indicated for attention deficit hyperactivity disorder) to treat cannabis withdrawal was investigated in a small open-label study which showed that doses of 20 to 80 mg a day for 11 weeks tended to reduce marijuana use in cannabis-dependent individuals (p = 0.06) (Tirado et al. 2008). However, the potential benefit of this medication was negated by adverse gastrointestinal effects (nausea, vomiting, dyspepsia, and loose stools) in the majority of patients.
An open-label study reported that the anxiolytic agent buspirone did not significantly decrease cannabis withdrawal symptoms (McRae-Clark et al. 2009).
Divalproex, a mood stabilizer, when given once a day (1500 mg/day for 29 days), decreased marijuana craving during abstinence but increased ratings of anxiety, irritability, and tiredness (Haney et al. 2004). In a clinical double-blind trial with divalproex (500–2000 mg/day for 6 weeks), there was no effect on any withdrawal measures in the treatment groups in comparison with the placebo group. There was, however, increased incidence of divalproex-related adverse reactions and poor patient compliance during the trial (Levin et al. 2004).
Lithium has been tested for effectiveness in the treatment of cannabis withdrawal. Following results from preclinical studies (Cui et al. 2001), a small (n = 9), community-based, open-label study of the effects of lithium (600–900 mg/day for 6 days) on non-treatment-seeking individuals meeting DSM-IV criteria for cannabis dependence found a reduction in withdrawal signs in 50% of the patients (Bowen et al. 2005). Another study also reported that lithium (500 mg, 2x/day for 7 days) reduced the incidence of cannabis withdrawal symptoms, including depression and anxiety, with relatively few adverse effects (Winstock et al. 2009).
A case study with varying doses of the atypical antipsychotic quetiapine (100–1200 mg for 6 months) given to cannabis users with schizophrenia or bipolar disorder reported reduced cannabis use in these patients over the course of treatment (Potvin et al. 2004). However, the results of the study were complicated by concurrent treatments with anti-depressants, gabapentin, or methadone in some patients.
Conclusions
Research has firmly established the existence of a clinically significant and distinct cannabis withdrawal syndrome, characterized by anger, aggression, irritability, anxiety or nervousness, decreased appetite or weight loss, restlessness, and sleep difficulties with strange dreams. Dependent individuals may continue to use marijuana to avoid these and other withdrawal symptoms.
Laboratory animal models of cannabinoid withdrawal have been useful not only for characterizing and investigating the neurobiology of cannabinoid dependence but also for assessing potential pharmacological agents for therapeutic use. However, not all positive results in preclinical testing translate to clinical success, probably because of the wide variety of symptoms, both physical and psychological, observed in humans.
The clinical studies described in the preceding section indicate that cannabinoid substitutes, such as THC, show the greatest promise to treat cannabis withdrawal. However, although THC reliably reduces withdrawal responses in cannabinoid-dependent humans and laboratory animals, this drug is also primarily responsible for marijuana’s pharmacological effects and thus raises concern about the long-term outcome of this type of substitution therapy. Indeed, repeated THC administration has been well established to produce dependence. In contrast, inhibition of the endocannabinoid catabolic enzyme FAAH reduces the severity of cannabinoid withdrawal in animal models of THC dependence and, unlike THC, FAAH inhibitors do not appear to have reinforcing properties or dependence liability.
Given the relatively mild nature of the withdrawal syndrome and the political and public perception of cannabis dependence as a public health concern, there would have to be negligible abuse potential and side effects associated with any pharmacotherapeutic option. Further clinical studies are necessary to ascertain whether endocannabinoid catabolic enzyme inhibitors are effective for reducing withdrawal in cannabis-dependent individuals with minimal adverse impacts.
Acknowledgments
This research was supported by National Institutes of Health grants (P01DA009789, R01DA015197, and T32AA007456).
Footnotes
Abbreviations that appear ≥3x throughout this article: 2-AG, 2-arachidonylglycerol lipase; CB, cannabinoid; FAAH, fatty acid amide hydrolase; MAGL, monoacylglycerol lipase; THC, Δ9-tetrahydrocannabinol
Contributor Information
Divya Ramesh, Graduate student in the Department of Pharmacology and Toxicology at Virginia Commonwealth University (VCU) in Richmond.
Joel E. Schlosburg, Postdoctoral researcher in the Committee on the Neurobiology of Addictive Disorders at the Scripps Research Institute in La Jolla, California.
Jason M. Wiebelhaus, Graduate student in the Department of Pharmacology and Toxicology at VCU.
Aron H. Lichtman, Professor in the Department of Pharmacology and Toxicology and the Institute of Drug and Alcohol Studies at VCU.
