Back to Journals » Substance Abuse and Rehabilitation » Volume 15

Pharmacological Treatments for Methamphetamine Use Disorder: Current Status and Future Targets

Authors Yates JR 

Received 3 June 2024

Accepted for publication 15 August 2024

Published 30 August 2024 Volume 2024:15 Pages 125—161

DOI https://doi.org/10.2147/SAR.S431273

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Rajendra Badgaiyan



Justin R Yates

Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA

Correspondence: Justin R Yates, Email [email protected]

Abstract: The illicit use of the psychostimulant methamphetamine (METH) is a major concern, with overdose deaths increasing substantially since the mid-2010s. One challenge to treating METH use disorder (MUD), as with other psychostimulant use disorders, is that there are no available pharmacotherapies that can reduce cravings and help individuals achieve abstinence. The purpose of the current review is to discuss the molecular targets that have been tested in assays measuring the physiological, the cognitive, and the reinforcing effects of METH in both animals and humans. Several drugs show promise as potential pharmacotherapies for MUD when tested in animals, but fail to produce long-term changes in METH use in dependent individuals (eg, modafinil, antipsychotic medications, baclofen). However, these drugs, plus medications like atomoxetine and varenicline, may be better served as treatments to ameliorate the psychotomimetic effects of METH or to reverse METH-induced cognitive deficits. Preclinical studies show that vesicular monoamine transporter 2 inhibitors, metabotropic glutamate receptor ligands, and trace amine-associated receptor agonists are efficacious in attenuating the reinforcing effects of METH; however, clinical studies are needed to determine if these drugs effectively treat MUD. In addition to screening these compounds in individuals with MUD, potential future directions include increased emphasis on sex differences in preclinical studies and utilization of pharmacogenetic approaches to determine if genetic variances are predictive of treatment outcomes. These future directions can help lead to better interventions for treating MUD.

Keywords: methamphetamine use disorder, pharmacotherapy, clinical trial, animal models

Introduction

Methamphetamine (METH) is a synthetic and potent psychostimulant drug that increases attention and arousal, suppresses appetite, and produces euphoria.1–3 Although these effects are often reinforcing, METH can cause life-threatening hyperthermia and heart arrythmias,4,5 and it can produce symptoms that mimic schizophrenia.6,7 Another major problem associated with METH is the development of tolerance with repeated administration and the emergence of aversive withdrawal symptoms such as depressed mood and lethargy upon cessation of use.8–10 Long-term METH use can negatively impact one’s health,11 as individuals that use METH are at risk for dental problems (“meth mouth”), heart disease, stroke, and contraction of a sexually transmitted infection (STI).12–15 Some of the adverse health effects of METH are compounded by its manufacturing process. METH can be synthesized from pseudoephedrine, a nasal decongestant, or from phenyl-2-propanone (P2P), a precursor to pseudoephedrine, using hazardous chemicals like lye and camping fuel.16 Some clandestine labs now synthesize ephedrine and P2P using commercially available materials, which introduces additional impurities,17 thus increasing the risk of adverse health effects.

There are several routes of administration associated with METH: oral, intravenous, intranasal, and inhalation. When ingested orally, peak subjective effects occur approximately 3 h after use; other routes of administration lead to rapid onset of subjective effects, occurring less than 15 min when injected or used intranasally and occurring within 20 min when smoked.18 Because METH is lipophilic, it quickly crosses the blood-brain barrier, where it can exert its psychoactive effects.19 METH also enters most organs, with high concentrations observed in lungs, liver, and kidneys.20 METH is metabolized primary by the hepatic cytochrome P450 2D6 enzyme,21 leading to 4-hydroxymethamphetamine and amphetamine (AMPH) as major metabolites,22,23 before being eliminated from the body through urine.24 Compared to other stimulants like cocaine, the half-life of METH is considerably longer, approximately 7–12 h depending on the dose and the route of administration.18,25–27

METH increases extracellular concentrations of the monoamine neurotransmitters dopamine, norepinephrine, and serotonin via several mechanisms. METH is a vesicular monoamine transporter 2 (VMAT-2) reverser, causing monoamine concentrations to increase in the presynaptic terminal button of neurons.28 Depending on the dose used, METH can reverse the direction of monoamine transporters, inhibit dopamine transporters, and/or inhibit the enzyme monoamine oxidase (MAO).29,30 Regardless, excess monoamine levels in the cytoplasm are released into the synapse, resulting in METH’s wide-ranging physiological and subjective effects.18 Increased dopaminergic activity in the mesocorticolimbic pathway underlies the reinforcing effects of METH,31 although reduced dopamine release is observed in brain regions associated with reward (eg, nucleus accumbens) after chronic use, which may explain why tolerance develops to the reinforcing effects of METH.32

Since the 2010s, METH use has increased significantly throughout the world.33–36 More problematic is the increase in methamphetamine use disorder (MUD) and corresponding increase in overdose deaths.34 MUD, like other substance use disorders (SUDs), is characterized by several criteria according to agencies like the American Psychiatric Association37 and the International Classification of Diseases.38 Included in these criteria are using larger amounts of METH or using METH for longer periods of time than intended; persistent desire or unsuccessful efforts to control METH use; spending increasing amounts of time trying to obtain METH, use METH, and/or recover from METH’s effects; craving to use METH; failing to fulfill other responsibilities due to METH use; continued use of METH despite negative consequences; developing tolerance to METH’s effects; and experiencing withdrawal symptoms upon cessation of use.37 Recurrence of substance use following abstinence (“relapse”) is another prominent feature of SUDs.39

The socioeconomic costs of MUD are high as lost productivity, increased crime and incarceration rates, and premature mortality are consequences of the increasing popularity of this drug.40–42 As MUD rates continue to increase, finding effective treatment strategies to help individuals reduce METH use is imperative, especially given that no pharmacological treatments currently exist for MUD. The purpose of the current review is to discuss the pharmacological treatments that have been screened in various assays related to METH dependence and MUD. The review is not limited to clinical studies using METH-dependent individuals. Instead, results of preclinical studies and human studies involving both dependent and nondependent individuals will be discussed to provide a comprehensive review of how certain treatments may be effective in (1) reducing METH intake in individuals, (2) decreasing the likelihood of resuming METH use following a treatment intervention, (3) ameliorating METH-induced cognitive impairments, and/or (4) serving as a complimentary treatment for other interventions (eg, cognitive-behavioral therapy).

