<<

Yonsei Medical Journal Vol. 47, No. 2, pp. 167 - 178, 2006

Pharmacotherapy for Dependence: Anticraving for Relapse Prevention

Young-Chul Jung and Kee Namkoong

Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea.

Alcohol dependence is a chronic disorder that results from as traffic accidents in which the driver is intoxi- a variety of genetic, psychosocial, and environmental factors. cated, and to set boundaries for injudicious Relapse prevention for alcohol dependence has traditionally alcohol use in Korea. These socio-cultural changes involved psychosocial and psychotherapeutic interventions. Pharmacotherapy, however, in conjunction with behavioral have probably been successful in lowering the therapy, is generating interest as another modality to prevent prevalence of alcohol abuse but the prevalence of relapse and enhance abstinence. Naltrexone and acamprosate alcohol dependence seems to have been less af- are at the forefront of the currently available pharmacologi- fected.2 In a recent Korean epidemiological study,3 cal options. Naltrexone is an opioid and it was established that 10.20% of the adult popu- is thought to reduce the rewarding effect of alcohol. lation has a lifetime prevalence of alcohol depen- Acamprosate normalizes the dysregulation of N-methyl-D- aspartate (NMDA)-mediated glutamatergic excitation that dence (15.97% of men and 4.64% of women) occurs in alcohol withdrawal and early abstinence.These which makes alcohol dependence the second most different mechanisms of action and different target neuro- common psychiatric disorder in Korea. transmitter systems may endow the two drugs with efficacy Treating alcohol dependence usually consists of for different aspects of alcohol use behavior. Since not all two phases: detoxification and rehabilitation. The patients seem to benefit from naltrexone and acamprosate, initial detoxification stage deals with acute with- there are ongoing efforts to improve the treatment outcomes drawal symptoms. The later rehabilitation stage by examining the advantages of combined pharmacotherapy and exploring the variables that might predict the response attempts to prevent relapse and develops a life- of the medications. In addition, novel medications are being style compatible with long-term abstinence. investigated to assess their efficacy in preventing relapse and Whereas detoxification is widely accepted as a increasing abstinence. pharmacotherapeutic domain, rehabilitation, in Key Words: Alcohol dependence, pharmacotherapy, naltrexone, clinical practice, has traditionally involved psy- acamprosate chosocial and psychotherapeutic interventions consisting of individual and group psycho- therapy, cognitive-behavioral treatments, and self- INTRODUCTION directed groups such as Alcoholics Anonymous. Although psychosocial treatments have shown Alcohol dependence is a chronic disorder that effectiveness in reducing alcohol consumption and results from a variety of genetic, psychosocial, and maintaining abstinence, 40 to 70% of patients still environment factors.1 Over the last 20 years, there relapse to drinking within a year following treat- 4 has been considerable progress in efforts to reduce ment. As a part of the efforts to improve the the enormous alcohol related costs to society, such treatment outcomes for alcohol dependence, phar- macotherapy is being investigated as another modality to enhance abstinence and prevent Received March 28, 2006 relapse, complementing psychosocial interven- Reprint address: requests to Dr. Kee Namkoong, Department of Psychiatry, Yonsei University College of Medicine, 134 Shinchon- tions. dong, Seodaemun-gu, Seoul 120-752, Korea. Tel: 82-2-2228-1620, The rationale of using pharmacotherapy for Fax: 82-2-313-0891, E-mail: [email protected] alcohol dependence is based on several prem-

Yonsei Med J Vol. 47, No. 2, 2006 Young-Chul Jung and Kee Namkoong ises.5,6 First, advances in neurobiology have novel pharmacotherapies currently under inves- identified the neurobehavioral effects of alcohol tigation. and their associated neurotransmitter systems which are related to the development of depen- dence and, at the same time, are potential targets THE NEUROBIOLOGY OF ALCOHOL DEPEN- for pharmacological approaches. Second, recent DENCE genetic studies have confirmed that alcohol dependence is a heterogeneous condition. While For many years, it has been suggested that some gene variations predispose people to alcohol alcohol exerts its neurobiological effects mainly by dependence, others confer protection. Third, ani- increasing membrane fluidity, altering the func- mal models of alcohol dependence relapse have tion of macro-molecules in the cell membrane. demonstrated that pharmacologic agents can New evidence, however, indicates that alcohol reduce alcohol consumption and have proven to binds to hydrophobic pockets of proteins, modu- be fairly predictive of similar responses in human lating their function by changing their 3-dimen- patients. Fourth, medications have improved the sional structure. Proteins that are particularly treatment of other addictive disorders such as sensitive to this effect include ion-channels, neuro- for nicotine dependence and metha- transmitter receptors, and enzymes involved in done for heroin dependence, encouraging phar- signal transduction.9 Neurotransmitters with not- macotherapies for the treatment of alcohol depen- able sensitivity to this effect include , dence. , gamma-aminobutyric-acid (GABA), glu- To date, three medications - disulfiram, naltrex- tamic acid, adenosine, neuropeptide Y, norep- one, and acamprosate - have been approved by inephrine, cannabinoid receptors, and opioid the U.S. Food and Drug Administration (FDA) for peptides.10 These neurotransmitter systems are treatment of alcohol dependence (Table 1). Only involved in the different components of alcohol about 20% of eligible patients receive them, dependence and are therefore targets for however.7 Unfortunately, medications are still dis- pharmacotherapeutic interventions. paraged as a “crutch” and some still stick to the old adage, “You can't treat a drug problem with The brain reward system: dopamine and endo- a drug.”8 genous opioid This article discusses (1) the neurobiological basis of alcohol dependence; (2) the efficacy and Considerable evidence has emerged suggesting safety of disulfiram, naltrexone and acamprosate, that the dopamine system plays a central role in the approved pharmacotherapies for alcohol de- the biology of alcoholism. Mesolimbic dopamine pendence; (3) the ongoing issues for improving A10 neurons are activated by alcohol, resulting in the effectiveness of pharmacotherapy, and (4) the a release of the neurotransmitter in the nucleus

Table 1. Medications for Relapse Prevention of Alcohol Dependence

US-FDA Grade of strength Clinical recommendations approved? of evidence Disulfiram Yes B (Fair) Not recommended for routine use in primary care

Naltrexone Yes FDA-approved options for treatment of alcohol dependence in A (Good) Acamprosate Yes conjunction with behavioral therapy SSRI No Recommended for patients with comorbid depressive disorders

Ondansetrone No I (Insufficient) Recommended to reduce drinking frequency and increase Topiramate No abstinence.