References
- Aceto MD, Scates SM, Lowe JA, Martin BR. Cannabinoid precipitated withdrawal by the selective cannabinoid receptor antagonist, SR 141716A. Eur J Pharmacol. 1995;282:R1–R2. doi: 10.1016/0014-2999(95)00447-s. [DOI] [PubMed] [Google Scholar]
- Aceto MD, Scates SM, Lowe JA, Martin BR. Dependence on delta-9-tetrahydrocannabinol: Studies on precipitated and abrupt withdrawal. J Pharmacol Exp Ther. 1996;278:1290–1295. [PubMed] [Google Scholar]
- Aceto MD, Scates SM, Razdan RK, Martin BR. Anandamide, an endogenous cannabinoid, has a very low physical dependence potential. J Pharmacol Exp Ther. 1998;287:598–605. [PubMed] [Google Scholar]
- Aceto MD, Scates SM, Martin BB. Spontaneous and precipitated withdrawal with a synthetic cannabinoid, WIN 55212-2. Eur J Pharmacol. 2001;416:75–81. doi: 10.1016/s0014-2999(01)00873-1. [DOI] [PubMed] [Google Scholar]
- Ahn K, McKinney MK, Cravatt BF. Enzymatic pathways that regulate endocannabinoid signaling in the nervous system. Chem Rev. 2008;108:1687–1707. doi: 10.1021/cr0782067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- APA [American Psychiatric Association] DSM-IV: Diagnostic and Statistical Manual of Mental Disorders. 4. Arlington VA: 1994. [Google Scholar]
- Anggadiredja K, Yamaguchi T, Tanaka H, Shoyama Y, Watanabe S, Yamamoto T. Prostaglandin E2 attenuates SR141716A-precipitated withdrawal in tetrahydrocannabinol-dependent mice. Brain Res. 2003;966:47–53. doi: 10.1016/s0006-8993(02)04169-0. [DOI] [PubMed] [Google Scholar]
- Arendt M, Rosenberg R, Foldager L, Sher L, Munk-Jorgensen P. Withdrawal symptoms do not predict relapse among subjects treated for cannabis dependence. Am J Addict. 2007;16:461–467. doi: 10.1080/10550490701640985. [DOI] [PubMed] [Google Scholar]
- Beardsley PM, Martin BR. Effects of the cannabinoid CB1 receptor antagonist, SR141716A, after Δ9-tetrahydrocannabinol withdrawal. Eur J Pharmacol. 2000;387:47–53. doi: 10.1016/s0014-2999(99)00792-x. [DOI] [PubMed] [Google Scholar]
- Beardsley PM, Balster RL, Harris LS. Dependence on tetrahydrocannabinol in rhesus monkeys. J Pharmacol Exp Ther. 1986;239:311–319. [PubMed] [Google Scholar]
- Bonn-Miller MO, Moos RH. Marijuana discontinuation, anxiety symptoms, and relapse to marijuana. Addict Behav. 2009;34:782–785. doi: 10.1016/j.addbeh.2009.04.009. [DOI] [PubMed] [Google Scholar]
- Bortolato M, Mangieri RA, Fu J, Kim JH, Arguello O, Duranti A, Tontini A, Mor M, Tarzia G, Piomelli D. Antidepressant-like activity of the fatty acid amide hydrolase inhibitor URB597 in a rat model of chronic mild stress. Biol Psychiatry. 2007;62:1103–1110. doi: 10.1016/j.biopsych.2006.12.001. [DOI] [PubMed] [Google Scholar]
- Bowen R, McIlwrick J, Baetz M, Zhang X. Lithium and marijuana withdrawal. Can J Psychiatry. 2005;50:240–241. doi: 10.1177/070674370505000410. [DOI] [PubMed] [Google Scholar]
- Breivogel CS, Childers SR, Deadwyler SA, Hampson RE, Vogt LJ, Sim-Selley LJ. Chronic delta9-tetrahydrocannabinol treatment produces a time-dependent loss of cannabinoid receptors and cannabinoid receptor-activated G proteins in rat brain. J Neurochem. 1999;73:2447–2459. doi: 10.1046/j.1471-4159.1999.0732447.x. [DOI] [PubMed] [Google Scholar]
- Breivogel CS, Scates SM, Beletskaya IO, Lowery OB, Aceto MD, Martin BR. The effects of delta9-tetrahydrocannabinol physical dependence on brain cannabinoid receptors. Eur J Pharmacol. 2003;459:139–150. doi: 10.1016/s0014-2999(02)02854-6. [DOI] [PubMed] [Google Scholar]
- Budney AJ, Hughes JR. The cannabis withdrawal syndrome. Curr Opin Psychiatry. 2006;19:233–238. doi: 10.1097/01.yco.0000218592.00689.e5. [DOI] [PubMed] [Google Scholar]
- Budney AJ, Novy PL, Hughes JR. Marijuana withdrawal among adults seeking treatment for marijuana dependence. Addiction. 1999;94:1311–1322. doi: 10.1046/j.1360-0443.1999.94913114.x. [DOI] [PubMed] [Google Scholar]
- Budney AJ, Hughes JR, Moore BA, Novy PL. Marijuana abstinence effects in marijuana smokers maintained in their home environment. Arch Gen Psychiatry. 2001;58:917–924. doi: 10.1001/archpsyc.58.10.917. [DOI] [PubMed] [Google Scholar]
- Budney AJ, Moore BA, Vandrey RG, Hughes JR. The time course and significance of cannabis withdrawal. J Abnorm Psychol. 2003;112:393–402. doi: 10.1037/0021-843x.112.3.393. [DOI] [PubMed] [Google Scholar]
- Budney AJ, Vandrey RG, Hughes JR, Moore BA, Bahrenburg B. Oral delta-9-tetrahydrocannabinol suppresses cannabis withdrawal symptoms. Drug Alcohol Depend. 2007;86:22–29. doi: 10.1016/j.drugalcdep.2006.04.014. [DOI] [PubMed] [Google Scholar]
