Dennis M. Fisher, M.D., Editor

ANESTHESIOLOGISTS administer many drugs that alter mood in patients during the perioperative period. These drugs include inhaled general anesthetic agents, barbiturates, benzodiazepines, opioids, [small alpha, Greek]2-adrenergicagents, local anesthetic agents, antihistamines, and anticholinergic agents. This article discusses abuse liability and the estimated prevalence of abuse of these drugs. Because the practice of anesthesiology involves manipulation of consciousness via psychotropic agents, an understanding of the abuse potential of these consciousness-altering agents may be helpful.

Abuse liability is defined as the potential or likelihood that a drug will be used for nonmedical (recreational) purposes. The most commonly used testing methods for abuse liability assess the reinforcing (or rewarding) effects of a drug, its subjective effects, and its discriminative stimulus effects.

Three methods are used by psychopharmacologists to assess the reinforcing or rewarding effects of drugs: self-administration, preference, and conditioned place preference. With self-administration, the test animal (or human) is given access to a drug and a nondrug (placebo) so that consumption of each can be compared. In animal studies, the animal presses a lever for access to a drug or its vehicle (usually saline), which is typically delivered from an infusion pump to an intravenous catheter. If the response rate when the drug is available is higher than the response rate when the vehicle is available, the drug is said to function as a reinforcer. Although most studies of drug self-administration involve animals as subjects, numerous studies of self-administration have been conducted with humans. The concordance in results between self-administration studies using animals and humans is high, validating the use of animals to estimate the potential for a drug's abuse in humans. [1-3] 

The preference or choice procedure is similar to a self-administration procedure, except that the testee may have more options: drug versus placebo, drug versus drug, or drug versus a nondrug reinforcer (such as food or saccharin). The first option measures whether a drug is a reinforcer, and the second and third options measure the relative reinforcing efficacy of one drug compared with another or with a nondrug substance.

There are several variants to the conditioned place preference procedure. In one variant, an animal is placed in a two-compartment chamber. Each compartment is distinguishable from the other by one or more features (e.g., color of the walls, texture of the floors). When the animal has habituated to the chamber, the conditioning process begins. In a series of alternating trials, the animal is injected with a drug and placed in the drug compartment or injected with saline and placed in the other compartment, so that the effects of the drug become associated with one of the compartments. After the conditioning trials, the drug-free animal is placed in a neutral area of the chamber, and the relative amount of time spent in the drug compartment is measured. If the animal spends more time in the drug compartment than in the nondrug compartment, the drug is said to have rewarding effects.

To measure subjective effects in humans, a drug or placebo is administered, and the person is asked periodically to report subjective feelings. The medium of self-report varies and includes visual analog scales, adjective rating checklists, and standardized questionnaires to measure mood or specific drug-induced changes in subjective effects. Three questionnaires frequently used by psychopharmacologists include the Profile of Mood States (POMS), [4]the Addiction Research Center Inventory (ARCI), and the Single Dose Questionnaire (SDQ). [5]A shortened version of the original ARCI 550-item inventory [6]consists of 49 items [7]grouped into five different subscales representing subjective effects associated with different drugs: the Pentobarbital-Chlorpromazine Alcohol Group for sedation; the Amphetamine scale and the Benzedrine Group scale for intellectual efficiency and psychomotor stimulation; the Morphine-Benzedrine Group scale for euphoria; and the Lysergic Acid Diethylamide Scale indicative of dysphoria or psychotomimetic effects.

The discriminative stimulus effects of a drug are internal, or interoceptive, cues produced by the drug that are presumably mediated by the same neurochemical substrate(s) that mediate the subjective effects of a drug in humans. [8]To measure drug discrimination in animals, the animal is trained to emit a certain behavioral response after receiving drug X and another response after receiving drug Y. One of the drugs is typically the placebo and represents the nondrug response. For example, when an animal is given drug X (called the training drug), it is trained to press one of two levers (e.g., the left lever) in a test chamber. When the animal is given drug Y (e.g., placebo), it is trained to press the right lever in the chamber. A correct response (responding on the left lever when drug X is given and responding on the right lever when drug Y is given) is reinforced (usually with food pellets), whereas inaccurate responses (e.g., responding on the left lever when drug Y is given) during the discrimination training phase are not reinforced. The key stage of the drug discrimination procedure is the testing phase, which involves giving a drug other than drug X. If the animal responds on the left lever, the test drug is said to substitute for drug X and to have similar discriminative stimulus effects to those of drug X. If the animal presses the right lever, the test drug does not have similar discriminative stimulus effects to that of drug X. The drug discrimination procedure is highly specific; typically only those drugs with pharmacologic similarities to the training drug result in drug-appropriate responses, and drugs with pharmacologic dissimilarities result in nondrug or placebo-appropriate responses. [8]Drug discrimination is a valid screening tool for abuse liability in that a drug that has similar discriminative stimulus effects to that of a training drug with abuse potential (e.g., heroin) is also likely to have abuse potential.

In general, those drugs that are reinforcing, that have a pleasant spectrum of subjective effects, or that share discriminative stimulus effects with known drugs of abuse are said to have abuse liability. Those drugs that do not have these effects are said to have negligible or minimal abuse liability. There is no system universally agreed on by psychopharmacologists to rank the degree of abuse liability of a drug. The degree of abuse liability depends on many factors, including the doses tested, the route of administration, the species tested, and the test method used (self-administration testing vs. subjective effects testing). When enough scientific data have been collected on a drug using several different assays and species, however, it is generally possible to place a drug into one of three abuse liability categories: substantial, minimal, or equivocal. A drug with substantial abuse liability is one for which there is concordance across studies, assays, and species that indicates abuse liability. Conversely, a drug with minimal abuse liability is one for which there is concordance across studies, assays, and species that indicates negligible abuse liability. A drug with equivocal abuse liability is a drug in which the degree of abuse liability is not clear. It may be that one procedure indicates substantial abuse liability and another procedure suggests minimal abuse liability, or it may be that even using the same species and assay there are disagreements among studies, with some suggesting substantial abuse liability and others suggesting something less than substantial. Finally, a drug with equivocal abuse liability may show less than substantial effects but greater than minimal in a given procedure. For example, using subjective effects testing, a drug may produce a spectrum of pleasant and dysphoric effects. Or, in a self-administration procedure, the drug may generate responses at a slightly higher rate than that generated by presentation of the drug vehicle; by definition the drug functions as a reinforcer, but its efficacy may be weak. These drugs can be classified as having equivocal abuse liability.

Fentanyl and its analogs, alfentanil, sufentanil, and more recently remifentanil, have been tested for abuse liability. Fentanyl and sufentanil produced a conditioned place preference in rats, [9]and fentanyl is self-administered by rats and primates, [10,11]indicating that these drugs are reinforcers. Fentanyl and sufentanil share discriminative stimulus effects with other [micro sign] opioids that are abused, including morphine and heroin. [12]When fentanyl (dose range, 0.4-1.6 mg/70 kg) was administered to non-physically dependent opioid abusers, they reported increased euphoria scores on the ARCI, identified the drug as “dope,” and reported on the SDQ that they liked the drug effects. [13]In another study that assessed fentanyl (0.125 and 0.250 mg/70 kg) and alfentanil (1 and 2 mg/70 kg, intravenously) in nondependent opioid abusers, a similar spectrum of pleasant subjective effects was found with both drugs. [14]Remifentanil, at several sub-anesthetic doses, was compared with fentanyl in nondependent opioid abusers in an abuse liability study. [15]The drugs had similar euphoric effects, although the duration of effect was longer with fentanyl, which is consistent with its pharmacokinetic profile. The investigators concluded that “a drug abuser seeking a longer-lasting drug effect might select fentanyl over remifentanil, but these data do not rule out remifentanil abuse when briefer or repeated effects are desired.”[15] 

Abuse liability studies can and do include non-drug-abusing humans. Such studies give important information regarding the overall abuse liability in the general population, most of whom are not drug abusers. There is evidence that fentanyl has abuse liability in non-drug abusers. In two studies in human subjects, participants reported liking fentanyl, [16,17]but in another study some participants liked its effects, some found the effects to be neutral, and others disliked its effects. [18]In a self-administration study, non-drug abusers were more likely to choose fentanyl than saline in the presence of a painful stimulus. [19]Again, there was intersubject variability on the measure of reinforcing effects, with some choosing fentanyl over placebo even in the absence of pain. Therefore, fentanyl appears to have abuse potential in some non-drug abusers.

Morphine and heroin (3,6-diacetylmorphine) also have been assessed for abuse liability. Although heroin, the most commonly abused opioid in the world, [20]is not available for clinical use in the United States, it is in medical use in some countries, including the United Kingdom. In laboratory self-administration studies performed with animals, morphine and heroin are readily self-administered [21]and function as rewards in conditioned place preference procedures. [22,23]Both drugs substitute for each other in drug discrimination studies and also share discriminative stimulus effects with other full [micro sign] agonists. [24,25]The reinforcing effects of heroin and morphine have been established in nondependent opioid abusers. [26,27]After administration of morphine or heroin, opioid abusers had increased scores for euphoria on the ARCI, identified the drugs as “dope,” and reported liking them on the SDQ. [28,29]Non-drug-abusing volunteers, however, reported dislike of the drug effects. [30]In other studies, the unpleasant effects of morphine, such as heaviness, lethargy, and drowsiness, were reported more frequently than euphoria. [31,32]Thus, morphine and heroin have substantial abuse liability in opioid abusers and minimal abuse liability in non-drug abusers.

Meperidine (pethidine) has the same abuse liability profile as that or morphine and heroin; in animals, it is self-administered [21]and has discriminative stimulus effects in common with other full [micro sign] agonists. [33]In opioid abusers, it increased euphoria scores on the ARCI and drug-liking ratings and was identified as “dope” on the SDQ. [29]The drug's profile of subjective effects differs from that of morphine. Meperidine increased sedation scores on the ARCI and “sleepy” ratings on the SDQ, and some study participants identified the drug as a barbiturate. The increased feelings of sedation may be attributable to the putative anticholinergic effects of meperidine. [34]In non-drug abusers, meperidine also produced higher sedation ratings than did morphine. [35]It is note-worthy that meperidine tended to produce more consistent liking ratings among healthy volunteers than did morphine.

Hydromorphone, in both dependent and nondependent opioid abusers, resembles morphine in its effects, [36,37]and its abuse potential is considered to be the same as that of morphine. [38]Animal and human abuse liability tests have established that codeine, hydrocodone, and propoxyphene have an abuse potential similar to that of other [micro sign] opioid agonists. Codeine and propoxyphene function as reinforcers [21]and have morphine-like discriminative stimulus effects. [24]In opioid-abusing humans, all three opioids have a positive spectrum of subjective effects. [39-41]No systematic studies have been conducted with oxycodone, but it is probable that its abuse liability is similar to that of other [micro sign] agonists.

The opioids nalbuphine, butorphanol, and pentazocine are mixed agonists-antagonists. These drugs are self-administered by nondependent animals, [42-44]and in drug discrimination studies they resemble the full [micro sign] opioid agonists. [45-47]In animals that are physically dependent on [micro sign] opioids, the drugs are not self-administered but rather precipitate withdrawal. [48-50]The subjective effects of these drugs in physically dependent abusers resemble those of opioid withdrawal. [51-53]Therefore, abuse potential in physically dependent opioid abusers is minimal. The drugs appear to have equivocal abuse potential in opioid abusers who are not physically dependent on opioids. [38]In this population, the drugs produced greater dysphoria and sedation scores on the ARCI than did morphine-like drugs, especially at high doses. [38]At lower doses, however, euphoria scores and liking ratings were increased. In drug discrimination studies using humans, the mixed agonist-antagonist drugs were more likely to substitute for each other (e.g., nalbuphine for butorphanol) than for morphine-like agonists. [37,54]In contrast, in animal studies, the [micro sign] agonists and mixed agonists-antagonists readily substitute for each other. This discrepancy has not been explained adequately.