SSRI, Serotonin-Specific Reuptake Inhibitor.

Yonsei Med J Vol. 47, No. 2, 2006 Pharmacotherapy of Alcohol Dependence accumbens and mediating positive reinforcement stable abstinence after acute detoxification is so and reward.11 It is postulated that repeated al- difficult to achieve. Antiglutamamatergic agents, cohol use sensitizes the system, so that behavioral such as NMDA antagonists and anticonvulsant stimuli associated with alcohol also cause the agents, have been proposed to reduce the moti- release of dopamine and facilitate additional vation for drinking by suppressing symptoms of alcohol use.12 This sensitization may account for alcohol withdrawal. Recent data suggest that the craving and preoccupation with alcohol that NMDA antagonists may have other beneficial are the hallmarks of alcohol dependence. effects in alcohol dependent patients, such as sub- The endogenous opioid system seems to play a stituting for deficits in negative feedback signals modulatory role on the dopaminergic system, or reducing the development of tolerance/sensi- whereby activation of opiate receptors stimulates tization to alcohol.15,16 the release of dopamine in the brain. Alcohol consumption increases the release of endorphins (which are endogenous opioid peptides) in the CURRENTLY APPROVED AGENTS FOR brain, thus indirectly activating the dopaminergic ALCOHOL DEPENDENCE reinforcement/reward system.13 It has been postu- lated that individual differences in the sensitivity Aversive agents: disulfiram of endogenous opioid systems may underlie in- dividual differences in the intensity of alcohol The first agent to be approved for treatment of craving and the risk of becoming alcohol depen- alcohol dependence was disulfiram. This sub- dent. stance was serendipitously discovered to be an agent causing alcohol aversion in Ohio rubber Subclinical withdrawal symptoms: glutamate workers in 1939. The major metabolic pathway for and GABA alcohol metabolism is a two-step enzymatic pro- cess (ethanol acetaldehyde acetic acid). Disul- The facilitation of inhibitory GABAergic and the firam is an irreversible inhibitor that blocks the inhibition of excitatory glutamatergic neurotrans- second stage of alcohol metabolism, causing an mission are important targets for the acute effects accumulation of toxic intermediate acetaldehyde, of alcohol. Potentiation of GABAergic inhibition is which results in hypotension, flushing, nausea, widely accepted as the underlying cause of the and vomiting. The objective of disulfiram treat- acute sedative effects of alcohol. Long-term adap- ment is thus to create an aversion to alcohol, tive changes to the sedative effects of alcohol in rather than to modulate its neurochemical effects. these two neurotransmitter systems are thought to Although many studies have been performed with underlie the development of alcohol dependence. disufiram, controlled clinical trials demonstrated After chronic exposure to alcohol, there is a com- inconsistent findings for alcohol drinking out- pensatory up-regulation of the glutamatergic comes between disulfiram and placebo, and have system (and down-regulation of the GABA failed to clearly establish the therapeutic benefit of system) in an attempt to balance alcohol's inhibi- this treatment in enhancing abstinence.17 How- tory action. The result is an increased tolerance for ever, it is difficult to conclude the efficacy of the alcohol.14 When alcohol is abruptly withdrawn, treatment through classical double-blind, placebo- however, a state of hyper-excitability emerges. controlled trials, since it is the psychological deter- This is perceived by the patient as a disagreeable rent effect of the drug rather than its biological state of arousal, anxiety and sleeplessness and is effect that is useful.18 Fuller et al.19 observed no the core of the negative affective state which the significant differences in abstinence rates or in the alcoholic patient will drink to relieve. These time to first relapse among groups taking placebo, plastic changes in the brain, brought about by 1 mg/day disulfiram (an inactive dose) or 250 change in protein synthesis, are only slowly rever- mg/day disulfiram (the standard dose). The pa- sible. This may explain the persistence of negative tients receiving 250 mg disulfiram, however, had craving during alcohol withdrawal and why fewer drinking days once they relapsed than did

Yonsei Med J Vol. 47, No. 2, 2006 Young-Chul Jung and Kee Namkoong the other two groups. drugs do not have pharmacological or phar- The disulfiram dosage is usually 250 mg per macokinetic interaction with alcohol and so the day with a maximum of 500 mg per day. While serious side effects associated with disulfiram are normal results of alcohol consumption after taking not troublesome when using naltrexone or disulfiram are palpitations, flushing, nausea, acamprosate. vomiting and headaches, more severe reactions The important difference between naltrexone could include myocardial infarction, congestive and acamprosate are mainly attributed to their heart failure, respiratory depression, and death. mechanisms of action. Naltrexone is an anta- The use of disulfiram appears to be most useful gonist at the opioid receptors, which are known in adherent patients, in special high-risk patients, to mediate the rewarding effects of alcohol, and and when administration is supervised. is thus thought to reduce the desire or craving for rewarding. Although less well defined, acam- Anticraving agents: naltrexone and acamprosate prosate normalizes the dysregulation of NMDA- mediated glutamatergic excitation that occurs in Naltrexone and acamprosate are at the fore- alcohol withdrawal and early abstinence. This front of currently available pharmacological effect probably attenuates the desire or craving options and they share many important features20 for reduce of tension. These different mecha- (Table 2). Most of the concerns in using nisms of actions may endow the two drugs with medications for relapse prevention are probably efficacy for different components of drinking. related to the possible intrinsic dependence potential. However, there is no evidence that Opioid antagonist: naltrexone naltrexone or acamprosate have developed either tolerance or withdrawal symptoms, including The USA FDA's approval of naltrexone for the rebound drinking when treatment is ceased, and treatment of alcohol dependence was based nor do they have any overt psychoactive effects mainly on two small, double-blind, placebo-con- on the central nervous system. In addition, both trolled trials demonstrating a reduced rate of