Methods for Screening the Efficacy of Potential Pharmacotherapies for MUD

Animal research is valuable for screening potential pharmacotherapies to determine if they are (a) safe to administer and (b) efficacious at attenuating the behavioral effects of METH. Additionally, animal research allows one to isolate the neural mechanisms underlying treatment-induced alterations in METH’s rewarding properties. To this end, numerous assays are used to study the neurobehavioral actions of potential pharmacotherapies.

Although tolerance develops to the rewarding properties of METH, individuals show behavioral sensitization following repeated METH use. Particularly problematic is the emergence of stereotypies.43 In humans, common METH-induced stereotypies include excoriation, jaw clenching, and bruxism.44,45 Repeated METH exposure increases behavioral sensitization and stereotypies in animals such as circling, biting, head bobbing, sniffing, and grooming.46–50 Preclinical studies are primarily used to determine if a potential pharmacotherapy attenuates METH-induced locomotor sensitization and/or stereotypies. This research is valuable as behavioral sensitization is reported to be a risk factor for future drug taking in animals.51–53

Conditioned place preference (CPP) allows one to measure the conditioned rewarding effects of a stimulus such as METH.54,55 Although CPP can be measured in humans,56 no current studies exist in which METH was used. As such, discussion of METH CPP is limited to animal subjects in the current review. In CPP, the potential pharmacotherapy can be administered before each METH treatment to determine if acquisition of METH CPP can be blunted. Alternatively, the putative pharmacotherapy can be administered before the posttest; this measures the ability of the treatment to attenuate the expression of METH CPP. Detailed discussion of CPP as a tool for screening pharmacotherapies is detailed elsewhere.57

Self-administration uses operant conditioning principles to measure the direct reinforcing effects of a stimulus.58 There are numerous self-administration paradigms that model distinct aspects of addictive-like behaviors, including short-access models (eg, < 3 h)59–62 and extended access models (eg, 6+ h).63–65 Extended access to drug often leads to escalation of drug intake, a defining trait of SUDs.66 Progressive ratio schedules measure the reinforcing efficacy of METH by requiring the subject to emit more responses after each reinforcer delivery;67,68 a break point is calculated, indicating the highest response ratio completed by the subject.69 Self-administration can be assessed in humans that are not currently seeking treatment for their substance use.70,71

Individuals with a SUD have access to numerous reinforcers that compete with substances like METH. Choice procedures are useful for modeling preference for drug reinforcement over alternative reinforcers. In animal studies, subjects respond on one manipulandum to earn a drug infusion and respond on a separate manipulandum to earn an alternative reinforcer like food.72,73 Several methods can be used with human participants. Individuals can choose between drug and placebo or drug and an alternative reinforcer such as money.74,75 Choice procedures can also be used to compare preference for the same drug delivered via different routes of administration.76

In addition to using self-administration procedures, clinical pharmacology studies often have individuals rate their subjective experiences of a drug administered alone or in combination with a potential pharmacotherapy.77 Subjective effects of METH include perceived physiological changes (eg, feeling stimulated), and pleasurable/aversive feelings. Clinical trials recruit individuals with a confirmed MUD, giving some of the participants the potential pharmacotherapy while giving other participants a placebo.78 Variables such as drug-free urine samples and dropout rates are then compared between the drug treatment and placebo groups.

Preclinical research often models recurrence of substance use with the reinstatement paradigm. Reinstatement can be assessed with both CPP and drug self-administration following extinction training in which drug is no longer paired with a specific environmental context (CPP) or delivered after completion of each response requirement (drug self-administration).79–82 Incubation of craving is similar to the reinstatement paradigm, except that animals are not given extinction training; instead, they are given a single extinction session followed by a period of forced abstinence in which animals are kept in their home cage (eg, 30 days), followed by a second extinction session.83

Methodology for Literature Review

Between March and May 2024, multiple searches were conducted in PubMed, using combinations of terms, including those for the potential pharmacotherapies discussed in this review; “methamphetamine” and “methamphetamine use disorder” (including alterations to the word methamphetamine like “METH” and “methylamphetamine”); “withdrawal”; “subjective effects”; “self-administration”; “progressive ratio”; “conditioned place preference” or “CPP”; “reinstatement”; “incubation of craving”; “relapse”; “locomotor activity”; “stereotypy” or “stereotypies”. Literature reviews and meta-analyses were excluded, and article titles and abstracts were screened for relevance. Additional articles were found by reviewing the reference sections of articles found during the literature search.

Potential Pharmacotherapies for MUD

Numerous drugs have been screened in assays measuring the physiological, the cognitive, and/or the reinforcing/rewarding effects of METH. Many of the drugs discussed below are already approved to treat other psychological and/or physical conditions while some have been tested in animals only. The treatments reviewed in the current paper are organized by their mechanism of action.

Pharmacotherapies for Attention-Deficit/Hyperactivity Disorder (ADHD)

Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterized by increased hyperactivity/impulsivity and/or inattention and is linked to hypoactive catecholamine neurotransmission.84 The primary treatment options are the psychostimulants d-AMPH and methylphenidate (MPH). Like METH, AMPH causes monoamines, specifically the catecholamines norepinephrine and dopamine, to be released into the synapse through their respective transporters, inhibits VMAT-2 functioning, and blocks activity of MAO; instead of reversing the flow of catecholamine transporters, MPH inhibits the reuptake of dopamine and norepinephrine, but there is evidence that MPH leads to a redistribution of VMAT-2.85

Giving METH-dependent individuals AMPH or MPH is a form of substitution treatment.86 Substitution treatment is already used for other drugs (eg, tobacco)87 and is an important component of harm reduction.88 Ideally, providing METH-dependent individuals with a stimulant drug that can be taken in a safer manner than inhalation or injection can help minimize the health risks associated with these routes of administration (eg, contraction of an STI from sharing needles). However, caution is needed when providing individuals AMPH or MPH given that they have misuse potential and can be diverted (eg, dissolved and then snorted or injected).89,90

AMPH blunts some of the subjective effects of METH,77 but results from animal studies are not as promising. Neither AMPH nor MPH affect METH self-administration in monkeys,91,92 while self-administration of MPH potentiates the reinforcing effects of METH in rats.93 These findings suggest that individuals maintained on AMPH or MPH may self-administer more METH to counteract the diminished subjective effects experienced during substitution treatment. Instead of targeting both dopamine and noradrenergic systems, an alternative approach is to use atomoxetine, a selective norepinephrine transporter inhibitor and non-stimulant treatment for ADHD. Although atomoxetine does not alter the subjective effects of METH,94 it reduces METH craving and METH-positive urine samples in individuals receiving methadone maintenance therapy.95 Preclinical studies assessing cocaine self-administration are encouraging as atomoxetine decreases relapse-like behavior in rodents.96–98

Modafinil

Modafinil is an anti-narcoleptic drug that acts as a weak dopamine transporter (DAT) inhibitor.99 Modafinil decreases METH self-administration and attenuates drug-seeking behavior in rodents.100–103 These effects may not be completely mediated by inhibition of DAT, as administration of the highly selective DAT inhibitor GBR 12909 potentiates METH-induced increases in locomotor activity and reinstatement of METH seeking.104,105 Indeed, modafinil increases glutamate and histamine levels and decreases GABA levels.106 Drugs targeting the glutamatergic and GABAergic systems will be discussed later in this review.