The subjective effects of intravenous butorphanol, nalbuphine, and pentazocine have been characterized and compared with those of morphine in non-opioid abusing volunteers. [55-57]The three mixed agonist-antagonist drugs produced some degree of drug liking soon after injection, but morphine did not. Butorphanol was more sedative than morphine; nalbuphine and morphine produced similar subjective effects. Pentazocine produced more dysphoria than did morphine, which is consistent with the greater incidence of psychotomimesis (i.e., unpleasant auditory, visual, and other bodily sensations) with pentazocine in the clinical population. [58]Transnasal butorphanol increased sedation but not drug liking in opioid abusers and in nonabusers, suggesting that this route of administration of butorphanol has a low potential for abuse in the general population. [59,60] 

Three opioids, buprenorphine, dezocine, and tramadol, are partial [micro sign] agonists. In physically dependent humans, buprenorphine and dezocine are thought to have low abuse liability because these drugs precipitate a mild withdrawal syndrome. [61,62]In non-physically dependent animals and humans with a history of opioid abuse, however, the drugs function as reinforcers, [50,63]share discriminative stimulus effects with full [micro sign] opioid agonists, [50,64]and have a positive subjective effects profile. [54,65]Tramadol also has substantial noradrenergic and serotonergic activity. [66]The abuse potential of this drug has not been studied as thoroughly as that of other opioids. Tramadol is self-administered in rhesus monkeys. [67]In nondependent opioid abusers, the drug has minimal subjective effects but is identified as “dope”;[68]in dependent opioid users it has no subjective, behavioral, or physiologic effects. [69] 

The abuse liability of full [micro sign] agonists in animals and humans is high. The actual abuse of opioids is low in the United States population but still affects hundreds of thousands of people, has remained relatively constant over the last 30 yr, and poses a significant public health problem. All of the full [micro sign] opioids, including the opioid/acetaminophen or opioid/aspirin preparations, are abused. The lifetime, past-year, and past-month incidence figures of nonmedical use of heroin reported by people >or= to 12 yr surveyed by the National Household Survey on Drug Abuse (NHSDA) were 1.2%, 0.2%, and 0.1%, respectively. [70]Why full [micro sign] opioids such as morphine and meperidine are not abused to any great extent by opioid abusers is unknown. One potential reason may be that these drugs are not easily diverted from pharmaceutical supply houses and hospitals, given their scheduling status, and therefore they are not readily available in the drug-abusing community. Differences in metabolic pathways may account for the differential abuse; meperidine has an active metabolite, normeperidine, which with chronic high doses can cause excitation and toxicity in the central nervous system. including tremors and convulsions. [71]Heroin, in contrast, does not have toxic metabolites. Another factor to consider is pharmacokinetic properties; the “rush” with morphine is not as immediate as with heroin because morphine crosses the blood-brain barrier more slowly. [72]Drugs with a more rapid onset of effect tend to be more reinforcing, and abused, than drugs with a slower onset of effect. [73] 

Abuse of fentanyl is associated with high morbidity and mortality. [74-77]One subgroup in the general population likely to abuse fentanyl and sufentanil is anesthesiology personnel, [78]possibly because they have ready access to these drugs. Other potential reasons include the difficulty in detecting abuse of fentanyl/sufentanil with urine toxicology screening (i.e., many clinical laboratories do not test for fentanyl and its analogs because fentanyl does not cross-react with commercially available opioid immunoassays, and concentrations of fentanyl in fluids and tissues in fatal intoxications are near the lower limit of detection for even the most sensitive analytical techniques because of its potency [79]); the rapid onset of effects (the “rush”) with fentanyl; its calming and euphoric effects; and the relative ease of titrating effect and duration of effect, given the short-acting nature of the drugs. Abuse of fentanyl might increase in the coming years with the advent of new formulations in which injection will the advent of new formulations in which injection will no longer be necessary (oral transmucosal fentanyl citrate, transdermal fentanyl patch). Although there are no confirmed cases of abuse of oral transmucosal fentanyl citrate to date, there is a case report of a person inhaling fentanyl from a patch. [80]Whether remifentanil will pose a serious threat as an abused drug among anesthesiology personnel is unknown. It is possible that the short-lived effects of remifentanil, unless prolonged by a continuous infusion, would deter abuse. If remifentanil supplants fentanyl for medical procedures, however, its greater availability may increase illicit use. Remifentanil, unlike other fentanyl-related agents, requires dilution and can be diverted for personal use more easily during the diluting process. Finally, because remifentanil is relatively new, the amount needed for each patient are not easily determined from the small scientific literature base currently available. A larger amount may be prepared for a case than is actually needed, again making diversion easier.

In laboratory animals, the mixed opioid agonist-antagonist drugs have high abuse liability, but in opioid abusers they tend to produce sedation and dysphoria, indicating a lower probability of abuse. Only a few case reports of abuse exist. [81-84]In drug abusers who depend physically on opioids, withdrawal symptoms would probably be precipitated by use of these drugs. The partial agonist, buprenorphine, has a profile of high abuse liability in animals and in abusers who are not physically dependent on opioids. Abuse of buprenorphine by dependent and nondependent opioid abusers has been reported in several countries, including the United Kingdom, New Zealand, and India. [85-89].

Midazolam and diazepam have been assessed for abuse liability. Rodents and primates self-administer midazolam, and it induces conditioned place preference, demonstrating reinforcing effects. [90-92]In drug discrimination studies with animals, midazolam shares similar discriminative stimulus characteristics with other benzodiazepines and also, in rodents, with the barbiturates. [93,94]No studies with humans have assessed the abuse liability of midazolam systematically. Before midazolam was developed, diazepam was used preoperatively and for conscious sedation procedures because of its amnestic effects. The popularity of diazepam has declined, possibly because its carrier, propylene glycol, is painful and because midazolam is an available alternative. As with midazolam, diazepam functions as a reinforcer in animals. [91,95]Diazepam shares discriminative stimulus effects with other benzodiazepines and some barbiturates. [96,97]Several studies have characterized the abuse liability of diazepam in sedative abusers and non-drug-abusing volunteers. [98,99]In sedative abusers, diazepam was chosen over placebo in choice tests. Relative to placebo, it increased euphoria scores on the ARCI and drug-liking ratings. [100]In nonanxious volunteers who characterized their alcohol consumption as light, diazepam did not function as a reinforcer and did not generate positive subjective effects. [101]In nonanxious moderate drinkers, however, the drug functioned as a reinforcer and increased drug-liking ratings and mood scores for friendliness and elation on the POMS. [101,102]Thus, quantity of alcohol normally consumed can modulate the reinforcing effects of benzodiazepines. Finally, in volunteers with anxiety disorders, diazepam functioned as a reinforcer in two studies [103,104]but not in two others. [105,106] 

Other benzodiazepines used in anesthetic practice include lorazepam, alprazolam, triazolam, flunitrazepam, and temazepam. Flunitrazepam is not available for clinical use in the United States but is available elsewhere. These drugs have relatively short durations of action, which make them suitable for premedication, and they are used as hypnotic, anxiolytic, and sedative agents. [107-111]Several preclinical and human studies (testing subjective and reinforcing effects) have established that alprazolam, lorazepam, triazolam, and flunitrazepam have abuse potential. [112-119]The amount of information on the abuse liability of temazepam is limited, [120,121]although there are reports of its abuse. [122,123] 

Diazepam and other short-acting benzodiazepines function as reinforcers in animals, in sedative-abusing humans, in moderate drinkers, and perhaps in anxious individuals. Humans who find benzodiazepines to be reinforcing also report a profile of positive subjective effects (i.e., liking of drug effects). More testing of abuse liability must be conducted in sedative abusers and non-drug-abusing volunteers with midazolam and temazepam.

The epidemiology of benzodiazepine abuse is difficult to assess. The NHSDA provides only aggregate data regarding nonmedical use of benzodiazepines and barbiturates. The lifetime, past-year, and past-month incidence of nonmedical use of sedatives and tranquilizers reported by people >or= to 12 yr surveyed were 6.6%, 1.3%, and 0.6%, respectively. [70]The decreasing use of barbiturates suggests that most people abusing sedative-hypnotic agents abuse benzodiazepines instead. [98]Benzodiazepines likely to be abused are shorter acting and have a rapid onset, including all of those discussed in the previous section. [98] 

The people most likely to abuse benzodiazepines are those who also abuse other substances, i.e., polydrug abusers. [98,124-126]Some patients on methadone maintenance ingest a benzodiazepine after their methadone dose, reportedly to achieve a “high” state that is unachievable with either drug alone. [127]People who abuse opioids or who are undergoing opioid withdrawal also self-medicate with benzodiazepines to prevent or ameliorate some signs and symptoms of withdrawal (e.g., difficulty in sleeping, anxiety). [128]The relationship between alcoholism and abuse of benzodiazepines is far from clear. There is some evidence of greater use of benzodiazepines by this subgroup compared with the general population. [129,130]Strong evidence that the benzodiazepines are abused by this subgroup is lacking, although anxious alcoholics may take the drug for anxiolysis. [131] 

Finally, there has been some concern about the over-prescription of benzodiazepines for anxiety and sleep disorders. Most people who have prescriptions for benzodiazepines for anxiety or sleep disorders take them for <or= to 1 month. [132,133]Although a significant minority of patients take benzodiazepines for >or= to 1 yr (e.g., two patient surveys presented >or= to 12-month use rates of 15% and 25%[134,135]), their use patterns (e.g., lack of dose escalation) indicate that they are taking the drug for the prescribed reason, anxiolysis or insomnia. [136,137]Several reviews of whether benzodiazepines are overprescribed and abused in the general population have concluded that there is little evidence for either. [136-140] 

Barbiturates

Thiopental and methohexital are the two barbiturates most frequently used in anesthesia practice. Primates self-administer methohexital, demonstrating that it has reinforcing effects. [141]Thiopental has not been tested for its reinforcing efficacy. In drug discrimination procedures, barbiturates share discriminative stimulus effects with each other [142]and with benzodiazepines. [96]The abuse liability of thiopental and methohexital has not been studied in humans. Sedative abusers preferred pentobarbital, another shorter-acting barbiturate, to placebo. [143,144]Pentobarbital increased drug-liking and euphoria scores on the ARCI. [143-145]Non-drug-abusing volunteers, however, did not prefer pentobarbital to placebo; further, the drug did not generate positive subjective effects in them. [146] 

Although information on the abuse liability of thiopental and methohexital is limited, the information that has been presented shows that short-acting barbiturates have abuse potential. The abuse potential is greater in sedative abusers than in non-drug-abusing volunteers.

The actual incidence of barbiturate abuse is difficult to quantify. In the 1970s the incidence of barbiturate abuse was appreciable, [147]but epidemiologic surveys in various countries indicate that barbiturate abuse has since declined. [148-150]. For example, in the Monitoring of the Future survey, sponsored by the National Institute on Drug Abuse, the percent of 12th grade students who reported barbiturate use in the past month decreased by 64%, from 4.7% in 1975 to 1.7% in 1994. [150]This decline in barbiturate abuse appears to track the decline in prescription use of barbiturates, as newer and safer sedative-hypnotic agents have become available over the past two decades. [151] 

Propofol

Little research has been done on the abuse liability of propofol or its effects relative to other drug classes. In two animal studies, [92,152]subanesthetic and anesthetic doses of propofol induced a conditioned place preference; thus, propofol and its after-effects (recovery) had rewarding properties. No studies have examined the effects of propofol in drug abusers. Several studies with non-drug-abusing patients and healthy volunteers have yielded mixed results. One study showed no measurable subjective effects from propofol [153]; in several other studies, patients experienced positive mood states. [154-156]There was considerable intersubject variability in the volunteer studies, with some participants showing a positive response, others a neutral response, and others a negative response to the drug. [157,158]In a study that assessed the reinforcing effects of propofol (0.6 mg/kg, intravenously) with a preference procedure, 50% of the participants consistently chose the drug and reported liking it, whereas the other 50% consistently chose the placebo and reported either unpleasant acute or residual effects from propofol. [159]The investigators concluded that in some non-drug-abusing volunteers, propofol functions as a reinforcer.

It is difficult to categorize the abuse liability of propofol broadly in the absence of crucial studies that examine its reinforcing and discriminative stimulus effects in sedative abusers. Such studies are needed, given the positive profile of effects propofol produces in animals and in non-drug-abusing volunteers and patients. [91,152,154-156,159]Of the two published case reports regarding abuse of propofol, one involved an anesthesiologist. [160,161]Several factors mitigate against its use: its evanescent effect (unless the drug is delivered by an infusion pump) and the possibility of thrombophlebitis and pain on injection from its lipid carrier.

Ketamine

Ketamine, a dissociative anesthetic agent, is a noncompetitive antagonist at the N-methyl-D-aspartate receptor. Rodents and primates self-administer ketamine. [162-164]A compound closely related to ketamine, phencyclidine, was preferred in a conditioned place preference procedure in one study, [165]but in another study it produced a place aversion. [166]Ketamine shares discriminative stimulus effects with phencyclidine and other excitatory amino acid antagonists. [167,168]Ketamine has not been studied in drug abusers for its abuse liability, which is noteworthy, given the similarity of its pharmacologic profile with phencyclidine and hallucinogens. The subjective effects of ketamine have been studied in healthy volunteers and somewhat less systematically in patients, with consistent results. Reported effects include derealization, the perception that the environment around oneself is unreal; depersonalization, the belief that one no longer exists or exists outside of one's body; thought disorders; acute or residual visual hallucinations; and pleasant or unpleasant dreams. [169-173]Some individuals report flashback-like effects weeks or months after receiving ketamine.

Ketamine appears to have abuse liability based on its reinforcing and discriminative stimulus effects in animals. There were case reports of ketamine abuse in the 1980s [174,175]and, more recently, reports of ketamine use during all-night dancing or “rave” parties. [176]The incidence of ketamine abuse is low but may be increasing. Hallucinogen use, including use of the noncompetitive N-methyl-D-aspartate antagonist phencyclidine, increased from 1994 to 1995. [70] 

The volatile inhaled anesthetic agents, isoflurane, enflurane, halothane, desflurane, and sevoflurane, have not been tested for their reinforcing effects. Ether, however, functioned as a reinforcer in macaque monkeys. [177]In two drug discrimination studies in mice, halothane shared discriminative stimulus effects with the training drugs, ethanol [178]and pentobarbital. [179]This substitution suggests that at least some of the subjective effects of halothane, ethanol, and pentobarbital are mediated by the same neurochemical pathway. The subjective effects of subanesthetic concentrations of isoflurane have been studied in non-drug-abusing volunteers. [180]Isoflurane produced sedative-like effects, with substantial intersubject variability on drug liking.