Table 2. Comparison of the Characteristics of Naltrexone and Acamprosate

Naltrexone Acamprosate Efficacy parameters Increased abstinence Maybe Yes Decreased heavy drinking Yes Maybe Longer-term efficacy No Yes Sustained efficacy post-treatment No Yes Onset of action Rapid Slow Compliance Variable Good Contingent on psychosocial intervention Variable Independent Safety parameters Interaction with alcohol No No Intrinsic dependence potential No No Overall safety profile Good Good Hepatic impact Yes No Clinically relevant drug interactions Yes No

Yonsei Med J Vol. 47, No. 2, 2006 Pharmacotherapy of Alcohol Dependence relapse to heavy drinking, reduced craving, and nausea (10%), headache (7%), anxiety (2%) and less frequent drinking in naltrexone-treated sedation (2%).36 Naltrexone has been shown to patients.22,23 During the following years, several have dose-related hepatotoxicity, although gen- more trials have followed and three meta-analyses erally this occurs at doses of 300 mg per day, have concluded that naltrexone is efficacious in higher than those recommended for treatment of the treatment of alcohol dependence.24-26 The most alcohol dependence. The drug is contraindicated consistent finding obtained with naltrexone is an in patients with hepatitis or liver failure, and a increased time to first relapse (typically defined as monthly check of hepatic transaminase levels is more than 5 drinks/day in males, 4 drinks/day in recommended, for the first three months and females). The decrease in relapse rate, however, every three months thereafter. has not been observed in all studies. Such dif- ferences in results are seen even in the largest NMDA/ GABA : Acamprosate trial.27 Several factors may explain the discrep- ancies in results of the different clinical trials of Acamprosate was investigated in nearly 20 con- naltrexone. The animal models of relapse indicate trolled, published trials with about 4000 patients that naltrexone blocks cue-induced relapse but is and these studies have produced consistent results less effective against stress-induced relapse.28 The showing that acamprosate treatment is superior to effect of naltrexone on relapse may, to some placebos in maintaining abstinence.37 In all but extent, be dependent on associated psycho- three clinically controlled published studies, the therapy, since naltrexone was found to be more proportion of acamprosate-treated patients ab- effective in patients receiving training in coping staining at the end of the study was twice as high skills than in those receiving supportive therapy as patients receiving placebos. In addition, two alone.29 Compliance may be a limiting factor in studies38,39 evaluated long-term abstinence for 1- naltrexone treatment.30-32 Monti et al.32 demon- ear after the end of the treatment period, and both strated a significant treatment outcome only when showed that treatment effects were maintained. A non-compliant subjects were excluded from the systematic metaanalysis37 in which clinical data analysis. A large multi-site, double blind, placebo- from 17 trials were reanalyzed concluded that the controlled trial with an injectable, sustained-re- treatment effect could increase with time. lease formation, (a strategy to improve compli- Since most of the trials were undertaken in ance) demonstrated that relapse to heavy drinking Europe, it was considered important to evaluate decreased in patients receiving depot preparation the efficacy and safety of acamprosate in different compared to placebo.33 ethnic groups, given the emerging role of genetic A conceptual framework for integrating the and cultural issues in drug treatment. In a study clinical data on naltrexone has been proposed by by Namkoong et al.,40 however, acamprosate did Sinclair,34 who suggested that naltrexone is useful not show any treatment benefits when compared for preventing relapse rather than at maintaining to the placebo. This negative finding might be ex- absolute abstinence. Thus, the contingency of plained by the sample characteristics (i.e., a more drinking alcohol and taking naltrexone is impor- severe form of alcohol dependence, a lower level tant in bringing to light treatment effects. In a of social support, a short interval between the last recent systematic meta-analysis of 24 placebo-con- drink and the first ), the dosage issues trolled trials, including a total of 2861 patients, of acamprosate, the short study period (8 weeks), short-term naltrexone therapy significantly de- and the variable concomitant psychosocial treat- creased relapse rate (relative risk 0.64), but did not ment. enhance absolute abstinence (relative risk 0.91).35 Acamprosate is available in 333-mg enteric For the first 90 days of abstinence, when the coated tablets. Dosing is determined by weight ( risk of relapse is greatest, the recommended 60 kg: 1998 mg, < 60 kg: 1332 mg). It is not meta- dosage of naltrexone is a single dose of 50 mg per bolized but is eliminated by renal excretion, and day but doses of 25 mg to 100 mg daily are some- should therefore be given cautiously with renal times used. The most common side effects are impairment. Acamprosate is well tolerated with

