The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Reviews and OverviewsFull Access

Medication Development for Addictive Disorders: The State of the Science

Abstract

In 1989, the National Institute on Drug Abuse (NIDA) established its Medications Development Program. This program has concentrated on developing pharmacotherapies for opiate and cocaine dependence and, more recently, for methamphetamine and cannabis dependence. The major goals of this program are to optimize existing treatments and to expand treatment options for physicians and patients. This review will concentrate on the development of pharmacotherapies for the following substance abuse disorders: opiate, cocaine, methamphetamine, and cannabis dependence. Left untreated, opiate and stimulant dependence are responsible for significant morbidity and mortality. For example, use of illicit opiates is associated with an increased risk of hepatitis C infection, HIV infection, and other medical consequences, e.g., an overdose. The NIDA Medications Development Program has had success in developing, with pharmaceutical partners, levomethadyl acetate, buprenorphine, and buprenorphine/naloxone for opiate dependence. Moreover, several marketed medications have shown promise in reducing cocaine use. Of interest, these medications likely operate through diverse neurochemical mechanisms, suggesting that combination therapy may be a rational next step that could increase treatment gains further in cocaine-dependent patients. The Medications Development Program has also identified multiple neuronal mechanisms that are altered by chronic administration of drugs of abuse. Advances in neuroscience have identified changes in conditioned cueing, drug priming, stress-induced increases in drug intake, and reduced frontal inhibitory mechanisms as all being possible for the development of, maintenance of, and possible relapse to, addiction. Potential medications that modulate these mechanisms are highlighted.

The National Institute on Drug Abuse (NIDA), a component of the National Institutes of Health, is responsible for research into the causes of and treatments for drug addiction. As part of this effort, the Medications Development Program was established in 1989 with a primary mission to develop medications for opiate and cocaine dependence. For a decade, the Medications Development Program concentrated on these disorders. This review will report the progress made in developing medications for these disorders. More recently, it has increased its focus to include developing medications for methamphetamine and cannabis dependence. Because of space limitations, we will not review medication development for methamphetamine and cannabis disorders.

Opiate Dependence

According to the Office of National Drug Control Policy, there are almost a million long-term opiate users in the United States. Opiate dependence is associated with significant all-causes mortality and significant morbidity. The major pharmacotherapeutic agent for the management of opiate dependence is methadone. Although methadone is not a new medication developed under the NIDA Medications Development Program, a brief review of its effects will serve to illustrate several points: 1) that dose effects for opiates can be seen in both clinical pharmacology studies and clinical trials and 2) that reduction of opiate use and increased retention in clinical trials may be surrogates for the reduction in mortality and morbidity seen in epidemiological studies.

Methadone is an orally active, long-acting synthetic opiate that was recognized in the 1960s as having the potential to treat opiate dependence (1, 2). A clinical pharmacology study demonstrated that increasing doses of methadone affected abstinence signs and symptoms and reduced drug craving. These investigators also reported that doses of methadone ranging from 80–120 mg/day produced a blockade of the effects of intravenously administered heroin, hydromorphone, and methadone. The term “agonist blockade” was coined to describe this phenomenon.

Initial clinical trials using a variety of designs showed a benefit of methadone to reduce opiate use and retain patients in treatment (37). These studies also showed a collateral benefit to reduce recidivism (3), criminal activity (4), and mortality (7). Further clinical trials showed a dose effect to reduce opiate use and increase retention in treatment (812). An orderly dose effect has been reported for comparisons of 50 mg/day versus 20 mg/day doses of methadone (8), 60 mg/day versus 20 mg/day (9), 80–100 mg/day versus lower doses (10), 80 mg/day versus 30 mg/day (11), and 60–100 mg/day versus 20 mg/day (12). In the 1990s, an increase in average methadone dose in opiate treatment programs was noted (13). Notwithstanding the higher dose trend, patient dosing should be individualized.

Methadone has been shown to reduce opiate-related mortality. A U.S. study (14) reported that untreated heroin addicts had a yearly mortality rate of 8.3%, whereas those in methadone treatment had a mortality rate of 0.8%. A Swedish study (7) reported a sixfold reduction in mortality in methadone-treated opiate addicts. Finally, an Australian study (15) reported a fourfold reduction in the risk of dying for patients in methadone programs.

Methadone has been shown to reduce morbidity insofar as it reduces the risk of infectious disease. In a retrospective study (16), the duration of methadone maintenance therapy was inversely correlated with the prevalence of HIV. In a prospective study (17), a sevenfold reduction in the incidence of HIV was seen in a methadone-treated group versus an untreated cohort that agreed to be a part of the study but refused treatment.

Levomethadyl acetate (LAAM) is the alpha-acetyl congener of methadone. Its principal difference from methadone is its conversion to the active metabolites norLAAM and dinorLAAM (1821). Initial studies were performed in the 1970s with a dosing regimen of three times a week (22, 23). LAAM was originally thought to be a pro drug, but recent evidence suggests that LAAM has agonist properties (24). A large phase III safety trial led to its approval by the Food and Drug Administration (FDA) in 1993 (25). Dosing recommendations on the product label suggested that induction doses could range from 20 to 40 mg/day and that maximum thrice-weekly dosing regimens of 30/130/180 mg or 140/140/140 mg were permissible. Alternate-day dosing was also added to the labeling. Postmarketing studies (26) noted that a rapid-induction regimen of 25 to 100 mg/day in 17 days yielded the greatest reduction in illicit opiate use, but a higher side effect profile was encountered. A dose effect of LAAM on illicit opiate use was reported (27), with the 100/100/140 mg thrice-weekly regimen giving the greatest reduction in opiate use. The FDA issued a “black box” warning for LAAM because of postmarketing surveillance reports of QTc prolongation in ECGs, with several reports of torsades de pointes, a polymorphic life-threatening ventricular arrhythmia (28).

Buprenorphine is a mu opiate partial agonist (29) that was originally marketed as an analgesic product. Investigators conducting an abuse liability study in human volunteers reported that subcutaneously administered buprenorphine has less subjective effects than morphine, a lesser withdrawal syndrome, and an ability to block the subjective responses of up to 120 mg doses of morphine (30). Subsequent work established that the sublingual route was preferable to oral dosing because of high first-pass effects (31, 32). The first outpatient treatment study to our knowledge (9) compared 8 mg of sublingual buprenorphine liquid to 20 and 60 mg doses of orally administered methadone in a randomized, double-blind, double-dummy study. Retention and decreased illicit opiate use in the buprenorphine group were superior to the response seen in the group that was receiving 20 mg/day of methadone.

NIDA and Reckitt & Colman (now ReckittBenckiser) received a Cooperative Research and Development Award to develop buprenorphine as a treatment for opiate dependence. Under this agreement, a second study conducted with the liquid formulation was performed in a multisite trial in which opiate-addicted patients were randomly assigned to 1, 4, 8, and 16 mg/day of buprenorphine (33). The a priori comparison was the effects of 1 mg/day versus 8 mg/day on illicit opiate use, retention, and opiate craving. The 8 mg/day dose group had significant reductions in illicit opiate use, reduced craving, and had better retention.

