<<

The new england journal of medicine

review article

mechanisms of disease Addiction

Jordi Camí, M.D., Ph.D., and Magí Farré, M.D., Ph.D.

rug addiction is a chronic, relapsing disorder in which From the Institut Municipal d’Investigació compulsive drug-seeking and drug-taking behavior persists despite serious Mèdica (J.C., M.F.), Universitat Pompeu d Fabra (J.C.), and Universitat Autònoma de negative consequences. Addictive substances induce pleasant states (euphoria Barcelona (M.F.) — all in Barcelona, in the initiation phase) or relieve distress. Continued use induces adaptive changes in Spain. Address reprint requests to Dr. the central nervous system that lead to tolerance, physical dependence, sensitization, Camí at Institut Municipal d’Investigació Mèdica, Doctor Aiguader 80, E-08003 Bar- craving, and relapse (Table 1). The addictive discussed here are , cannab- celona, Spain, or at [email protected]. inoids, ethanol, cocaine, amphetamines, and nicotine. The World Health Organization1 and the American Psychiatric Association2 use the N Engl J Med 2003;349:975-86. term “substance dependence” rather than “drug addiction.” “Drug addiction,” however, Copyright © 2003 Massachusetts Medical Society. emphasizes the behavioral connotation of the term and is less likely to be confused with physical dependence.3 We use both terms interchangeably in this review. The Amer- ican Psychiatric Association’s definition of substance dependence2 requires a patient to meet at least three of the seven criteria listed in Table 1. Tolerance and physical depend- ence reflect physiological adaptation to the effects of a drug, whereas the remaining cri- teria define uncontrollable drug consumption. However, tolerance and physical depend- ence are neither necessary nor sufficient for a diagnosis of substance dependence. Substance abuse2 or harmful use,1 a less severe disorder, may result in dependence. Theories of addiction have mainly been developed from neurobiologic evidence and data from studies of learning behavior and memory mechanisms. They overlap in some aspects and are not mutually exclusive. None of them alone can explain all aspects of ad- diction. It is not our purpose to present a detailed assessment of these theories, especial- ly because of the complexity of the problem. Generally, addictive drugs can act as posi- tive reinforcers (producing euphoria) or as negative reinforcers (alleviating symptoms of withdrawal or dysphoria). Environmental stimuli (cues) associated with drug use it- self can also induce a conditioned response (withdrawal or craving) in the absence of the drug.4,5 Koob and Le Moal6,7 have proposed that the organism tries to counteract the effects of a given drug through a vicious circle in which the hedonic set point (the point at which pleasure is achieved) continually changes in response to the administration of the sub- stance. They argue that drug addiction results from dysregulation of the reward mecha- nism and subsequent allostasis, the ability to achieve stability through change. Robinson and Berridge8,9 emphasize the dissociation between the incentive value of the drug (“wanting”) and its pleasurable or hedonic effects (“liking”), so that the brain system involved in the reward mechanism becomes hypersensitized to both the direct effects of the drug and associated stimuli that are not directly attributable to the drug. This hyper- sensitization causes pathologic wanting, or craving, independently of the presence of withdrawal symptoms and leads to compulsive drug-seeking and drug-taking behavior. Although liking progressively decreases, drugs become pathologically wanted (craving). Complementary to this incentive–sensitization theory,8,9 compulsive drug-seeking and drug-taking behavior is facilitated by difficulties in decision making and the ability to judge the consequences of one’s own actions. These cognitive difficulties have been linked to deficits in the activation of areas in the .10,11 An overlap in

n engl j med 349;10 www.nejm.org september 4, 2003 975

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

The new england journal of medicine

drug through the blood–brain barrier, water solu- Table 1. Definitions of Terms Used in Drug Addiction. bility facilitates the injection of a drug, volatility fa- Craving (formerly called psychological dependence) is an intense desire to re- vors the inhalation of drugs in vapor form, and heat experience the effects of a psychoactive substance. Craving is the cause of resistance favors smoking of the drug.13 Character- relapse after long periods of abstinence. istics such as rapid onset and intensity of effect in- Physical or physiological dependence is an outdated term that refers to physi- 14,15 cal tolerance and the withdrawal syndrome. crease the potential for abuse ; therefore, sub- stances that rapidly reach high levels in the brain are Priming refers to a new exposure to a formerly abused substance. This expo- sure can precipitate rapid resumption of abuse at previous levels or at usually preferred (e.g., flunitrazepam is preferred higher levels. over triazolam, and smoking “crack” cocaine is Relapse is a resumption of drug-seeking or drug-taking behavior after a period preferred to intranasal administration).16,17 A short of abstinence. Priming, environmental cues (people, places, or things as- half-life (e.g., that of heroin) produces more abrupt sociated with past drug use), and stress can trigger intense craving and cause a relapse. and intense syndromes of withdrawal than does a 13 Reward is a stimulus that the brain interprets as intrinsically positive or as long half-life (e.g., that of methadone). something to be attained. Sensitization is the increase in the expected effect of a drug after repeated ad- personality and psychiatric disorders ministration (e.g., increased locomotor activation after the administration Personality traits and mental disorders are major of psychostimulants). Sensitization also refers to persistent hypersensitiv- conditioning factors in drug addiction. Risk-taking ity to the effect of a drug in a person with a history of exposure to that drug (or to stress). Sensitization is one of the neurobiologic mechanisms in- or novelty-seeking traits favor the use of addictive volved in craving and relapse. drugs.18 Polydrug use is frequent among those Substance abuse is characterized by recurrent and clinically significant ad- with drug addiction, and many fulfill the criteria verse consequences related to the repeated use of substances, such as fail- for dependence on or abuse of (or both) more than ing to fulfill major role obligations, use of drugs in situations in which it is 19 physically hazardous, occurrence of substance-related legal problems, and one substance. Psychiatric disorders, particularly continued drug use despite the presence of persistent or recurrent social , , depression, and at- or interpersonal problems. tention-deficit–hyperactivity disorder, are associat- Substance dependence is a cluster of cognitive, behavioral, and physiological ed with an increased risk of abuse. A dual diagnosis symptoms indicating that a person is continuing to use a substance de- spite having clinically significant substance-related problems. For sub- (substance abuse and mental disorder) has unfavor- 20 stance dependence to be diagnosed, at least three of the following must be able implications for management and outcome. present: symptoms of tolerance; symptoms of withdrawal; the use of a substance in larger amounts or for longer periods than intended; persist- genetic factors ent desire or unsuccessful attempts to reduce or control use; the spending of considerable time in efforts to obtain the substance; a reduction in im- Genetic factors that influence the metabolism and portant social, occupational, or recreational activities because of drug use; the effects of drugs contribute to the risk of addic- and continued use of a substance despite attendant health, social, or eco- 21 nomic problems. tion. Men whose parents were alcoholics have an increased likelihood of alcoholism even when they Withdrawal syndrome is a constellation of signs and symptoms that follows the abrupt discontinuation or reduction in the use of a substance or after were adopted at birth and raised by parents who blockage of the actions of a substance with antagonists (e.g., naloxone in were not alcoholic, and they also have a reduced sen- heroin addiction). The syndrome can also be produced by cues associated sitivity to alcohol that predicts the development of with substance use (conditioned withdrawal). Symptoms tend to be the 22 opposite of those produced after short-term exposure to a drug. With- alcoholism. Carriers of an allele of aldehyde dehy- drawal is one of the causes of compulsive drug-taking behavior and short- drogenase that encodes an isoenzyme with reduced term relapse. activity are less likely to abuse alcohol owing to the presence of increased levels of acetaldehyde, which is responsible for aversive effects.23 A Leu7Pro poly- memory mechanisms and the mechanisms of drug morphism of the neuropeptide Y gene has been cor- addiction has also been proposed.12 related with increased alcohol consumption,24 and single-nucleotide polymorphisms of the gene en- factors influencing drug coding the µ correlates with an in- abuse and dependence creased likelihood of heroin abuse.25 A deficiency in the cytochrome P-450 2D6 gene blocks the enzy- pharmacologic and physicochemical matic conversion of codeine to morphine, thereby properties of drugs preventing codeine abuse.26 Pharmacologic and physicochemical properties of With regard to nicotine dependence, subjects drugs are important factors in how drugs are con- with defective cytochrome P-450 2A6 *2 and *4 sumed. Liposolubility increases the passage of a alleles, which impair the metabolism of nicotine,