- Budney AJ, Vandrey RG, Hughes JR, Thostenson JD, Bursac Z. Comparison of cannabis and tobacco withdrawal: Severity and contribution to relapse. J Substance Abuse Treat. 2008;35:362–368. doi: 10.1016/j.jsat.2008.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cabral GA, Marciano-Cabral F. Cannabinoid receptors in microglia of the central nervous system: Immune functional relevance. J Leukoc Biol. 2005;78:1192–1197. doi: 10.1189/jlb.0405216. [DOI] [PubMed] [Google Scholar]
- Chung T, Martin CS, Cornelius JR, Clark DB. Cannabis withdrawal predicts severity of cannabis involvement at 1-year follow-up among treated adolescents. Addiction. 2008;103:787–799. doi: 10.1111/j.1360-0443.2008.02158.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clapper JR, Mangieri RA, Piomelli D. The endocannabinoid system as a target for the treatment of cannabis dependence. Neuropharmacology. 2009;56 (Suppl 1):235–243. doi: 10.1016/j.neuropharm.2008.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Compton D, Aceto M, Lowe J, Martin B. In vivo characterization of a specific cannabinoid receptor antagonist. SR141716A: Inhibition of Δ9-tetrahydrocannabinol-induced responses and apparent agonist activity. J Pharmacol Exp Ther. 1996;277:586–594. [PubMed] [Google Scholar]
- Cook SA, Lowe JA, Martin BR. CB1 receptor antagonist precipitates withdrawal in mice exposed to delta9-tetrahydrocannabinol. J Pharmacol Exp Ther. 1998;285:1150–1156. [PubMed] [Google Scholar]
- Copersino ML, Boyd SJ, Tashkin DP, Huestis MA, Heishman SJ, Dermand JC, Simmons MS, Gorelick DA. Quitting among non-treatment-seeking marijuana users: Reasons and changes in other substance use. Am J Addict. 2006;15:297–302. doi: 10.1080/10550490600754341. [DOI] [PubMed] [Google Scholar]
- Cornelius JR, Chung T, Martin C, Wood DS, Clark DB. Cannabis withdrawal is common among treatment-seeking adolescents with cannabis dependence and major depression, and is associated with rapid relapse to dependence. Addict Behav. 2008;33:1500–1505. doi: 10.1016/j.addbeh.2008.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Costa B, Giagnoni G, Colleoni M. Precipitated and spontaneous withdrawal in rats tolerant to anandamide. Psychopharmacology (Berl) 2000;149:121–128. doi: 10.1007/s002139900360. [DOI] [PubMed] [Google Scholar]
- 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]
- Darmani NA, Pandya DK. Involvement of other neurotransmitters in behaviors induced by the cannabinoid CB1 receptor antagonist SR 141716A in naive mice. J Neural Transm. 2000;107:931–945. doi: 10.1007/s007020070043. [DOI] [PubMed] [Google Scholar]
- Dawes MA, Liguori A, Dougherty DM. Cannabis withdrawal among adolescent cannabis users in an outpatient research setting. Am J Addict. 2006;15:485–486. doi: 10.1080/10550490601000637. [DOI] [PubMed] [Google Scholar]
- Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, Gibson D, Mandelbaum A, Etinger A, Mechoulam R. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992;258:1946–1949. doi: 10.1126/science.1470919. [DOI] [PubMed] [Google Scholar]
- Diana M, Melis M, Muntoni AL, Gessa GL. Mesolimbic dopaminergic decline after cannabinoid withdrawal. Proc Natl Acad Sci U S A. 1998;95:10269–10273. doi: 10.1073/pnas.95.17.10269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elsohly MA, Slade D. Chemical constituents of marijuana: The complex mixture of natural cannabinoids. Life Sci. 2005;78:539–548. doi: 10.1016/j.lfs.2005.09.011. [DOI] [PubMed] [Google Scholar]
- Falenski KW, Thorpe AJ, Schlosburg JE, Cravatt BF, Abdullah RA, Smith TH, Selley DE, Lichtman AH, Sim-Selley LJ. FAAH−/− mice display differential tolerance, dependence, and cannabinoid receptor adaptation after delta 9-tetrahydrocannabinol and anandamide administration. Neuropsychopharmacology. 2010;35:1775–1787. doi: 10.1038/npp.2010.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Felder CC, Glass M. Cannabinoid receptors and their endogenous agonists. Annu Rev Pharmacol Toxicol. 1998;38:179–200. doi: 10.1146/annurev.pharmtox.38.1.179. [DOI] [PubMed] [Google Scholar]
- Fredericks AB, Benowitz NL. An abstinence syndrome following chronic administration of delta-9-tetrahydrocannabinol in rhesus monkeys. Psychopharmacology (Berl) 1980;71:201–202. doi: 10.1007/BF00434412. [DOI] [PubMed] [Google Scholar]
- Gaoni Y, Mechoulam R. Isolation, structure, and partial synthesis of an active constituent of hashish. J Am Chem Soc. 1964;86:1646–1647. [Google Scholar]