Modafinil somewhat blunts the positive subjective effects of METH and ameliorates METH-induced increases in systolic blood pressure,107,108; but see109 but it does not significantly reduce choice for METH in a self-administration paradigm nor reduce METH use, craving, attentional bias toward METH-paired stimuli, or severity of dependence in METH-dependent individuals.107–113 There is some evidence that modafinil can be used in conjunction with cognitive-behavioral therapy to reduce METH use.114 At the cellular level, modafinil protects against METH-induced neuroinflammation, dopamine toxicity, and cell death in the striatum,115 which is consistent with the inhibited dopamine release observed following selective blockade of DAT.116,117 The neuroprotective effects of modafinil may account for its ability to reverse working and verbal memory deficits observed in both METH-dependent individuals and rodents.112,118–120 Modafinil also improves learning in an associative learning task in METH-dependent individuals and improves inhibitory control in individuals with higher baseline rates of METH use.121,122

Antidepressant Drugs

The primary mechanism of action of antidepressants is to increase serotonin and norepinephrine levels. MAO inhibitors (MAOIs) prevent the metabolism of monoamines.123 Tricyclics/tetracyclics prevent the reuptake of serotonin/norepinephrine, but they interact with multiple molecular targets (eg, histamine receptors, cholinergic receptors, adrenergic receptors). Commonly prescribed today, selective serotonin reuptake inhibitors (SSRIs) and serotonin/norepinephrine reuptake inhibitors (SNRIs) also prevent the reuptake of norepinephrine and/or serotonin, but they have fewer “off-site” targets.

Due to the side effect profile of MAOIs, with cardiovascular effects being particularly problematic, individuals treated with MAOIs need to maintain a restrictive diet to avoid a life-threatening hypertensive crisis.124 As such, MAOIs are rarely tested in individuals with MUD. Selegiline (ie, deprenyl), a MAO-B inhibitor, fails to attenuate the positive subjective effects of METH, instead potentiating individuals’ self-reported “bad effects” following METH administration.125 This is somewhat concerning as aversive experiences, in conjunction with the dietary restrictions associated with MAOIs, may lead to lower compliance in METH-dependent individuals.

The tricyclic antidepressant imipramine increases treatment retention rates, but it does not significantly alter drug craving, self-reported use frequency, or drug-positive urine samples in stimulant-dependent individuals.126 One important consideration is that most of the participants in this study were dependent on cocaine (151 out of 183 participants), with the rest (32/183) being dependent on METH. Similar results are obtained when a sample of METH-dependent individuals is used: increased treatment retention but no effects on drug craving or frequency of METH use.127 Preclinical research demonstrates that desipramine increases dopamine release following METH administration while decreasing dopamine levels following administration of other amphetamines.128 Additionally, imipramine, as well as clomipramine, potentiates METH-induced stereotypies in rats.129,130 These preclinical findings may provide an account for the inability of imipramine to reduce the reinforcing effects of METH in dependent individuals.

In contrast to tricyclics, tetracyclics block METH-induced increases in locomotor activity and locomotor sensitization in animals.131,132 The tetracyclic antidepressant maprotiline reduces METH-induced stereotypies in rodents.133 Mirtazapine, when administered after a conditioning session, prevents the expression of METH CPP,134 and decreases cue-induced reinstatement in rats.135 Mirtazapine also decreases METH use in individuals with MUD,136,137 including cisgender men and transgender women who have sex with men,136 although it is ineffective in facilitating retention in a METH withdrawal program.138 This latter finding suggests that mirtazapine may not be beneficial for individuals actively going through withdrawal. Like MAOIs, using tetracyclic medications for long-term treatment of MUD is challenging given their side-effect profile. As tricyclic and tetracyclic antidepressants have multiple mechanisms of action, individuals can experience a wide range of side effects, including dry mouth, urinary difficulties, weight gain, drowsiness, and headaches.139

Similar to tetracyclics, SSRIs differentially alter locomotor responses to METH. Citalopram exacerbates locomotor activity following METH administration,140 with this effect appearing to be influenced by increased dopamine, but not serotonin, depletion in the nigrostriatal pathway.141 However, fluoxetine and paroxetine attenuate METH-induced locomotor sensitization,142,143 and they decrease METH CPP.143,144 Clinical trials with sertraline show that SSRIs can be contraindicated for individuals with MUD, as individuals treated with sertraline alone have lower treatment retention rates, experience more adverse events, and increase their METH use.145,146

Concerning SNRIs, duloxetine ameliorates METH-induced cognitive deficits,147,148 and venlafaxine blocks reinstatement of METH CPP.149 Clinical trials have not tested the efficacy of SNRIs for the treatment of METH dependence, although trials for cocaine dependence have largely been unsuccessful.150–152; but see.153,154

Bupropion is a dual-purpose medication, prescribed to treat both depressive disorders and nicotine dependence; compared to other antidepressant drug classes, bupropion is unique in that it inhibits dopamine and norepinephrine transporters and upregulates VMAT-2 expression.155 The ability of bupropion to inhibit reuptake of dopamine and to target VMAT-2 may make it a promising pharmacotherapy for MUD. To this end, bupropion blunts the positive subjective effects of METH,156 and it decreases METH self-administration in monkeys.91; but see157 Unfortunately, bupropion also decreases self-administration of sucrose in rats,158–160 suggesting that long-term bupropion treatment may lead to increased anhedonia in individuals. Indeed, anhedonia is observed in healthy individuals given bupropion.161 Bupropion can also lead to significant adverse events, including tachycardia, seizures, and suicidal ideations.162–164