976 n engl j med 349;10 www.nejm.org september 4, 2003

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

mechanisms of disease smoke fewer cigarettes and are less likely to be de- subject. Although there is evidence that long-term pendent than subjects who are homozygous for use of cannabis impairs memory,37,38 the cause of these alleles.27 A single-nucleotide polymorphism the marijuana amotivational syndrome — loss of in the gene encoding fatty acid amide hydrolase, a energy and drive to work — remains unclear.34 major endocannabinoid-inactivating , has G-protein–coupled CB1 receptors recently been associated with both an increased like- (Fig. 1 and 2), which are richly distributed in basal lihood of recreational use of illegal drugs and prob- ganglia and cerebral-cortex regions, are implicated lem use of drugs or alcohol.28 The minor (A1) allele in cannabinoid abuse and addiction. In contrast to of the TaqIA D2 receptor gene has been other neurotransmitters, endocannabinoids act as linked to severe alcoholism; polysubstance, psycho- retrograde messengers at many central synapses. stimulant abuse or dependence; and opioid and nic- They are released from postsynaptic neurons and 29 otine dependence. Advances in genomic scanning activate CB1 receptors on presynaptic neurons, in- (for quantitative trait loci) will allow the identifica- hibiting the release of neurotransmitters.39 Natural tion of allelic variants that contribute to the vulner- ligands of CB1 receptors (anandamide, 2-arachido- ability to addiction.30 nylglycerol, and noladin ether) have a shorter period of action than synthetic or plant-derived cannab- 39 drug effects and inoids. Selective synthetic and antago- mechanisms of action nists of CB1 receptors are currently being developed for medical purposes.40,41 opioids Short-term administration of heroin or morphine ethanol produces euphoria, sedation, and a feeling of tran- When ethanol is given at low doses or initially dur- quility. Repeated administration rapidly produces ing acute ethanol intoxication, it is perceived as a tolerance and intense physical dependence. Over- stimulant owing to the suppression of central in- dose can cause lethal respiratory depression. Nu- hibitory systems, but as the plasma levels of ethanol merous reports have documented impairments in increase, sedation, motor incoordination, ataxia, health related to long-term heroin use.31,32 and impaired psychomotor performance appear.42 Opioids activate specific receptors (µ, d, and k) The withdrawal syndrome (seizures and delirium that couple the G protein (Fig. 1 and 2). Knockout tremens) may be severe and clinically challenging. mice lacking the µ receptor neither exhibit the be- The long-term effects of ethanol consumption have havioral effects induced by opioids nor become been extensively reviewed elsewhere.43,44 physically dependent when given opioids (Table 2). Ethanol modifies the activity of serotonin The µ receptor has also been implicated in mediat- (5-hydroxytryptamine3 [5-HT3]) receptors, nicotin- ing or modulating the rewarding effect of other ic receptors, g-aminobutyric acid type A (GABAA) drugs of abuse (e.g., ). Mice in which receptors, and the N-methyl-d-aspartate (NMDA) different receptors (CB1 cannabinoid and D2 dopa- subtype of glutamate receptors. Ethanol acutely in- mine receptors) and transporters (dopamine) have hibits binding to the d-opioid receptor, and long- been knocked out have been used to demonstrate term exposure to ethanol increases the density of the effect of systems other than the opioid on opi- µ and d receptors.45 Its actions on nearly all recep- oid-induced pharmacologic responses.33 tors are the result of a direct interaction with the re- ceptor protein.46 cannabinoids The use of marijuana or hashish produces feelings cocaine and amphetamines of relaxation and well-being and impairs cognitive Short-term administration of psychostimulants function and performance of psychomotor tasks. such as amphetamine produces euphoria, a feeling Overdose can induce panic attack and .34 of well-being, and alertness as well as increased A high incidence of cannabis consumption has been arousal, concentration, and motor activity. These reported among patients with schizophrenia.35 substances increase blood pressure and the pulse Symptoms of withdrawal — restlessness, irritabil- rate and induce the release of corticotropin-releas- ity, and insomnia — are subtle and appear in heavy ing factor, corticotropin, and cortisol.47-49 Long- consumers.36 The long-term effects of high doses term use may cause irritability, aggressive and ster- of cannabinoids is a complex and controversial eotyped behavior, and paranoid-like psychosis.

n engl j med 349;10 www.nejm.org september 4, 2003 977

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

The new england journal of medicine

transmitters to the synapse; inhibit the uptake of Neurotransmitters dopamine, norepinephrine, and serotonin by mem- Acetylcholine, g-aminobutyric brane transporters; and act as mild inhibitors of Drugs of abuse acid, glutamate, serotonin Nicotine, , monoamine oxidase (Fig. 2). Amphetamine and barbiturates, ethanol, methamphetamine seem to be more selective for dopamine and norepinephrine than for serotonin transporters, but MDMA and designer amphet- amines are more selective for the serotonin trans- Channel porter.55