- Georgotas A, Zeidenberg P. Observations on the effects of four weeks of heavy marihuana smoking on group interaction and individual behavior. Compr Psychiatry. 1979;20:427–432. doi: 10.1016/0010-440x(79)90027-0. [DOI] [PubMed] [Google Scholar]
- Gerard CM, Mollereau C, Vassart G, Parmentier M. Molecular cloning of a human cannabinoid receptor which is also expressed in testis. Biochem J. 1991;279 (Pt 1):129–134. doi: 10.1042/bj2790129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gobbi G, Bambico FR, Mangieri R, Bortolato M, Campolongo P, Solinas M, Cassano T, Morgese MG, Debonnel G, Duranti A, Tontini A, Tarzia G, Mor M, Trezza V, Goldberg SR, Cuomo V, Piomelli D. Antidepressant-like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc Natl Acad Sci U S A. 2005;102:18620–18625. doi: 10.1073/pnas.0509591102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez S, Fernandez-Ruiz J, Di Marzo V, Hernandez M, Arevalo C, Nicanor C, Cascio MG, Ambrosio E, Ramos JA. Behavioral and molecular changes elicited by acute administration of SR141716 to delta9-tetrahydrocannabinol-tolerant rats: An experimental model of cannabinoid abstinence. Drug Alc Depend. 2004;74:159–170. doi: 10.1016/j.drugalcdep.2003.12.011. [DOI] [PubMed] [Google Scholar]
- Haney M, Ward AS, Comer SD, Foltin RW, Fischman MW. Abstinence symptoms following oral THC administration to humans. Psychopharmacology (Berl) 1999;141:385–394. doi: 10.1007/s002130050848. [DOI] [PubMed] [Google Scholar]
- Haney M, Ward AS, Comer SD, Hart CL, Foltin RW, Fischman MW. Bupropion SR worsens mood during marijuana withdrawal in humans. Psychopharmacology (Berl) 2001;155:171–179. doi: 10.1007/s002130000657. [DOI] [PubMed] [Google Scholar]
- Haney M, Hart CL, Ward AS, Foltin RW. Nefazodone decreases anxiety during marijuana withdrawal in humans. Psychopharmacology (Berl) 2003;165:157–165. doi: 10.1007/s00213-002-1210-3. [DOI] [PubMed] [Google Scholar]
- Haney M, Hart CL, Vosburg SK, Nasser J, Bennett A, Zubaran C, Foltin RW. Marijuana withdrawal in humans: Effects of oral THC or divalproex. Neuropsychopharmacology. 2004;29:158–170. doi: 10.1038/sj.npp.1300310. [DOI] [PubMed] [Google Scholar]
- 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. Psychopharmacology (Berl) 2008;197:157–168. doi: 10.1007/s00213-007-1020-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hasin DS, Keyes KM, Alderson D, Wang S, Aharonovich E, Grant BF. Cannabis withdrawal in the United States: Results from NESARC. J Clin Psychiatry. 2008;69:1354–1363. doi: 10.4088/jcp.v69n0902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herkenham M, Lynn AB, Little MD, Johnson MR, Melvin LS, DeCosta BR, Rice KC. Cannabinoid receptor localization in the brain. Proc Natl Acad Sci U S A. 1990;87:1932–1936. doi: 10.1073/pnas.87.5.1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hermann D, Lemenager T, Gelbke J, Welzel H, Skopp G, Mann K. Decision making of heavy cannabis users on the Iowa Gambling Task: Stronger association with THC of hair analysis than with personality traits of the Tridimensional Personality Questionnaire. Eur Addict Res. 2009;15:94–98. doi: 10.1159/000189788. [DOI] [PubMed] [Google Scholar]
- Howlett AC, Mukhopadhyay S. Cellular signal transduction by anandamide and 2-arachidonoylglycerol. Chem Phys Lipids. 2000;108:53–70. doi: 10.1016/s0009-3084(00)00187-0. [DOI] [PubMed] [Google Scholar]
- Huang P, Liu-Chen LY, Unterwald EM, Cowan A. Hyperlocomotion and paw tremors are two highly quantifiable signs of SR141716-precipitated withdrawal from delta 9-tetrahydrocannabinol in C57BL/6 mice. Neurosci Lett. 2009;465:66–70. doi: 10.1016/j.neulet.2009.08.073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang P, Liu-Chen LY, Kirby LG. Anxiety-like effects of SR141716-precipitated delta9-tetrahydrocannabinol withdrawal in mice in the elevated plus-maze. Neurosci Lett. 2010;475:165–168. doi: 10.1016/j.neulet.2010.03.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huestis MA. Pharmacokinetics and metabolism of the plant cannabinoids, delta 9-tetrahydrocannabinol, cannabidiol and cannabinol. Handb Exp Pharmacol. 2005:657–690. doi: 10.1007/3-540-26573-2_23. [DOI] [PubMed] [Google Scholar]
- 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. Arch Gen Psychiatry. 2001;58:322–328. doi: 10.1001/archpsyc.58.4.322. [DOI] [PubMed] [Google Scholar]