Nitrous oxide, at concentrations ranging from 15-70%, functions as a reinforcer in primates. [181,182]In the studies that demonstrated reinforcement, different concentrations of nitrous oxide or oxygen (placebo) were delivered contingent on pressing a lever. The lever was pressed more when nitrous oxide was available than when oxygen (placebo) was available. In the only drug discrimination study involving nitrous oxide, it substituted for the training drug, ethylketocyclazocine, a selective [small kappa, Greek] opioid agonist, but not for morphine. [183]The investigators concluded that the discriminative stimulus effects of nitrous oxide are more [small kappa, Greek]-like than [micro sign]-like. This conclusion is consistent with that from some studies of nitrous oxide in humans, which reported dysphoric effects such as hallucinations, unpleasant memories, and confusion. [184,185]In other studies with the same subject population, however, pleasant effects were reported, [186-189]effects that are atypical of [small kappa, Greek] agonists in humans. The reinforcing effects of nitrous oxide in concentrations use in dental procedures (20-40%) have been studied in non-drug-abusing volunteers. Nitrous oxide did not function as a reinforcer in most individuals tested. [190-193]Nitrous oxide functioned as a reinforcer in moderate alcohol drinkers, however. [194]As with the benzodiazepine diazepam, the effects of nitrous oxide were more reinforcing in non-drug-abusing moderate drinkers than in lighter drinkers. [101,102] 

The primary population that abuses volatile inhaled anesthetic agents includes people who work in medical settings. [195]Accidental deaths have been documented with enflurane and halothane, and evidence indicated that the anesthetic agent was used for intoxication rather than life-ending purposes. [196-198]Causes of death have included airway obstruction, cardiac arrhythmias, and hepatitis. [199]An interesting case of abuse of isoflurane was reported during the war between the United States and Iraq in 1990 and 1991. Most of the United States troops were stationed in Saudi Arabia, where alcohol was not allowed because of Muslim conventions. An Air Force pharmacist was convicted in 1991 of not only distributing sleeping pills and amphetamines to enlisted men but also “committing conduct unbecoming an officer” by sniffing isoflurane in the presence of an enlisted man. He stated during the court martial trial that isoflurane became a common substitute for beer. During the war, one soldier died from a myocardial infarction, apparently after having used isoflurane.[double dagger] During “normal” conditions, a drug such as isoflurane may not be abused because other more familiar drugs are available. If the familiar drugs are no longer accessible, however, a drug that produces similar subjective effects may be substituted in its place. The most recent NHSDA estimated lifetime recreational use of volatile anesthetic agents to be 0.3% of the population aged 12 yr and older. [70] 

Nitrous oxide, although readily accessible in the operating room and the dental office, does not appear to be abused to any great extent by medical and dental personnel. [200]The drug is used recreationally via small cylindrical cartridges used to make whipping cream or by the procurement of large gas cylinders from a medical setting or industrial supply store. The gas is placed in balloons and people achieve a transient state of intoxication by breathing and rebreathing from the balloon. The most recent NHSDA estimated lifetime recreational use of nitrous oxide to be 2.2% of the population aged 12 yr and older. [70] 

Cocaine is used in intranasal surgery to provide anesthesia and, because of its vasoconstrictive effects, to decrease bleeding and shrink congested mucous membranes. [201]The abuse potential of cocaine has received as much, if not more, attention than opioids in the past decade. Cocaine functions as a reinforcer, as demonstrated in drug self-administration studies in rats [7]and rhesus monkeys. [202]The drug is thought to be a very strong reinforcer in that animals expend a tremendous amount of energy to “earn” cocaine [203]and, in certain situations, forgo food to the point of starvation. [204]Conditioned place preference procedures mirror the results of self-administration studies; cocaine consistently has rewarding properties. [205]Cocaine has discriminative stimulus effects in common with a number of other psychomotor stimulants and dopaminergic agonists, including d-amphetamine, methamphetamine, and apomorphine. [206,207]The reinforcing and subjective effects of cocaine have been studied in cocaine abusers. Intranasal, smoked, and intravenous cocaine are self-administered at a rate higher than placebo. [208,209]Subjective effects include increased euphoria scores on the ARCI and increased ratings for “good drug effects.”[210] 

Procaine and the other commonly used ester local anesthetic agents chloroprocaine and tetracaine bear a structural similarity to cocaine and are used frequently for topical and regional anesthesia. Procaine and chloroprocaine were positive reinforcers when delivered intravenously to rhesus monkeys. [211,212]Tetracaine was self-administered only occasionally and appears to be a marginal reinforcer. [213]Procaine also serves as a discriminative stimulus in rats, and cocaine has partially or fully substituted for procaine. [214,215]The similarity of discriminative stimulus effects with procaine and cocaine and their reinforcing effects in animals suggest that procaine has abuse potential in humans. In the only study to assess the abuse liability of procaine in cocaine abusers, some of the subjective effects of procaine were similar and others were dissimilar to those produced by cocaine. Procaine did not differ from placebo on ARCI or POMS scores, but three of four individuals identified procaine doses of 48 and 96 mg as cocaine. [216] 

Lidocaine, an amide local anesthetic drug, is used as a topical and regional agent and intravenously as an antiarrhythmic agent. An intranasal formulation is used to treat migraine headaches. [217]Intravenously administered lidocaine did not function as a reinforcer in primates. [214]In drug discrimination studies with rats, lidocaine shared discriminative stimulus effects with procaine, indicating that lidocaine might have some abuse potential. [214]In one study, intranasally administered lidocaine produced subjective responses similar to those produced by cocaine in stimulant abusers, [218]but the study has been criticized because it assessed only one subjective measure, that of “high.” In another study, neither euphoria scores from the ARCI nor liking ratings increased after intravenous administration of lidocaine in humans with histories of cocaine abuse. [219]Studies with lidocaine suggest that other local anesthetic agents from the amide class that are used for regional anesthesia, including bupivacaine and mepivacaine, may have low abuse liability.

Cocaine, regardless of route of administration, has clear abuse liability as measured in animal and human studies. The actual epidemiology of cocaine abuse mirrors its abuse liability. The lifetime, past-year, and past-month incidence of nonmedical use of cocaine reported by people >or= to 12 yr surveyed by the NHDSA were 10.3%, 1.7%, and 0.7%, respectively. [70]Clearly, there continues to be a significant public health problem with cocaine use in this country. The abuse of cocaine among medical personnel, however, appears to be no higher than in the general population. [220]Its common formulation as a cocaine suspension in medical settings and its limited availability decreases the likelihood of intrahospital abuse of this drug. Based on studies in animals, procaine and the other drugs of the ester class have a high abuse potential in theory, although actual abuse of these local anesthetic agents are low. One reason for the lack of procaine abuse may be its relatively short elimination half-life (7.7 min [221]vs. 40 min for cocaine [222]); a procaine abuser would have to administer the drug frequently to maintain an intoxicated state. [216] 

Clonidine has been studied for its reinforcing and discriminative stimulus effects. Primates pressed a lever to obtain clonidine at a higher rate than for saline. [223]In rats, clonidine had rewarding effects in conditioned place preference trials [224]and substituted for morphine in a drug discrimination study. [225]Yet the reinforcing efficacy of clonidine was not confirmed in opioid abusers who were undergoing methadone detoxification. [226]In oral form, clonidine is used to ameliorate withdrawal symptoms during heroin detoxification. [227,228]There have been only a few case reports of abuse of clonidine in the literature. [229]Patients in methadone maintenance programs reportedly used clonidine to boost the euphoric effects of diazepam, which is sometimes prescribed for symptoms of opioid withdrawal. Clonidine also may be used in times of illicit opioid scarcity. One high-risk group identified as likely to abuse clonidine is opioid-dependent women who also use cocaine. [230] 

Dexmedetomidine, another [small alpha, Greek]2selective agonist being evaluated as an adjuvant to general anesthesia, has not been tested for abuse liability in animals or humans. Healthy volunteers and patients reported sedative effects from the drug. [231,232]The patients also reported anxiolysis. [232] 

Several animal studies have tested anticholinergic agents for their abuse liability. Intravenously administered scopolamine functions as a reinforcer in rats. [233]Atropine and scopolamine substitute for one another in drug discrimination studies, but typically do not substitute for drugs from other classes, such as morphine, imipramine, amphetamine, and pentobarbital. [24,234,235]Numerous studies have characterized the subjective effects of anticholinergic drugs in healthy volunteers. In general, scopolamine tends to have more marked effects on mood than does atropine, most likely because of the greater penetration by scopolamine of the blood-brain barrier. [236]Supratherapeutic doses of atropine produce subjective effects, however. At lower doses, anticholinergic agents produce sedation, dizziness, dry mouth, and bradycardia; decreased arousal and energy; and blurred vision (because of dilation of the eye). [237-240]At higher doses, a central anticholinergic syndrome, which includes psychotomimetic effects (delirium, hallucinations, confusion, and restlessness), can occur. [241,242]There have been some case reports of anticholinergic abuse, primarily among patients taking antipsychotic medication. Anticholinergic agents such as trihexyphenidyl and benztropine are prescribed for amelioration of certain side effects of antipsychotic and antiparkinsonian agents (e.g., extrapyramidal side effects). Patients took supratherapeutic doses, presumably for the psychotomimetic and euphoric effects. [243-245]There are also case reports of people co-abusing anticholinergic agents with other drugs (including alcohol) to enhance the psychoactive effects of these substances. [246] 

Intravenous self-administration of H1-antagonists, including diphenhydramine, has been demonstrated in monkeys, [247]indicating that H (1) antihistamines have reinforcing effects in primates. In a drug discrimination study, diphenhydramine substituted for cocaine in three of four pigeons. [248]Abuse liability of H1antihistamines has not been shown in humans. In humans who abused sedatives, oral diphenhydramine in doses up to 400 mg did not increase drug-liking ratings or euphoria scores on the ARCI, relative to placebo. In contrast, ARCI scores for sedation and dysphoria were increased, and some self-reported side effects were restlessness, agitation, irritability, and vomiting. [249]In a sample of non-drug-abusing volunteers, 25 and 50 mg of orally administered tripelennamine were not chosen more often than placebo, indicating a lack of reinforcing effects. Study participants reported disliking its sedative effects. [250]There is one study of opioid abusers who reported euphoria after parenterally administered tripelennamine, [251]but there are only a few case reports in the medical literature on antihistamine abuse. [252,253]In the 1970s in several regions of the United States, tripelennamine was injected concurrently with pentazocine ("T's" and “blues”) to produce a heroin/cocaine-like high. [254]The abuse of “T's” and “blues” declined precipitously when the manufacturer of oral pentazocine added naloxone to the formulation. [254,255]In nondependent opioid abusers, this new formulation antagonized the effects of pentazocine, and in dependent opioid abusers, it precipitated opioid withdrawal. Were tripelennamine functioning as a reinforcer, as suggested by Lange and Jasinski, [251]abuse of tripelennamine alone should have continued in opioid abusers. It apparently has not. In summary, although animal studies suggest an abuse liability of antihistamines, studies in humans suggest, at best, equivocal abuse potential, and the epidemiologic data concerning actual abuse of antihistamines are more concordant with the human studies.

In baboons, ephedrine was self-administered, but in an unstable and erratic pattern. [256]In rats, ephedrine shared discriminative stimulus effects with amphetamine, methamphetamine, and cocaine. [257,258]In opioid abusers, subcutaneous ephedrine increased euphoria scores on the ARCI and drug-liking ratings, in the same manner as amphetamine and methamphetamine. [7]The reinforcing properties of ephedrine have been examined via a choice procedure in non-drug-abusing volunteers [259]; choice of ephedrine did not exceed chance levels. The profile of subjective effects predicted a low abuse potential for this drug in this subpopulation; it included increased visual analog scale ratings of “high,”“stimulated,” and “anxious” without increasing drug-liking ratings. Consistent with the abuse liability of this drug among opioid abusers, however, there are reports of ephedrine abuse in the literature. In Japan, over-the-counter cough syrup containing ephedrine is abused. [260]Some over-the-counter stimulants include ephedrine, and these drugs are abused by truck drivers and athletes, presumably for their stimulant effects. [261,262]The abuse of methamphetamine, which is synthesized in clandestine laboratories from ephedrine, [263]is increasing in the United States. [264,265] 

Droperidol and metoclopramide are dopamine antagonists. The primary side effects of droperidol include anxiogenesis, restlessness, and dizziness [266]; those of metoclopramide include restlessness, dry mouth, and drowsiness. [267]No abuse liability testing has been conducted on these older generation antiemetic agents, and there are no published cases of abuse of these drugs. The newer generation 5-hydroxytryptamine3antagonist, ondansetron induced neither a preference nor an aversion for the side of the chamber paired with the drug in the conditioned place preference paradigm. [268]The drug appears to be devoid of subjective effects at therapeutic and supratherapeutic doses. [269]No cases of ondansetron abuse have been published. Several studies in animals [270-273]and humans [274-276]have examined ondansetron and other 5-hydroxytryptamine3antagonists as potential treatments of opioid, alcohol, and stimulant abuse with mixed results. The amount of interest in the past several years directed at 5-hydroxytryptamine3compounds for disorders ranging from drug abuse to generalized anxiety disorder suggests that these compounds will continue to be studied for their therapeutic efficacy for some time. [277] 

(Table 1) categorizes the different anesthetic drugs based on their putative abuse liability. Full mu opioid agonists, benzodiazepines, and cocaine clearly have abuse potential and are abused. Other drugs such as mixed agonist-antagonist opioids and antihistamines have equivocal abuse potential, and epidemiologic data indicate a low incidence of abuse of these drugs. Finally, there are gaps in the literature in which the abuse potential of drugs such as ketamine, inhaled general anesthetic agents, and propofol would be worthy of testing, especially in drug abusers.