Extracellular Gate other addictive substances Nicotine binds to neuronal nicotinic acetylcholine receptors,56 and barbiturates and benzodiazepines bind and modulate the ion-channel–gated GABAA Intracellular receptors (Fig. 1 and 2).57 The psychotomimetic Ca2+, Na+, K+, Cl¡ actions of dissociative anesthesics (phencyclidine and ) are mediated by their noncompeti- Figure 1. Ionotropic Mechanisms of Action of Drugs of Abuse. tive antagonism at the NMDA-sensitive glutamate Drugs of abuse are usually receptor agonists, such as endogenous neuro- receptor (ligand-gated ).58 Lysergide or transmitters, that act on two different types of membrane receptors: ionotro- mescaline (classic hallucinogens) are partial ago- pic (shown in this figure) and metabotropic (shown in Fig. 2). Ionotropic 58 receptors (ligand-gated ion channels) mediate fast synaptic transmission. nists at 5-HT2a receptors, whereas in- The neurotransmitter or the drug binds to the receptor, which undergoes a duces their effects through the activation of k-opi- conformational change, opening the gate and allowing ions to enter the cyto- oid receptors.59 plasm and causing depolarization or polarization of the membrane and acti- vation of various proteins. Nicotine binds to nicotinic receptors, which contain a sodium channel. Benzodiazepines, barbiturates, and ethanol neurobiology bind to g-aminobutyric acid (GABA) type A receptors, facilitating the entry of chloride. Ethanol and phencyclidine inhibit N-methyl-d-aspartate–sensitive animal models glutamate receptors, which contain calcium and sodium channels. Phencycli- Various animal behavioral paradigms have been dine also acts as an antagonist. used to study the neuronal substrates involved in ad- diction, especially euphoria and rewarding effects, including self-stimulation, self-administration, and Whereas signs of withdrawal can be mild (depres- conditioned place-preference models (Fig. 3).60 In sion, lack of energy, and insomnia), craving is very the place-preference model, the rewarding proper- intense.50 So-called designer derivatives of am- ties of a compound are associated with the particular phetamine (3,4-methylenedioxymethamphetamine characteristics of a given environment (place); after [MDMA], or “ecstasy”) produce euphoria51,52 and conditioning, the animal prefers to spend more time increased empathy (an “entactogenic” effect), but in the environment associated with the drug. Mo- some derivatives have hallucinogenic effects. Acute lecular studies have identified regulatory processes intoxication with a psychostimulant can cause cer- that occur after drug administration at the level of ebral hemorrhage, hyperthermia and heat stroke, receptors, membrane transporters, and their asso- the serotonin syndrome, panic, and psychosis. The ciated signaling proteins. Useful strains of mice serotonin syndrome is characterized by altered have been developed by genetically disrupting drug mental status, autonomic instability, and neuro- targets (receptors and transporters) or proteins in muscular abnormalities resulting in hyperthermia. the pathways of these targets. Genetic alterations So-called designer derivatives of amphetamine may are generally present throughout development in have toxic effects on dopamine and serotonin neu- these mice. Therefore, when the phenotypes of in- rons.53,54 terest are absent, the effect of a drug could actually Cocaine is a potent blocker of the dopamine-, reflect compensatory changes in other neurobiolog- norepinephrine-, and serotonin-uptake transport- ic systems.33,61 To avoid this limitation, conditioned ers. Amphetamines have a more complex mecha- and tissue-specific knockout animals have been de- nism of action. Amphetamines cause neuronal veloped.62 storage vesicles in the cytoplasm to release neuro- For some drugs, the risk of abuse in humans can

978 n engl j med 349;10 www.nejm.org september 4, 2003

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

mechanisms of disease

Neurotransmitters Endogenous opioids, endogenous cannabinoids, Ion channel dopamine, norepinephrine, serotonin

Drugs of abuse Second-messenger– Amphetamines, cocaine generating enzyme Gate Drugs of abuse Opioids, cannabinoids, hallucinogens, phencyclidine

P

Receptor Second Protein kinase messenger GDP

GTP

Transcription factors (CREB, FosB) Pharmacologic effects

Figure 2. Metabotropic Mechanisms of Action of Drugs of Abuse. Metabotropic (G-protein–coupled) receptors mediate slow synaptic transmission. G proteins are trimeric structures composed of two functional units: an a subunit that catalyzes GTPase activity (converting guanosine triphosphate [GTP] to guanosine diphosphate [GDP]), and a b–g dimer that interacts with the a subunit when bound to GDP (inactive state). The binding of the activates a nearby G protein. The a subunit bound to GTP subsequently dissociates from its b and g subunits. Both can activate or inhibit ( or phospholipase C) that synthesize second messengers such as cyclic adenosine monophosphate (cAMP), cyclic guanosine monophosphate, inositol tri- phosphate, and diacylglycerol. In addition, the b and g subunits directly regulate calcium-, sodium-, and potassium-ion channels. Second messengers also regulate ion channels by activating protein kinases, which phosphorylate (P) such channels. Protein kinases induce pharmacologic effects and produce changes in transcription factors such as cAMP- responsive element–binding protein (CREB) and ∆FosB. Opioids bind to opioid receptors (which reduce cAMP levels), and cannabinoids bind to cannabinoid receptors. Classic hallucinogens are partial agonists of serotonin receptors. Am- phetamines and cocaine have an indirect action on receptors, increasing the synaptic levels of dopamine, norepineph- rine, and serotonin (facilitating release and inhibiting , respectively). These neurotransmitters activate different subtypes of , , and receptors. be predicted with the use of tests based on behavior- neuroanatomical substrates al paradigms in animals. These methods, however, The neuronal pathways of drug addiction are com- are not systematically used during the development ponents of the mesocorticolimbic dopamine sys- of centrally acting drugs,63 probably because the tems that originate in neurons in the ventral teg- methods are not required by regulatory agencies. mental area (Fig. 4).64 All drugs of abuse act on this

n engl j med 349;10 www.nejm.org september 4, 2003 979

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved.

The new england journal of medicine

the withdrawal syndrome associated with opioids, Table 2. Inactivation of Opioid Receptors in Knockout Mice. cannabinoids, ethanol, psychostimulants, and nic- Opioid Self- Place- otine, there is a substantial decrease in That Withdrawal Administration Preference levels in the .66 Is Knocked Out Syndrome Model* Model* Analgesia Dopamine binds to a G-protein–coupled recep- µ Receptor No No No No tor with two main subtypes, D1-like receptors (D1 d Receptor Yes Unknown Unknown Yes and D5) and D2-like receptors (D2, D3, and D4). k Receptor Yes (reduction Unknown Yes Yes D1-like receptors activate adenylyl cyclase, whereas in symptoms) D2-like receptors inhibit the enzyme (Fig. 1 and 2). A membrane dopamine transporter moves the re- * The model is defined in Figure 3. leased neurotransmitter from the extracellular space back into the presynaptic neuron (uptake). Animals system at different levels. The mesolimbic circuit can be taught to self-administer D1-like and D2-like includes projections from cell bodies of the ventral receptor agonists.55,67 tegmental area to limbic structures, such as the nucleus accumbens, , and . Dopamine and Opioids This circuit has been implicated in acute reinforcing Opioids release dopamine mainly by an indirect effects, memory, and conditioned responses linked mechanism that decreases the activity of GABA- to craving and the emotional and motivational inhibitory interneurons in the ventral tegmental changes of the withdrawal syndrome. The mesocor- area. Rodents can be taught to self-administer tical dopamine circuit includes projections from the µ-receptor agonists into both the ventral tegmental ventral tegmental area to the prefrontal cortex, orb- area and the nucleus accumbens. Stimulation of itofrontal cortex, and anterior cingulate. It is in- k receptors decreases dopamine levels in the nucle- volved in the conscious experience of the effects of us accumbens and produces aversive responses. Ro- drugs, drug craving, and the compulsion to take dents will continue to self-administer opioid into drugs. The mesolimbic and the mesocortical dopa- the nucleus accumbens even in the presence of do- mine circuits operate in parallel and interact with paminergic lesions or after a dopamine antagonist each other and with other areas — forming the so- has been given. Reward and physical dependence on called extended amygdala — by means of projec- opioids are mediated by the activation of µ receptors tions from the GABA neurons of the nucleus accum- (Table 2), since reinforcement is blocked by selec- bens to the ventral tegmental area and prefrontal tive receptor antagonists. Mice in which the µ recep- cortex and projections from the pre- tor has been knocked out do not exhibit place pref- frontal cortex to the nucleus accumbens and ventral erence or withdrawal signs after the administration tegmental area.11,12,64 of morphine.68