Subsequent to the study by Ling et al. (33), it was decided to develop a sublingual tablet and to add naloxone, a narcotic antagonist, to one of the formulations. The rationale for adding naloxone was to produce a less abusable, less divertable tablet. The dose ratio of buprenorphine to naloxone was chosen from data gathered in clinical pharmacology studies in opiate-dependent subjects maintained with morphine (34), methadone (35), or buprenorphine (36). In the first study (34), the subjects maintained on a dosage of 60 mg/day of morphine sulfate were randomly administered one of six medication treatments intravenously in a counterbalanced order: morphine, buprenorphine, buprenorphine/naloxone at 8:1, buprenorphine/naloxone at 4:1, buprenorphine/naloxone at 2:1, and placebo. Subjective measures of positive and negative effects were assessed for the first hour after dosing. The 4:1 ratio was chosen because it produced significant attenuation of buprenorphine’s effects without producing significant withdrawal signs. The 2:1 ratio was aversive because it produced withdrawal on four measures and was the only dose combination that the subjects reported that they would not pay money for.

A randomized, double-blind comparison of the effects of tablet formulations of 16 mg/day of buprenorphine at 16/4, buprenorphine/naloxone or placebo, was carried out in a multicenter trial (37). The placebo-controlled portion of the trial was carried out for a 1-month treatment duration. Subjects in either buprenorphine dose group had reduced opiate use and reduced craving versus the placebo group. Thereafter, all subjects were given open-label buprenorphine/naloxone for 11 months. Other subjects participating at new sites were given 1 year of open-label buprenorphine/naloxone.

In October 2002, the FDA approved sublingual buprenorphine tablets and buprenorphine/naloxone tablets for the management of opiate dependence (38). Buprenorphine was concurrently changed to a Schedule III drug on the U.S. Controlled Substances Act (39). According to the Drug Abuse Treatment Act of 2000, qualified physicians can prescribe Schedule III, IV, and V opiates for the treatment of opiate dependence, providing that the FDA approves the medications for that purpose.

Cocaine Dependence

According to the Office of National Drug Control Policy, there are over 3 million long-term cocaine users in the United States. Cocaine abuse and dependence are associated with mortality and significant morbidity (40). There are no FDA-approved medications for the treatment of cocaine dependence. The NIDA Medications Development Program has test-marketed medications for their potential efficacy to reduce cocaine use. Several medications have shown efficacy or preliminary evidence of efficacy. These results will be highlighted.

Disulfiram, an inhibitor of sulfhydryl-containing enzymes, is marketed as a treatment for alcohol dependence. Two open-label studies showed reduction in cocaine intake after reductions in alcohol use in disulfiram-medicated patients (41). Clinical pharmacology studies of the interaction of disulfiram and cocaine have been reported (42, 43). Responses to intranasal cocaine (1 or 2 mg/kg) were altered in subjects treated with either 250 or 500 mg/day of disulfiram (42). Subjects receiving disulfiram reported dysphoria, anxiety, and paranoia after cocaine administration.

Several investigators have reported disulfiram-related decreases in cocaine use in outpatient studies. Doses of 250 and 500 mg/day were reported to reduce cocaine use in subjects using cocaine/alcohol (44). Two other studies showed that disulfiram’s efficacy might be direct rather than indirect (through reduction in concurrent alcohol use) because they were performed with cocaine-abusing, opiate-dependent patients (45, 46). Carroll et al. (47) reported the results of a two-by-two design (drug/placebo-by-cognitive behavior therapy/interpersonal psychotherapy). The psychotherapy treatments were administered weekly for 12 weeks. The disulfiram-cognitive behavior therapy group reduced their drug use compared to the placebo-cognitive behavior therapy and the disulfiram-interpersonal psychotherapy groups. This study suggests an interaction between disulfiram and the type of psychotherapy given (47). A multicenter trial is planned as a follow-up to these studies.

Similar results (with a medication interacting with one type of behavioral therapy and not another) have been reported in a relapse-prevention trial with naltrexone (48). Abstinent, formerly dependent, cocaine-addicted patients showed less relapse when their treatment was combined with a relapse-prevention behavioral therapy but not another behavioral therapy.

Baclofen, a γ-aminobutyric acid (GABA) B agonist (60 mg/day), has been reported to reduce the craving for cocaine in a clinical pharmacology setting (49). Reduction of cocaine use has been reported in baclofen-treated patients in a randomized, placebo-controlled trial (50). Of interest, the patients with the highest levels of cocaine use during the baseline period benefited the most from this medication. A follow-up trial is ongoing to attempt to verify this finding. Another GABA-ergic medication, tiagabine, has been reported to reduce cocaine use at a dose of 24 mg/day (51). A follow-up trial has been completed, and the results will be forthcoming.

Investigators at the University of Pennsylvania have devised a scale that captures the intensity of cocaine withdrawal, the Cocaine Selective Severity Scale (52). This scale, used in conjunction with baseline drug urinalysis testing, predicts treatment efficacy, measured by the ability to maintain 3 weeks of continuous abstinence with 87% accuracy (53). Moreover, patients with high Cocaine Selective Severity Scale scores showed a differential positive response to amantadine (54) or propranolol (55) in outpatient studies.

Another use of this scale is to determine whether medications for relapse prevention will differentially affect patients with high or low Cocaine Selective Severity Scale scores. A trial was conducted with topiramate (200 mg/day maximum dose) in cocaine-dependent patients with low Cocaine Selective Severity Scale scores (<21). The patients who were abstinent during the 2-week baseline period had less return to cocaine use that those randomly assigned to placebo or the patients who continued to use cocaine during the baseline period (56). (Topiramate is a marketed antiepileptic medication that has GABA-enhancing and glutamate-inhibiting properties.) This suggests that topiramate may be beneficial, with patients capable of achieving some level of abstinence through behavioral therapy.

Modafinil is another medication of interest. It is a newer stimulant that increases daytime alertness in narcoleptic patients (57). Preclinical assessment suggests that the stimulatory action is nondopaminergic (5860). Initial assessments of abuse liability in human subjects suggest that the abuse liability is minimal (6163). Moreover, an initial report of cases of stimulant abusers treated with modafinil noted that one user described less craving for amphetamines, and another reported decreased craving and less use of cocaine (64). Clinical laboratory testing of the potential interactive effects of 200 and 400 mg/day doses of modafinil and cocaine in cocaine-experienced subjects was performed (65). No additive effects on cardiovascular parameters were noted. Additionally, the subjects reported a dampening of the subjective effects of cocaine on the Addiction Research Center Inventory (ARCI) Amphetamine Scale (a measure of drug euphoria).

Modafinil also has effects on neuropsychological tests that may be germane to the treatment of substance abuse disorders. In a neuropsychological test battery, modafinil selectively improved performance in normal volunteers (66). Notably, a positive effect on prepotent inhibition was seen in a go/no-go test. This suggests that impulsive responding was reduced. The authors suggested that modafinil might possibly benefit patients with attention deficit hyperactivity disorder (ADHD). A subsequent follow-up study in an adult ADHD group (67) verified that increased accuracy of responding with concomitant response delays was indicative of an inhibition of impulsive responding. Such response delays may allow for other cognitive processes to enter into the decision process.

In summary, modafinil has three effects that provide a rationale for outpatient testing in a cocaine-addicted population:

1.

Its stimulatory quality may reduce anergia and anhedonia and other symptoms of cocaine withdrawal.