There are several issues remaining that are pertinent to testing of abuse liability and the epidemiology of abuse of anesthetic drugs. We raise these issues in question form. We believe such questions have clinical relevance to the anesthesia profession and deserve attention.

1. Why do some anesthetic drugs have abuse potential and others do not, i.e., what characteristics do abused drugs have that are missing from drugs that are rarely abused?

Abused drugs tend to induce a state of euphoria or extreme well-being. This euphoria is thought to be intimately related to the abuse potential of a drug. [278-281]Researchers have attempted to unravel the neurochemical underpinnings of the euphorigenic effects of such diverse drugs as cocaine, heroin, and diazepam. One theory is that these drugs, through different mechanisms of action, stimulate the mesolimbic dopamine system, [282,283]considered to be the reward center of the brain. [284]Drugs that are used recreationally by humans, including alcohol and tobacco, tend to stimulate this system. Lesioning this system decreases self-administration of drugs such as nicotine, cocaine, and heroin. [285,286]Another important reward mechanism appears to be activation of the endogenous opioid system. When this system is deactivated by opiate receptor antagonists, self-administration of opioids and other drugs (e.g., alcohol) can be decreased, suggesting that the reinforcing effects of some drugs are mediated by activation of opiate receptors. [287-291] 

2. Why does abuse potential of a drug not necessarily predict its abuse?

Morphine has high abuse potential but is not abused to a large extent within or outside the medical community. Barbiturates have abuse potential, but the incidence of their abuse has decreased over the past 20 yr. There are many reasons for the less-than-expected abuse of a drug. The abuse of drugs is not linked only to pharmacology but to a number of other factors. [292]One key factor is availability. The manufacture of barbiturates by pharmaceutical companies has decreased over the past 20 yr because of the decrease in prescription use of this drug class. Therefore, even though barbiturates may be diverted for nonprescribed uses, the drugs are scarce, creating less opportunity for abuse. Another factor is relative availability. Morphine, which has the potential for abuse, is not abused to a great extent because heroin is readily available and is preferred to morphine. If availability of heroin were to decrease markedly, it is likely that abuse of other full mu agonists such as morphine would increase greatly. Finally, such factors as drug pricing and fads are often involved in the apparent dissociation between the abuse liability of a drug and its actual abuse. [293] 

3. Why do patients who receive “drugs of abuse” peri- and post-operatively not subsequently seek out these drugs, i.e., why isn't there a greater incidence of iatrogenic addiction (drug seeking prompted by exposure to a drug for a medically valid reason)?

We speculate about the answer to this question. Drug abuse is “caused” by a number of variables, such as familial problems, peer pressure, mental and physical stressors, economic distress, personality disorders, genetic predisposition, and psychiatric morbidity. [294,295]Exposure to a drug is certainly a necessary precursor to drug abuse, but exposure alone is seldom sufficient. In almost all cases of drug abuse, other predisposing factors contribute to the abuse. In addition to not having these predisposing factors, most patients would have difficulty procuring opioids and barbiturates for illicit purposes. Finally, there is the issue of whether patients who receive psychotropic abusable drugs during or after surgery experience pleasant subjective effects. In a sterile medical setting or in the presence of the stress of pain during drug administration, it may not be possible to experience euphoric effects. In a recent laboratory study, pain decreased the intensity of the subjective effects of morphine in volunteers. [296]Pain also may antagonize pleasant effects of drugs given during the peri- and postoperative period in much the same way that pain antagonizes the respiratory depressant effects of opioids. [297]We contend, however, that patients with a history of drug abuse could be at increased risk for drug relapse by exposure to drugs of abuse perioperatively. Although definitive clinical studies have not been conducted in this area, our speculation is based on animal and human studies showing that passive administration of a drug, after the self-administration of that drug ceased, can “reinstate” responding to that drug. [298,299]Thus, a patient who had abused opioids in the past but is currently abstinent may crave or abuse opioids after receiving them during perioperative care. [300]Further research, retrospective and prospective, is needed on this interesting and clinically important issue.

4. Because drugs used in anesthesia practice can reinstate drug craving and perhaps drug seeking, should the perioperative drug treatment of patients who are active or recovering alcoholics and drug abusers differ from treatment for non-drug abusers?

Some procedures can be performed with regional anesthesia, obviating the need for sedation or analgesia with benzodiazepines, opioids, or inhaled and intravenous anesthetic agents. The advent of many different kinds of nonsteroidal antiinflammatory drugs also makes it possible to choose a nonopioid analgesic regimen with no known abuse potential. The first consideration of an anesthesiologist in choosing an analgesic, however, should be achieving an optimal degree of pain relief. If opioids are indicated, the drugs should be used in effective doses, because underdosing can lead to drug craving, increased anxiety, and, most obviously, pain. [301]The interested reader is referred to other articles that address the special needs of drug abusers during the perioperative period. [301-308] 

5. Why is drug abuse not epidemic among anesthesiology personnel who work daily with drugs with substantial abuse liability?

In most instances, mere availability of a drug is not in itself sufficient to lead to abuse of a drug. Other factors, including genetic predisposition, comorbidity, deprivation of other reinforcers, and stressors, are usually necessary. One could argue, however, that mere availability might play a role in drug abuse among anesthesiologists in that the percentage of anesthesiologists in drugs abuse programs for physicians is greater than the percentage of physicians who are anesthesiologists (12.1% vs. 3.9%, respectively, in one study [309]). This statistic suggests a greater propensity for drug abuse by anesthesiologists than by physicians in other medical subspecialties. The percentage of anesthesiologists who are drug (or alcohol) abusers has not been estimated, however, nor has the area of how incidence of drug abuse among anesthesiologists compares with that in the general population.

6. Should storage/accountability practices be changed for anesthetic drugs?

In many hospitals, drug accountability procedures have become more sophisticated (e.g., satellite pharmacies within an operating room) so that drug diversion for personal use is more difficult. The question is whether storage and accountability should be changed for the so-called benign drugs. We have some concerns about ketamine because it has rewarding effects in animals [162-164]; shares discriminative stimulus effects with another drug of abuse, phencyclidine [167,168]; and is known to be abused. [174-176]We also have some concerns about propofol, which has rewarding effects in animals. [92,152]Some non-drug-abusing volunteers chose it over placebo in a laboratory study, [159]and there have been cases of abuse of propofol, albeit isolated. [160,161]Aside from ketamine and propofol, however, there is little evidence of substantial abuse of antihistamines, anticholinergic agents, or lidocaine. The drugs that have substantial potential for abuse (e.g., opioids and cocaine) are stored, accounted for, and used appropriately in most hospitals.

The purpose of this article was to discuss the abuse liability and abuse of drugs that anesthesiologists use in their practice. One major point to be gleaned from this article is that the abuse liability and abuse of drugs used in anesthesia practice vary within and across drug classes. A second point is that the pharmacology (mechanism of action) of a drug is only one determinant of its abuse; other factors include characteristics of the person and the environment. Finally, several issues were raised to serve as catalysts for further collaborative research between the disciplines of anesthesiology and psychopharmacology.

[double dagger] Military Pharmacist Guilty of Sharing Drugs in Gulf. Miami Herald, August 2, 1991.