VMAT-2 Inhibitors

Studies examining the efficacy of VMAT-2-selective drugs have been limited to animals. The plant-derived lobeline decreases METH self-administration and METH-induced stereotypies in rodents.165,166 Because lobeline is not entirely selective for VMAT-2,167 lobelane, an analog of lobeline, was next tested.168,169 Like lobeline, lobelane decreases METH self-administration in rats, but tolerance develops following repeated lobelane treatment.170 Lobelane analogs now show promise in reducing dependence-like behavior in rodents, as they decrease METH self-administration, reinstatement of METH seeking, METH CPP, and METH-induced hyperactivity, with no tolerance observed following repeated administration.171–177

Altering Tyrosine Levels

Tyrosine is a non-essential amino acid synthesized from phenylalanine that serves as a vital precursor to dopamine. Decreasing tyrosine levels or preventing the conversion of tyrosine to L-DOPA (l-3,4-dihydroxyphenylalanine; immediate precursor to dopamine) limits METH-induced increases in dopamine release,178 thus diminishing the reinforcing effects of METH. Administration of the tyrosine hydroxylase inhibitor alpha-methyl-p-tyrosine protects against METH’s neurotoxic and hyperthermic effects,179–182 and attenuates METH-induced dopamine depletion.183,184 Only one study has examined the effects of tyrosine depletion on METH effects in humans, with subjective effects and METH-induced mania decreasing following tyrosine depletion.185 There is evidence that tyrosine depletion decreases cue- and drug-induced craving for cocaine, but fails to affect cocaine self-administration in non-dependent cocaine users.186 One major concern associated with tyrosine depletion is increased apathy and decreased contentment.187

Monoaminergic Receptor Ligands

By limiting tyrosine activity, dopamine is not the only neurotransmitter that is decreased; norepinephrine levels decrease following alpha-methyl-p-tyrosine administration,188 most likely because dopamine is the direct precursor of norepinephrine.189 Instead of targeting monoamine synthesis or metabolism, one approach is to use monoaminergic receptor ligands to block the effects of METH in individuals.

Each monoaminergic system has its own receptor types, with all but one coupled to a G protein (ie, metabotropic receptor).190–192 Dopamine D1-like (D1 and D5) receptors, all three noradrenergic beta receptor subtypes (1–3), and serotonin 5-HT4, 5-HT6, and 5-HT7 receptors are Gs-coupled, leading to increased adenyl cyclase activity in the neuron, resulting in increased neuronal excitation.190–192 Noradrenergic alpha-1 and serotonin 5-HT2 receptors are also excitatory, but they are coupled to a Gq protein, which activates a different intracellular signaling pathway involving phospholipase C and protein kinases.190,192 Dopamine D2-like (D2, D3, and D4), noradrenergic alpha-2, and serotonin 5-HT1 and 5-HT5 receptors are inhibitory. Stimulation of these receptors leads to decreased activity of adenyl cyclase.190–192 Finally, the 5-HT3 receptor is ionotropic, consisting of five subunits surrounding an ion channel. When a ligand binds to the receptor, the ion channel opens, allowing sodium ions to enter the neuron.192

Buspirone

Buspirone is an anxiolytic that acts as a partial agonist at serotonin 5-HT1A receptors and as an antagonist at dopamine D2-like receptors.193 Buspirone decreases reinstatement of METH seeking in rats,194 but it fails to alter choice of METH over food in monkeys and fails to affect the subjective or the reinforcing effects of intranasal METH in humans.195,196 In fact, buspirone increases some positive subjective effects of oral METH.197 These results raise concerns about the use of buspirone as a treatment for MUD.

Antipsychotic Medications

Historically used to treat psychiatric conditions like schizophrenia, antipsychotic medications exert their effects by acting as dopamine D2 receptor antagonists, with atypical antipsychotics also acting as serotonin, specifically 5-HT2A, and noradrenergic receptor antagonists.198 Some antipsychotics, such as aripiprazole, act as partial agonists at dopamine D2-like receptors and 5-HT1A receptors in addition to antagonizing 5-HT2A receptors.199

Antipsychotics decrease METH-induced hyperactivity and block locomotor sensitization following repeated METH administration;200–211 but see.212 The atypical antipsychotic risperidone increases METH self-administration in monkeys.157 Aripiprazole produces biphasic effects when a fixed ratio schedule is used, decreasing self-administration when lower doses of METH are used, but increasing self-administration when a higher dose of METH is used.213 The increased self-administration observed in monkeys and in rats may not necessarily indicate that antipsychotics increases the reinforcing effects of METH; instead, animals may increase their responding to achieve a hedonic set point that is similar to when pretreated with vehicle. When a progressive ratio schedule is used, aripiprazole decreases the reinforcing efficacy of METH.213

Neither haloperidol nor risperidone blunt METH’s stimulant effects in humans;214 yet, individuals maintained on risperidone decrease their use of METH,215,216 but these results are somewhat difficult to interpret due to the small number of individuals that completed treatment (eg, only 12 of 53 participants completed an 8-week treatment of risperidone injections).216 Aripiprazole blunts some positive subjective effects of METH and decreases METH self-administration in non-dependent individuals,217–219 but it fails to decrease METH use in dependent individuals.220,221 As aripiprazole increases treatment retention,221 it may be better served as a treatment given in addition to non-pharmacological-based interventions. However, evidence indicates that individuals given antipsychotics “off-label” may develop a hypersensitive dopaminergic system, which can increase drug cravings and worsen stimulant use disorder.222

One benefit of antipsychotics is they ameliorate the psychotomimetic-like effects of METH. Antipsychotics block behavioral disturbances in rodents that model schizophrenia-like behavior (eg, prepulse inhibition deficits) and abolish METH-induced self-injurious behavior in mice.223–225 Clinically, antipsychotics reduce METH-induced psychosis and sedate individuals experiencing METH toxicity.222,226–231 Related to METH toxicity, antipsychotics may be useful for reversing METH-induced hyperthermia.232