2.

Its effects on blunting of craving and the subjective response to cocaine may prevent cocaine priming and multiple-use episodes.

3.

Its effect on cognitive processes to reduce impulsive responding may allow more time for an addict to bring other cognitive systems into the decision-making process of whether to use or not use cocaine.

Outpatient studies assessing the safety and efficacy of modafinil in cocaine-dependent patients are ongoing.

Neurobiological Mechanisms and Pharmacotherapy for Addictions

Multiple neurobiological mechanisms are thought to be involved in the addiction to cocaine. Five mechanisms will be highlighted: 1) the effects of cocaine on the dopamine system, 2) conditioned cueing, 3) cocaine-induced priming, 4) cocaine and stress, and 5) frontal cortex inhibition mechanisms.

Appetitive processes linked to dopamine and mesolimbic-mesocortical pathways have been implicated in the reinforcing actions of cocaine (6870). Cocaine is a dopamine transporter inhibitor that is capable of increasing the extracellular concentration of dopamine severalfold (7173). Supraphysiological concentrations of dopamine may influence the propensity for repeating the cocaine-taking experience. Moreover, there is laboratory evidence that prolonged cocaine administration may result in a hypodopaminergic state (74).

The “agonist” therapy approach, i.e., the use of a similar drug to mimic certain aspects of cocaine, was first tried in NIDA’s attempt to develop a laboratory-based medication for cocaine dependence. Desirable compounds of this type would have reduced abuse liability, such as transporter inhibitors with a slower onset and a longer duration of action. One of the first compounds of this type to come to our attention was GBR 12909. GBR 12909 was reported to increase extracellular dopamine concentrations in the rhesus monkey, with a peak effect at 1 hour and a sustained duration (71). Similar results were seen in squirrel monkeys (73). GBR 12909 has been evaluated in the monkey and found to block cocaine self-administration (7577), especially under conditions in which the unit dose of cocaine was low or the response requirements for cocaine were high (78).

Studies in human subjects have shown that GBR 12909 (vanoxerine) is not a psychomotor stimulant (79). NIDA has recently stopped its phase I studies in cocaine-experienced subjects because of the appearance of rate-dependent QTc prolongation in five of five subjects given 75 mg/day of GBR 12909 for 11 days (Cantilena, personal communication).

A second approach that is derived from the dopamine hypothesis of cocaine’s action is the possible development of dopamine “stabilizers,” which may be partial agonists or “partial” mixed-action antagonists. Compounds of this type are mild stimulants in animals but can reduce the locomotor stimulant effects of amphetamine (80). The interpretation at the time was that autoreceptor antagonists preferentially inhibited presynaptic dopamine receptors, but at higher doses and at higher levels of dopamine stimulation, they also block postsynaptic receptors. Thus, in low-dopaminergic states, such compounds would increase dopamine release but act as antagonists under conditions of higher dopaminergic tone. Other compounds of this type (AJ76 and UH-232) antagonized cocaine depression of ventral-tegmental area firing rates and reduced cocaine-induced locomotor activity when injected simultaneously (UH-232) or 30 minutes after cocaine administration (AJ76) (81). AJ76 also antagonized cocaine self-administration (82). A third potential autoreceptor antagonist, DS-121, was reported to produce a dose-dependent (high dose) antagonism of amphetamine or cocaine discrimination (83) and antagonized cocaine self-administration in the rat (84).

Dopamine D2 partial agonists can also function as both agonists and antagonists and may also have stabilizing effects. Terguride, a partial agonist at D2 receptors, reversed amphetamine withdrawal signs in animals (working for a sweetened solution) (85) and also reduced amphetamine self-administration under a progressive ratio schedule (86). Of interest, quinpirole, a full D2 agonist, did not antagonize amphetamine self-administration. These data suggest that these “stabilizer” compounds may have different properties than full agonists or full antagonists.

The first marketed medication with a similar mechanism that has been dubbed a dopamine “stabilizer” (87, 88) is aripiprazole, which is approved for the treatment of schizophrenia. Its efficacy has been reviewed, and it is believed to have “mixed” actions or different efficacy at different receptor subtypes (89). Neuroimaging studies have show that clinically relevant doses occupy 95% of the dopamine receptors without producing extrapyramidal side effects (90). Given its characterization as a dopamine system stabilizer, NIDA is interested in evaluating its effects in both cocaine- and methamphetamine-dependent patients.

Conditioned cueing can be characterized as an intersection of Pavlovian and instrumental conditioning processes, whereby previously neutral stimuli paired to drug experiences develop both motivational and reinforcing significance. For a review of the behavioral concepts and laboratory approaches to studying this phenomenon, refer to reviews by Everitt et al. and Everitt and Robbins (91, 92). Second-order schedules of drug reinforcement are used to study the behavior, its neural substrates, and its pharmacological modulation. Lesioning studies in animals have differentiated the involvement of nuclei, such as the basolateral amygdala, in responding for cues, as opposed to a role in primary reinforcement. Human imaging studies have reported that conditioned cues activate the amygdala, anterior cingulate, lateral orbitofrontal cortex, rhinal cortex, and right hemispheric dorsolateral prefrontal cortex (9395). Conditioned cueing may be of relevance to human addiction, and rodent models may be of value in discovering pharmacological modulators of conditioned cues.

Multiple neurotransmitter systems may be involved in behavioral responses to conditioned cues. Diverse neurotransmitter systems, such as GABA B (49), the endocannabinoid system (96), the dopamine D3 system (97), and ionotropic glutamate antagonists (98, 99) have been implicated. Cannabinoid antagonists, D3 partial agonists and antagonists, and AMPA antagonists are being sought by NIDA for testing in conditioned-cue paradigms.

Drug priming is another neural mechanism of interest (100). Priming is defined as the response to reintroduction of a drug, in the form of subsequent increased drug intake, in a formerly dependent drug user. This behavior can be modeled in the laboratory by using noncontingent administration of cocaine in animals trained to self-administer cocaine. Noncontingent administration can also provoke reinstatement in animals after extinction of self-administration (101).

There is evidence of both dopaminergic and glutamatergic influences on the effects of drug priming. Systemic administration of a D1 agonist blocked cocaine priming-induced reinstatement (102), and priming could be blocked by microinjections of the dopamine antagonist flupenthixol into the medial prefrontal cortex. Reinstatement induced by microinjections of cocaine into the medial prefrontal cortex can be blocked by administration of CNQX, an AMPA/kainate antagonist, into the nucleus accumbens, suggesting that the glutamatergic pathway from the medial prefrontal cortex to the nucleus accumbens plays an important role (103). Similarly, Cornish and Kalivas (104) demonstrated a facilitatory role for glutamate acting at AMPA receptors in the effects of priming. Systemic administration of cocaine or the direct infusion of AMPA or dopamine into the nucleus accumbens can produce reinstatement of drug-seeking behavior in rats, and intra-accumbens administration of CNQX blocked the priming effects of AMPA, dopamine, and cocaine. In contrast, intra-accumbens administration of fluphenazine was only successful in blocking the effects of intra-accumbens dopamine on reinstatement, suggesting the importance of glutamate in priming-induced reinstatement.