1.
Johanson CE, Balster RL: A summary of the results of a drug self-administration study using substitution procedures in rhesus monkeys. Bull Narc 1978; 30:43-54
2.
Brady JV: Animal models for assessing drugs of abuse. Neurosci Biobehav Rev 1991; 15:35-43
3.
Griffiths RR, Bigelow GE, Henningfield JE: Similarities in animal and human drug-taking behavior, Advances in Substance Abuse, Behavioral and Biological Research. Volume 1. Edited by Mello NK. Greenwich, JAI Press, 1980, pp 1-90
4.
McNair D, Lorr M, Droppleman L: Profile of Mood States. San Diego, Educational and Industrial Testing Service, 1972
5.
Fraser HF, van Horn GG, Martin WR, Wolbach AB, Isbell H: Methods for evaluating abuse liability. (A) “Attitude” of opiate addicts towards opiate-like drugs. (B) A short-term “direct” addiction test. J Pharmacol Exp Ther 1961; 133:371-87
6.
Haertzen CA: Development of scales based on patterns of drug effects, using the Addiction Research Center Inventory (ARCI). Psychol Rep 1966; 18:163-94
7.
Martin WR, Sloan JW, Sapira JD, Jasinski DR: Physiologic, subjective, and behavioral effects of amphetamine, methamphetamine, ephedrine, phenmetrazine, and methylphenidate in man. Clin Pharmacol Ther 1971; 12:245-58
8.
Holtzman SG: Discriminative stimulus effects of drugs: Relationship to potential for abuse, Modern Methods in Pharmacology. Volume 6. New York, Wiley-Liss, 1990, pp 193-210
9.
Mucha RF, Herz A: Motivational properties of kappa and mu opioid receptor agonists studied with place and taste preference conditioning. Psychopharmacology 1985; 86:274-80
10.
van Ree JM, Slangen JL, de Wied D: Intravenous self-administration of drugs in rats. J Pharmacol Exp Ther 1978; 204:547-57
11.
Woods JH, Young AM, Herling S: Classification of narcotics on the basis of their reinforcing, discriminative, and antagonist effects in rhesus monkeys. Fed Proc 1982; 41:221-7
12.
Miksic S, Shearman G, Lal H: Generalization study with some narcotic and nonnarcotic drugs in rats trained for morphine-saline discrimination. Psychopharmacology 1978; 60:103-4
13.
Gorodetzky CW, Martin WR: A comparison of fentanyl, droperidol, and morphine. Clin Pharmacol Ther 1966; 6:731-9
14.
Greenwald MK, June HL, Stitzer ML, Marco AP: Comparative clinical pharmacology of short-acting mu opioids in drug abusers. J Pharmacol Exp Ther 1996; 277:1228-36
15.
Baylon GJ, Kaplan HL, Gorthy L, Somer GR, Busto UE, Sellers EM: Comparative abuse liability of fentanyl and remifentanil, Problems of Drug Dependence 1995, NIDA Research Monograph Series 162. Edited by Harris LS. Rockville, U. S. Department of Health and Human Services, 1996, p 275
16.
Pfeiffer A, Brantl V, Herz A, Emrich HM: Psychotomimesis mediated by kappa opiate receptors. Science 1986; 233:774-6
17.
Hoehe M, Duka T, Doenicke A: Human studies on the mu opiate receptor agonist fentanyl: Neuroendocrine and behavioral responses. Psychoneuroendocrinology 1988; 13:397-408
18.
Zacny JP, Lichtor JL, Zaragoza JG, de Wit H: Subjective and behavioral responses to intravenous fentanyl in healthy volunteers. Psychopharmacology 1992; 107:319-26
19.
Zacny JP, McKay MA, Toledano AY, Marks S, Young CJ, Klock PA, Apfelbaum JL: The effects of a cold-water immersion stressor on the reinforcing and subjective effects of fentanyl in healthy volunteers. Drug Alcohol Depend 1996; 42:133-42
20.
Jaffe J: Drug addiction and drug abuse, Goodman and Gilman's The Pharmacological Basis of Therapeutics. 8th edition. Edited by Gilman AG, Rall TW, Nies AS, Taylor P. New York, Pergamon Press, 1990, pp 522-73
21.
Young AM, Swain HH, Woods JH: Comparison of opioid agonists in maintaining responding and in suppressing morphine withdrawal in rhesus monkeys. Psychopharmacology 1981; 74:329-35
22.
Hand TH, Stinus L, Le Moal M: Differential mechanisms in the acquisition and expression of heroin-induced place preference. Psychopharmacology 1989; 98:61-7
23.
Gaiardi M, Bartoletti M, Bacchi A, Gubellini C, Costa M, Babbini M: Role of repeated exposure to morphine in determining its affective properties: Place and taste conditioning studies in rats. Psychopharmacology 1991; 103:183-6
24.
Shannon HE, Holtzman SG: Further evaluation of the discriminative stimulus effects of morphine in the rat. J Pharmacol Exp Ther 1977; 201:55-66
25.
Schaefer GJ, Holtzman SG: Discriminative effects of morphine in the squirrel monkey. J Pharmacol Exp Ther 1977; 201:67-75
26.
Lamb RJ, Preston KL, Schindler CW, Meisch RA, Davis F, Katz JL, Henningfield JE, Goldberg SR: The reinforcing and subjective effects of morphine in post-addicts: A dose-response study. J Pharmacol Exp Ther 1991; 259:1165-73
27.
Mello NK, Mendelson JH, Kuehnle JC, Sellers MS: Operant analysis of human heroin self-administration and the effects of naltrexone. J Pharmacol Exp Ther 1981; 216:45-54
28.
Martin WR, Fraser HF: A comparative study of physiological and subjective effects of heroin and morphine administered intravenously in postaddicts. J Pharmacol Exp Ther 1961; 133:388-99
29.
Jasinski DR, Preston KL: Evaluation of tilidine for morphine-like subjective effects and euphoria. Drug Alcohol Depend 1986; 18:273-92
30.
Lasagna L, von Felsinger JM, Beecher JK: Drug-induced mood changes in man. JAMA 1955; 157:1006-20
31.
Bourke DL, Rosenberg M, Allen PD: Physostigmine: Effectiveness as an antagonist of respiratory depression and psychomotor effects caused by morphine or diazepam. Anesthesiology 1984; 61:523-8
32.
Zacny JP, Lichtor JL, Flemming D, Coalson DW, Thompson WK: A dose-response analysis of the subjective, psychomotor and physiological effects of intravenous morphine in healthy volunteers. J Pharmacol Exp Ther 1994; 268:1-9
33.
Bartoletti M, Gaiardi M, Gubellini C, Bacchi A, Babbini M: Time-dependent generalization of morphine stimulus properties to meperidine: Antagonism by naloxone. Pharmacol Biochem Behav 1989; 34:429-31
34.
Batterman RC: Clinical effectiveness and safety of a new synthetic analgesic drug, demerol. Arch Intern Med 1943; 71:345-56
35.
Zacny JP, Lichtor JL, Binstock W, Coalson DW, Cutter T, Flemming DC, Glosten B: Subjective, behavioral and physiological responses to intravenous meperidine in healthy volunteers. Psychopharmacology 1993; 111:306-14
36.
Preston KL, Bigelow GE, Bickel W, Liebson IA: Three-choice drug discrimination in opioid-dependent humans: Hydromorphone, naloxone and saline. J Pharmacol Exp Ther 1987; 243:1002-9
37.
Preston KL, Bigelow GE, Bickel WK, Liebson IA: Drug discrimination in human postaddicts: Agonist-antagonist opioids. J Pharmacol Exp Ther 1989; 250:184-96
38.
Preston KL, Jasinski DR: Abuse liability studies of opioid agonist-antagonists in humans. Drug Alcohol Depend 1991; 28:49-82
39.
Fraser HF, Martin WR, Wolbach AB, Isbell H: Addiction liability of an isoquinoline analgesic, 1-(p-chlorophenethyl)-2-methyl-6,7-dimethoxy-1,2,3,4,-tetrahydroisoquinoline. Clin Pharmacol Ther 1961; 2:287-99
40.
Kay DC, Gorodetzky CW, Martin WR: Comparative effects of codeine and morphine in man. J Pharmacol Exp Ther 1967; 156:101-6
41.
Jasinski DR, Martin WR: Assessment of the dependence-producing properties of dihydrocodeinone and codoxime. Clin Pharmacol Ther 1967; 8:266-70
42.
Lukas SE, Brady JV, Griffiths RR: Comparison of opioid self-injection and disruption of schedule-controlled performance in the baboon. J Pharmacol Exp Ther 1986; 238:924-31
43.
Lukas SE, Griffiths RR, Brady JV: Buprenorphine self-administration by the baboon: Comparison with other opioids, Problems of Drug Dependence 1982, NIDA Monograph Series No. 43. Edited by Harris LS. Rockville, National Institute on Drug Abuse, 1983, pp 178-83
44.
Yanagita T, Katoh S, Wakasa Y, Oinuma N: Dependence potential of buprenorphine studied in rhesus monkeys, Problems of Drug Dependence 1981, National Institute on Drug Abuse Research Monograph Series No. 41. Edited by Harris LS. Rockville, National Institute on Drug Abuse, 1982, pp 208-14
45.
Picker MJ, Benyas S, Horwitz JA, Thompson K, Mathewson C, Smith MA: Discriminative stimulus effects of butorphanol: Influence of training dose on the substitution patterns produced by mu, kappa, and delta opioid agonists. J Pharmacol Exp Ther 1996; 279:1130-41
46.
Gerak LR, France CP: Discriminative stimulus effects of nalbuphine in rhesus monkeys. J Pharmacol Exp Ther 1996; 276:523-31
47.
Shannon HE, Holtzman SG: Morphine training dose: A determinant of stimulus generalization to narcotic antagonists in the rat. Psychopharmacology 1979; 61:239-44
48.
Villareal JE: The effects of morphine agonists and antagonists on morphine-dependent rhesus monkeys, Agonists and Antagonist Actions of Narcotic Analgesic Drugs. Proceedings of the Symposium of the British Pharmacological Society. Edited by Kosterlitz HW, Collier HOJ, Villareal JE. London, Macmillan, 1972, pp 73-93
49.
Pircio AW, Gylys JA, Cavanagh RL, Buyniski JP, Bierwagen ME: The pharmacology of butorphanol, a 3,14-dihydroxymorphinan narcotic antagonist analgesic. Arch Intern Pharmacodyn Ther 1976; 220:231-57
50.
Woods JH, Gmerek DE: Substitution and primary dependence studies in animals. Drug Alcohol Depend 1985; 14:233-47
51.
Strain EC, Preston KL, Liebson IA, Bigelow GE: Precipitated withdrawal by pentazocine in methadone-maintained volunteers. J Pharmacol Exp Ther 1993; 267:624-34
52.
Preston KL, Bigelow GE, Liebson IA: Butorphanol-precipitated withdrawal in opioid-dependent human volunteers. J Pharmacol Exp Ther 1988; 246:441-8
53.
Preston KL, Bigelow GE, Liebson IA: Antagonist effects of nalbuphine in opioid-dependent human volunteers. J Pharmacol Exp Ther 1989; 248:929-37
54.
Preston KL, Bigelow GE: Drug discrimination assessment of agonist-antagonist opioids in humans: A three-choice saline-hydromorphone-butorphanol procedure. J Pharmacol Exp Ther 1994; 271:48-60
55.
Zacny JP, Lichtor JL, Thapar P, Coalson DW, Flemming D, Thompson W: Comparing the subjective, psychomotor, and physiological effects of intravenous butorphanol and morphine in healthy volunteers. J Pharmacol Exp Ther 1994; 270:579-88
56.
Zacny JP, Conley K, Marks S: Comparing the subjective, psychomotor, and physiological effects of intravenous nalbuphine and morphine in healthy volunteers. J Pharmacol Exp Ther 1997; 280:1159-69
57.
Zacny JP, Hill JL, Black ML, Sadeghi P: Comparing the subjective, psychomotor, and physiological effects of intravenous pentazocine and morphine in normal volunteers. J Pharmacol Exp Ther 1998;286:1197-207
58.
Brogden RN, Speight TM, Avery GS: Pentazocine: A review of its pharmacological properties, therapeutic efficacy and dependence liability. Drugs 1973; 5:6-91
59.
Zacny JP, Lichtor JL, Klafta JM, Alessi R, Apfelbaum JL: The effects of transnasal butorphanol on mood and psychomotor functioning in healthy volunteers. Anesth Analg 1996; 82:931-5
60.
Preston KL, Sullivan JT, Testa M, Jasinski DR: Psychopharmacology and abuse potential of transnasal butorphanol. Drug Alcohol Depend 1994; 35:159-67
61.
Walsh SL, Preston KL, Bigelow GE, Stitzer ML: Acute administration of buprenorphine in humans: Partial agonist and blockade effects. J Pharmacol Exp Ther 1995; 274:361-72
62.
Strain EC, Preston KL, Liebson IA, Bigelow GE: Opioid antagonist effects of dezocine in opioid-dependent humans. Clin Pharmacol Ther 1996; 60:206-17
63.
Young AM, Stephens KR, Hein DW, Woods JH: Reinforcing and discriminative stimulus properties of mixed agonist-antagonist opioids. J Pharmacol Exp Ther 1984; 229:118-26
64.
Schaefer GJ, Holtzman SG: Morphine-like stimulus effects in the monkey: Opioids with antagonist properties. Pharmacol Biochem Behav 1981; 14:241-5
65.
Jasinski DR, Preston KL: Assessment of dezocine for morphine-like subjective effects and miosis. Clin Pharmacol Ther 1985; 38:544-8
66.
Lee CR, McTavish D, Sorkin EM: Tramadol: A preliminary review of its pharmacodynamic and pharmacokinetic properties and therapeutic potential in acute and chronic pain states. Drugs 1993; 46:313-40
67.
Yanagita T: Drug dependence potential of 1-(m-methoxyphenyl)-2-(dimethylaminomethyl)-cyclohexan-1-ol hydrochloride (tramadol) tested in monkeys. Arzneimittelforschung 1978; 28:158-63
68.
Preston KL, Jasinski DR, Testa M: Abuse potential and pharmacological comparison of tramadol and morphine. Drug Alcohol Depend 1991; 27:7-17
69.
Cami J, Lamas X, Farre M: Acute effects of tramadol in methadone-maintained volunteers. Drugs 1994; 47(suppl):39-43
70.
Preliminary Estimates From the 1995 National Household Survey on Drug Abuse. Rockville, Substance Abuse and Mental Health Services Administration, 1996
71.
Reisine T, Pasternak G: Opioid analgesics and antagonists, Goodman and Gilman's The Pharmacological Basis of Therapeutics. 9th edition. Edited by Hardman JG, Limbird LE, Gilman AG. New York, McGraw-Hill, 1996, pp 521-56