the leading role of the dopamine pathway Dopamine and Cannabinoids Both natural rewards (food, drink, and sex) and ad- ∆9-Tetrahydrocannabinol and other cannabinoids dictive drugs stimulate the release of dopamine increase the efflux of dopamine in the nucleus ac- from neurons of the presynaptic ventral tegmental cumbens and cell firing in the ventral tegmental area into the nucleus accumbens, causing eupho- area by their actions on CB1 receptors in glutamater- ria and reinforcement of the behavior. In the case gic and GABA-ergic neurons associated with the of natural rewards, there is a very rapid adaptive nucleus accumbens and ventral tegmental area.69 change, or habituation, after a few experiences, and Laboratory animals cannot be taught to self-admin- the novelty or unexpectedness of the reward seems ister ∆9-tetrahydrocannabinol, and spontaneous to play a major part in the initial response. The re- symptoms of withdrawal do not appear after such sponse to addictive drugs is not influenced by ha- drugs are stopped. Highly potent, short-acting syn- bituation, and each dose of the drug stimulates the thetic cannabinoid agonists (e.g., Win 55,212-2) in- release of dopamine.65 Moreover, dopamine medi- duce self-administration behavior in rodents.70 Se- ates the hedonic consequences of a reinforcing stim- lective cannabinoid antagonists (e.g., SR 141716A) ulus, promoting associative learning about the stim- precipitate a withdrawal syndrome in cannabinoid- ulus or anticipating its rewarding effects.65 During dependent animals, but at doses that are difficult

980 n engl j med 349;10 www.nejm.org september 4, 2003

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved. mechanisms of disease to extrapolate to the consumption of plant-derived cannabis in humans.71 A Electric Pump dispensing stimulator drug or saline Dopamine and Ethanol Ethanol raises dopamine levels in the nucleus ac- cumbens by an indirect mechanism: it increases the Computer firing of dopamine neurons in the ventral tegmental area by activating GABAA receptors or by inhibiting NMDA receptors. Alcohol administration induces Lever specific, long-lasting adaptive changes in the com- position of NMDA receptor subunits, which en- hances the function of NMDA receptors. These find- ings are the basis of the experimental and clinical B use of NMDA antagonists for treating the ethanol- Drug withdrawal syndrome and reducing alcohol intake and relapse rate.43,72 Opioid and serotonin recep- tors seem to have a role in the reinforcing effects of ethanol. In rats, naltrexone decreases the rate of self- administration of ethanol, and selected 5-HT3 an- tagonists block the release of dopamine induced by ethanol and reduce alcohol consumption.45,72

Dopamine and Cocaine and Amphetamines Saline Cocaine and amphetamines increase synaptic do- pamine levels by inhibiting the activity of dopamine transporters. Imaging of the brain has shown that cocaine and amphetamines increase extracellular dopamine levels in the and that euphoria is related to the occupancy of dopamine transporters by cocaine and amphetamines.73 Lesions in dopa- mine pathways, inhibition of dopamine synthesis, and dopamine antagonists markedly attenuate the rate of self-administration of cocaine in rats. Dopa- mine-transporter–knockout mice are insensitive to the locomotor stimulatory effects and are less sen- sitive than normal mice to the behavioral effects of ? psychostimulants, but they will still self-administer cocaine and amphetamine. This effect may indicate a contribution of the serotonin and norepinephrine systems to the maintenance of the rewarding prop- Drug-tested mouse prefers chamber erties of cocaine.55,61 in which drug was given

Figure 3. Animal Behavioral Paradigms Used to Study Positive Reinforcing Dopamine and Nicotine (Rewarding) Actions of Drugs. Nicotine exerts its positive reinforcing effects by act- In Panel A, animals are trained to press a lever to obtain a drug or saline (self- ing on a4b2 nicotinic acetylcholine receptors locat- administration) or to receive intracranial current in brain-rewarding loci (self- ed on the somatodendritic membranes of the dopa- stimulation). In Panel B, mice are placed in a box with two discrete chambers, mine cells of the ventral tegmental area and possibly or environments, and are then repeatedly injected with a drug in one environ- by sensitizing a7 nicotinic acetylcholine receptors ment and with saline in the other environment. In a drug-free state, the animal is allowed access to both environments, and the amount of time spent in each located on glutamate terminals. Experiments with environment is recorded. A positively reinforcing effect of the drug is apparent mutant mice lacking b2 nicotinic acetylcholine re- if the mouse spends more time in the environment in which the drug was ad- ceptors confirm the primary role of this receptor in ministered (place preference) than in the one in which saline was administered. promoting the self-administration of nicotine.74,75

n engl j med 349;10 www.nejm.org september 4, 2003 981

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved. The new england journal of medicine

Amphetamines, cocaine, opioids, cannabinoids, phencyclidine

GLU 5-HT Amygdala GLU Opioid DA DA Prefrontal GABA cortex GABA Opioid GABA DA GABA Nucleus accumbens NE DA Locus ceruleus 5-HT Opioid Raphe nucleus Opioids, ethanol, Ventral tegmental area barbiturates, benzodiazepines

Figure 4. Neural Reward Circuits Important in the Reinforcing Effects of Drugs of Abuse. As shown in the rat brain, mesocorticolimbic dopamine (DA) systems originating in the ventral tegmental area include projections from cell bodies of the ventral tegmental area to the nucleus accumbens, amygdala, and prefrontal cortex; glutamatergic (GLU) projections from the prefrontal cortex to the nucleus accumbens and the ventral tegmental area; and projections from the g-aminobutyric acid (GABA) neurons of the nucleus accumbens to the prefrontal cortex. Opi- oid interneurons modulate the GABA-inhibitory action on the ventral tegmental area and influence the firing of norepi- nephrine (NE) neurons in the locus ceruleus. Serotonergic (5-HT) projections from the raphe nucleus extend to the ventral tegmental area and the nucleus accumbens. The figure shows the proposed sites of action of the various drugs of abuse in these circuits.