Yonsei Med J Vol. 47, No. 2, 2006 Young-Chul Jung and Kee Namkoong limited side effects, most commonly transient There are several possible explanations for the diarrhea (10%) and headache (20%). Like naltrex- superior efficacy of the combination treatment.43 one and disulfiram, acamprosate is FDA pregn- First, there may be subgroups that respond selec- ancy category C, i.e., there have been adverse tively to naltrexone or acamprosate and thus the effects on the fetus in animal studies but no added benefit of combination therapy would be human trials have been performed. merely explained by the recruitment of additional responder patients. A hypothesis of such patient subgroups may be that 'reward' craving drinkers IMPROVING THE EFFECTIVENESS OF would respond better to naltrexone and 'relief' PHARMACOTHERPY craving drinkers would respond better to acam- prosate (Fig. 1). Attempts to find variables that Combination Pharmacotherapy: Naltrexone plus might predict the response of medications will be Acamprosate discussed later. Second, the combination produces a synergic anticraving effect as the two drugs Combining naltrexone and acamprosate in the interfere with distinct biological aspects of the treatment of alcohol dependence is an attractive craving process. Third, pharmacokinetic interac- concept for several reasons. Since naltrexone and tion might underlie the observed treatment acamprosate have different mechanisms of action benefits, whereby bioavailability might be en- and different target neurotransmitter systems, pre- hanced by co-administration of the other drug. sumably, they affect different aspects of alcohol The ongoing COMBINE study44 plans to recruit use behavior. (Naltrexone decreases alcohol con- 1,375 subjects at 11 sites to examine treatment sumption and acamprosate stabilizes abstinence.) interactions between naltrexone, acamprosate and Pharmacokinetic and behavioral assessments of two behavioral interventions (medical manage- combining naltrexone and acamprosate have ment and combined behavioral interventions). The found the combination to be safe. data from COMBINE will answer important ques- Kiefer et al.41 performed a randomized, double- tions regarding the effectiveness of naltrexone and blind, placebo-controlled, clinical trial of 160 al- acamprosate both alone and in combination, as cohol-dependent patients and assessed the effi- well as that of psychosocial treatment. cacy of naltrexone and acamprosate, as mono- therapy and in combination. It was demonstrated that the proportion of patients remaining abso- lutely abstinent at the end of the 12-week treat- ment period was around twice as high in the combination therapy group than in the mono- therapy group (placebo 25%, naltrexone alone 65%, acamprosate alone 50%, combination therapy 73%). Even though further relapse occurred during the follow-up period, the relative treat- ment benefits between the three treatment groups and the placebo group was maintained at the end of the 3-month open label phase. There was, however, no significant difference between the three treatment groups (placebo 20%, naltrexone alone 47%, acamprosate alone 46%, combination therapy 66%).42 Although combination therapy was generally well tolerated, the incidence of diar- rhea (13.8%) and nausea (5.6%) was significantly Fig. 1. A representation of the neuroadaptive model of greater than in the monotherapy groups, perhaps craving and possible mechanisms of naltrexone and due to a pharmacokinetic interaction. acamprosate.

Yonsei Med J Vol. 47, No. 2, 2006 Pharmacotherapy of Alcohol Dependence

The matching hypothesis: the three pathway jective reports of stimulation following drinking. model of craving Lesch et al.51 conducted a long-term, prospec- tive study to assess the efficacy of acamprosate Since not all patients seem to benefit from the based on the four subtypes of alcohol dependent- different anticraving drugs, the use of matching patients. Consistent with their matching hypo- procedures might be important in improving the thesis, based on the three pathway model of effectiveness of treatment with anticraving com- Verheul et al.,48 acamprosate differentially re- pounds.45 The type of craving could possibly be duced alcohol intake in those patients who use an important predictor or matching variable with alcohol to counteract withdrawal symptoms (Type anticraving compounds. In general, craving I), and in patients who use alcohol as a conflict- mainly refers to the strong desire or urge to ex- solving and anxiety reducing agent (Type II), but perience the effect of a previously experienced not in patients who ingest alcohol to self-medicate psychoactive substance.46 Despite the simplicity of affective disorders (Type III) or patients with a this definition, a wide variety of craving concepts history of cerebral impairment that precedes the are used in the research and clinical field. Verheul development of alcohol dependence (Type IV). A et al.47 have proposed a novel three pathway recent study including the pooled data of 1,485 model of craving in alcoholics. This model sug- alcohol-dependent patients from seven rando- gests that craving is likely to result from distinct mized, controlled trials comparing acamprosate psychobiological mechanisms and that the efficacy and a placebo, directly tested the matching hypo- of different anti-craving compounds is associated thesis that acamprosate would be more effective with individual differences in craving: (1) The in patients with high physical dependence at reward pathway suggests that craving or desire baseline, negative family history of alcoholism, for the rewarding, stimulating and/or enhancing late age of onset, serious anxiety symptomatology effects of alcohol might result from either dopami- at baseline, severe craving at baseline, and female nergic/opioidergic dysregulation or a personality gender.52 In contrast to the expectations, the authors style characterized by reward seeking and/or found that none of these theoretically relevant hedonism. (2) The relief pathway suggests that clinical matching variables predicted the treatment craving or desire for the reduction of tension, effectiveness of acamprosate. Similarly, another arousal or withdrawal might result from either pooled analysis of 11 trials by Sass et al.53 did not GABAergic/glutamatergic dysregulation or a per- find any demographical, psychopathological or sonality style characterized by stress reactivity, biological predictors for acamprosate efficacy. anxiety sensitivity, and/or hyperarousability. (3) It should be noted that the three-pathway The obsessive pathway can be defined as a lack model by Verheul et al.48 and related matching of control over intrusive thoughts about drinking hypotheses might not cover all the possible mech- resulting in impaired functioning. This pathway anisms of action of anti-craving medications. For of craving might result from serotonin deficiency example, a recent study showed that naltrexone or a personality style characterized by low con- not only includes an opioid-receptor blockade, but straint or disinhibition. that it indirectly increased hypothalamic pituitary Some studies found that patients with high adrenocortical (HPA) activity, resulting in higher levels of alcohol craving are most likely to benefit adrenocorticotropic hormone (ACTH) and cortisol from naltrexone treatment.48,49 No distinction, levels, which may in turn be partially responsible however, was made between reward, relief and for the effect of naltrexone on drinking and obsessive craving. Palfai et al.50 conducted a study craving.54 among hazardous drinkers and suggested that naltrexone may be particularly effective in re- Pharmacogenomics: μ-Opioid Receptor Polymor- ducing the cue-elicited positive reinforcement of phism alcohol for those with high positive alcohol out- come expectancies. Positive outcome expectancies Investigating the role of genetics in predicting also moderated the effects of naltrexone on sub- treatment outcomes is one promising area of