The contribution of glutamate to reinstatement was also studied in cocaine self-administering rats and their yoked comparison subjects. Glutamate concentrations increased in the nucleus accumbens only in response to reinstatement of lever pressing for cocaine. In contrast, dopamine concentrations increased in both groups of rats. Reversible inhibition of the frontal cortex by infusion of the GABA B agonist, baclofen, prevented the increase in accumbal glutamate, suggesting that the glutamatergic prefrontal cortex connection to the accumbens is a major mediator of cocaine-primed reinstatement (105). The neuronal pathways thought to be involved in cocaine addiction have been reviewed by Kalivas (106). Reciprocal glutamatergic connections between the amygdala and the medial prefrontal cortex also suggest that some components of drug-associated cueing may be mediated through a medial prefrontal cortex-accumbal pathway. For these reasons, NIDA has great interest in evaluating AMPA antagonists or medications with an AMPA antagonist component for the prevention of relapse.

Stress is another factor thought to increase the propensity of alcohol and drug users to relapse (107111). There are multiple models of stress in laboratory animals, and to some extent, the effects of drugs on stress are model specific and stressor specific (112). In the intermittent foot shock stress model, stress has been shown to reinstate response in animals trained to self-administer cocaine (113, 114), heroin (115, 116), nicotine (117), alcohol (118, 119), and cannabinoids (120). Central noradrenergic and extrahypothalamic corticotropin-releasing factor (CRF) have been shown to mediate the effects of intermittent foot shock stress on reinstatement. Lofexidine (121) and CRF antagonists (122) have been shown to modulate the noradrenergic and CRF components of the response to stress-induced reinstatement of cocaine self-administration.

Finally, the effects drugs of abuse on frontal cortex inhibitory processes and drug abuse as compulsive behavior are just beginning to be understood and modeled in the laboratory (123). As our understanding of the deleterious effects of long-term drug abuse on frontal lobe function and cognitive decision-making processes evolves (see reference 124 for a review), medications that affect these processes through cognitive enhancement (e.g., modafinil) will be evaluated for their efficacy in the treatment of drug abuse.

In summary, LAAM, buprenorphine, and buprenorphine/naloxone have been developed for the treatment of opiate dependence. The buprenorphine products may be prescribed by qualified physicians, representing a shift toward the medical management of opiate addiction in the office-based setting. Progress in developing medications for cocaine dependence has been realized. The “first generation” of medications for cocaine dependence are in confirmatory testing in clinical trials or in the planning stages of confirmatory trials. Molecular targets derived from neuroscience will form the second generation of products for the treatment of cocaine dependence. Insofar as these products may affect processes that may, to one extent or another, affect all substance dependence disorders, the “second generation” of products may be developed for multiple addictions.

Presented in part at the 157th annual meeting of the American Psychiatric Association, New York, May 1–6, 2004. Received Sept. 3, 2004; revision received Nov. 30, 2004; accepted Dec. 10, 2004. From the Division of Pharmacotherapies and Medical Consequences of Drug Abuse, National Institute on Drug Abuse, NIH, Department of Health and Human Services. Address corresponding and reprint requests to Dr. Vocci, Division of Pharmacotherapies and Medical Consequences of Drug Abuse, 6001 Executive Blvd., Rm. 4133, MSC 9551, Bethesda, MD 20892-9551; (e-mail).

References

1. Dole VP, Nyswander M: A medical treatment of diacetylmorphine (heroin) addiction: a clinical trial with methadone hydrochloride. JAMA 1965; 193:646–650Crossref, MedlineGoogle Scholar

2. Dole VP, Nyswander ME, Kreek MJ: Narcotic blockade. Arch Intern Med 1966; 118:304–309Crossref, MedlineGoogle Scholar

3. Dole VP, Robinson JW, Orraga J, Towns E, Searcy P, Caine E: Methadone treatment of randomly selected criminal addicts. N Engl J Med 1969; 280:1372–1375Crossref, MedlineGoogle Scholar

4. Newman RG, Whitehill WB: Double blind comparison of methadone and placebo maintenance treatment of narcotic addicts in Hong Kong. Lancet 1978; 8141:484–488Google Scholar

5. Gronbladh L, Gunne L: Methadone-assisted rehabilitation of Swedish heroin addicts. Drug Alcohol Depend 1989; 24:31–37Crossref, MedlineGoogle Scholar

6. Gunne L, Gronbladh L: The Swedish Methadone Maintenance Program, in The Social and Medical Aspects of Drug Abuse. Edited by Serban G. Jamaica, NY, Spectrum Publications, 1984, pp 205–213Google Scholar

7. Gronbladh L, Ohlund LS, Gunne LM: Mortality in heroin addiction: impact of methadone treatment. Acta Psychiatr Scand 1990; 82:223–227Crossref, MedlineGoogle Scholar

8. Strain EC, Stitzer ML, Liebson I, Bigelow GE: Dose-response effects of methadone in the treatment of opiate dependence. Ann Intern Med 1993; 119:23–27Crossref, MedlineGoogle Scholar

9. Johnson RE, Jaffe JH, Fudala PJ: A controlled trial of buprenorphine treatment for opioid dependence. JAMA 1992; 267:2750–2755Crossref, MedlineGoogle Scholar

10. Strain EC, Bigelow GE, Liebson IA, Stitzer ML: Moderate- vs high-dose methadone in the treatment of opiate dependence: a randomized trial. JAMA 1999; 281:1000–1005Crossref, MedlineGoogle Scholar

11. Ling W, Wesson DR, Charuvastra C, Klett CJ: A controlled trial comparing buprenorphine and methadone maintenance in opioid dependence. Arch Gen Psychiatry 1996; 53:401–407Crossref, MedlineGoogle Scholar

12. Johnson RE, Chatuape MA, Strain EC, Walsh SL, Stitzer ML, Bigelow GE: A comparison of levomethadyl acetate, buprenorphine, and methadone for opioid dependence. N Engl J Med 2000; 343:1290–1297Crossref, MedlineGoogle Scholar

13. D’Aunno T, Pollack HA: Changes in methadone treatment practices: results from a national panel study, 1988–2000. JAMA 2002; 288:850–856Crossref, MedlineGoogle Scholar

14. Gearing MF: Methadone maintenance in the treatment of heroin addicts in New York City: a ten year overview, in Neurotoxicology. Edited by Roizen L, Shiraki H, Grcevic N. New York, Raven Press, 1977, pp 77–79Google Scholar

15. Capelhorn JR, Dalton MS, Haldar F, Petranas AM, Nisbet JG: Methadone maintenance and addicts’ risk of fatal heroin overdose. Subst Use Misuse 1996; 31:1177–1196Google Scholar

16. Novick D, Kreek MJ, Des Jarlais DC, Spira TJ, Khuri ET, Ragunath J, Kalyanaraman VS, Gelb AM, Miescher A: Antibody to LAV, the putative agent of AIDS, in parenteral drug abusers and methadone-maintained patients: therapeutic, historical, and ethical aspects. NIDA Res Monogr 1986; 67:318–320MedlineGoogle Scholar

17. Metzger DS, Woody CE, McLellan AR, O’Brien CP, Druley P, Navaline H, DePhillips D, Druley P, Abrutyn E: Human immunodeficiency virus seroconversion among intravenous drug users in- and out-of-treatment: an 18 month prospective follow-up. J Acquir Immune Defic Syndr 1993; 6:1049–1056MedlineGoogle Scholar