- 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. Psychopharmacology (Berl) 2007;194:505–515. doi: 10.1007/s00213-007-0861-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hutcheson DM, Tzavara ET, Smadja C, Valjent E, Roques BP, Hanoune J, Maldonado R. Behavioural and biochemical evidence for signs of abstinence in mice chronically treated with delta-9-tetrahydrocannabinol. Br J Pharmacol. 1998;125:1567–1577. doi: 10.1038/sj.bjp.0702228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnston LD, O’Malley PM, Bachman JG, Schulenberg JE. College Students and Adults Ages 19–50 NIH Publication No 10-7585. II. Bethesda: National Institute on Drug Abuse; 2010. Monitoring the Future: National Survey Results on Drug Use, 1975–2009. [Google Scholar]
- Jones RT, Benowitz N. The 30-day trip: Clinical studies of cannabis tolerance and dependence. In: Braude MC, Szara S, editors. Pharmacology of Marihuana. New York: Raven Press; 1976. pp. 627–642. [Google Scholar]
- Jones RT, Benowitz NL, Herning RI. Clinical relevance of cannabis tolerance and dependence. J Clin Pharmacol. 1981;21:143S–152S. doi: 10.1002/j.1552-4604.1981.tb02589.x. [DOI] [PubMed] [Google Scholar]
- Justinova Z, Mangieri RA, Bortolato M, Chefer SI, Mukhin AG, Clapper JR, King AR, Redhi GH, Yasar S, Piomelli D, Goldberg SR. Fatty acid amide hydrolase inhibition heightens anandamide signaling without producing reinforcing effects in primates. Biol Psychiatry. 2008;64:930–937. doi: 10.1016/j.biopsych.2008.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kathuria S, Gaetani S, Fegley D, Valino F, Duranti A, Tontini A, Mor M, Tarzia G, Rana GL, Calignano A, Giustino A, Tattoli M, Palmery M, Cuomo V, Piomelli D. Modulation of anxiety through blockade of anandamide hydrolysis. Nat Med. 2003;9:76–81. doi: 10.1038/nm803. [DOI] [PubMed] [Google Scholar]
- Kaymakcalan K, Ayhan IH, Tulunay FC. Naloxone-induced or postwithdrawal abstinence signs in Δ9-tetrahydrocannabinol-tolerant rats. Psychopharmocology. 1977;55:243–249. doi: 10.1007/BF00497855. [DOI] [PubMed] [Google Scholar]
- Klein TW, Newton C, Larsen K, Lu L, Perkins I, Nong L, Friedman H. The cannabinoid system and immune modulation. J Leukoc Biol. 2003;74:486–496. doi: 10.1189/jlb.0303101. [DOI] [PubMed] [Google Scholar]
- Kouri EM, Pope HG., Jr Abstinence symptoms during withdrawal from chronic marijuana use. Exp Clin Psychopharmacol. 2000;8:483–492. doi: 10.1037//1064-1297.8.4.483. [DOI] [PubMed] [Google Scholar]
- Kouri EM, Pope HG, Jr, Lukas SE. Changes in aggressive behavior during withdrawal from long-term marijuana use. Psychopharmacology (Berl) 1999;143:302–308. doi: 10.1007/s002130050951. [DOI] [PubMed] [Google Scholar]
- Ledent C, Valverde O, Cossu G, Petitet F, Aubert JF, Beslot F, Bohme GA, Imperato A, Pedrazzini T, Roques BP, Vassart G, Fratta W, Parmentier M. Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice. Science. 1999;283:401–404. doi: 10.1126/science.283.5400.401. [DOI] [PubMed] [Google Scholar]
- Leite JR, Carlini EA. Failure to obtain “cannabis-directed behavior” and abstinence syndrome in rats chronically treated with Cannabis sativa extracts. Psychopharmacologia. 1974;36:133–145. doi: 10.1007/BF00421785. [DOI] [PubMed] [Google Scholar]
- 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 Addict. 2004;13:21–32. doi: 10.1080/10550490490265280. [DOI] [PubMed] [Google Scholar]
- Levin FR, Brooks DJ, Bisaga A, Raby W, Rubin E, Aharonovich E, Nunes EV. Severity of dependence and motivation for treatment: Comparison of marijuana- and cocaine-dependent treatment seekers. J Addict Dis. 2006;25:33–41. doi: 10.1300/J069v25n01_06. [DOI] [PubMed] [Google Scholar]
- Levin FR, Mariani JJ, Brooks DJ, Pavlicova M, Cheng W, Nunes EV. Dronabinol for the treatment of cannabis dependence: A randomized, double-blind, placebo-controlled trial. Drug Alcohol Depend. 2011;116:142–150. doi: 10.1016/j.drugalcdep.2010.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lichtman AH, Martin BR. Cannabinoid tolerance and dependence. Handb Exp Pharmacol. 2005;168:691–717. doi: 10.1007/3-540-26573-2_24. [DOI] [PubMed] [Google Scholar]
- Lichtman AH, Wiley JL, LaVecchia KL, Neviaser ST, Arthrur DB, Wilson DM, Martin BR. Acute and chronic cannabinoid effects: Characterization of precipitated withdrawal in dogs. Eur J Pharmacol. 1998a;357:139–148. doi: 10.1016/s0014-2999(98)00558-5. [DOI] [PubMed] [Google Scholar]
- Lichtman AH, Wiley JL, LaVecchia KL, Neviaser ST, Arthur DB, Wilson DM, Martin BR. Effects of SR 141716A after acute or chronic cannabinoid administration in dogs. Eur J Pharmacol. 1998b;357:139–148. doi: 10.1016/s0014-2999(98)00558-5. [DOI] [PubMed] [Google Scholar]