Yonsei Med J Vol. 47, No. 2, 2006 Young-Chul Jung and Kee Namkoong research since a genetic basis of for alcoholism days. The finding of a genotype that predicts (heritability rate, 50-60%) is well established.55 success with naltrexone, if replicated, will provide Monterossso et al.49 explored the predictors of a pharmacogenetic tool to enhance the matching naltrexone response in a double-blind, placebo- of patients to treatment and encourage the search controlled trial and demonstrated significant in- for additional functional polymorphisms. teractions between treatment condition and family history of alcohol problems. Some studies have investigated individual differences with respect to THE DEVELOPMENT OF NOVEL DRUGS sensitivity in the hypothalamic pituitary adrenal FOR ALCOHOL DEPENDENCE axis to naltrexone administration. King et al.56 examined the neuroendocrine and mood re- agents and (5-HT3 an- sponses to oral naltrexone as a function of the tagonist) biological family history of alcoholism. The results demonstrated the potential biological bases of During the last two decades, a number of drugs altered opioidergic sensitivity in those persons acting on serotonergic neurotransmission have with an assumed greater inherited vulnerability been studied in alcohol dependence, since sero- for future alcoholism. Meanwhile, studies of tonin is widely implicated in a variety of consum- family history as a predictor of treatment response matory behaviors and impulsivity. These agents have led to speculation that naltrexone may func- are either selective serotonin reuptake inhibitors tion differently in genetically predisposed indi- or receptor agonist/antagonists. Most of this viduals. work, however, has used small samples with rela- Naltrexone has a high affinity for the μ-opioid tively short treatment periods. Selective serotonin receptor, which is hypothesized to be the principal reuptake inhibitors, despite their effectiveness in site of action of the drug. It has been hypothesized animals, have shown inconsistent or disappoin- that sequence variation in the gene encoding the ting results in humans60-65 and so the usefulness μ-receptor may result in a receptor with altered is still controversial.17 Meanwhile, no evidence of expression, structure, or function, and as a conse- clinical efficacy in alcohol-dependent has been 66,67 quence, may increase or decrease an individual's obtained with ritanserin (5-HT2 anatagonist). In susceptibility to substance dependence.57 Oslin et addition, a meta-analysis of studies performed 58 al. examined the association between two with bupropion (5-HT1 partial agonist) concluded specific polymorphisms of the gene encoding the that any efficacy of bupropion was secondary to μ-opioid receptor and treatment outcomes, mea- an anxiolytic effect, rather than on drinking sured over 12 weeks, in alcohol dependent perse.68 patients who were prescribed naltrexone or a Of the numerous serotonergic drugs which placebo. Patients with one or two copies of the have been suggested as pharmacotherapies for Asp40 allele treated with naltrexone had signifi- alcohol dependence treatment, ondansetron, a cantly lower rates of relapse (26.1% versus 47.9%; 5-HT3 antagonist that is FDA-approved as an p = 0.0044) and took a longer time to return to antiemetic, appears to be the most promising.69 heavy drinking than those homozygous for the The 5-HT3 receptor is involved in the expression Asn40 allele. There were no differences in relapse of alcohol's rewarding effects. Behavioral phar- rates or abstinence rates between the two geno- macological studies show that many of the reward type groups among those assigned to placebo. effects of alcohol are mediated by interactions 59 Meanwhile, Kim et al. reported that the allele between DA and the 5-HT3 receptor in the mid- 70,71 frequency of the Asp40 allele was 39.7% in the brain and cortex. 5-HT3 receptors are densely Korean alcohol dependent group, which is con- distributed in the terminals of mesocorticolimbic sistent with previous studies demonstrating a DA-containing neurons where they regulate DA higher Asp40 allele frequency in the Asian popu- release in these brain regions. Following a pre- lation. Within the alcohol dependent group, the vious clinical trial,72 Johnson et al.73 evaluated Asp40 allele was associated with more drinking ondansetron as a treatment for alcohol depen-

Yonsei Med J Vol. 47, No. 2, 2006 Pharmacotherapy of Alcohol Dependence dence in a 12-week, double-blind, placebo-con- was equally effective in both early-onset and trolled trial of 321 patients. The early-onset, al- late-onset alcohol dependence.78 The study used cohol dependent group treated with ondansetron an escalating dose of 25 to 300 mg per day. Hyper- (particularly 4 μg/kg b.i.d.) reported fewer drinks sensitivity to the drug was the only known contra- per day and fewer drinks per drinking days, indication. No serious adverse events occurred. while the late-onset group treated with ondan- Paresthesia was the most common adverse effect, setron did not differ from those treated with which includes dizziness, somnolence, diplopia placebo. It is interesting that while serotonine and nausea. reuptake inhibitors have little effect on drinking among early-onset alcoholics, ondansetron, with functionally opposite effects in the serotonergic CONCLUSION system, is effective for the early-onset subtype. Sufficient evidence exists that early-onset alcoholics Important advances have been made in the are more prone to serotonergic dysfunction than development of pharmacotherapy in alcohol late-onset alcoholics.74,75 dependence and it is experiencing a major shift in direction. There is clear evidence of the effi- Mood stabilizers/ Anticonvulsants and Topira- cacy and safety of natrexone and acamprosate, mate (GABA agonist) and still more novel medications are under investigation. In addition, important questions Mood stabilizers and anticonvulsants decrease remain regarding the optimal dose and duration alcohol consumption in experimental animals. of treatment, the role of combinations of medi- Clinical trials, however, have not provided clear cations, and the treatment subtypes of alcohol evidence of the efficacy of treatment for alcohol dependent patients. dependence. The controlled trials of lithium did not demonstrate efficacy in either non-depressed REFERENCES or depressed alcohol-dependent patients and Garbutt et al.17 concluded that lithium lacks effi- 1. Morse RM, Flavin DK. The definition of alcoholism. cacy in the treatment of primary alcohol depen- The Joint Committee of the National Council on Alcoholism and Drug Dependency and the Amercian dence. More promising are results with non- Society of Addiction Medicine to Study the Definition benzodiazepine anticonvulsants such as carba- and Criteria for the Diagnosis of Alcoholism. JAMA mazepinem, valproate, gabapentin, vigabatrin and 1992;268:1012-4. topiramate.76 2. Jung YC, Namkoong K. Two alcoholisms: Abuse and Topiramate, although only approved by the Dependence: Nosology issues from the epidemiological studies of alcoholism in Korea. Psychiatr Invest 2005;2: FDA for seizure disorders, was evaluated in the 40-52. treatment of alcohol dependence because of its 3. Cho MJ, Hahm BJ, Kim JK, Park KK, Chung EK, Suh effects on GABAergic and glutamatergic systems. TW, et al. Korean Epidemiologic Catchment Area It has been shown to augment GABA function (KECA) Study for Psychiatric Disorders: Prevalence of and inhibit specific glutamatergic pathways at the Specific Psychiatric Disorders. J Korean Neuropsychiatr AMPA/kinate receptors. This may decrease the Assoc 2004;43:470-80. 4. Finney JW, Hahn AC, Moos RH. The effectiveness of cortical expression of alcohol reward by de- inpatient and outpatient treatment for alcohol abuse: 77 creasing the midbrain dopamine function. In a the need to focus on mediators and moderators of 12-week double-blind, placebo-controlled trial of setting effects. Addiction 1996;91:1773-96. 150 alcohol dependent patients, topiramate was 5. Swift RM. Drug therapy in alcohol dependence. N Engl more effective than placebo in initiating absti- J Med 1999;340:1482-90. 6. Kenna GA, McGeary JE, Swift RM. Pharmacotherapy, nence and in reducing self-reported drinks per pharmacogenomics, and the future of alcohol depen- day, drinks per drinking days, and heavy dence treatment, Part 2. Am J Health Syst Pharm 2004; drinking days. Compared with a placebo, it also 61:2272-388. significantly reduced craving as assessed with 7. Williams SH. Medications for treating alcohol depen- Obsessive Compulsive Drinking Scale. Topiramate dence. Am Fam Physician 2005;72:1775-80.