18. Billings RE, Booker R, Smits S, Peland A, McMahon RE: Metabolism of acetyl methadol: a sensitive assay for non-acetyl methadol and identification of a new metabolite. J Med Chem 1975; 16:305–306CrossrefGoogle Scholar

19. McMahon RE, Calp HW, Marshal FJ: The metabolism of alpha-dl-acetyl methadol in the rat: the identification of a probable active metabolite. J Pharmacol Exp Ther 1965; 149:436–445MedlineGoogle Scholar

20. Nickander R, Booher R, Miles H: Alpha-l-acetylmethadol and its n-demethylated metabolites have potent opiate action in the guinea pig ileum. Life Sci 1974; 14:2011–2017Crossref, MedlineGoogle Scholar

21. Kaiko RF, Inturissi CE: Disposition of acetylmethadol in relation to its pharmacological action. Clin Pharmacol Ther 1975; 18:96–103Crossref, MedlineGoogle Scholar

22. Ling W, Charuvastra VC, Kaim SC, Klett CJ: Methadyl acetate and methadone as maintenance treatments for heroin addicts. Arch Gen Psychiatry 1976; 33:709–720Crossref, MedlineGoogle Scholar

23. Ling W, Klett CJ, Gillis RD: A cooperative clinical study of methadyl acetate. Arch Gen Psychiatry 1978; 35:345–353Crossref, MedlineGoogle Scholar

24. Walsh SJ, Johnson RE, Cone EJ, Bigelow GE: Intravenous and oral l-alpha-acetylmethadol: pharmacodynamics and pharmacokinetics in humans. J Pharmacol Exp Ther 1998; 285:71–82MedlineGoogle Scholar

25. Fudala PJ, Vocci F, Montgomery A, Trachtenberg AI: Levomethadyl acetate (LAAM) for the treatment of opiate dependence: a multisite, open label study of LAAM safety and an evaluation of the product labeling and treatment regulations. J Maint Addict 1997; 1:9–39CrossrefGoogle Scholar

26. Jones HE, Strain EC, Bigelow GE, Walsh SL, Stitzer ML, Eissenberg TE, Johnson RE: Induction with levomethadyl acetate: safety and efficacy. Arch Gen Psychiatry 1998; 55:729–736Crossref, MedlineGoogle Scholar

27. Eissenberg T, Bigelow GE, Strain EC, Walsh SL, Brooner RK, Stitzer ML, Johnson RE: Dose-related efficacy of levo-methadyl acetate for treatment of opiate dependence. JAMA 1997; 227:1945–1951CrossrefGoogle Scholar

28. Kreek MJ, Vocci FJ: History and current status of opioid maintenance treatments: blending conference session. J Subst Abuse Treat 2002; 23:93–105Crossref, MedlineGoogle Scholar

29. Martin WR, Eades CG, Thompson JA, Huppler RE, Gilbert PE: The effects of morphine- and nalorphine-like drugs in the nondependent and morphine-dependent chronic spinal dog. J Pharmacol Exp Ther 1976; 197:517–532MedlineGoogle Scholar

30. Jasinski DR, Pevnick JS, Griffiths JD: Human pharmacology and abuse potential of the analgesic buprenorphine. Arch Gen Psychiatry 1978; 35:501–516Crossref, MedlineGoogle Scholar

31. Jasinski DR, Fudala PJ, Johnson RE: Sublingual versus subcutaneous buprenorphine in opiate abusers. Clin Pharmacol Ther 1989; 45:513–519Crossref, MedlineGoogle Scholar

32. Bickel WK, Stitzer ML, Bigelow GE, Liebson IA, Jasinski DR, Johnson RE: Buprenorphine: dose-related blockade of opioid challenge effect in opioid dependent humans. J Pharmacol Exp Ther 1988; 247:47–53MedlineGoogle Scholar

33. Ling W, Charuvastra C, Collins JF, Batki S, Brown LS Jr, Kintaudi P, Wesson DR, McNicholas L, Tusel DJ, Malkerneker U, Renner JA Jr, Santos E, Casadonte P, Fye C, Stine S, Wang RI, Segal D: Buprenorphine maintenance treatment of opiate dependence: a multicenter, randomized clinical trial. Addiction 1998; 93:475–486Crossref, MedlineGoogle Scholar

34. Mendelson J, Jones RT, Welm S, Baggott M, Fernandez I, Melby AK, Nath RP: Buprenorphine and naloxone combinations: the effects of three dose ratios in morphine-stabilized, opiate-dependent volunteers. Psychopharmacology (Berl) 1999; 141:37–46Crossref, MedlineGoogle Scholar

35. Mendelson J, Jones RT, Welm S, Brown J, Batki S: Buprenorphine and naloxone interactions in methadone maintenance patients. Biol Psychiatry 1997; 41:1095–1101Crossref, MedlineGoogle Scholar

36. Harris D, Jones RT, Welm S, Upton R, Lin E, Mendelson J: Buprenorphine and naloxone co-administration in opiate-dependent patients stabilized on sublingual buprenorphine. Drug Alcohol Depend 2000; 61:85–94Crossref, MedlineGoogle Scholar

37. Fudala PJ, Bridge TP, Herbert S, Williford WO, Chiang CN, Jones K, Collins J, Raisch D, Casadonte P, Goldsmith RJ, Ling W, Malkerneker U, McNicholas L, Renner J, Stine S, Tusel D: Office-based treatment of opiate addiction with a sublingual-tablet formulation of buprenorphine and naloxone. N Engl J Med 2003; 349:949–958Crossref, MedlineGoogle Scholar

38. US Food and Drug Administration Center for Drug Evaluation and Research Drug Information: Subutex (buprenorphine hydrochloride) and Suboxone tablets (buprenorphine hydrochloride and naloxone hydrochloride). http://www.fda.gov/cder/drug/infopage/subutex_suboxone/default.htmGoogle Scholar

39. Sapienza F: Schedules of controlled substances: rescheduling of buprenorphine from Schedule V to Schedule III. Fed Regist 2002; 67:62354–62370MedlineGoogle Scholar

40. Mortality Data From the Drug Abuse Warning Network. Rockville, Md, Substance Abuse and Mental Health Services Administration, 2002Google Scholar

41. Higgins ST, Budney AJ, Bickel WK, Hughes JR, Foerg F: Disulfiram therapy in patients abusing cocaine and alcohol (letter). Am J Psychiatry 1993; 150:675–676MedlineGoogle Scholar

42. Hameedi FA, Rosen MI, McCance-Katz EF, McMahon TJ, Price LH, Jatlow PI, Woods SW, Kosten TR: Behavioral, physiological, and pharmacological interaction of cocaine and disulfiram in humans. Biol Psychiatry 1995; 37:560–563Crossref, MedlineGoogle Scholar

43. McCance EF, Kosten TR, Jatlow P: Chronic disulfiram treatment effects on intranasal cocaine administration. Biol Psychiatry 1998; 43:540–543Crossref, MedlineGoogle Scholar

44. Carroll KM, Rounsaville BJ, Gordon LT, Nich C, Jatlow P, Bisighini RM, Gawin FH: Psychotherapy and pharmacotherapy for ambulatory cocaine abusers. Arch Gen Psychiatry 1994; 51:177–187Crossref, MedlineGoogle Scholar