Dopamine Receptor Ligands

As dopaminergic dysfunction in the mesocorticolimbic pathway is heavily implicated in SUDs,233 blocking dopamine receptors represents a potential mechanism for attenuating the physiological and/or the reinforcing effects of METH. The mixed dopamine receptor antagonist levo-tetrahydropalmatine decreases METH self-administration and METH-induced reinstatement.234 The protective effects of levo-tetrahydropalmatine appear to be driven by blockade of D1-like receptors, as D1-like, but not D2-like, receptor antagonists decrease METH self-administration and reinstatement of METH-seeking behavior.105,235,236 However, both D1-like and D2-like antagonists block METH-induced hyperactivity and stereotypies, as well as the development of behavioral sensitization following repeated METH administration.131,209,224,237–241 Likewise, blocking either dopamine D1-like or D2-like receptors attenuates the acquisition and the expression of METH CPP in rodents,242–248 although some work shows that D2-like receptors are uninvolved in the acquisition of METH CPP.243,248 Another seemingly paradoxical finding is that while a dopamine D2 receptor partial agonist decreases METH-induced hyperactivity,209 D2-like agonists increase METH seeking in a reinstatement model.249

Dopamine D1-like and D2-like receptors have complex interactions with one another in striatal pathways. Projections from striatal D2-containing medium spiny neurons (MSNs) to D1-containing MSNs are more common than projections from D1-like MSNs to D2-like MSNs, and synaptic connections formed by D2-like MSNs are stronger than those formed by D1-like MSNs.250 Upregulation of D2-like MSNs in nucleus accumbens also increases motivation for reinforcement via disinhibition of the ventral pallidum,251 which mirrors increased motivation for reinforcement following direct stimulation of D1-like receptors in nucleus accumbens.252 Thus, D2-like receptor antagonists can produce similar results as D1-like receptor antagonists by preventing the inhibition of inhibitory neurons that typically regulate the release of dopamine.

As D2-like receptor antagonists have fewer extrapyramidal side effects compared to D1-receptor antagonists,198 there is growing interest in targeting these receptors for SUDs, particularly the dopamine D3 receptor.253 Selective dopamine D3 receptor antagonists attenuate METH-induced behavioral sensitization,254 ameliorate prepulse inhibition deficits following METH administration,255 prevent METH-induced hyperthermia,256 blunt METH CPP,254 reduce intracranial self-stimulation following METH administration,257,258 decrease the reinforcing efficacy of METH,257,259–261 and block reinstatement of METH-seeking behavior.257,259,260,262 While dopamine D3 receptor partial agonists attenuate METH-induced intracranial self-stimulation and METH self-administration,245,261 one selective D3 receptor partial agonist has no effect on METH self-administration in monkeys in a choice procedure.195 Despite this last finding, the results with dopamine D3 receptor antagonists are promising and merit further testing.

Currently, there is little work examining the effects of selective dopamine D4 receptor ligands on dependence-like behaviors despite evidence showing that this receptor may be an important target for preventing recurrence of stimulant use.263 A selective dopamine D4 receptor antagonist blocks METH-induced hyperactivity,264 but has no effect on the development of behavioral sensitization.265 There is recent evidence that D4 receptor partial agonists decrease cocaine self-administration in rats;266 thus, investigating how D4 receptor ligands influence MUD-like behaviors is warranted.

Noradrenergic Receptor Ligands

Alpha-1 receptor agonists are used as decongestants and hypotension medications,267,268 and beta receptor agonists are commonly prescribed for pulmonary-related conditions.269 Alpha-1 and beta receptor antagonists are used to decrease hypertension.270,271 Alpha-2 receptor agonists are used as sedatives in animal surgery,272 with clonidine and guanfacine used as ADHD medications.273 Alpha-2 receptor antagonists are often used to reverse the sedative effects of alpha-2 agonists,274 with yohimbine being used to model stress-induced reinstatement of drug-seeking behavior, including for METH.275,276 As METH increases norepinephrine levels, one approach to blunting the physiological/behavioral effects of METH is to block alpha-1/beta receptors and/or to stimulate alpha-2 receptors.

Research examining the efficacy of noradrenergic receptor ligands is limited to animals. Alpha-1 and beta receptor antagonists ameliorate METH-induced hyperthermia and conditioned hyperactivity.179,277–279 Similarly, an alpha-1 receptor inverse agonist attenuates conditioned hyperactivity following repeated METH treatments.280 Although the effects of alpha-1/beta receptor antagonists on the reinforcing effects of METH are unknown at this point, there is evidence that blocking alpha-1 receptors decreases cocaine self-administration;281–283 but see.284 Conversely, results with beta receptor antagonists are mixed, with some studies showing decreased cocaine self-administration,285,286 no change in cocaine self-administration,282 or increased cocaine self-administration.283 While ineffective on their own, combination treatment of an alpha-1 receptor antagonist and a beta receptor antagonist attenuates cue-induced reinstatement of cocaine seeking.287

Alpha-2 receptor agonists attenuate METH-induced hyperactivity and stereotypies,288 METH’s hyperthermic effects,289 and METH CPP,290 although they do not show much efficacy in blunting the direct reinforcing effects of psychostimulants;282,291 but see.292,293 Alpha-2 receptor agonists reduce cue- and stress-induced cocaine seeking,287,294–296 suggesting they may be an important treatment for individuals with MUD that are currently or have recently completed a treatment intervention.

Serotonin Receptor Ligands

Similar to dopamine receptor antagonists and noradrenergic alpha-2 receptor agonists, administration of 5-HT1A agonists and antagonists, 5-HT2A receptor antagonists, 5-HT2C agonists, and 5-HT5A antagonists reduces METH-induced hyperactivity.131,209,297–299 METH-induced stereotypies are similarly blunted by 5-HT1A agonists.130 Conversely, 5-HT2C antagonists potentiate METH-induced locomotor activity.209 Concerning METH reward, a 5-HT1B agonist attenuates the expression, but not acquisition, of METH CPP,300 and 5-HT3 antagonists block METH CPP.301

Psychedelic hallucinogens are receiving interest as potential pharmacotherapies for conditions like posttraumatic stress disorder and alcohol use disorder.302,303 The major psychedelic hallucinogens are naturally derived and include lysergic acid diethylamide (LSD), psilocybin, mescaline, and N,N-dimethyltryptamine (DMT). Psychedelic hallucinogens are agonists at 5-HT2A receptors, although they bind to multiple 5-HT receptor subtypes, with LSD also acting as an agonist at dopaminergic and noradrenergic receptors.304 Results from the few studies examining the efficacy of psychedelic hallucinogens are mixed. LSD fails to alter intracranial self-stimulation following METH injection.305 Psilocin, an active metabolite of psilocybin, decreases METH-induced hyperlocomotion and CPP but does not affect reinstatement of METH CPP.306 Additional work further diminishes the idea of targeting the 5-HT2A receptor as a pharmacotherapy for MUD as the 5-HT2A inverse agonist/antagonist pimavanserin fails to reduce preference for METH over food in monkeys.307

Lorcaserin is a 5-HT2C agonist that was previously used as a weight-loss drug.308 Lorcaserin reduces stimulant use in METH-dependent individuals,309 but the appeal of lorcaserin as a viable treatment for MUD is reduced by the finding that individuals choose cocaine more frequently compared to money when treated with lorcaserin despite reporting decreased drug craving.310 Additionally, results from a double-blind, placebo-controlled clinical trial show no benefits of lorcaserin for the treatment of cocaine use disorder.311 Finally, lorcaserin is no longer on the market in the United States due to its potential carcinogenic effects.312 Given that a 5-HT2C inverse agonist, but not 5-HT2C antagonists, attenuates METH-seeking in rodents,313 this may provide a novel approach to treating METH dependence as opposed to stimulating these receptors.