- Lichtman AH, Fisher J, Martin BR. Precipitated cannabinoid withdrawal is reversed by delta-9-tetrahydrocannabinol or clonidine. Pharmacol Biochem Behav. 2001a;69:181–188. doi: 10.1016/s0091-3057(01)00514-7. [DOI] [PubMed] [Google Scholar]
- Lichtman AH, Sheikh SM, Loh HH, Martin BR. Opioid and cannabinoid modulation of precipitated withdrawal in delta-9-tetrahydrocannabinol and morphine-dependent mice. J Pharmacol Exp Ther. 2001b;298:1007–1014. [PubMed] [Google Scholar]
- Lichtman AH, Leung D, Shelton C, Saghatelian A, Hardouin C, Boger D, Cravatt BF. Reversible inhibitors of fatty acid amide hydrolase that promote analgesia: Evidence for an unprecedented combination of potency and selectivity. J Pharmacol Exp Ther. 2004;311:441–448. doi: 10.1124/jpet.104.069401. [DOI] [PubMed] [Google Scholar]
- Long JZ, Li W, Booker L, Burston JJ, Kinsey SG, Schlosburg JE, Pavon FJ, Serrano AM, Selley DE, Parsons LH, Lichtman AH, Cravatt BF. Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid behavioral effects. Nat Chem Biol. 2009;5:37–44. doi: 10.1038/nchembio.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346:561–564. doi: 10.1038/346561a0. [DOI] [PubMed] [Google Scholar]
- Matsuda LA, Bonner TI, Lolait SJ. Localization of cannabinoid receptor mRNA in rat brain. J Comp Neurol. 1993;327:535–550. doi: 10.1002/cne.903270406. [DOI] [PubMed] [Google Scholar]
- McMahon LR. Discriminative stimulus effects of the cannabinoid CB1 antagonist SR 141716A in rhesus monkeys pretreated with Δ9-tetrahydrocannabinol. Psychopharmacology (Berl) 2006;188:306–314. doi: 10.1007/s00213-006-0500-6. [DOI] [PubMed] [Google Scholar]
- McMahon LR, France CP. Discriminative stimulus effects of the cannabinoid antagonist, SR 141716A, in delta-sup-9-tetrahydrocannabinol-treated rhesus monkeys. Exp Clin Psychopharmacol. 2003;11:286–293. doi: 10.1037/1064-1297.11.4.286. [DOI] [PubMed] [Google Scholar]
- McMillan DE, Dewey WL, Turk RF, Harris LS, McNeil JH. Blood levels of 3H-Δ9-tetrahydrocannabinol and its metabolites in tolerant and nontolerant pigeons. Biochem Pharmacol. 1973;22:383–397. doi: 10.1016/0006-2952(73)90419-x. [DOI] [PubMed] [Google Scholar]
- McRae AL, Brady KT, Carter RE. Buspirone for treatment of marijuana dependence: A pilot study. Am J Addict. 2006;15:404. doi: 10.1080/10550490600860635. [DOI] [PubMed] [Google Scholar]
- 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 Depend. 2009;105:132–138. doi: 10.1016/j.drugalcdep.2009.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski N, Schatz A, Gopher A, Almog S, Martin B, Compton D, Pertwee R, Griffin G, Bayewitch M, Barg J, Vogel Z. Identification of an endogenous 2-monoglyceride, present in canine gut that binds to cannabinoid receptors. Biochem Pharmacol. 1995;50:83–90. doi: 10.1016/0006-2952(95)00109-d. [DOI] [PubMed] [Google Scholar]
- Mennes CE, Ben Abdallah A, Cottler LB. The reliability of self-reported cannabis abuse, dependence and withdrawal symptoms: Multi-site study of differences between general population and treatment groups. Addict Behav. 2009;34:223–226. doi: 10.1016/j.addbeh.2008.10.003. [DOI] [PubMed] [Google Scholar]
- 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]
- Nowlan R, Cohen S. Tolerance to marijuana: Heart rate and subjective “high. Clin Pharmacol Ther. 1977;22:550–556. doi: 10.1002/cpt1977225part1550. [DOI] [PubMed] [Google Scholar]
- Oliva JM, Ortiz S, Palomo T, Manzanares J. Behavioural and gene transcription alterations induced by spontaneous cannabinoid withdrawal in mice. J Neurochem. 2003;85:94–104. doi: 10.1046/j.1471-4159.2003.01627.x. [DOI] [PubMed] [Google Scholar]
- Panagis G, Vlachou S, Nomikos GG. Behavioral pharmacology of cannabinoids with a focus on preclinical models for studying reinforcing and dependence-producing properties. Curr Drug Abuse Rev. 2008;1:350–374. doi: 10.2174/1874473710801030350. [DOI] [PubMed] [Google Scholar]
- Peters EN, Hughes JR. Daily marijuana users with past alcohol problems increase alcohol consumption during marijuana abstinence. Drug Alcohol Depend. 2010;106:111–118. doi: 10.1016/j.drugalcdep.2009.07.027. [DOI] [PubMed] [Google Scholar]
- Piomelli D. The endogenous cannabinoid system and the treatment of marijuana dependence. Neuropharmacology. 2004;47 (Suppl 1):359–367. doi: 10.1016/j.neuropharm.2004.07.018. [DOI] [PubMed] [Google Scholar]
- Potvin S, Stip E, Roy JY. The effect of quetiapine on cannabis use in 8 psychosis patients with drug dependency. Can J Psychiatry. 2004;49:711. doi: 10.1177/070674370404901020. [DOI] [PubMed] [Google Scholar]