45. George TP, Chawarski MC, Pakes J, Carroll KM, Kosten TR, Schottenfeld RS: Disulfiram versus placebo for cocaine dependence in buprenorphine-maintained subjects: a preliminary trial. Biol Psychiatry 2000; 47:1080–1086Crossref, MedlineGoogle Scholar

46. Petrakis IL, Carroll KM, Nich C, Gordon LT, McCance-Katz EF, Frankforter T, Rounsaville BJ: Disulfiram treatment for cocaine dependence in methadone-maintained subjects. Addiction 2000; 95:219–228Crossref, MedlineGoogle Scholar

47. Carroll KM, Fenton LR, Ball SA, Nich C, Frankforter TL, Shi J, Rounsaville BJ: Efficacy of disulfiram and cognitive behavioral therapy in cocaine dependent outpatients: a randomized placebo-controlled trial. Arch Gen Psychiatry 2004; 61:264–272Crossref, MedlineGoogle Scholar

48. Schmitz JM, Stotts AL, Rhoades HM, Grabowski J: Naltrexone and relapse prevention treatment for cocaine-dependent patients. Addict Behav 2001; 26:167–180Crossref, MedlineGoogle Scholar

49. Brebner K, Childress AR, Roberts DCS: A potential role for GABA(B) agonists in the treatment of psychostimulant addiction. Alcohol Alcohol 2002; 37:478–484Crossref, MedlineGoogle Scholar

50. Shoptaw S, Yang X, Rotherman-Fuller EJ, Hsieh YC, Kintaudi PC, Charuvastra YC, Ling W: Randomized placebo-controlled trial of baclofen for cocaine dependence: preliminary effects for individuals with chronic patterns of cocaine use. J Clin Psychiatry 2003; 64:1440–1448Crossref, MedlineGoogle Scholar

51. Gonzalez G, Sevarino K, Sofouglu M, Poling J, Oliveto A, Gonsai K, George TP, Kosten TR: Tiagabine increases cocaine-free urines in cocaine-dependent methadone-treated patients: results of a randomized pilot study. Addiction 2003; 98:1625–1632Crossref, MedlineGoogle Scholar

52. Kampman K, Volpicelli J, McGinnis DE, Alterman AI, Weinrieb RM, D’Angelo L, Epperson LE: Reliability and validity of the Cocaine Selective Severity Scale. Addict Behav 1998; 23:449–461Crossref, MedlineGoogle Scholar

53. Kampman K, Volpicelli J, Mulvaney F, Rukstalis M, Alterman AI, Pettinati H, Weinrieb RM, O’Brien CP: Cocaine withdrawal severity and urine toxicology results from treatment entry predict outcome in medications trials for cocaine dependence. Addict Behav 2002; 27:251–260Crossref, MedlineGoogle Scholar

54. Kampman KM, Volpicelli JR, Alterman AI, Cornish J, O’Brien CP: Amantadine in the treatment of cocaine-dependent patients with severe cocaine withdrawal symptoms. Am J Psychiatry 2000; 157:2052–2054LinkGoogle Scholar

55. Kampman K, Volpicelli J, Mulvaney F, Alterman AI, Cornish J, Gariti P, Cnaan A, Poole S, Muller E, Acosta T, Luce D, O’Brien C: Effectiveness of propranolol for cocaine dependence treatment may depend on cocaine withdrawal symptom severity. Drug Alcohol Depend 2001; 63:69–78Crossref, MedlineGoogle Scholar

56. Kampman KM, Pettinati H, Lynch KG, Dackis C, Sparkman T, Weigley C, O’Brien CP: A pilot trial of topiramate for the treatment of cocaine dependence. Drug Alcohol Depend 2004; 75:233–240Crossref, MedlineGoogle Scholar

57. Moldofsky H, Broughton RJ, Hill JD: A randomized trial of long-term continued efficacy and safety of modafinil in narcolepsy. Sleep Med 2000; 1:109–116Crossref, MedlineGoogle Scholar

58. Ferraro L, Antonelli T, O’Connor WT, Tanganelli S, Rambert FA, Fuxe K: Modafinil: an antinarcoleptic drug with a different neurochemical profile to d-amphetamine and dopamine uptake blockers. Biol Psychiatry 1997; 42:1181–1183Crossref, MedlineGoogle Scholar

59. Simon P, Hemet C, Ramassamy C, Costentin J: Non-amphetaminic mechanism of stimulant locomotor effect of modafinil in mice. Eur Neuropsychopharmacol 1995; 5:509–514Crossref, MedlineGoogle Scholar

60. Lin JS, Hou Y, Jouvet M: Potential brain neuronal targets for amphetamine-, methylphenidate-, and modafinil-induced wakefulness, evidenced by c-fos immunocytochemistry in the cat. Proc Natl Acad Sci USA 1996; 93:14128–14133Crossref, MedlineGoogle Scholar

61. Rush CR, Kelly TH, Hays LR, Baker RW, Wooten AF: Acute behavioral and physiological effects on modafinil in drug abusers. Behav Pharmacol 2002; 13:105–115Crossref, MedlineGoogle Scholar

62. Jasinski DR: An evaluation of the abuse potential of modafinil using methylphenidate as a reference. J Psychopharmacol 2000; 14:53–60Crossref, MedlineGoogle Scholar

63. Warot D, Corruble E, Payan C, Weil JS, Peuch AJ: Subjective effects of modafinil: a new central adrenergic stimulant in healthy volunteers: a comparison with amphetamine, caffeine, and placebo. Eur Psychiatry 1993; 8:201–208Google Scholar

64. Malcolm R, Book SW, Moak D, DeVane L, Czepowicz V: Clinical applications of modafinil in stimulant abusers: low abuse potential. Am J Addict 2002; 11:247–249Crossref, MedlineGoogle Scholar

65. Dackis CA, Lynch KG, Yu E, Samaha FF, Kampman KM, Cornish JW, Rowan A, Poole S, White L, O’Brien CP: Modafinil and cocaine: a double-blind, placebo-controlled drug interaction study. Drug Alcohol Depend 2003; 70:29–37Crossref, MedlineGoogle Scholar

66. Turner DC, Robbins TW, Clark L, Aron AR, Dowson J, Sahakian BJ: Cognitive enhancing effects of modafinil in healthy volunteers. Psychopharmacology (Berl) 2003; 165:260–269Crossref, MedlineGoogle Scholar

67. Turner DC, Clark L, Dowson J, Robbins TW, Sahakian BJ: Modafinil improves cognition and response inhibition in adult attention-deficit/hyperactivity disorder. Biol Psychiatry 2004; 55:1031–1040Crossref, MedlineGoogle Scholar

68. Ritz MC, Lamb RJ, Goldberg SR, Kuhar MJ: Cocaine receptors on dopamine transporters are related to self-administration of cocaine. Science 1987; 237:1219–1223Crossref, MedlineGoogle Scholar

69. Bradberry CW: Acute and chronic dopamine dynamics in a non-human primate model of recreational cocaine use. J Neurosci 2000; 20:7109–7115Crossref, MedlineGoogle Scholar