the opioid pathway long-term drug use The opioid system is another pathway in the brain and neuroadaptation involved in the rewarding effects of addictive drugs.71 The importance of this system in addiction tolerance and withdrawal to cannabinoids was brought out by studies in which Tolerance leads to modifications of drug use to ob- opioid-receptor antagonists were found to attenuate tain desired effects, by increasing the dose, reducing the self-administration of cannabinoids and precip- the intervals between doses, or both. Withdrawal itate behavioral signs of withdrawal in rats treated compels addicts to resume drug use to prevent or with cannabinoid agonists for long periods.76 Opi- reduce physical symptoms and dysphoria. Both tol- oid antagonists reduce the consumption and self- erance and withdrawal increase compulsive drug- administration of ethanol in animals, but naltrexone seeking and drug-taking behavior and are essential does not reduce the rate of relapse or alcohol con- in maintaining the addiction. Sensitization, howev- sumption in patients with alcoholism.77 By con- er, is also important. Molecular adaptations related trast, µ-receptor–knockout mice cannot be induced to tolerance and dependence have been studied ex- to self-administer alcohol.72 The opioid system also tensively in animal models of opioid and cocaine ad- inhibits the self-administration of nicotine in ani- diction, and the results can be extrapolated to other mals. Indeed, naloxone precipitates withdrawal in substances.12,79,80 rats that receive nicotine for long periods and re- duces cigarette consumption in smokers.74 In addi- Opioid Tolerance tion, nicotine-induced conditioned place preference Short-term administration of opioid activates the 78 is abolished in µ-receptor–knockout mice. µ-opioid Gai/o-coupled receptor, which inhibits

982 n engl j med 349;10 www.nejm.org september 4, 2003

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved. mechanisms of disease adenylyl cyclase, lowers cyclic adenosine mono- duction of dynorphin, which activates k receptors in phosphate (cAMP) levels, decreases cAMP-depend- the neurons of the ventral tegmental area and de- ent protein kinase A activity, and reduces phos- creases the release of dopamine in the nucleus ac- phorylation of cytoplasmic and nuclear targets, cumbens. These changes contribute to the negative including cAMP–responsive element–binding pro- emotions (dysphoria and anhedonia) present dur- tein (CREB), a transcription factor. Activation of ing the early phases of abstinence.66,79 µ receptors can also cause the phosphorylation of some mitogen-activated protein kinases, which in Stress Systems turn phosphorylate CREB and other transcription Drug administration and withdrawal activate central factors.12,79,81,82 Decreases in the number of opioid and peripheral stress systems.88 Short-term admin- receptors have been related in some reports to the istration elevates peripheral glucocorticoid levels development of opioid tolerance. Continuous stim- and central corticotropin-releasing factor levels. ulation desensitizes opioid receptors, which be- These hormonal elevations have been related to the come phosphorylated by G-protein–coupled recep- rewarding properties of drug use. During withdraw- tor kinases; b-arrestins then bind to the receptors, al, an increase in corticotropin-releasing factor in causing them to be internalized by the neuron.83 the amygdala has been related to stress and negative Other studies, however, have found that opioids that effects of abstinence.47,80 cause internalization of the opioid receptors are in- efficient in initiating tolerance.84 molecular mechanisms of sensitization and relapse Opioid Withdrawal Long-term administration of addictive drugs pro- Chronic activation of opioid receptors produces ef- duces alterations in the brain that increase vulnera- fects opposite to those of acute activation. It up- bility to relapse and facilitate craving even months regulates cAMP signaling pathways by increasing or years after successful detoxification. Factors in- the activity of adenylyl cyclases (subtypes I and VII), volved in relapse and craving include acute reexpo- cAMP-dependent protein kinase A, and tyrosine sure to the drug or drug-priming, exposure to envi- hydroxylase. In addition, chronic activation of opi- ronmental stimuli previously paired with drug use oid receptors increases the phosphorylation of or conditioned drug cues, and exposure to environ- CREB and ∆FosB, factors regulating gene transcrip- mental stressors (Table 1). The extent of sensitiza- tion.66,79,85 These changes correlate with the man- tion varies with different drugs and is responsible ifestations of the withdrawal syndrome. for responses, craving, and relapse. Up-regulation of cAMP is a homeostatic re- sponse to the inhibition of the locus ceruleus by opi- Functional Changes oids and a key mechanism in withdrawal. The locus Repeated administration of opioids, psychostimu- ceruleus is a noradrenergic nucleus that regulates lants, or nicotine sensitizes laboratory animals to the arousal, responses to stress, and the activity of the stimulant or rewarding effects, or both, of these ad- autonomic nervous system. Tolerance to the inhib- dictive substances. Behavioral sensitization is asso- itory effects of opioids occurs in the locus ceruleus ciated with marked and long-lasting alterations in during long-term administration. When opioid lev- the functional activity of the mesocorticolimbic els fall, the firing rates of neurons in the locus are dopamine system, particularly in glutamate and unopposed and lead to adrenergic overactivation. dopamine transmission in the nucleus accum- A direct relation between overactivity of the locus bens.12,67 Elevated levels of the R1 subunit of ceruleus neurons and the somatic expression of opi- a-amino-3-hydroxy-5-methyl-4-isoxazolepropion- oid withdrawal has been demonstrated.86 Mecha- ic acid (AMPA) subtype of glutamate receptors nisms other than noradrenergic neurons may also seems to be involved in sensitization and relapse.89 participate in opioid withdrawal because destruc- Stress or a single dose of addictive substances with tion of the locus ceruleus does not alter naloxone- different molecular mechanisms of action (cocaine, precipitated or spontaneous opioid withdrawal.87 amphetamine, morphine, ethanol, or nicotine) pro- In the nucleus accumbens, a similar up-regula- duces similar degrees of enhancement in the tion of the cAMP pathway, including activation of strength of glutamate excitatory synapses (AMPA- CREB, occurs after long-term administration of opi- sensitive glutamate receptors) on dopamine neu- oids, ethanol, or cocaine. CREB increases the pro- rons in the ventral tegmental area. The administra-

n engl j med 349;10 www.nejm.org september 4, 2003 983

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved. The new england journal of medicine