Ondansetron is a serotonin 5-HT3 receptor antagonist that is used as an antiemetic.314 Ondansetron potentiates METH-induced locomotor activity,131 which is interesting as a different 5-HT3 antagonist reduces METH-induced hyperactivity.315 Clinically, ondansetron is no more effective than placebo at reducing METH use, withdrawal symptoms, craving, or dependence severity.316 Ondansetron does reverse the anorexic-like effects of METH,317 which can be valuable for helping stimulate appetite in those with MUD.

Trace Amine-Associated Receptor Agonists

Trace amines are chemicals that are structurally related to monoamines and are derived from phenylalanine.318 For example, phenylalanine is converted to phenethylamine by aromatic L-amino acid decarboxylase, the same enzyme that converts L-DOPA into dopamine. Other trace amines include tyramine, octopamine, and synephrine. Trace amines bind to the trace amine-associated receptor 1 (TAAR 1), which regulates monoaminergic neurons. Specifically, trace amines modulate monoamine transporter function.319,320 Because METH acts as an agonist at TAAR 1,319,321–323 there is interest in determining if TAAR 1 agonists can act as a form of substitution therapy for MUD. This research is limited to preclinical models so far, but results are promising. TAAR 1 agonists reduce METH-induced dopamine release in nucleus accumbens, attenuate METH-induced locomotor sensitization, decrease METH drug self-administration, blunt the reinforcing efficacy of METH, and block drug-induced reinstatement of METH seeking.324–326 Another benefit of TAAR 1 agonists is that they can ameliorate motor impulsivity caused by acute METH administration and abrupt cessation of METH.327

Cholinergic Drugs

As numerous neurotransmitter systems are implicated in addiction,328 testing the efficacy of non-monoaminergic drugs for METH dependence is needed. Acetylcholine is an important neurotransmitter that regulates muscle contractions and is implicated in learning and cognition.329,330 Systemic administration of METH increase acetylcholine levels in ventral tegmental area and striatum of rodents,331,332 regions of the brain involved in the addiction process.333 Cholinergic receptors control dopamine release in nucleus accumbens when animals are presented with drug-paired cues,334 suggesting that the cholinergic system may be an important target for preventing drug-seeking behavior.

There are two major types of cholinergic receptors: nicotinic and muscarinic. Nicotinic receptors are ionotropic and consist of five subunits; stimulation of nicotinic receptors leads to sodium influx into the neuron. Muscarinic receptors are metabotropic and can be excitatory (M1, M3, and M5) or inhibitory (M2 and M4).335

Nicotinic Receptor Ligands

Nicotine is an agonist at nicotinic receptors and is the primary psychoactive chemical in tobacco. One interesting finding is that as craving for nicotine increases, craving for and use of stimulants decrease.336 Similarly, cue-induced drug seeking is enhanced in animals following antagonism of nicotinic receptors.334 These results indicate that nicotine may be useful as a form of substitution treatment for MUD. Indeed, nicotine fully substitutes for METH in a drug-discrimination paradigm in animals while METH acts as a partial substitute for nicotine only.337

Repeated administration of nicotine during extinction training decreases reinstatement of METH seeking.338 Yet, other research raises concerns about the viability of long-term nicotine administration as a treatment for MUD. First, repeated nicotine exposure fails to alter the acquisition, the extinction, and the reinstatement of METH CPP.339 Chronic, but not acute, administration of nicotine augments locomotor activity following METH treatment.340,341; but see342 While acute administration of nicotine transiently decreases METH self-administration, repeated nicotine administration fails to alter METH self-administration.343 In fact, chronic nicotine treatment enhances METH self-administration in adult females.344 Acute nicotine administration potentiates reinstatement of METH-seeking behavior,343,345 although this effect is not observed in rats treated with nicotine during adolescence.346 Nicotine may be beneficial for ameliorating cognitive deficits resulting from METH use. Nicotine administration reverses METH-induced deficits in novel object recognition, memory, and prepulse inhibition and attenuates METH-induced increases in risky choice.347–350

Varenicline is a partial agonist at ⍺4β2 and an agonist at ⍺7 nicotinic receptors and is used to for tobacco dependence as a form of substitution treatment.351 One concern is that varenicline decreases responding for both METH and saline/food,174,352; but see353 suggesting that this treatment may increase anhedonia in individuals; furthermore, low doses of varenicline potentiate reinstatement of METH-seeking, although this effect may reflect an increase in general locomotor activity as responding increases on a manipulandum previously paired with saline,352,353 There is evidence that varenicline blunts the positive subjective effects of METH,354 but clinical research shows that varenicline is ineffective at decreasing METH use in dependent individuals.355 Like nicotine, varenicline may be useful for reversing cognitive impairments resulting from METH use, as it reverses METH-induced increases in risky choice in rodents.350 Instead of using an agonist or partial agonist at nicotinic receptors, one approach is to use a selective ⍺4β2 nicotinic receptor desensitizing compound. To this end, an ⍺4β2 nicotinic receptor desensitizing compound decreases METH self-administration in rats.356

Ibogaine is a naturally derived psychoactive substance found in certain plants that has high affinity for nicotinic acetylcholine receptors, adrenergic ⍺2 receptors, and the serotonin transporter.357 Ibogaine blocks METH-induced hyperthermia,358 but it increases METH-induced stereotypies.359 The ⍺3β4 nicotinic receptor antagonist 18-methoxycoronaridine (18-MC), a synthetic analog of ibogaine, decreases METH self-administration,61,360,361 but it increases locomotor activity in rats chronically treated with METH and potentiates METH-induced stereotypies like ibogaine.359 These results are concerning as they suggest that blocking nicotinic receptors can exacerbate problematic behaviors like skin picking in individuals with MUD.