72.
Oldendorf WH, Hyman S, Braun L, Oldendorf SZ: Blood-brain barrier: Penetration of morphine, codeine, heroin, and methadone after carotid injection. Science 1972; 178:984-6
73.
de Wit H, Dudish S, Ambre J: Subjective and behavioral effects of diazepam depend on its rate of onset. Psychopharmacology 1993; 112:324-30
74.
Hays LR, Stillner V, Littrell R: Fentanyl dependence associated with oral ingestion. Anesthesiology 1992; 77:819-20
75.
Silsby HD, Kruzich DJ, Hawkins MR: Fentanyl citrate abuse among health care professionals. Mil Med 1984; 149:227-8
76.
Marquardt KA, Tharratt RS: Inhalation abuse of fentanyl patch. J Toxicol Clin Toxicol 1994; 32:75-8
77.
DeSio JM, Bacon DR, Peer G, Lema MJ: Intravenous abuse of transdermal fentanyl therapy in a chronic pain patient. Anesthesiology 1993; 79:1139-41
78.
Silverstein JH, Silva DA, Iberti TJ: Opioid addiction in anesthesiology. Anesthesiology 1993; 79:354-75
79.
Poklis A: Fentanyl: A review for clinical and analytical toxicologists. J Toxicol Clin Toxicol 1995; 33:439-47
80.
Marquardt KA, Tharratt RS: Inhalation abuse of fentanyl patch. J Toxicol Clin Toxicol 1994; 32:75-8
81.
Austin RP: Diversion of butorphanol (letter). Am J Hosp Pharm 1983; 40:1306
82.
Smith SG, Davis MW: Nonmedical use of butorphanol and diphenhydramine (letter). JAMA 1984; 252:1010
83.
Evans WS, Bowen JN, Giordano FL, Clark B: A case of stadol dependence (letter). JAMA 1985; 253:2191-2
84.
Wagner JM, Cohen S: Fibrous myopathy from butorphanol injections. J Rheumatol 1991; 18:1934-5
85.
Chowdhury AN, Chowdhury S: Buprenorphine abuse: Report from India. Br J Addict 1990; 85:1349-50
86.
Singh RA, Mattoo SK, Malhotra A, Varma VK: Cases of buprenorphine abuse in India. Acta Psychiatr Scand 1992; 86:46-8
87.
Robinson GM, Dukes PD, Robinson BJ, Cooke RR, Mahoney GN: The misuse of buprenorphine and a buprenorphine-naloxone combination in Wellington, New Zealand. Drug Alcohol Depend 1993; 33:81-6
88.
San L, Torrens M, Castillo C, Porta M, de la Torre R: Consumption of buprenorphine and other drugs among heroin addicts under ambulatory treatment: Results from cross-sectional studies in 1988 and 1990. Addiction 1993; 88:1341-9
89.
Torrens M, San L, Cami J: Buprenorphine versus heroin dependence: Comparison of toxicologic and psychopathologic characteristics. Am J Psychiatry 1993; 150:822-4
90.
Falk JL, Tang M: Midazolam oral self-administration. Drug Alcohol Depend 1985; 15:151-63
91.
Griffiths RR, Lukas SE, Bradford LD, Brady JV, Snell JD: Self-injection of barbiturates and benzodiazepines in baboons. Psychopharmacology 1981; 75:101-9
92.
Pain L, Oberling P, Sandner G, Di Scala G: Effect of midazolam on propofol-induced positive affective state assessed by place conditioning in rats. Anesthesiology 1997; 87:935-43
93.
Woudenberg F, Slangen JL: Discriminative stimulus properties of midazolam: Comparison with other benzodiazepines. Psychopharmacology 1989; 97:466-70
94.
Sannerud CA, Ator NA: Drug discrimination analysis of midazolam under a three-level procedure: I. Dose-dependent differences in generalization and antagonism. J Pharmacol Exp Ther 1995; 272:100-11
95.
Spyracki G, Fibiger HC: A role for the mesolimbic dopamine system in the reinforcing properties of diazepam. Psychopharmacology 1988; 94:133-7
96.
Ator NA, Griffiths RR: Differential generalization to pentobarbital in rats trained to discriminate lorazepam, chlordiazepoxide, diazepam or triazolam. Psychopharmacology 1989; 98:20-30
97.
Evans SM, Johanson CE: Discriminative stimulus properties of midazolam in the pigeon. J Pharmacol Exp Ther 1989; 248:29-38
98.
Griffiths RR: Wolf B: Relative abuse liability of different benzodiazepines in drug abusers. J Clin Psychopharmacol 1990; 10:237-43
99.
de Wit H, Griffiths RR: Testing the abuse liability of anxiolytic and hypnotic drugs in humans. Drug Alcohol Depend 1991; 28:83-111
100.
Griffiths RR, McLeod DR, Bigelow GE, Liebson IA, Roache JD, Nowowieski P: Comparison of diazepam and oxazepam: Preference, liking, and extent of abuse. J Pharmacol Exp Ther 1984; 229:501-8
101.
de Wit H, Pierri J, Johanson CE: Reinforcing and subjective effects of diazepam in nondrug-abusing volunteers. Pharmacol Biochem Behav 1989; 33:205-13
102.
Evans SM, Griffiths RR, de Wit H: Preference for diazepam, but not buspirone, in moderate drinkers. Psychopharmacology 1996; 123:154-63
103.
Chutuape MA, de Wit H: Preferences for ethanol and diazepam in anxious individuals: An evaluation of the self-medication hypothesis. Psychopharmacology 1995; 121:91-103
104.
Roache JD, Stanley MA, Creson DL, Meisch RA, Shah NN: Diazepam reinforcement in anxious patients. Exp Clin Psychopharmacol 1996; 4:308-14
105.
de Wit H, Uhlenhuth EH, Hedeker D, McCracken SG, Johanson CE: Lack of preference for diazepam in anxious volunteers. Arch Gen Psychiatry 1986; 43:533-41
106.
McCracken SG, de Wit H, Uhlenhuth EH, Johanson CE: Preference for diazepam in anxious adults. J Clin Psychopharmacol 1990; 10:190-6
107.
Short TG, Galletly DC: Acute tolerance from benzodiazepine night sedation. Anaesthesia 1991; 46:929-31
108.
Franssen C, Hans P, Brichant JF, Noirot D, Lamy M: Comparison between alprazolam and hydroxyzine for oral premedication. Can J Anaesth 1993; 40:13-7
109.
Kontinen VK, Maunuksela EL, Sarvela J: Premedication with sublingual triazolam compared with oral diazepam. Can J Anaesth 1993; 40:829-34
110.
Bailie R, Christmas L, Price N, Restall J, Simpson P, Wesnes K: Effects of temazepam premedication on cognitive recovery following alfentanil-propofol anaesthesia. Br J Anaesth 1989; 63:68-75
111.
Mattila MA, Salmela J, Vaananen A, Kylmamaa T: Midazolam vs lorazepam and placebo as hypnotic premedication before surgery: A controlled, double-blind study. Drugs Exp Clin Res 1985; 11:841-4
112.
Griffiths RR, Lamb RJ, Sannerud CA, Ator NA, Brady JV: Self-injection of barbiturates, benzodiazepines and other sedative-anxiolytics in baboons. Psychopharmacology 1991; 103:154-61
113.
Funderburk FR, Griffiths RR, McLeod DR, Bigelow GE, Mackenzie A, Liebson IA, Nemeth-Coslett R: Relative abuse liability of lorazepam and diazepam: An evaluation in recreational drug users. Drug Alcohol Depend 1988; 22:215-22
114.
Troisi JR, Critchfield TS, Griffiths RR: Buspirone and lorazepam abuse liability in humans: Behavioral effects, subjective effects and choice. Behav Pharmacol 1993; 4:217-30
115.
Busto U, Kaplan HL, Zawertailo L, Sellers EM: Pharmacologic effects and abuse liability of bretazenil, diazepam, and alprazolam in humans. Clin Pharmacol Ther 1994; 55:451-63
116.
Mumford GK, Rush CR, Griffiths RR: Abecamil and alprazolam in humans: Behavioral, subjective and reinforcing effects. J Pharmacol Exp Ther 1995; 272:570-80
117.
Evans SM, Funderburk FR, Griffiths RR: Zolpidem and triazolam in humans: Behavioral and subjective effects and abuse liability. J Pharmacol Exp Ther 1990; 255:1246-55
118.
Farre M, Teran MT, Cami J: A comparison of the acute behavioral effects of flunitrazepam and triazolam in healthy volunteers. Psychopharmacology 1996; 125:1-12
119.
Bond A, Seijas D, Dawling S, Lader M: Systemic absorption and abuse liability of snorted flunitrazepam. Addiction 1994; 89:821-30
120.
de la Garza R, Johanson CE: Discriminative stimulus properties of intragastrically administered d-amphetamine and pentobarbital in rhesus monkeys. J Pharmacol Exp Ther 1987; 243:955-62
121.
Rush CR, Higgins ST, Hughes JR, Bickel WK: A comparison of the acute behavioral effects of triazolam and temazepam in normal volunteers. Psychopharmacology 1993; 112:407-14
122.
Green ST, Goldberg DJ, Christie PR, Frischer M, Thomson A, Carr SV, Taylor A: Female streetworker-prostitutes in Glasgow: A descriptive study of their lifestyle. AIDS Care 1993; 5:321-35
123.
Ralston GE, Taylor JA: Temazepam abuse (letter). Addiction 1993; 88:423
124.
O'Brien CP: Drug addiction and drug abuse, Goodman and Gilman's The Pharmacological Basis of Therapeutics. 9th edition. Edited by Hardman JG, Limbird LE, Gilman AG. New York, McGraw-Hill, 1996, pp 557-78
125.
Cole JO, Chiarello RJ: The benzodiazepines as drugs of abuse. J Psychiatr Res 1990; 24(suppl 2):135-44
126.
San L, Tato J, Torrens M, Castillo C, Farre M, Cami J: Flunitrazepam consumption among heroin addicts admitted for in-patient detoxification. Drug Alcohol Depend 1993; 32:281-6
127.
Stitzer ML, Griffiths RR, McLellan AT, Grabowski J, Hawthorne JW: Diazepam use among methadone maintenance patients: Patterns and dosages. Drug Alcohol Depend 1981; 8:189-99
128.
Perera KM, Jenner FA: Some characteristics distinguishing high and low dose users of benzodiazepines. Br J Addict 1987; 82:1329-34
129.
Busto U, Simpkins J, Sellers EM, Sisson B, Segal R: Objective determination of benzodiazepine use and abuse in alcoholics. Br J Addict 1983; 78:429-35
130.
Wiseman SM, Spencer-Peet J: Prescribing for alcoholics: A survey of drugs taken prior to admission to an alcoholism unit. Practitioner 1985; 229:88-9
131.
Ciraulo DA, Sands BF, Shader RI: Critical review of liability for benzodiazepine abuse among alcoholics. Am J Psychiatry 1988; 145:1501-6
132.
Woods JH, Katz JL, Winger GD: Abuse liability of benzodiazepines. Pharmacol Rev 1987; 39:251-413
133.
Woods JH, Katz JL, Winger G: Benzodiazepines: Use, abuse, and consequences. Pharmacol Rev 1992; 44:151-347
134.
Balter, MB, Uhlenhuth EH: New epidemiologic findings about insomnia and its treatment. J Clin Psychiatry 1992; 53(suppl 12):34-9
135.
Mellinger GD, Balter MB, Uhlenhuth EH: Prevalence and correlates of the long-term regular use of anxiolytics. JAMA 1984; 251:375-9
136.
Woods JH, Katz JL, Winger G: Abuse and therapeutic use of benzodiazepines and benzodiazepine-like drugs, Psychopharmacology: The Fourth Generation of Progress. Edited by Bloom FE, Kupfer DJ. New York, Raven Press, 1995, pp 1777-91
137.
Uhlenhuth EH, de Wit H, Balter MB, Johanson CE, Mellinger GD: Risks and benefits of long-term benzodiazepine use. J Clin Psychopharmacol 1988; 8:161-7
138.
Allgulander C: Addiction on prescribed sedative-hypnotics. Human Psychopharmacol 1996; 11(suppl 1):S49-54
139.
Greenblatt DJ, Shader RI, Abernethy DR: Drug therapy: Current status of benzodiazepines. N Engl J Med 1983; 309:410-6
140.
Rickels K: Are benzodiazepines overused and abused? Br J Clin Pharmacol 1981; 11(suppl 1): 71S-83S
141.
Winger G, Stitzer ML, Woods JH: Barbiturate-reinforced responding in rhesus monkeys: Comparisons of drugs with different durations of action. J Pharmacol Exp Ther 1975; 195:505-14
142.
Winger G, Herling S: Discriminative stimulus effects of pentobarbital in rhesus monkeys: tests of stimulus generalization and duration of action. Psychopharmacology 1982; 76:172-6
143.
Griffiths RR, Bigelow GE, Liebson I, Kaliszak JE: Drug preference in humans: Double-blind choice comparison of pentobarbital, diazepam, and placebo. J Pharmacol Exp Ther 1980; 215:649-61
144.
Griffiths RR, Troisi JR, Silverman K, Mumford GK: Multiple-choice procedure: An efficient approach for investigating drug reinforcement in humans. Behav Pharmacol 1993; 4:3-13
145.
Sullivan JT, Jasinski DR, Johnson RE: Single-dose pharmacodynamics of diazepam and pentobarbital in substance abusers. Clin Pharmacol Ther 1993; 54:645-53
146.
Cole-Harding S, de Wit H: Self-administration of pentobarbital in light and moderate alcohol drinkers. Pharmacol Biochem Behav 1992; 43:563-9
147.
Ray O: Drugs, Society, and Human Behavior. 3rd edition. St. Louis, Mosby, 1983, pp 317-9
148.
Patterson EW, Myers G, Gallant DM: Patterns of substance abuse on a college campus: A 14-year comparison study. Am J Drug Alcohol Abuse 1988; 14:237-46
149.
Obafunwa JO, Busuttil A: Deaths from substance overdose in the Lothian and Borders region of Scotland (1983-1991). Human Exp Toxicol 1994; 13:401-6
150.
National Institute on Drug Abuse: National Survey Results on Drug Use from the Monitoring the Future Study. Volume 1. Rockville, U. S. Department of Health and Human Services, 1995, p 269
151.
Rall TW: Hypnotics and sedatives: Ethanol, Goodman and Gilman's The Pharmacological Basis of Therapeutics. 8th edition. Edited by Gilman AG, Rall TW, Nies AS, Taylor P. New York, Pergamon Press, 1990, p 345-82
152.
Pain L, Oberling P, Sandner G, Di Scala G: Effect of propofol on affective state as assessed by place conditioning paradigm in rats. Anesthesiology 1996; 85:121-8
153.
Whitehead C, Sanders LD, Oldroyd G, Haynes TK, Marshall RW, Rosen M, Robinson JO: The subjective effects of flow-dose propofol: A double-blind study to evaluate dimensions of sedation and consciousness with low-dose propofol. Anaesthesia 1994; 49:490-6