- Preuss UW, Watzke AB, Zimmermann J, Wong JW, Schmidt CO. Cannabis withdrawal severity and short-term course among cannabis-dependent adolescent and young adult inpatients. Drug Alcohol Depend. 2010;106:133–141. doi: 10.1016/j.drugalcdep.2009.08.008. [DOI] [PubMed] [Google Scholar]
- Rinaldi-Carmona M, Barth F, Heaulme M, Shire D, Calandra B, Congy C, Martinez S, Maruani J, Neliat G, Caput D. SR141716A, a potent and selective antagonist of the brain cannabinoid receptor. FEBS Lett. 1994;350:240–244. doi: 10.1016/0014-5793(94)00773-x. [DOI] [PubMed] [Google Scholar]
- Rodriguez de Fonseca F, Carrera M, Navarro M, Koob K, Weiss F. Activation of corticotropin-releasing factor in the limbic system during cannabinoid withdrawal. Science. 1997;276:2050–2054. doi: 10.1126/science.276.5321.2050. [DOI] [PubMed] [Google Scholar]
- Romero J, Garcia-Palomero E, Castro JG, Garcia-Gil L, Ramos JA, Fernandez-Ruiz JJ. Effects of chronic exposure to delta-9-tetrahydrocannabinol on cannabinoid receptor binding and mRNA levels in several rat brain regions. Brain Res Mol Brain Res. 1997;46:100–108. doi: 10.1016/s0169-328x(96)00277-x. [DOI] [PubMed] [Google Scholar]
- Rubino T, Patrini G, Massi P, Fuzio D, Vigano D, Giagnoni G, Parolaro D. Cannabinoid-precipitated withdrawal: A time-course study of the behavioral aspect and its correlation with cannabinoid receptors and G protein expression. J Pharmacol Exp Ther. 1998;285:813–819. [PubMed] [Google Scholar]
- Rubino T, Vigano D, Massi P, Spinello M, Zagato E, Giagnoni G, Parolaro D. Chronic delta-9-tetrahydrocannabinol treatment increases cAMP levels and cAMP-dependent protein kinase activity in some rat brain regions. Neuropharmacology. 2000;39:1331–1336. doi: 10.1016/s0028-3908(99)00196-3. [DOI] [PubMed] [Google Scholar]
- SAMHSA [Substance Abuse and Mental Health Services] NSDUH Ser H-34, DHHS Publ No SMA 08-4343. Rockville MD: Office of Applied Studies; 2008. Results from the 2007 National Survey on Drug Use and Health: National Findings. [Google Scholar]
- SAMHSA. DHHS Publ No SMA 11-4618. Rockville MD: Center for Behavioral Health Statistics and Quality; 2011. Drug Abuse Warning Network, 2008: National Estimates of Drug-Related Emergency Department Visits. [Google Scholar]
- 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;11:342–352. doi: 10.1208/s12248-009-9110-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schlosburg JE, Blankman JL, Long JZ, Nomura DK, Pan B, Kinsey SG, Nguyen PT, Ramesh D, Booker L, Burston JJ, Thomas EA, Selley DE, Sim-Selley LJ, Liu QS, Lichtman AH, Cravatt BF. Chronic mono-acylglycerol lipase blockade causes functional antagonism of the endocannabinoid system. Nat Neurosci. 2010;13:1113–1119. doi: 10.1038/nn.2616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith NT. A review of the published literature into cannabis withdrawal symptoms in human users. Addiction. 2002;97:621–632. doi: 10.1046/j.1360-0443.2002.00026.x. [DOI] [PubMed] [Google Scholar]
- Stewart JL, McMahon LR. Rimonabant-induced delta-9-tetrahydrocannabinol withdrawal in rhesus monkeys: Discriminative stimulus effects and other withdrawal signs. J Pharmacol Exp Ther. 2010;334:347–356. doi: 10.1124/jpet.110.168435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K, Yamashita A, Waku K. 2-Arachidonoyglycerol: A possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Comm. 1995;215:89–97. doi: 10.1006/bbrc.1995.2437. [DOI] [PubMed] [Google Scholar]
- Swift W, Hall W, Teesson M. Characteristics of DSM-IV and ICD-10 cannabis dependence among Australian adults: Results from the National Survey of Mental Health and Wellbeing. Drug Alcohol Depend. 2001;63:147–153. doi: 10.1016/s0376-8716(00)00197-6. [DOI] [PubMed] [Google Scholar]
- Taylor DA, Fennessy MR. Relationship between body temperature and brain monoamines during the development of tolerance to delta9-tetrahydrocannabinol in the rat. Psychopharmacology (Berl) 1978;56:279–285. doi: 10.1007/BF00432850. [DOI] [PubMed] [Google Scholar]
- Taylor DA, Fennessy MR. Time-course of the effects of chronic delta 9-tetrahydrocannabinol on behaviour, body temperature, brain amines and withdrawal-like behaviour in the rat. J Pharm Pharmacol. 1982;34:240–245. doi: 10.1111/j.2042-7158.1982.tb04235.x. [DOI] [PubMed] [Google Scholar]