Yonsei Med J Vol. 47, No. 2, 2006 Young-Chul Jung and Kee Namkoong

8. O'Brien CP. Anticraving medications for relapse pre- maintenance treatment of alcohol dependence: a vention: a possible new class of psychoactive medica- meta-analysis of randomized controlled trials. Alcohol tions. Am J Psychiatry 2005;162:1423-31. Alcohol 2001;36:544-52. 9. Gordis E. The neurobiology of alcohol abuse and alco- 26. Srisurapanont M, Jarusuraisin N. Opioid antagonist for holism: building knowledge, creating hope. Drug alcohol dependence. Cochrane Database Syst Rev 2002; Alcohol Dependence 1998;51:9-11. (2):CD001867. 10. Koob GF, Roberts AJ, Schulteis G, Parsons LH, Heyser 27. Krystal JH, Cramer JA, Krol WF, Kirk GF, Rosenheck CJ, Hyytia P, et al. Neurocircuitry targets in ethanol RA; Veterans Affairs Naltrexone Cooperative Study 425 reward and dependence. Alcohol Clin Exp Res 1998;22: Group. Naltrexone in the treatment of alcohol depen- 3-9. dence. N Eng J Med 2001;345:1734-9. 11. Gessa GL, Muntoni F, Collu M, Vargiu L, Mereu G. 28. Liu X, Weiss F. Addictive effect of stress and grug cues Low doses of ethanol activate dopaminergic neurons in on reinstatement of ethanol seeking: exacerbation by the ventral tegmental area. Brain Res 1985;348:201-03. history of dependence and role of concurrent activation 12. Robinson TE, Berridge KC. The neural basis of drug of corticotropin-releasing factor and opioid mechanism. craving: an incentive-sensitization theory of addiction. J Neurosci 2002;22:7856-61. Brain Res Rev 1993;18:247-91. 29. Balladin J, Berglund M, Borg S, Mansson M, Bendtsen 13. Benjamin D, Grant E, Pohorecky LA. Naltrexone re- P, Franck J, et al. A 6-month controlled naltrexone verses ethanol-induced dopamine release in the study: combined effect with cognitive behavioral nucleus accumbens in awake, freely moving rats. Brain therapy in outpatient treatment of alcohol dependence. Res 1993;621:137-40. Alcohol Clin Exp Res 2003;27:1142-9. 14. Grobin AC, Matthews DB, Devaud LL, Morrow AL. 30. Anton RF, Moak DH, Waid LR, Latham PK, Malcolm The role of GABAA receptors in the acute and chronic RJ, Dias JK. Naltrexone and cognitive behavioral effects of ethanol. Psychopharmacology 1998;139:2-19. therapy of outpatients alcoholics: results of a placebo- 15. Quertemont E, Brabant C, De Witte P. Acamprosate controlled trial. Am J Psychiatry 1999;156:1758-64. reduces context-dependent ethanol effects. Psychophar- 31. Chick J, Anton R, Checinski K, Croop R, Drummond macology 2002;164:10-8. DC, Farmer R, et al. A multicentre, randomized, 16. Kiefer F, Jahn H, Tarnaske T, Helwig H, Briken P, double-blind, placebo-controlled trial of naltrexone in Holzbach R, et al. Comparing and combining naltrex- the treatment of alcohol dependence or abuse. Alcohol one and acamprosate in relapse prevention of alcohol- Alcohol 2000;35:587-93. ism: a double-blind, placebo-controlled study. Arch 32. Monti PM, Rohsenow DJ, Swift RM, Gulliver SB, Colby Gen Psychiatry 2003;60:92-9. SM, Mueller TI, et al. Naltrexone and cue exposure 17. Garbutt JC, West Sl, Carey TS, Lohr KN, Crews FT. with coping and communication skills training for Pharmacological treatment of alcohol dependence: a alcoholics: treatment process and 1-year outcomes. review of the evidence. JAMA 1999;281:1318-25. Alcohol Clin Exp Res 2001;25:1634-47. 18. Kiefer F, Mann K. New achievements and pharmaco- 33. Garbutt JC, Kranzler HR, O'Malley SS, Gastfriend DR, therapeutic approaches in the treatment of alcohol Pettinati HM, Silverman BL, et al. Efficacy and tolera- dependence. Eur J Pharmacol 2005;526:163-71. bility of long-acting injectable naltrexone for alcohol 19. Fuller RK, Brachey l, Brightwell DR, Derman RM, dependence: a randomized controlled trial. JAMA Emrick CD, Iber FL, et al. Disulfiram treatment of alco- 2005;293:1617-25. holism. A Veterans Administration Cooperative Study. 34. Sinclair JD. Evidence about the use of naltrexone for JAMA 1996;256:1449-55. different ways of using it in the treatment of alcohol- 20. Mason BJ. Acamprosate and naltrexone treatment for ism. Alcohol Alcohol 2001;36:2-10. alcohol dependence: an evidence-based risk-benefits 35. Srisurapanont M, Jarusuraisin N. Naltrexone for the assessment. Eur Neuropyschopharmacol 2003;13:469-75. treatment of alcoholism: a meta-analysis of randomized 21. Savin S. Saivin S, Hulot T, Chabac S, Potgieter A, controlled trials. Int J Neuropsychopharmacol. 2005;8: Durbin P, Houin G.et al. Clinical pharmacokinetics of 267-80. acamprosate. Clin. Pharmacokinet. 1998;35:331-45. 36. Croop RS. The safety profile of naltrexone in the treat- 22. Volpicelli JR, Alterman AI, Hayashida M, O'Brien CP. ment of alcoholism. Results from a multicenter usage Naltrexone in the treatment of alcohol dependence. study. The Naltrexone Usage Study Group. Arch Gen Arch Gen Psychiatry 1992;49:876-80. Psychiatry 1997;54:1130-5. 23. O'Malley SS, Jaffe AJ, Chang G, Schottenfeld RS, Meyer 37. Mann K, Lehert P, Morgan MY. The efficacy of RE, Rounsaville B. Naltrexone and coping skills acamprosate in maintaining abstinence in alcohol de- therapy for alcohol dependence. A controlled study. pendent individuals: results of meta-analysis. Alcohol Arch Gen Psychiatry 1992;49:881-7. Clin Exp Res 2004;28:51-63 24. Kranzler HR, Van Kirk J. Efficacy of naltrexone and 38. Sass H, Soyka M, Mann K, Zieglgansberger W. Relapse acamprosate for alcoholism treatment: a meta-analysis. prevention by acamprosate. Results from a placebo Alcohol Clin Exp Res 2001;25:1335-41. controlled study in alcohol dependence. Arch Gen 25. Streeton C, Whelan G. Naltrexone, a relapse prevention Psychiatry 1996;53:673-80.