tion of a glucocorticoid antagonist blocked the ac- The glutamate system of the brain is responsible tions of stress but not those of addictive substances. for the long-term plasticity associated with learn- These changes can have a role in behavioral sensi- ing and memory. It is therefore not surprising that tization.90 the same glutamatergic mechanisms also underlie Sensitization is associated with long-lasting addiction-related behavior.12,66,79,95,96 Behavioral adaptive changes in the patterns of expression of sensitization can persist for weeks or months and genes of the terminal mesolimbic dopamine sys- is augmented by environmental cues (e.g., people, tems,79 in particular the activation of proteins be- places, or paraphernalia associated with past drug longing to the family of transcription factor activator use).12 These cues and sensitization contribute to protein 1, such as Fos proteins. In self-administra- relapse. Interestingly, the processes involved in sen- tion models, long-term treatment with morphine, sitization overlap the neuronal substrates and path- cocaine, or nicotine increases the expression of the ways involved in the rewarding properties of drug family of Fos-related transcription factors, particu- abuse.7,9 larly the extremely stable isoforms of ∆FosB. The persistent increase in the expression of Fos genes comments in mesolimbic structures is in part a consequence of the activation of the cAMP cascade through Advances in the neurobiology of drug addiction D1 dopamine receptors in the ventral tegmental have led to the identification of neuronal substrates area.85,91,92 responsible for the rewarding effects of prototypical drugs of abuse, which are crucial to the addictive Structural Changes process. There is increasing evidence that prolonged Exposure to addictive drugs can cause long-lasting exposure to drugs of abuse produces long-lasting structural changes in neurons. Opioids decrease the effects in cognitive and drug-rewarding circuits. size and the caliber of dendrites and soma of dopa- For this reason, addiction should be considered a mine neurons of the ventral tegmental area.93 Re- chronic medical illness.97,98 Symptoms of with- peated use of cocaine or amphetamine increases the drawal can be treated, and maintenance therapy is number of dendritic branch points and spines on available for most drugs of abuse,43,98-101 but the both medium spiny neurons in the nucleus accum- development of long-term strategies based on med- bens and pyramidal neurons in the medial prefron- ication, psychosocial support, and continued mon- tal cortex.94 Neurotrophic factors seem to be re- itoring97,98 is a challenging clinical goal. sponsible for these changes. Modifications in the Supported in part by grants from Generalitat de Catalunya density of dendritic spines and neurotrophic factors (2001SGR00407), Fondo de Investigación Sanitaria (98/0181 and 01/1336), and Biomed 2 (BMH4-CT98-2267). have also been implicated in long-term potentiation We are indebted to Rafael Maldonado, M.D., Ph.D., for valuable and long-term depression by AMPA-sensitive gluta- scientific review and helpful comments, and to Marta Pulido, M.D., mate receptors. for editorial assistance.

references 1. World Health Organization. The ICD-10 pothesis of reward: past and current status. ral cortices of cocaine patients. Biol Psychia- classification of mental and behavioral dis- Trends Neurosci 1999;22:521-7. try 2002;51:134-42. orders: clinical descriptions and diagnostic 6. Koob GF, Le Moal M. Drug abuse: he- 11. Goldstein RZ, Volkow ND. Drug addic- guidelines. Geneva: World Health Organi- donic homeostatic dysregulation. Science tion and its underlying neurobiological basis: zation, 1992. 1997;278:52-8. neuroimaging evidence for the involvement 2. American Psychiatric Association. Di- 7. Idem. Drug addiction, dysregulation of of the frontal cortex. Am J Psychiatry 2002; agnostic and statistical manual of mental reward, and allostasis. Neuropsychophar- 159:1642-52. disorders, 4th ed., text revision: DSM-IV-TR. macology 2001;24:97-129. 12. Hyman SE, Malenka RC. Addiction and Washington, D.C.: American Psychiatric As- 8. Robinson TE, Berridge KC. Incentive- the brain: the neurobiology of compulsion sociation, 2000. sensitization and addiction. Addiction 2001; and its persistence. Nat Rev Neurosci 2001; 3. Maddux JF, Desmond DP. Addiction or 96:103-14. 2:695-703. dependence? Addiction 2000;95:661-5. 9. Idem. Addiction. Annu Rev Psychol 2003; 13. Farre M, Cami J. Pharmacokinetic con- 4. Stolerman I. Drugs of abuse: behavioural 54:25-53. siderations in abuse liability evaluation. Br J principles, methods and terms. Trends Phar- 10. Franklin TR, Acton PD, Maldjian JA, et al. Addict 1991;86:1601-6. macol Sci 1992;13:170-6. Decreased gray matter concentration in the 14. Mumford GK, Evans SM, Fleishaker JC, 5. Spanagel R, Weiss F. The dopamine hy- insular, orbitofrontal, cingulate, and tempo- Griffiths RR. Alprazolam absorption kinet-