70. Pontieri FE, Tanda G, Di Chiari G: Intravenous cocaine, morphine, and amphetamine preferentially increase extra-cellular dopamine in the “shell” as compared with the “core” of the rat nucleus accumbens. Proc Natl Acad Sci USA 1995; 92:12304–12308Crossref, MedlineGoogle Scholar

71. Tsukada H, Harada N, Nishiyama S, Ohba H, Kakiuchi T: Dose response and duration effects of acute administrations of cocaine and GBR112909 on dopamine synthesis and transporter in the conscious monkey brain: PET studies combined with microdialysis. Brain Res 2000; 860:141–148Crossref, MedlineGoogle Scholar

72. Bradberry CW, Barrett-Larimore RL, Jatkow P, Rubino SR: Impact of self-administered cocaine and cocaine cues on extracellular dopamine in mesolimbic and sensorimotor striatum in rhesus monkeys. J Neurosci 2000; 20:3874–3883Crossref, MedlineGoogle Scholar

73. Czoty PW, Justice JB Jr, Howell LL: Cocaine-induced changes in extracellular dopamine determined by microdialysis in awake squirrel monkeys. Psychopharmacology (Berl) 2000; 48:299–306CrossrefGoogle Scholar

74. Parsons LH, Smith AD, Justice JB: Basal extracellular dopamine is decreased in the rat nucleus accumbens during abstinence from chronic cocaine. Synapse 1991; 9:60–65Crossref, MedlineGoogle Scholar

75. Glowa J, Wojnicki FHE, Matecka D, Bacher J, Mansbach R, Balster R, Rice KC: Effects of dopamine reuptake inhibitors on food and cocaine-maintained responding, I: dependence on the unit dose of cocaine. Exp Clin Psychopharmacol 1995; 3:219–231CrossrefGoogle Scholar

76. Glowa J, Wojnicki FHE, Matecka D, Rice K, Rothman RB: Effects of dopamine reuptake inhibitors on food and cocaine-maintained responding, II: comparisons with other drugs and repeated administrations. Exp Clin Psychopharmacol 1995; 3:232–239CrossrefGoogle Scholar

77. Lindsey KP, Wilcox KM, Votaw JR, Goodman MW, Plisson C, Carroll FI, Rice KC, Howell LL: Effects of dopamine transporter inhibitors on cocaine self-administration in rhesus monkeys: relationship to transporter occupancy determined by positron emission tomography neuroimaging. J Pharmacol Exp Ther 2004; 309:959–969Crossref, MedlineGoogle Scholar

78. Stafford D, Rice KC, Lewis DB, Glowa JR: Response requirements and unit dose modify the effects of GBR 12909 on cocaine-maintained behavior. Exp Clin Psychopharmacol 2000; 8:539–548Crossref, MedlineGoogle Scholar

79. Sogaard U, Michalow J, Butler B, Lund LA, Ingersen SH, Skrumsager BK, Rafaelsen OJ: A tolerance study of single and multiple dosing of the selective dopamine uptake inhibitor GBR 12909 in healthy volunteers. Int Clin Psychopharmacol 1990; 5:237–251Crossref, MedlineGoogle Scholar

80. Sonesson C, Waters N, Svensson K, Carlsson A, Smith MW, Piercey MF, Meier E, Wikstrom H: Substituted 3-phenylpiperidines: new centrally acting dopamine autoreceptor antagonists. J Med Chem 1993; 36:3188–3196Crossref, MedlineGoogle Scholar

81. Piercey MF, Lum JT, Hoffman WE, Carlsson A, Ljung E, Svensson K: Antagonism of cocaine’s pharmacological effects by the stimulant dopaminergic antagonists, (+)-AJ76 and (+)-UH232. Brain Res 1992; 588:217–222Crossref, MedlineGoogle Scholar

82. Richardson NR, Piercey MF, Svensson K, Collins RJ, Myers JE, Roberts DCS: Antagonism of cocaine self-administration by the preferential dopamine autoreceptor antagonist (+)-AJ76. Brain Res 1993; 619:15–21Crossref, MedlineGoogle Scholar

83. Clark D, Exner M, Furmidge LJ, Svensson K, Sonesson C: Effects of the dopamine autoreceptor antagonist (-)-DS121 on the discriminative stimulus properties of d-amphetamine and cocaine. Eur J Pharmacol 1995; 275:67–74Crossref, MedlineGoogle Scholar

84. Smith A, Piercey MF, Roberts DCS: Effect of (-)-DS121 and (+)-UH232 on cocaine self-administration in rats. Psychopharmacology (Berl) 1995; 120:93–98Crossref, MedlineGoogle Scholar

85. Orsini C, Koob G, Pulvirenti L: Dopamine partial agonist reverses amphetamine withdrawal in rats. Neuropsychopharmacology 2001; 25:789–792Crossref, MedlineGoogle Scholar

86. Izzo E, Orsini C, Koob G, Pulvirenti L: A dopamine partial agonist and antagonist block amphetamine self-administration in a progressive ratio schedule. Pharmacol Biochem Behav 2001; 68:701–708Crossref, MedlineGoogle Scholar

87. Stahl S: Dopamine system stabilizers, aripiprazole, and the next generation of antipsychotics, part 1: “Goldilocks” actions at dopamine receptors. J Clin Psychiatry 2001; 62:841–842Crossref, MedlineGoogle Scholar

88. Stahl S: Dopamine system stabilizers, aripiprazole, and the next generation of antipsychotics, part 2: illustrating their mechanism of action. J Clin Psychiatry 2001; 62:923–924Crossref, MedlineGoogle Scholar

89. Keck PE Jr, McElroy SL: Aripiprazole: a partial dopamine D2 receptor agonist antipsychotic. Expert Opin Investig Drugs 2003; 12:655–662Crossref, MedlineGoogle Scholar

90. Grunder G, Carlsson A, Wong DF: Mechanism of new antipsychotic medications. Arch Gen Psychiatry 2003; 60:974–977Crossref, MedlineGoogle Scholar

91. Everitt BE, Cockinson A, Robbins TW: The neuropsychological basis of addictive behavior. Brain Res Rev 2001; 36:129–138Crossref, MedlineGoogle Scholar

92. Everitt BE, Robbins TW: Second-order schedules of drug reinforcement in rats and monkeys: measurement of reinforcing efficacy and drug-seeking behavior. Psychopharmacology (Berl) 2000; 153:17–30Crossref, MedlineGoogle Scholar

93. Grant S, London ED, Newlin D, Villemagne VL, Xiang L, Contoreggi C, Phillips RL, Kimes AS, Margolin A: Activation of memory circuits during cocaine cue-elicited craving. Proc Natl Acad Sci USA 1996; 93:12040–12045Crossref, MedlineGoogle Scholar

94. Childress AR, McElgin W, Mozley PD, O’Brien CP: Limbic activation during cue-induced craving for cocaine and for natural rewards (abstract). Biol Psychiatry 1999; 45:53SGoogle Scholar

95. Bonson KR, Grant SJ, Contoreggi CS, Links JM, Metcalfe J, Weyl HL, Kurian V, Ernst M, London ED: Neural systems and cue-induced cocaine craving. Neuropsychopharmacology 2002; 26:376–386Crossref, MedlineGoogle Scholar