Yonsei Med J Vol. 47, No. 2, 2006 Pharmacotherapy of Alcohol Dependence

39. Whitworth AB, Fischer F, Lesch OM, Nimmerrichter A, self-administration in alcohol-dependent subjects and Oberbauer H, Platz T, et al. Comparison of acampro- activates the hypothalamic-pituitary-adrenocortical axis. sate and placebo in long-term treatment of alcohol Psychopharmacology(Berl) 2002;160:19-29. dependence. Lancet 1996;347:1438-42. 55. Heath AC. Genetic influences on alcoholism risk: a 40. Namkoong K, Lee BO, Lee PG, Choi MJ, Lee E. review on adoption and twin studies. Alcohol Health Acamprosate in Korean alcohol-dependent patients: a Res World 1995;19:166-71. multi-centre, randomized, double blind, placebo con- 56. King AC, Schluger J, Gunduz M, Borg L, Perret G, Ho trolled study. Alcohol Alcohol 2003;38:135-41. A, et al. Hypothalamic-pituitary-adrenocortical (HPA) 41. Keifer F, Jahn H, Tarnaske T, Helwig H, Briken P, axis response and biotranformation of oral naltrexone: Holzbach R, et al. Comparing and combining naltrex- preliminary examination of relationship to family his- one and acamprosate in relapse prevention of alcohol- tory of alcoholism. Neuropsychopharmacology 2002;26: ism: a double-blind, placebo-controlled study. Arch 778-88. Gen Psychiatry 2003;60:92-9. 57. Lichtermann D, Franke P, Maier W, Rao ML. Phar- 42. Kiefer F, Andersohn F, Otte C, Wolf K, Jahn H, macogenetics and addiction to opiates. Eur J Pharmacol Wiedemann K. Long-term effects of pharmacotherapy 2000;410:269-79. on relapse prevention in alcohol dependence. Acta 58. Oslin DW, Berrettini W, Kranzler HR, Pettinati H, Neuropsychiatr 2004;16:233-8. Gelernter J, Vopicelli JR, et al. A functional polymor- 43. Kiefer F, Wiedemann K. Combined therapy: what does phism of the mu-opioid receptor gene is associated acamprosate and naltrexone combination tell us? Alco- with naltrexone response in alcohol-dependent hol Alcohol 2004;39:542-7. patients. Neuropsychopharmacology 2003;28:1546-52. 44. COMBINE study group. Testing combined pharmaco- 59. Kim SG, Kim CM, Kang DH, Kim YJ, Byun WT, Kim therapies and behavioral interventions in alcohol SY, et al. Association of functional opioid receptor dependence (the COMBINE study): rationale and genotypes with alcohol dependence in Koreans. methods. Alcohol Clin Exp Res 2003;27:1107-22. Alcohol Clin Exp Res 2004;28:986-90. 45. Ooteman W, Verheul R, Naassila M, Daoust M, 60. Gorelick DA, Paredes A. Effects of on Schippers GM, Koeter MWJ, et al. Patient-treatment alcohol consumption in male alcoholics. Alcohol Clin matching with anti-craving medications in alcohol Exp Res 1992;16:261-5. dependent patients: A review on phenotypic, endo- 61. Kabel DI, Petty F. A placebo-controlled, double-blind phenotypic and genetic indicators. J Substance Use study of fluozetine in severe alcohol dependenc: ad- 2005;10:75-96. junctive pharmacotherapy during and after inpatient 46. Anton RF. What is craving? Models and implications treatment. Alcohol Clin Exp Res 1996;20:780-4. for treatment. Alcohol Res Health 1999;23:165-73. 62. Kranzler HR, Burleson JA, Korner P, Del Boca FK, 47. Verheul R, van den Brink W, Geerlings P. A three- Bohn MJ, Brown J, et al. Placebo-controlled trial of pathway psychobiological model of craving for alcohol. fluoxetine as an adjunct to relapse prevention in Alcohol Alcohol 1999;34:197-222. alcoholics. Am J Psychiatry 1995;152:391-7. 48. Volpicelli JR. Naltrexone in the alcohol dependence. 