984 n engl j med 349;10 www.nejm.org september 4, 2003

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved. mechanisms of disease ics affects abuse liability. Clin Pharmacol tion: current approaches and future pros- of cocaine abuse and dependence. N Engl J Ther 1995;57:356-65. pects. Drugs 2002;62:1331-43. Med 1996;334:965-72. 15. Roset P, Farre M, de la Torre R, et al. 32. Fiellin DA, O’Connor PG. Office-based 51. Cami J, Farre M, Mas M, et al. Human Modulation of rate of onset and intensity of treatment of opioid-dependent patients. of 3,4-methylenedioxymeth- drug effects reduces abuse potential in N Engl J Med 2002;347:817-23. amphetamine (“ecstasy”): psychomotor healthy males. Drug Alcohol Depend 2001; 33. Kieffer BL, Gavériaux-Ruff C. Exploring performance and subjective effects. J Clin 64:285-98. the opioid system by gene knockout. Prog Psychopharmacol 2000;20:455-66. 16. Farre M, Teran MT, Roset PN, Mas M, Neurobiol 2002;66:285-306. 52. Harris DS, Baggott M, Mendelson JH, Torrens M, Cami J. Abuse liability of fluni- 34. Hall W, Solowij N. Adverse effects of Mendelson JE, Jones RT. Subjective and hor- trazepam among methadone-maintained cannabis. Lancet 1998;352:1611-6. monal effects of 3,4-methylenedioxymeth- patients. Psychopharmacology (Berl) 1998; 35. Zammit S, Allebeck P, Andreasson S, amphetamine (MDMA) in humans. Psycho- 140:486-95. Lundberg I, Lewis G. Self reported cannabis pharmacology (Berl) 2002;162:396-405. 17. Hatsukami DK, Fischman MW. Crack use as a risk factor for schizophrenia in 53. Ernst T, Chang L, Leonido-Yee M, Speck cocaine and cocaine hydrochloride: are the Swedish conscripts of 1969: historical co- O. Evidence for long-term neurotoxicity as- differences myth or reality? JAMA 1996;276: hort study. BMJ 2002;325:1199. sociated with methamphetamine abuse: a 1580-8. 36. Budney AJ, Hughes JR, Moore BA, Novy 1H MRS study. Neurology 2000;54:1344-9. 18. Helmus TC, Downey KK, Arfken CL, PL. Marijuana abstinence effects in marijua- 54. Ricaurte GA, Yuan J, Hatzidimitriou G, Henderson MJ, Schuster CR. Novelty seek- na smokers maintained in their home envi- Cord BJ, McCann UD. Severe dopaminergic ing as a predictor of treatment retention for ronment. Arch Gen Psychiatry 2001;58:917- neurotoxicity in primates after a common heroin dependent cocaine users. Drug Alco- 24. recreational dose regimen of MDMA (“ec- hol Depend 2001;61:287-95. 37. Solowij N, Stephens RS, Roffman RA, et stasy”). Science 2002;297:2260-3. 19. Leri F, Bruneau J, Stewart J. Understand- al. Cognitive functioning of long-term heavy 55. Everitt BJ, Wolf ME. Psychomotor stim- ing polydrug use: review of heroin and co- cannabis users seeking treatment. JAMA ulant addiction: a neural systems perspec- caine co-use. Addiction 2003;98:7-22. 2002;287:1123-31. [Erratum, JAMA 2002; tive. J Neurosci 2002;22:3312-20. [Erratum, 20. Kavanagh DJ, McGrath J, Saunders JB, 287:1651.] J Neurosci 2002;22:1a.] Dore G, Clark D. Substance misuse in pa- 38. Bolla KI, Brown K, Eldreth D, Tate K, 56. Mansvelder HD, McGehee DS. Cellular tients with schizophrenia: epidemiology Cadet JL. Dose-related neurocognitive ef- and synaptic mechanisms of nicotine addic- and management. Drugs 2002;62:743-55. fects of marijuana use. Neurology 2002;59: tion. J Neurobiol 2002;53:606-17. 21. Crabbe JC. Genetic contributions to ad- 1337-43. 57. Smith DE, Wesson DR. Benzodiaz- diction. Annu Rev Psychol 2002;53:435-62. 39. Wilson RI, Nicoll RA. Endocannabinoid epines and other sedative-hypnotics. In: 22. Schuckit MA, Edenberg HJ, Kalmijn J, et signaling in the brain. Science 2002;296: Galanter M, Kleber HD, eds. The American al. A genome-wide search for genes that re- 678-82. Psychiatric Press textbook of substance late to a low level of response to alcohol. Al- 40. Maldonado R. Study of cannabinoid de- abuse treatment. 2nd ed. Washington, D.C.: cohol Clin Exp Res 2001;25:323-9. pendence in animals. Pharmacol Ther 2002; American Psychiatric Press, 1999:239-50. 23. Nestler EJ. Genes and addiction. Nat 95:153-64. 58. Abraham HD, McCann UD, Ricaurte Genet 2000;26:277-81. 41. Martin BR. Identification of the endoge- GA. Psychedelic drugs. In: Davis KL, Char- 24. Lappalainen J, Kranzler HR, Malison R, nous cannabinoid system through integra- ney D, Coyle JT, Nemeroff C, eds. Neuropsy- et al. A functional neuropeptide Y Leu7Pro tive pharmacological approaches. J Pharma- chopharmacology: the fifth generation of polymorphism associated with alcohol de- col Exp Ther 2002;301:790-6. progress. Philadelphia: Lippincott Williams pendence in a large population sample from 42. Holdstock L, de Wit H. Individual differ- & Wilkins, 2002:1545-56. the United States. Arch Gen Psychiatry ences in the biphasic effects of ethanol. Al- 59. Roth BL, Baner K, Westkaemper R, et al. 2002;59:825-31. cohol Clin Exp Res 1998;22:1903-11. Salvinorin A: a potent naturally occurring 25. Kreek MJ. Drug addictions: molecular 43. Swift RM. Drug therapy for alcohol de- nonnitrogenous kappa opioid selective ago- and cellular endpoints. Ann N Y Acad Sci pendence. N Engl J Med 1999;340:1482-90. nist. Proc Natl Acad Sci U S A 2002;99: 2001;937:27-49. 44. O’Connor PG, Schottenfeld RS. Patients 11934-9. 26. Kathiramalainathan K, Kaplan HL, Ro- with alcohol problems. N Engl J Med 1998; 60. Balster RL. Drug abuse potential evalua- mach MK, et al. Inhibition of cytochrome 338:592-602. tion in animals. Br J Addict 1991;86:1549- P450 2D6 modifies codeine abuse liability. 45. Weiss F, Porrino LJ. Behavioral neurobi- 58. J Clin Psychopharmacol 2000;20:435-44. ology of alcohol addiction: recent advances 61. Laakso A, Mohn AR, Gainetdinov RR, 27. Rao Y, Hoffmann E, Zia M, et al. Dupli- and challenges. J Neurosci 2002;22:3332-7. Caron MG. Experimental genetic approach- cations and defects in the CYP2A6 gene: 46. Zhang L, Hosoi M, Fuzuzawa M, Sun H, es to addiction. Neuron 2002;36:213-28. identification, genotyping, and in vivo ef- Rawlings RR, Weight FF. Distinct molecular 62. Mantamadiotis T, Lemberger T, Bleck- fects on smoking. Mol Pharmacol 2000;58: basis for differential sensitivity of the sero- mann SC, et al. Disruption of CREB function 747-55. tonin type 3A receptor to ethanol in the ab- in brain leads to neurodegeneration. Nat 28. Sipe JC, Chiang K, Gerber AL, Beutler E, sence and presence of agonist. J Biol Chem Genet 2002;31:47-54. Cravatt BF. A missense mutation in human 2002;277:46256-64. 63. Cami J. Human drug abuse liability: fatty acid amide hydrolase associated with 47. Sarnyai Z, Shaham Y, Heinrichs SC. The testing times ahead. Br J Addict 1991;86: problem drug use. Proc Natl Acad Sci U S A role of corticotropin-releasing factor in drug 1525-6. 2002;99:8394-9. addiction. Pharmacol Rev 2001;53:209-43. 64. Kalivas PW. Neurocircuitry of addiction. 29. Noble EP. D2 dopamine receptor gene 48. Farre M, de la Torre R, Gonzalez ML, et In: Davis KL, Charney D, Coyle JT, Nemeroff in psychiatric and neurologic disorders and al. Cocaine and alcohol interactions in hu- C, eds. Neuropsychopharmacology: the its phenotypes. Am J Med Genet 2003;116B: mans: neuroendocrine effects and cocaeth- fifth generation of progress. Philadelphia: 103-25. ylene metabolism. J Pharmacol Exp Ther Lippincott Williams & Wilkins, 2002:1357- 30. Uhl GR, Liu QR, Naiman D. Substance 1997;283:164-76. 66. abuse vulnerability loci: converging genome 49. Mello NK, Mendelson JH. Cocaine’s ef- 65. Di Chiara G. Drug addiction as dopa- scanning data. Trends Genet 2002;18:420- fects on neuroendocrine systems: clinical mine-dependent associative learning disor- 5. and preclinical studies. Pharmacol Biochem der. Eur J Pharmacol 1999;375:13-30. 31. Gonzalez G, Oliveto A, Kosten TR. Behav 1997;57:571-99. 66. Nestler EJ. Molecular basis of long-term Treatment of heroin (diamorphine) addic- 50. Mendelson JH, Mello NK. Management plasticity underlying addiction. Nat Rev