Muscarinic Receptor Ligands

Benztropine is a non-selective muscarinic receptor antagonist used to treat tremors associated with Parkinson’s disease.362 More recent studies using benztropine analogs show that they decrease METH-induced locomotor activity, METH self-administration, and reinstatement of METH seeking.363–365 One caveat to these findings is that one benztropine analog increases the reinforcing efficacy of METH as measured in a progressive ratio schedule.364

Scopolamine, another non-selective muscarinic receptor antagonist used to prevent motion sickness,366 prevents the development of METH-induced behavioral sensitization and stereotypies.367–369; but see370,371 While scopolamine does not affect METH CPP, the muscarinic M1 receptor antagonist trihexyphenidyl (an antispasmodic) blocks METH CPP.369 The selective muscarinic M1 receptor antagonist dicyclomine (an antispasmodic used to treat irritable bowel disease) does not alter the acute effects of METH on locomotor activity, but it potentiates behavioral sensitization.372

The muscarinic receptor agonist oxotremorine reverses both METH-induced hyperactivity and prepulse inhibition deficits,207 and the M1/M4 agonist xanomeline decreases METH-induced hyperactivity.373 Administration of muscarinic receptor agonists is difficult as they increase tremors and are used to model Parkinson’s disease.374,375 A potential future direction is to use partial agonists at muscarinic receptors. N-desmethylclozapine, a major metabolite of clozapine which acts as a partial agonist as muscarinic M1 receptors,376 prevents METH-induced psychotic-like effects.377 Whether muscarinic receptor partial agonists are efficacious in attenuating METH dependence-like behavior is currently unknown.

Acetylcholinesterase Inhibitors

A different potential cholinergic target is the inhibition of acetylcholinesterase, which metabolizes acetylcholine.378 Acetylcholinesterase inhibitors are currently used to treat Alzheimer’s disease.379 Despite blunting cocaine-induced behavioral sensitization and cocaine CPP, the acetylcholinesterase inhibitor donepezil fails to blunt these behaviors following METH administration.380 Donepezil also fails to alter memory impairments resulting from METH administration, but the weak acetylcholinesterase inhibitor galantamine attenuates METH-induced memory deficits.381 One positive findings is that both donepezil and galantamine decrease reinstatement of METH seeking.382,383 Rivastigmine modestly reduces some subjective effects of METH but is ineffective at reducing the direct reinforcing effects of METH and METH-induced deficits in cognition.384–387

Opioid Receptor Antagonists

Consisting of endorphins, dynorphins, and enkephalins, endogenous opioids bind to Gi-coupled metabotropic receptors (mu, kappa, delta, respectively).388 Given the influence of the endogenous opioid system to addiction,389 opioid receptor antagonists may be a viable molecular target for MUD. Naltrexone is used to treat opioid use disorder and alcohol use disorder.390 Naloxone is similar to naltrexone, but is most commonly used to reverse the effects of opioid overdose.391 As such, there is little research examining naloxone’s efficacy in METH dependence. The preclinical studies that have been conducted show that naloxone decreases stereotypical biting in mice but potentiates stereotypical sniffing and locomotor activity,392 and exacerbates METH-induced stereotypies in rats.393

Research with naltrexone is more promising, as it attenuates METH-induced behavioral sensitization, METH CPP, and METH-seeking behavior.392,394–398 The naltrexone analog nalmefene also decreases reinstatement of METH-seeking behavior.399 In humans, naltrexone fails to diminish the subjective effects and the direct reinforcing effects of METH in a non-clinical sample;400,401 but see.71 but there is evidence that naltrexone reduces subjective cue-induced craving in individuals with MUD and reduces the number of METH-using days in individuals with MUD or in individuals that engage in higher METH use.401–403 Naltrexone also decreases the ability of cue-induced craving to predict positive subjective effects.404 Importantly, the ability of naltrexone to blunt the subjective effects of METH is enhanced in individuals with lower executive functioning,405 indicating that neuropsychological tests should be performed in individuals to determine if naltrexone treatment will be effective.

The ability of naltrexone to selectively decrease the reinforcing effects of METH in dependent individuals may be related to a dysregulated dopaminergic system observed in these individuals. Individuals with MUD have increased resting state functional connectivity between brain regions like ventral striatum, amygdala, and hippocampus, which is blunted by naltrexone treatment.402 Naltrexone also reduces functional connectivity of regions like dorsal striatum and VTA with sensory-processing and motor-control regions of the brain that are associated with cue reactivity.406

Like naltrexone administered alone, combining naltrexone with other pharmacotherapies is ineffective at altering the subjective or the reinforcing effects of METH in non-dependent individuals,71,407 but it increases the percentage of drug-free urine samples in dependent individuals relative to placebo treatment, although the overall rate of success is moderately low.78 Combination treatment of naltrexone and bupropion is significantly more effective in gay/bisexual men compared to heterosexual men,408 an effect that is not observed when naltrexone is administered alone.409 As METH use is higher in LGBTQ+ individuals,410 naltrexone/bupropion provides a novel approach for treating individuals at increased risk for developing MUD.

In addition to naltrexone, future research may want to consider using drugs that are highly selective for one opioid receptor to better determine how endogenous opioids mediate the effects of METH in individuals. The mu opioid receptor antagonist β-funaltrexamine attenuates METH-induced increases in self-biting in mice, an effect that is not observed following selective blockade of kappa or delta receptors.392 The kappa receptor antagonist norbinaltorphimine dihydrochloride (nor-BNI) decreases escalation of METH intake in rats,411 while the non-selective delta opioid receptor antagonist naltrindole and the delta-2 opioid receptor antagonist naltriben reduce METH CPP.412

GABAergic Receptor Ligands

GABA is the major inhibitory neurotransmitter in the nervous system and binds to two receptor types: GABAA and GABAB.413 GABAA receptors are ionotropic, consisting of five subunits, and allow chloride to enter the neuron when activated. GABAB receptors are metabotropic, coupled to a Gi protein. Dysregulation of the GABA system is implicated in addiction.414 Furthermore, METH impairs signaling of GABAB receptors,415 and decreases immunoreactivity of inhibitory neurons in nucleus accumbens.416 The inhibition of GABAergic neurons, in turn, leads to further increases in dopamine release in the mesocorticolimbic pathway. Increasing GABAergic neurotransmission may be an effective treatment option for those with a MUD.