- Tirado CF, Goldman M, Lynch K, Kampman KM, O’Brien CP. Atomoxetine for treatment of marijuana dependence: A report on the efficacy and high incidence of gastrointestinal adverse events in a pilot study. Drug Alcohol Depend. 2008;94:254–257. doi: 10.1016/j.drugalcdep.2007.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tourino C, Maldonado R, Valverde O. MDMA attenuates THC withdrawal syndrome in mice. Psychopharmacology (Berl) 2007;193:75–84. doi: 10.1007/s00213-007-0772-5. [DOI] [PubMed] [Google Scholar]
- Tsou K, Patrick SL, Walker JM. Physical withdrawal in rats tolerant to delta 9-tetrahydrocannabinol precipitated by a cannabinoid receptor antagonist. Eur J Pharmacol. 1995;280:R13–R15. doi: 10.1016/0014-2999(95)00360-w. [DOI] [PubMed] [Google Scholar]
- Tzavara ET, Valjent E, Firmo C, Mas M, Beslot F, Defer N, Roques BP, Hanoune J, Maldonado R. Cannabinoid withdrawal is dependent upon PKA activation in the cerebellum. Eur J Neurosci. 2000;12:1038–1046. doi: 10.1046/j.1460-9568.2000.00971.x. [DOI] [PubMed] [Google Scholar]
- Valverde O, Maldonado R, Valjent E, Zimmer AM, Zimmer A. Cannabinoid withdrawal syndrome is reduced in pre-proenkephalin knockout mice. J Neurosci. 2000;20:9284–9289. doi: 10.1523/JNEUROSCI.20-24-09284.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Sickle MD, Duncan M, Kingsley PJ, Mouihate A, Urbani P, Mackie K, Stella N, Makriyannis A, Piomelli D, Davison JS, Marnett LJ, Di Marzo V, Pittman QJ, Patel KD, Sharkey KA. Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science. 2005;310:329–332. doi: 10.1126/science.1115740. [DOI] [PubMed] [Google Scholar]
- Vandrey RG, Budney AJ, Hughes JR, Liguori A. A within-subject comparison of withdrawal symptoms during abstinence from cannabis, tobacco, and both substances. Drug Alcohol Depend. 2008;92:48–54. doi: 10.1016/j.drugalcdep.2007.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verberne AJ, Taylor DA, Fennessy MR. Withdrawal-like behaviour induced by inhibitors of biogenic amine reuptake in rats treated chronically with delta 9-tetrahydrocannabinol. Psychopharmacology (Berl) 1980;68:261–267. doi: 10.1007/BF00428113. [DOI] [PubMed] [Google Scholar]
- Verberne AJ, Taylor DA, Fennessy MR. Attenuation of delta 9-tetrahydrocannabinol-induced withdrawal-like behaviour by delta 9-tetrahydrocannabinol. Psychopharmacology (Berl) 1981;73:97–98. doi: 10.1007/BF00431112. [DOI] [PubMed] [Google Scholar]
- Verberne AJ, Fennessy MR, Lewis SJ, Taylor DA. Involvement of brain histamine in delta 9-tetrahydrocannabinol tolerance and withdrawal. Pharmacol Biochem Behav. 1985;23:153–159. doi: 10.1016/0091-3057(85)90549-0. [DOI] [PubMed] [Google Scholar]
- Wallace GB, Cunningham EV. The Marihuana Problem in the City of New York: Sociological, Medical, Psychological and Pharmacological Studies. Lancaster PA: Jacques Cattell Press; 1944. [Google Scholar]
- Weinstein AM, Gorelick DA. Pharmacological treatment of cannabis dependence. Curr Pharm Des. 2011;17:1351–1358. doi: 10.2174/138161211796150846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wesley MJ, Hanlon CA, Porrino LJ. Poor decision-making by chronic marijuana users is associated with decreased functional responsiveness to negative consequences. Psychiatry Res. 2011;191:51–59. doi: 10.1016/j.pscychresns.2010.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitlow CT, Liguori A, Livengood LB, Hart SL, Mussat-Whitlow BJ, Lamborn CM, Laurienti PJ, Porrino LJ. Long-term heavy marijuana users make costly decisions on a gambling task. Drug Alcohol Depend. 2004;76:107–111. doi: 10.1016/j.drugalcdep.2004.04.009. [DOI] [PubMed] [Google Scholar]
- WHO [World Health Organization] [accessed on August 15, 2011];International Statistical Classification of Diseases and Related Health Problems. (10). 2007 Available online ( http://apps.who.int/classifications/apps/icd)
- Wiesbeck GA, Schuckit MA, Kalmijn JA, Tipp JE, Bucholz KK, Smith TL. An evaluation of the history of a marijuana withdrawal syndrome in a large population. Addiction. 1996;91:1469–1478. [PubMed] [Google Scholar]
- Williams EG, Himmelsbach CK, Wikler A, Rudle DC. Studies on marihuana and pyrahexyl compound. Public Health Rep. 1946;61:1059–1083. [PubMed] [Google Scholar]
- Wilson DM, Varvel SA, Harloe JP, Martin BR, Lichtman AH. SR 141716-Rimonabant precipitates withdrawal in marijuana-dependent mice. Pharmacol Biochem Behav. 2006;85:105–113. doi: 10.1016/j.pbb.2006.07.018. [DOI] [PubMed] [Google Scholar]
- Winsauer PJ, Lambert P, Moerschbaecher JM. Cannabinoid ligands and their effects on learning and performance in rhesus monkeys. Behav Pharmacol. 1999;10:497–511. doi: 10.1097/00008877-199909000-00008. [DOI] [PubMed] [Google Scholar]
- 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]