n engl j med 349;10 www.nejm.org september 4, 2003 985

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved. mechanisms of disease

Neurosci 2001;2:119-28. [Erratum, Nat Rev ment of alcohol dependence. N Engl J Med synaptic adaptation in dopamine neurons. Neurosci 2001;2:215.] 2001;345:1734-9. Neuron 2003;37:577-82. [Erratum, Neuron 67. Konipsky KL, Hyman SE. Molecular and 78. Berrendero F, Kieffer BL, Maldonado R. 2003;38:359.] cellular biology of addiction. In: Davis KL, Attenuation of nicotine-induced antinoci- 91. Kelz MB, Nestler EJ. DeltaFosB: a mo- Charney D, Coyle JT, Nemeroff C, eds. Neu- ception, rewarding effects, and dependence lecular switch underlying long-term neural ropsychopharmacology: the fifth genera- in mu-opioid receptor knock-out mice. plasticity. Curr Opin Neurol 2000;13:715- tion of progress. Philadelphia: Lippincott J Neurosci 2002;22:10935-40. 20. Williams & Wilkins, 2002:1367-79. 79. Nestler EJ. Molecular neurobiology of 92. Nestler EJ, Barrot M, Self DW. 68. De Vries TJ, Shippenberg TS. Neural addiction. Am J Addict 2001;10:201-17. DeltaFosB: a sustained molecular switch for systems underlying opiate addiction. 80. Shalev U, Grimm JW, Shaham Y. Neuro- addiction. Proc Natl Acad Sci U S A 2001;98: J Neurosci 2002;22:3321-5. biology of relapse to heroin and cocaine 11042-6. 69. Robbe D, Alonso G, Duchamp F, Bock- seeking: a review. Pharmacol Rev 2002;54: 93. Sklair-Tavron L, Shi WX, Lane SB, Har- aert J, Manzoni OJ. Localization and mecha- 1-42. ris HW, Bunney BS, Nestler EJ. Chronic nisms of action of cannabinoid receptors at 81. Taylor DA, Fleming WW. Unifying per- morphine induces visible changes in the the glutamatergic synapses of the mouse spectives of the mechanisms underlying the morphology of mesolimbic dopamine neu- nucleus accumbens. J Neurosci 2001;21: development of tolerance and physical de- rons. Proc Natl Acad Sci U S A 1996;93: 109-16. pendence to opioids. J Pharmacol Exp Ther 11202-7. 70. Fattore L, Cossu G, Martellotta CM, 2001;297:11-8. 94. Robinson TE, Gorny G, Mitton E, Kolb Fratta W. Intravenous self-administration of 82. Watts VJ. Molecular mechanisms for B. Cocaine self-administration alters the the cannabinoid CB1 receptor agonist WIN heterologous sensitization of adenylate cy- morphology of dendrites and dendritic 55,212-2 in rats. Psychopharmacology clase. J Pharmacol Exp Ther 2002;302:1-7. spines in the nucleus accumbens and neo- (Berl) 2001;156:410-6. 83. Nestler EJ, Landsman D. Learning about cortex. Synapse 2001;39:257-66. 71. Maldonado R, Rodriguez de Fonseca F. addiction from the genome. Nature 2001; 95. Berke JD, Hyman SE. Addiction, dopa- Cannabinoid addiction: behavioral models 409:834-5. mine, and the molecular mechanisms of and neural correlates. J Neurosci 2002;22: 84. He L, Fong J, von Zastrow M, Whistler memory. Neuron 2000;25:515-32. 3326-31. JL. Regulation of opioid receptor trafficking 96. Duman RS, Malberg J, Nakagawa S. 72. Spanagel R. Behavioral and molecular and morphine tolerance by receptor oligo- Regulation of adult neurogenesis by psy- aspects of alcohol craving and relapse. In: merization. Cell 2002;108:271-82. chotropic drugs and stress. J Pharmacol Exp Maldonado R, ed. Molecular biology of drug 85. Nestler EJ, Aghajanian GK. Molecular Ther 2001;299:401-7. addiction. Totowa, N.J.: Humana Press, and cellular basis of addiction. Science 97. McLellan AT, Lewis DC, O’Brien CP, 2002:295-313. 1997;278:58-63. Kleber HD. Drug dependence, a chronic 73. Volkow ND, Fowler JS. Application of 86. Rasmussen K, Beitner-Johnson DB, medical illness: implications for treatment, imaging technologies in the investigation of Krystal JH, Aghajanian GK, Nestler EJ. Opi- insurance, and outcomes evaluation. JAMA drug addiction. In: Davis KL, Charney D, ate withdrawal and the rat locus coeruleus: 2000;284:1689-95. Coyle JT, Nemeroff C, eds. Neuropsycho- behavioral, electrophysiological, and bio- 98. Kreek MJ, LaForge KS, Butelman E. pharmacology: the fifth generation of chemical correlates. J Neurosci 1990;10: Pharmacotherapy of addictions. Nat Rev progress. Philadelphia: Lippincott Williams 2308-17. Drug Discov 2002;1:710-26. [Erratum, Nat & Wilkins, 2002:1475-90. 87. Caille S, Espejo EF, Reneric JP, Cador M, Rev Drug Discov 2002;1:920.] 74. Merlo E, Heidbreder C, Mugnaini M, Koob GF, Stinus L. Total neurochemical le- 99. Johnson RE, Chutuape MA, Strain EC, Teneggi V. Molecular and behavioral aspects sion of noradrenergic neurons of the locus Walsh SL, Stitzer ML, Bigelow GE. A com- of nicotine dependence and reward. In: Mal- ceruleus does not alter either naloxone-pre- parison of levomethadyl acetate, buprenor- donado R, ed. Molecular biology of drug ad- cipitated or spontaneous opiate withdrawal phine, and methadone for opioid depend- diction. Totowa, N.J.: Humana Press, 2002: nor does it influence ability of clonidine to ence. N Engl J Med 2000;343:1290-7. 315-38. reverse opiate withdrawal. J Pharmacol Exp 100. Farre M, Mas A, Torrens M, Moreno V, 75. Picciotto MR, Corrigall WA. Neuronal Ther 1999;290:881-92. Cami J. Retention rate and illicit opioid use systems underlying behaviors related to nic- 88. Cami J, Gilabert M, San L, de la Torre R. during methadone maintenance interven- otine addiction: neural circuits and molecu- Hypercortisolism after opioid discontinua- tions: a meta-analysis. Drug Alcohol De- lar genetics. J Neurosci 2002;22:3338-41. tion in rapid detoxification of heroin ad- pend 2002;65:283-90. 76. Navarro M, Chowen J, Rocio A, et al. dicts. Br J Addict 1992;87:1145-51. 101. Fudala PJ, Bridge TP, Herbert S, et al. CB1 cannabinoid - 89. Carlezon WA Jr, Nestler EJ. Elevated lev- Office-based treatment of opiate addiction induced opiate withdrawal in morphine- els of GluR1 in the midbrain: a trigger for with a sublingual-tablet formulation of bu- dependent rats. Neuroreport 1998;9:3397- sensitization to drugs of abuse? Trends prenorphine and naloxone. N Engl J Med 402. Neurosci 2002;25:610-5. 2003;349:949-58. 77. Krystal JH, Cramer JA, Krol WF, Kirk 90. Saal D, Dong Y, Bonci A, Malenka RC. Copyright © 2003 Massachusetts Medical Society. GF, Rosenheck RA. Naltrexone in the treat- Drugs of abuse and stress trigger a common

986 n engl j med 349;10 www.nejm.org september 4, 2003

The New England Journal of Medicine Downloaded from nejm.org at UQ Library on April 4, 2017. For personal use only. No other uses without permission. Copyright © 2003 Massachusetts Medical Society. All rights reserved.