96. DeVries TJ, Shaham Y, Homberg JR, Crombag H, Schuurman K, Dieben J, Vanderschuren LJ, Schoffelmeer AN: A cannabinoid mechanism in relapse to cocaine seeking. Nat Med 2001; 7:1151–1154Crossref, MedlineGoogle Scholar

97. Vorel SR, Ashby CR Jr, Paul M, Liu X, Hayes R, Hagan JJ, Middlemiss DN, Stemp G, Gardner EL: Dopamine D3 receptor antagonism inhibits cocaine-seeking and cocaine-enhanced brain reward in rats. J Neurosci 2002; 22:9595–9603Crossref, MedlineGoogle Scholar

98. Di Ciano P, Everitt BJ: Dissociable effects of antagonism of NMDA and AMPA receptors in the nucleus accumbens core and shell on cocaine-seeking behavior. Neuropsychopharmacology 2001; 25:241–360CrossrefGoogle Scholar

99. Backstrom P, Hyytia P: Attenuation of cocaine-seeking behavior by the AMPA/kainate receptor antagonist CNQX in rats. Psychopharmacology (Berl) 2003; 166:69–76Crossref, MedlineGoogle Scholar

100. de Wit H: Priming effects with drugs and other reinforcers. Exp Clin Psychopharmacol 1996; 4:5–10CrossrefGoogle Scholar

101. de Wit H, Stewart J: Reinstatement of cocaine-reinforced responding in the rat. Psychopharmacology (Berl) 1981; 75:134–143Crossref, MedlineGoogle Scholar

102. Self D, Baarnhart WJ, Lehman DA, Nestler EJ: Opposite modulation of cocaine-seeking behavior by D1 and D2-like dopamine receptor agonists. Science 1996; 271:1586–1589Crossref, MedlineGoogle Scholar

103. Park WK, Bari AA, Jey AR, Anderson SM, Spealman RD, Rowlett JK, Pierce RC: Cocaine administered into the medial prefrontal cortex reinstates cocaine-seeking behavior by increasing AMPA receptor-mediated glutamate transmission in the nucleus accumbens. J Neurosci 2002; 22:2916–2925Crossref, MedlineGoogle Scholar

104. Cornish JL, Kalivas PW: Glutamate transmission in the nucleus accumbens mediates relapse in cocaine addiction. J Neurosci 2000; 20:1–5Crossref, MedlineGoogle Scholar

105. McFarland K, Lapish CC, Kalivas PW: Prefrontal glutamate release into the core of the nucleus accumbens mediates cocaine-induced reinstatement of drug-seeking behavior. J Neurosci 2003; 23:3531–3537Crossref, MedlineGoogle Scholar

106. Kalivas PW: Glutamate systems in cocaine addiction. Curr Opin Pharmacol 2004; 4:23–29Crossref, MedlineGoogle Scholar

107. Brown SA, Vik PW, Patterson TL, Grant I, Schuckit M: Stress vulnerability and adult alcohol relapse. J Stud Alcohol 1995; 56:538–545Crossref, MedlineGoogle Scholar

108. Sinha R, Fuse T, Aubin LR, O’Malley S: Psychological stress, drug-related cues, and cocaine craving. Psychopharmacology (Berl) 2000; 152:140–148Crossref, MedlineGoogle Scholar

109. Kosten TR, Rounsaville BJ, Kleber HD: A 2.5 year follow up of depression, life crises, and treatment effects on abstinence among opioid addicts. Arch Gen Psychiatry 1986; 43:733–739Crossref, MedlineGoogle Scholar

110. Kreek MJ, Koob GF: Drug dependence: stress and dysregulation of brain reward pathways. Drug Alcohol Depend 1998; 51:23–47Crossref, MedlineGoogle Scholar

111. McFall ME, Mackay PW, Donovan DM: Combat-related post-traumatic stress disorder and severity of substance abuse in Vietnam veterans. J Stud Alcohol 1992; 53:357–363Crossref, MedlineGoogle Scholar

112. Lu L, Shepard JD, Hall FS, Shaham Y: Effect of environmental stressors on opiate and psychostimulant reinforcement, reinstatement and discrimination in rats: a review. Neurosci Behav Rev 2003; 27:457–491Crossref, MedlineGoogle Scholar

113. Erb S, Shaham Y, Stewart J: The role of corticotropin-releasing factor and corticosterone in stress- and cocaine-induced relapse to cocaine seeking in rats. J Neurosci 1998; 18:5529–5536Crossref, MedlineGoogle Scholar

114. Ahmed SH, Koob GF: Cocaine- but not food-seeking behavior is reinstated by stress after extinction. Psychopharmacology (Berl) 1997; 132:289–295Crossref, MedlineGoogle Scholar

115. Shaham Y, Stewart S: Stress reinstates heroin self-administration behavior in drug-free animals: an effect mimicking heroin, not withdrawal. Psychopharmacology (Berl) 1995; 119:334–341Crossref, MedlineGoogle Scholar

116. Ahmed SH, Walker JR, Koob GF: Persistent increase in the motivation to take heroin in rats with a history of drug escalation. Neuropsychopharmacology 2000; 22:413–421Crossref, MedlineGoogle Scholar

117. Buczek Y, Le AD, Wang A, Stewart J, Shaham Y: Stress reinstates nicotine seeking but not sucrose solution seeking in rats. Psychopharmacology (Berl) 1999; 144:183–188Crossref, MedlineGoogle Scholar

118. Le AD, Harding S, Juzytsch W, Watchus J, Shalev U, Shaham Y: The role of corticotropin-releasing factor in stress-induced relapse to alcohol-seeking behavior in rats. Psychopharmacology (Berl) 2000; 150:317–324Crossref, MedlineGoogle Scholar

119. Martin-Fardon R, Ciccocioppo R, Massi M, Weiss F: Nociceptin prevents stress-induced ethanol- but not cocaine-seeking behavior in rats. Neuroreport 2000; 11:1939–1943Crossref, MedlineGoogle Scholar

120. DeVries AC, Taymans SE, Sundstrom JM, Pert A: Conditioned release of corticosterone by contextual stimuli associated with cocaine is mediated by corticotropin-releasing factor. Brain Res 1998; 786:39–46Crossref, MedlineGoogle Scholar

121. Highfield D, Yap J, Grimm JW, Shalev U, Shaham Y: Repeated lofexidine treatment attenuates stress-induced, but not drug cues: reinstatement of a heroin-cocaine mixture (speedball) seeking in rats. Neuropsychopharmacology 2001; 25:320–331Crossref, MedlineGoogle Scholar

122. Shaham Y, Erb S, Leung S, Buzbek Y, Stewart J: CP-1154, 526, a selective, non-peptide antagonist of the corticotropin releasing factor type 1 receptor attenuates stress-induced relapse to drug-seeking in cocaine and heroin-trained rats. Psychopharmacology (Berl) 1998; 137:184–190Crossref, MedlineGoogle Scholar

123. Vanderschuren L, Everitt B: Drug seeking becomes compulsive after prolonged cocaine self-administration. Science 2004; 305:1017–1019Crossref, MedlineGoogle Scholar

124. Lyvers M: “Loss of control” in alcoholism and drug addiction: a neuroscientific interpretation. Exp Clin Psychopharmacol 2002; 8:225–249CrossrefGoogle Scholar