63. Janiri L, Gobbi G, Mannelli P, Pozzi G, Serretti A, Lancet 1995;346:456. Tempesta E. Effects of fluoxetine at antidereesant doses 49. Monterosso JR, Flannery BA, Pettinati HM, Oslin DW, on short-term outcome of detoxified alcoholics. Int Clin Rukstalis M, O'Brein CP, et al. Predicting treatment Psychopharmacol 1996;11:109-17. response to naltrexone: The influence of craving and 64. Pettinati HM, Volpocelli JR, Luck G, Kranzler HR, family history. Am J Addiction 2001;10:258-68. Rukstalis MR, Cnaan A. Double-blind clinical trial of 50. Palfai T, Davidson D, Swift R. Influence of naltrexone sertraline treatment for alcohol dependence. J Clin on cue-elicited craving among hazardous drinkers: the Psychopharmacol 2001;21:143-53. moderational role of positive outcome experiences. Exp 65. Roy A. Placebo-controlled study of setaline in de- Clin Psychopharmacol 1999;7:266-73. pressed recently abstinent alcoholics. Biol Psychiatry 51. Lesch OM, Walter H. Subtypes of alcoholism and their 1998;44:633-7. role in therapy. Alcohol Alcohol 1996;31:63-7. 66. Johnson BA, Jansinski DR, Galloway GP, Kranzler H, 52. Verheul R, Lehert P, Geerlings PJ, Koeter MW, Van den Weinreib R, Anton RF, et al. Ritanserin in the treatment Brink W. Predictors of acamprosate efficacy: results of alcohol dependence-a multi-center clinical trial. from a pooled analysis of seven European trials in- Psychopharmacology 1996;128:206-15. cluding 1485 alcohol-dependent patients. Psychophar- 67. Wiesbeck GA, Weijers JG, Chick J, Naranjo CA, macology 2005;178:167-73. Boening J. Ritanserin in relapse prevention in abstinent 53. Sass H, Soyka M, Mann K, Zieglgansberger W. Relapse alcoholics: results from a placebo-controlled, double- prevention by acamprosate. Results from a placebo- blind internaltional multi-center trial. Ritanserin in controlled study on alcohol dependence. Arch Gen Alcoholism Work Group. Alcohol Clin Exp Res 1999;23: Psychiatry 1996;53:673-80. 230-5. 54. O' Malley SS, Krishnan-Sarin S, Farren C, Sinha R, 68. Malec TS, Malec EA, Dongier M. Efficacy of bupropion Kreek MJ. Naltrexone decreases craving and alcohol in alcohol dependence: a review. Alcohol Clin Exp Res

Yonsei Med J Vol. 47, No. 2, 2006 Young-Chul Jung and Kee Namkoong

1996;20:853-8. Mefford I, Linnoila M. Early-onset alcoholics have 69. LeMarquand D, Pihl RO, BenKelfat C. Serotonin and lower cerebrospinal fluid 5-hydroxyindoleacetic acid alcohol intake, abuse, and dependence: clinical evi- levels that late-onset alcoholics. Arch Gen Psychiatry dence. Biol Psychiatry 1994;36:326-37. 1996;53:211-6. 70. Barnes NM, Sharp T. A review of central 5-HT 75. Swann AC, Johnson BA, Cloninger CR, Chen YR. Rela- receptors and their function. Neuropharmacology 1999; tionships of plasma availability to course of 38:1083-152. illness and clinical features of alcoholism: a preliminary 71. Johnson BA, Cowen PJ. Alcohol induced reinforcement: study. Psychopharmacology(Berl) 1999;143:380-4. dopamine and 5-HT3 receptor interactions in animals 76. Book SW, Myrick H. Novel anticonvulsants in the and humans. Drug Dev Res 1993;30:153-69. treatment of alcoholism. Expert Opin Investing Drugs 72. Sellers EM, Toneatto T, Romach MK, Somer GR, Sobell 2005;14:371-6. LC, Sobell MB, et al. Clinical efficacy of the 5-HT3 an- 77. Johnson BA, Ait-Daoud N, Akhtar FZ, Ma JZ. Oral tagonist ondansetron in alcohol abuse and dependence. topiramate reduces the consequences of drinking and Alcohol Clin Exp Res 1995;18:879-85. improves the quality of life of alcohol-dependent in- 73. Johnson BA, Roache JD, Javors MA, Diclemente CC, dividuals: a randomized controlled trial. Arch Gen Cloninger CR, Prihoda TJ, et al. Ondansetron for Psychiatry 2004;61:905-12. reduction of drinking among biologically predisposed 78. Johnson BA, Ait-Daoud N, Bowden CL, DiClemente alcoholic patients: a randomized controlled trial. JAMA CC, Roache JD, Lawson K, et al. Oral topiramate for 2000;284:963-71. treatment of alcohol dependence: a randomised con- 74. Fils-Aime ML, Eckardt MJ, George DT, Brown GL, trolled trial. Lancet 2003;361:1677-85.

Yonsei Med J Vol. 47, No. 2, 2006