154.
Oxorn D, Orser B, Ferris LE, Harrington E: Propofol and thiopental anesthesia: A comparison of the incidence of dreams and perioperative mood alterations. Anesth Analg 1994; 79:553-7
155.
Monedero RP, Panadero AS, Garcia FP, Carrera JH, Catala JC, Arroyo JL: Comparative study between thiopental and propofol in short-duration anesthesia. Rev Esp Anestesiol Reanim 1991; 38:153-5
156.
Schaer H, Prochacka K: Recovery, amnesia and affective state following propofol in comparison with thiopental. Anaesthesist 1990; 39:306-12
157.
Zacny JP, Lichtor JL, Coalson DW, Finn RS, Uitvlugt AM, Glosten B, Flemming DC, Apfelbaum JL: Subjective and psychomotor effects of subanesthetic doses of propofol in healthy volunteers. Anesthesiology 1992; 76:696-702
158.
Zacny JP, Lichtor JL, Zaragoza JG, Coalson DW, Uitvlugt AM, Flemming DC, Binstock WB, Cutter T, Apfelbaum JL: Assessing the behavioral effects and abuse potential of propofol bolus injections in healthy volunteers. Drug Alcohol Depend 1993; 32:45-57
159.
Zacny JP, Lichtor JL, Thompson W, Apfelbaum JL: Propofol at a subanesthetic dose may have abuse potential in healthy volunteers. Anesth Analg 1993; 77:544-52
160.
Follette JW, Farley WJ: Anesthesiologist addicted to propofol. Anesthesiology 1992; 77:817-8
161.
Gundel H, Kuhs H: A case report on a 9-day-long abuse of propofol. Anasthesiol Intensivmed Notfallmed Schmerzther 1992; 27:181-2
162.
Moreton JE, Meisch RA, Stark L, Thompson T: Ketamine self-administration by the rhesus monkey. J Pharmacol Exp Ther 1977; 203:303-9
163.
Marquis KL, Gussio RP, Webb MG, Moreton JE: Cortical EEG changes during the self-administration of phencyclinoids. Neuropharmacology 1989; 28:1193-8
164.
Winger G, Palmer RK, Woods JH: Drug-reinforced responding: Rapid determination of dose-response functions. Drug Alcohol Depend 1989; 24:135-42
165.
Marglin SH, Milano WC, Mattie ME, Reid LD: PCP and conditioned place preferences. Pharmacol Biochem Behav 1989; 33:281-3
166.
Barr GA, Paredes W, Bridger WH: Place conditioning with morphine and phencyclidine: Dose dependent effects. Life Sci 1985; 36:363-8
167.
Beardsley PM, Anthony EW, Lopez OT: Phencyclidine established as a discriminative stimulus using ethanol as a reinforcer. Behav Pharmacol 1992; 3:497-505
168.
Butelman ER, France CP, Woods JH: The discriminative stimulus effects of MK-801 in pigeons. Behav Pharmacol 1991; 2:79-86
169.
Fine J, Finestone SC: Sensory disturbances following ketamine anesthesia: Recurrent hallucinations. Anesth Analg 1973; 52:428-30
170.
Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB Jr, Charney DS: Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans: Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 1994; 51:199-214
171.
Ghoneim MM, Hinrichs JV, Mewaldt SP, Petersen RC: Ketamine: Behavioral effects of subanesthetic doses. J Clin Psychopharmacol 1985; 5:70-7
172.
Malhotra AK, Pinals DA, Weingartner H, Sirocco K, Missar CD, Pickar D, Breier A: NMDA receptor function and human cognition: The effects of ketamine in healthy volunteers. Neuropsychopharmacology 1996; 14:301-7
173.
Moretti RJ, Hassan SZ, Goodman LI, Meltzer HY: Comparison of ketamine and thiopental in healthy volunteers: Effects on mental status, mood, and personality. Anesth Analg 1984; 63:1087-96
174.
Ahmed SN, Petchkovsky L: Abuse of ketamine (letter). Br J Psychiatry 1980; 137:303
175.
Felser JM, Orban DJ: Dystonic reaction after ketamine abuse. Ann Emerg Med 1982; 11:673-5
176.
Dotson JW, Ackerman DL, West LJ: Ketamine abuse. J Drug Issues 1995; 25:751-7
177.
Yanagita T, Takahashi S, Ishida K, Funamoto H: Voluntary inhalation of volatile anesthetics and organic solvents by monkeys. Jap J Clin Pharmacol 1970; 1:13-6
178.
Rees DC, Knisely JS, Breen TJ, Balster RL: Toluene, halothane, 1,1,1-trichloroethane and oxazepam produce ethanol-like discriminative stimulus effects in mice. J Pharmacol Exp Ther 1987; 243:931-7
179.
Rees DC, Knisely JS, Balster RL, Jordan S, Breen TJ: Pentobarbital-like discriminative stimulus properties of halothane, 1,1,1-trichloroethane, isoamyl nitrite, flurothyl and oxazepam in mice. J Pharmacol Exp Ther 1987; 241:507-15
180.
Zacny JP, Sparacino G, Hoffman PM, Martin R, Lichtor JL: The subjective, behavioral and cognitive effects of subanesthetic concentrations of isoflurane and nitrous oxide in healthy volunteers. Psychopharmacology 1994; 114:409-16
181.
Wood RW, Grubman J, Weiss B: Nitrous oxide self-administration by the squirrel monkey. J Pharmacol Exp Ther 1977; 202:491-9
182.
Grubman J, Woods JH: Schedule-controlled behavior maintained by nitrous oxide delivery in the rhesus monkey, Proceedings of a Symposium on Drugs as Reinforcers. International Congress Series 620. Tokyo, Excerpta Medica, 1981, pp 259-74
183.
Hynes MD, Hymson DL: Nitrous oxide generalizes to a discriminative stimulus produced by ethylketocyclazocine but not morphine. Eur J Pharmacol 1984; 105:155-9
184.
Block RI, Ghoneim MM, Hinrichs JV, Kumar V, Pathak D: Effects of a subanesthetic concentration of nitrous oxide on memory and subjective experience: Influence of assessment procedures and types of stimuli. Hum Psychopharmacol 1988; 3:257-65
185.
Mahoney FC, Moore PA, Baker EL, Letz R: Experimental nitrous oxide exposure as a model system for evaluating neurobehavioral tests. Toxicology 1988; 49:449-57
186.
Atkinson RM, Green JD, Chenoweth DE, Atkinson JH: Subjective effects of nitrous oxide: Cognitive, emotional, perceptual and transcendental experiences. J Psychedelic Drugs 1979; 11:317-30
187.
Block RI, Ghoneim MM, Kumar V, Pathak D: Psychedelic effects of a subanesthetic concentration of nitrous oxide. Anesth Prog 1990; 37:271-6
188.
Garfield JM, Garfield FB, Sampson J: Effects of nitrous oxide on decision-strategy and sustained attention. Psychopharmacologia 1975; 42:5-10
189.
Henrie JR, Parkhouse J, Bickford RG: Alteration of human consciousness by nitrous oxide as assessed by electroencephalography and psychological tests. Anesthesiology 1961; 22:247-59
190.
Dohrn CS, Lichtor JL, Coalson DW, Flemming D, Zacny JP: Reinforcing effects of extended inhalation of a low nitrous oxide concentration in humans. Pharmacol Biochem Behav 1993; 46:927-32
191.
Dohrn CS, Lichtor JL, Coalson DW, Uitvlugt A, de Wit H, Zacny JP: Reinforcing effects of extended inhalation of nitrous oxide in humans. Drug Alcohol Depend 1993; 31:265-80
192.
Yajnik S, Thapar P, Lichtor JL, Patterson T, Zacny JP: Effects of marijuana history on the subjective, psychomotor, and reinforcing effects of nitrous oxide in humans. Drug Alcohol Depend 1994; 36:227-36
193.
Zacny JP, Lichtor JL, Coalson DW, Alessi R, Goldsher G, Young CJ, Apfelbaum JL, Conley KM: Examining the subjective, psychomotor-impairing, and reinforcing effects of nitrous oxide in healthy volunteers: A dose-response analysis. Behav Pharmacol 1996; 7:194-9
194.
Cho AM, Coalson DW, Klock PA, Klafta JM, Marks S, Toledano AY, Apfelbaum JL, Zacny JP: The effects of alcohol history on the reinforcing, subjective and psychomotor effects of nitrous oxide in healthy volunteers. Drug Alcohol Depend 1997; 45:63-70
195.
Chalmers EM: Volatile substance abuse. Med J Aust 1991; 154:269-74
196.
Jacob B, Heller C, Daldrup T, Burrig KF, Barz J, Bonte W: Fatal accidental enflurane intoxication. J Forensic Sci 1989; 34:1408-12
197.
Hiroki T, Teruuchi T, Kurosa T, Kagiwara M: On the fatal cases of poisoning due to abuse of fluothane. Japan J Legal Med 1973; 27:243-7
198.
Kaplan HG, Bakken J, Quadracci L, Schubach W: Hepatitis caused by halothane sniffing. Ann Intern Med 1979; 90:797-8
199.
Yamashita M, Matsuki A, Oyama T: Illicit use of modern volatile anaesthetics. Can Anaesth Soc J 1984; 31:76-9
200.
Gillman MA: Nitrous oxide abuse in perspective. Clin Neuropharmacol 1992; 15:297-306
201.
Donlon JV: Anesthesia and eye, ear, nose, and throat surgery, Anesthesia. 4th edition. Edited by Miller RD. New York, Churchill Livingstone, 1994, pp 2175-96
202.
Woods JH, Schuster CR: Reinforcement properties of morphine, cocaine, and SPA as a function of unit dose. Int J Addict 1968; 3:231-7
203.
Yanagita T: An experimental framework for evaluation of dependence liability of various types of drugs in monkeys. Bull Narc 1973; 25:57-64
204.
Aigner TG, Balster RL: Choice behavior in rhesus monkeys: Cocaine versus food. Science 1978; 201:534-5
205.
Mucha RF, Van der Kooy D, O'Shaughnessy M, Bucenieks P: Drug reinforcement studied by the use of place conditioning in rat. Brain Res 1982; 243:91-105
206.
Woods JH, Winger GD, France CP: Reinforcing and discriminative stimulus effects of cocaine: Analysis of pharmacological mechanisms, Cocaine: Clinical and Biobehavioral Aspects. Edited by Fisher S, Raskin A, Uhlenhuth EH. New York, Oxford University Press, 1987, pp 21-65
207.
Colpaert FC, Niemegeers CJE, Janssen PAJ: Discriminative stimulus properties of cocaine: Neuropharmacological characteristics as derived from stimulus generalization experiments. Pharmacol Biochem Behav 1979; 10:535-46
208.
Fischman MW: The behavioral pharmacology of cocaine in humans, Cocaine: Pharmacology, Effects and Treatment of Abuse. National Institute of Drug Abuse Research Monograph No. 50. Edited by Grabowski J. Rockville, U. S. Department of Health and Human Services, 1984, pp 1543-53
209.
Foltin RW, Fischman MW: Self-administration of cocaine by humans: Choice between smoked and intravenous cocaine. J Pharmacol Exp Ther 1992; 261:841-9
210.
Fischman MW, Foltin RW: Self-administration of cocaine by humans: A laboratory perspective. Ciba Found Symp 1992; 166:165-73
211.
Ford RD, Balster RL: Reinforcing properties of intravenous procaine in rhesus monkeys. Pharmacol Biochem Behav 1977; 6:289-96
212.
Johanson CE: The reinforcing properties of procaine, choloroprocaine, and proparacaine in rhesus monkeys. Psychopharmacology 1980; 67:189-94
213.
Woolverton WL, Balster RL: Reinforcing properties of some local anesthetics in rhesus monkeys. Pharmacol Biochem Behav 1979; 11:669-72
214.
Woolverton WL, Balster RL: Behavioral pharmacology of local anesthetics: Reinforcing and discriminative stimulus effects. Pharmacol Biochem Behav 1982; 16:491-500
215.
Silverman PB, Schultz KA: Comparison of cocaine and procaine discriminative stimuli. Drug Dev Res 1989; 16:427-33
216.
Fischman MW, Schuster CR, Rajfer S: A comparison of the subjective and cardiovascular effects of cocaine and procaine in humans. Pharmacol Biochem Behav 1983; 18:711-6
217.
Kudrow L, Kudrow DB, Sandweiss JH: Rapid and sustained relief of migraine attacks with intranasal lidocaine: Preliminary findings. Headache 1995; 35:79-82
218.
Van Dyke CP, Jatlow P, Ungerer J, Barash P, Byck R: Cocaine and lidocaine have similar psychological effects after intranasal application. Life Sci 1979; 24:271-4
219.
Fischman MW, Schuster CR, Hatano Y: A comparison of the subjective and cardiovascular effects of cocaine and lidocaine in humans. Pharmacol Biochem Behav 1983; 18:123-7
220.
Lutsky I, Hopwood M, Abram SE, Cerletty JM, Hoffman RG, Kampine JP: Use of psychoactive substances in three medical specialties: Anaesthesia, medicine and surgery. Can J Anaesth 1994; 41:561-7
221.
Seifen, AB, Ferrari AA, Seifen EE, Thompson DS, Chapman J: Pharmacokinetics of intravenous procaine infusion in humans. Anesth Analg 1979; 58:382-6
222.
Javaid JI, Fischman MW, Schuster CR, Dekirmenjiian H, Davis JM: Cocaine plasma concentration: Relation to physiological and subjective effects in humans. Science 1978; 202:227-8
223.
Woolverton WL, Wessinger WD, Balster RL: Reinforcing properties of clonidine in rhesus monkeys. Psychopharmacology 1982; 77:17-23
224.
Asin KE, Wirtshafter D: Clonidine produces a conditioned place preference in rats. Psychopharmacology 1985; 85:383-5
225.
Krimmer EC, McGuire MS, Barry H III: Effects of the training dose on generalization of morphine stimulus to clonidine. Pharmacol Biochem Behav 1984; 20:669-73
226.
Preston KL, Bigelow GE, Liebson IA: Self-administration of clonidine, oxazepam and hydromorphone by patients undergoing methadone detoxification. Clin Pharmacol Ther 1985; 38:219-27
227.
Gold MS, Pottash AC, Sweeney DR, Kleber HD: Opiate withdrawal using clonidine: a safe, effective and rapid nonopiate treatment. JAMA 1980; 243:343-6
228.
Jasinski DR, Johnson RE, Kocher TR: Clonidine in morphine withdrawal: Differential effects on signs and symptoms. Arch Gen Psychiatry 1985; 42:1063-6
229.
Schaut J, Schnoll SH: Four cases of clonidine abuse. Am J Psychiatry 1983; 140:1625-7
230.
Anderson F, Paluzzi P, Lee J, Huggins G, Svikis D: Illicit use of clonidine in opiate-abusing pregnant women. Obstet Gynecol 1997; 90:790-4
231.
Mattila MJ, Mattila ME, Olkkola KT, Scheinin H: Effect of dexmedetomidine and midazolam on human performance and mood. Eur J Clin Pharmacol 1991; 41:217-23
232.
Aho M, Scheinin M, Lehtinen AM, Erkola O, Vuorinen J, Korttila K: Intramuscularly administered dexmedetomidine attenuates hemodynamic dynamic and stress hormone responses to gynecologic laparoscopy. Anesth Analg 1992; 75:932-9
233.
Glick SD, Guido RA: Scopolamine self-administration: Cholinergic involvement in reward mechanisms. Life Sci 1982; 31:909-13
234.
Overton DA: Discriminable effects of antimuscarinics: Dose response and substitution test studies. Pharmacol Biochem Behav 1977; 6:659-66
235.
Zhang L, Barrett JE: Imipramine as a discriminative stimulus. J Pharmacol Exp Ther 1991; 259:1088-93
236.
Brown JH: Atropine, scopolamine, and related antimuscarinic drugs, Goodman and Gilman's The Pharmacological Basis of Therapeutics. 8th edition. Edited by Gilman AG, Rall TW, Nies AS, Taylor P. New York, Pergamon Press, 1990, pp 150-65
237.
Parrott AC: The effects of transdermal scopolamine and four dose levels of oral scopolamine (0.15, 0.3, 0.6, and 1.2 mg) upon psychological performance. Psychopharmacology 1986; 89:347-54
238.
Nuotto E: Psychomotor, physiological and cognitive effects of scopolamine and ephedrine in healthy man. Eur J Clin Pharmacol 1983; 24:603-9
239.
Penetar DM, Henningfield JE: Psychoactivity of atropine in normal volunteers. Pharmacol Biochem Behav 1986; 24:1111-3
240.
Ghoneim MM, Mewaldt SP: Studies on human memory: The interactions of diazepam, scopolamine, and physostigmine. Psychopharmacology 1977; 52:1-6
241.
Sannita WG, Maggi L, Rosadini G: Effects of scopolamine (0.25-0.75 mg i.m.) on the quantitative EEG and the neuropsychological status of healthy volunteers. Neuropsychobiology 1987; 17:199-205
242.
Longo VG: Behavioral and electroencephalographic effects of atropine and related compounds. Pharmacol Rev 1966; 18:965-96
243.
Fisch RZ: Trihexyphenidyl abuse: Therapeutic implications for negative symptoms of schizophrenia? Acta Psychiatr Scand 1987; 75:91-4
244.
Smith JM: Abuse of antiparkinson drugs: A review of the literature. J Clin Psychiatry 1980; 41:351-4
245.
Marken PA, Stoner SC, Bunker MT: Anticholinergic drug abuse and misuse: Epidemiology and therapeutic implications. CNS Drugs 1996; 5:190-9
246.
Dilsaver SC: Antimuscarinic agents as substances of abuse: A review. J Clin Psychopharmacol 1988; 8:14-22
247.
Beardsley PM, Balster RL: The intravenous self-administration of antihistamines by rhesus monkeys. Drug Alcohol Depend 1992; 30:117-26
248.
Zacny JP: Discriminative stimulus effects of H1-antihistaminesin cocaine-trained pigeons. Behav Pharmacol 1990; 1:261-5
249.
Preston KL, Wolf B, Guarino JJ, Griffiths RR: Subjective and behavioral effects of diphenhydramine, lorazepam and methocarbamol: Evaluation of abuse liability. J Pharmacol Exp Ther 1992; 262:707-20
250.
Stern KN, Chait LD, Johanson CE: Reinforcing and subjective effects of oral tripelennamine in normal human volunteers. Behav Pharmacol 1989; 1:161-7
251.
Lange WR, Jasinski DR: The clinical pharmacology of pentazocine and tripelennamine (T's and Blues). Adv Alcohol Subst Abuse 1986; 5:71-83
252.
Feldman MD, Behar M: A case of massive diphenhydramine abuse and withdrawal from use of the drug (letter). JAMA 1986; 255:3119-20
253.
Glickman L: Diphenhydramine abuse and withdrawal (letter). JAMA 1986; 256:1894
254.
Senay EC: Clinical experience with T's and B's. Drug Alcohol Depend 1985; 14:305-312
255.
Poklis A: Decline in abuse of pentazocine/tripelennamine (T's and Blues) associated with the addition of naloxone to pentazocine tablets. Drug Alcohol Depend 1984; 14:135-40
256.
Griffiths RR, Brady JV, Bradford LD: Predicting the abuse liability of drugs with animal drug self-administration procedures: Psychomotor stimulants and hallucinogens, Advances in Behavioral Pharmacology. Volume 2. Edited by Thomas T, Dews PB. New York, Academic Press, 1979, pp 162-208
257.
Ando K, Yanagita T: Effects of an antitussive mixture and its constituents in rats discriminating methamphetamine from saline. Pharmacol Biochem Behav 1992; 41:783-8
258.
Gauvin DV, Moore KR, Youngblood BD, Holloway FA: The discriminative stimulus properties of legal, over-the-counter stimulants administered singly and in binary and ternary combinations. Psychopharmacology 1993; 110:309-19
259.
Chait LD: Factors influencing the reinforcing and subjective effects of ephedrine in humans. Psychopharmacology 1994; 113:381-7
260.
Seno E, Morita N, Saito S, Nakatani Y, Nakamura T, Sato S, Oda S: Social psychiatric study of the over-the-counter antitussive drug abuse: Shifts resulting from modification of the formulae. Seishin Shinkeigaku Zasshi 1996; 98:127-50
261.
Crouch DJ, Birky MM, Gust SW, Rollins DE, Walsh JM, Moulden JV, Quinlan KE, Beckel RW: The prevalence of drugs and alcohol in fatally injured truck drivers. J Forensic Sci 1993; 38:1342-53
262.
Smith DA, Perry PJ: The efficacy of ergogenic agents in athletic competition: II. Other performance-enhancing agents. Ann Pharmacother 1992; 26:653-9
263.
Allen AC, Kiser WO: Methamphetamine from ephedrine: I. Chloroephedrines and aziridines. J Forensic Med 1987; 32:753-62
264.
Huber A, Ling W, Shoptaw S, Gulati V, Brethen P, Rawson R: Integrating treatments for methamphetamine abuse: a psychosocial perspective. J Addict Dis 1997; 16:41-50
265.
Wolkoff DA: Methamphetamine abuse: An overview for health care professionals. Hawaii Med J 1997; 56:34-6
266.
Morrison JD, Clarke RSJ, Dundee JW: Studies of drugs given before anaesthesia: XXI. Droperidol. Br J Anaesth 1970; 42:730-5
267.
Dundee JW, Clarke RSJ, Howard PJ: Studies of drugs given before anaesthesia: XVII. Metoclopramide. Br J Anaesth 1974; 46:509-13
268.
Marshall JWB, Kelly ME, Randall JI, Costall B: The lack of effect of ondansetron in a mouse model of conditioned place preference (CPP). Br J Pharmacol 1994; 111(suppl):164P
269.
Doty P, Zacny JP, de Wit H: Effects of ondansetron pretreatment on acute responses to ethanol in social drinkers. Behav Pharmacology 1994; 5:461-9
270.
Lane JD, Pickering CL, Hooper ML, Fagan K, Tyers MB, Emmett-Oglesby MW: Failure of ondansetron to block the discriminative or reinforcing stimulus effects of cocaine in the rat. Drug Alcohol Depend 1992; 30:151-62
271.
Tomkins DM, Le AD, Sellers EM: Effect of the 5-HT3antagonist ondansetron on voluntary ethanol intake in rats and mice maintained on a limited access procedure. Psychopharmacology 1995; 117:479-85
272.
Grant KA: The role of 5-HT3receptors in drug dependence. Drug Alcohol Depend 1995; 38:155-71
273.
Borg PJ, Taylor DA: Voluntary oral morphine self-administration in rats: Effect of haloperidol or ondansetron. Pharmacol Biochem Behav 1994; 47:633-46
274.
Swift RM, Davidson D, Whelihan W, Kuznetsov O: Ondansetron alters human alcohol intoxication. Biol Psychiatry 1996; 40:514-21
275.
Grady TA, Broocks A, Canter SK, Pigott TA, Dubbert B, Hill JL, Murphy DL: Biological and behavioral responses to D-amphetamine, alone and in combination with the serotonin3receptor antagonist ondansetron, in healthy volunteers. Psychiatry Res 1996; 64:1-10
276.
Sell LA, Cowen PJ, Robson PJ: Ondansetron and opiate craving: A novel pharmacological approach to addiction. Br J Psychiatry 1995; 166:511-4
277.
Wilde MI, Markham A: Ondansetron: A review of its pharmacology and preliminary clinical findings in novel applications. Drugs 1996; 52:773-94
278.
Holtzman SG: Discriminative stimulus effects of drugs: Relationship to potential for abuse, Modern Methods in Pharmacology. Volume 6, Testing and Evaluation of Drugs of Abuse. New York, Wiley-Liss, 1990, pp 193-210
279.
Haertzen CA, Hockey JE: Addiction Research Center Inventory (ARCI): Measurement of euphoria and other drug effects, Methods of Assessing the Reinforcing Properties of Abused Drugs. Edited by Bozarth MA. New York, Springer-Verlag, 1987, pp 489-524
280.
Henningfield JE, Johnson RE, Jasinski DR: Clinical procedures for the assessment of abuse potential, Methods of Assessing the Reinforcing Properties of Abused Drugs. Edited by Bozarth MA. New York, Springer-Verlag, 1987, pp 573-90
281.
Jaffe JH, Jaffe FK: Historical perspectives on the use of subjective effects measures in assessing the abuse potential of drugs, Testing for Abuse Liability of Drugs in Humans. NIDA Research Monograph No. 92. Edited by Fischman MW, Mello NK, Rockville, U. S. Department of Health and Human Services, 1989, pp 43-72
282.
Wise RA, Bozarth MA: A psychomotor stimulant theory of addiction. Psychol Rev 1987; 94:469-92
283.
Di Chiara G, Imperato A: Drugs abused by humans preferentially increase synaptic dopamine concentration in the mesolimbic system of freely moving rats. Proc Natl Acad Sci U S A 1988; 85:5274-8
284.
Chen J: Dopaminergic mechanisms and brain reward. Semin Neurosci 1993; 5:315-20
285.
Zito KA, Vickers G, Roberts DCS: Disruption of cocaine and heroin self-administration following kainic acid lesions of the nucleus accumbens. Pharmacol Biochem Behav 1985; 23:1029-36
286.
Corrigall WA, Frankin KB, Coen KM, Clarke PB: The mesolimbic dopaminergic system is implicated in the reinforcing effects of nicotine. Psychopharmacology 1992; 107:285-9
287.
Myers RD, Borg S, Mossberg R: Antagonism by naltrexone of voluntary alcohol selection in the chronically drinking macaque monkey. Alcohol 1986; 3:383-8
288.
Le AD, Poulos CX, Quan B, Chow S: The effects of selective blockade of delta and mu opiate receptors on ethanol consumption by C57BL/6 mice in a restricted access paradigm. Brain Res 1993; 630:330-2
289.
Roberts DCS, Bennett SAL: Heroin self-administration in rats under a progressive ratio schedule of reinforcement. Psychopharmacology 1993; 111:215-8
290.
Stevens KE, Shiotsu G, Stein L: Hippocampal mu-receptors mediate opioid reinforcement in the CA3 region. Brain Res 1991; 545:8-16
291.
Harrigan SE, Downs DA: Continuous intravenous naltrexone effects on morphine self-administration in rhesus monkeys. J Pharmacol Exp Ther 1978; 204:481-6
292.
Balster RL: Drug abuse potential evaluation in animals. Br J Addict 1991; 86:1549-58
293.
Johanson CE, Woolverton WL, Schuster CR: Evaluating laboratory models of drug dependence, Psychopharmacology: The Third Generation of Progress. Edited by Meltzer HY. New York, Raven Press, pp 1617-25
294.
Woody GE, Urschel HC, Alterman A: The many paths to drug dependence, Vulnerability to Drug Abuse. Edited by Glantz M, Pickens R. Washington DC, American Psychological Association, 1992, pp 491-507
295.
Pickens R, Svikis D, McGue M, Lykken D, Heston M, Clayton P: Heterogeneity in the inheritance of alcoholism: A study of male and female twins. Arch Gen Psychiatry 1991; 48:19-28
296.
Conley KM, Toledano AY, Apfelbaum JL, Zacny JP: Modulating effects of a cold water stimulus on opioid effects in volunteers. Psychopharmacology 1997; 131:313-20
297.
Borgbjerg FM, Nielsen K, Franks J: Experimental pain stimulates respiration and attenuates morphine-induced respiratory depression: A controlled study in human volunteers. Pain 1996; 64:123-8
298.
de Wit H, Stewart J: Reinstatement of cocaine-reinforced responding in the rat. Psychopharmacology 1981; 75:134-43
299.
de Wit H, Stewart J: Drug reinstatement of heroin-reinforced responding in the rat. Psychopharmacology 1983; 79:29-31
300.
de Wit H: Priming effects with drugs and other reinforces. Exp Clin Psychopharmacol 1996; 4:5-10
301.
Savage SR: Addiction in the treatment of pain: Significance, recognition and management. J Pain Symptom Manage 1993; 8:265-78
302.
Voigt L: Anesthetic management of the cocaine abuse patient. Am Assoc Nurse Anesth J 1995; 63:438-43
303.
Latasch L, Christ R: Problems in anesthesia of drug addicts. Anaesthesist 1988; 37:123-39
304.
Wood PR, Soni N: Anaesthesia and substance abuse. Anaesthesia 1989; 44:672-80
305.
Rubenstein RB, Spira I, Wolff WI: Management of surgical problems in patients on methadone maintenance. Am J Surg 1976; 131:566-9
306.
Edwards R, Mosher VB: Alcohol abuse, anaesthesia, and intensive care. Anaesthesia 1980; 35:474-89
307.
Peacock JE: Anaesthesia and miscellaneous diseases, Anaesthesia. 2nd edition, volume 2. Edited by Nimmo WS, Rowbotham DJ, Smith G. London, Blackwell Scientific, 1984, pp 1130-47
308.
Vandam LD, Desai SP: Evaluation of the patient and preoperative evaluation, Clinical Anesthesia. Edited by Barash PG, Cullen BF, Stoelting RK. Philadelphia, J. B. Lippincott, 1989, pp 407-38
309.
Talbott GD, Gallegos KV, Wilson PO, Porter TL: The Medical Association of Georgia's Impaired Physicians Program: Review of the first 1000 physicians: analysis of specialty. JAMA 1987; 257:2927-30