Many anxiolytics potentiate activity of GABAA receptors by binding to the benzodiazepine site of the receptor.417 Clonazepam prevents the acquisition, but not the expression, of METH-induced locomotor sensitization in rats, an effect that is blocked by co-administration of the GABAA receptor antagonist flumazenil.418,419 Oxazepam attenuates METH CPP,420 and decreases METH-seeking behavior when combined with the steroidogenesis inhibitor metyrapone.421 Anxiolytics can differentially affect METH self-administration, with oxazepam decreasing METH self-administration across various doses and alprazolam potentiating self-administration at low unit doses and decreasing self-administration at higher doses.422 One potential future direction is to use GABAA negative allosteric modulators. One such modulator reduces intracranial self-stimulation and nucleus accumbens dopamine release following METH administration in mice.423

Baclofen is a GABAB receptor agonist prescribed to treat muscle spasms. GABA inhibits dopamine release in striatal regions.424 Baclofen exacerbates METH-induced stereotypies in mice,425 but it attenuates the acquisition and the expression of METH CPP,426 facilitates extinction of METH CPP,427 and decreases METH self-administration.67 Baclofen also reverses METH-induced impairments in prepulse inhibition and recognition memory.428–430 Overall, baclofen does not significantly alter METH use in individuals that are dependent on METH; however, there is some evidence that individuals that show better medication adherence decrease their use to a greater extent than placebo-treated individuals.431

Glutamatergic Targets

Glutamate is the major excitatory neurotransmitter in the mammalian brain. Like the GABAergic system, the glutamatergic system is composed of both ionotropic and metabotropic receptors.432 Kainate, AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid), and NMDA (N-methyl-D-aspartate) receptors consist of four subunits and allow sodium (and calcium in the case of NMDA receptors) into the neuron when activated. Metabotropic glutamate receptors (mGluRs) are divided into three groups: Group I (excitatory mGluR1 and mGluR5), Group II (inhibitory mGluR2, mGluR3, and mGluR4), and Group III (inhibitory mGluR6, mGluR7, and mGluR8).

Ionotropic Glutamate Receptor Ligands

The effects of selective kainate receptor ligands on METH-related behaviors are unknown in both animals and humans. However, riluzole, which non-selectively inhibits ionotropic glutamate receptors,433 prevents increased locomotor activity following acute METH administration and blocks METH-induced locomotor sensitization.434 The effects of riluzole do not appear to be influenced by antagonism of AMPA receptors, as an AMPA receptor antagonist fails to alter METH-induced behavioral sensitization or METH-induced stereotypies.46 Additionally, AMPA receptor antagonists do not alter METH self-administration or METH-seeking behavior.435,436

Memantine is an NMDA receptor uncompetitive antagonist (ie, channel blocker) that is used clinically to treat Alzheimer’s disease.437 Not surprisingly, memantine is efficacious in reversing memory impairments resulting from METH administration.438,439 However, memantine administration produces stereotypic sniffing and increases locomotor activity.440 While memantine decreases self-administration of higher doses of METH, it potentiates self-administration of a low unit dose of METH.441 Memantine also fails to alter the subjective effects of METH in non-dependent individuals,442 although the effects of memantine have not been tested in dependent individuals.

Like memantine, ketamine is an NMDA receptor channel blocker, but it is used as an anesthetic in animals and is now used as a rapidly acting antidepressant in humans.443,444 Specific to METH-dependence-like behavior, ketamine decreases self-administration in animals.445 Ketamine also decreases cocaine-induced hyperactivity in animals,446 and decreases cocaine self-administration in humans.447 One caveat to this latter finding is that preclinical research shows that ketamine attenuates the reinforcing effects of both cocaine and food,448 suggesting increased anhedonia. There is some evidence that ketamine blocks reinstatement of cocaine seeking in monkeys,449 but this study tested three subjects only. A recent clinical trial shows that combining a single ketamine infusion with mindfulness-based therapy helps improve abstinence.450

Beyond NMDA receptor channel blockers, drugs that selectively target NMDA receptors that contain a specific subunit provide a potential avenue for treating SUDs. NMDA receptors are composed of two GluN1 subunits and combinations of GluN2A, GluN2B, GluN2C, and GluN2D subunits. Glutamate binds to the GluN2 subunits while the co-agonist glycine binds to the GluN1 subunits.432 Rats prenatally exposed to METH have increased expression of GluN1 and GluN2B subunit, an effect that emerges during adulthood.451 METH treatment leads to differential GluN subunit expression in regions of the dorsal striatum, with upregulated GluN1 and GluN2A expression observed in dorsolateral striatum, and downregulated GluN2A and GluN2B expression observed in dorsomedial striatum.452 METH CPP also upregulates GluN2B-containing NMDA receptors in hippocampus,453 and METH self-administration increases GluN2B expression in medial prefrontal cortex.454

The GluN2B subunit has received increased attention as GluN2B-selective antagonists prevent reinstatement of nicotine and heroin seeking in animals.455,456 While the contribution of GluN2B-containing NMDA receptors to METH relapse-like behavior has not been elucidated, antagonism of GluN2B-containing NMDA receptors fails to alter the rate of extinction of METH self-administration.457 However, GluN2B-selective antagonists attenuate METH-induced locomotor activity and the acquisition and the expression of METH-induced behavioral sensitization.458,459 Additionally, one GluN2B-selective antagonist blocks the acquisition, but not the expression of METH CPP in rats.460

Metabotropic Glutamate Receptor Ligands

Similar to ionotropic glutamate receptors, METH alters the expression of mGluRs. Specifically, extended access to METH leads to decreased expression of mGluR2/3 in nucleus accumbens and striatum.461 As these receptors are inhibitory, mGluR2/3 agonists should be efficacious in blunting the effects of METH. Direct stimulation of mGluR2 or administration of mGluR2 positive allosteric modulators decrease METH-induced hyperactivity.462,463 An mGluR2 agonist selectively decreases the reinforcing efficacy of METH without altering food-maintained responding.464 However, while an mGluR2/3 agonist blunts reinstatement of METH-seeking, it also decreases reinstatement of food-seeking behavior.465 In contrast to mGluR2 agonists, an mGluR2 antagonist ameliorates depressive-like behavior resulting from METH withdrawal.466

Although there is no evidence for mGluR1 involvement in the incubation of METH craving,467 selective mGluR1 antagonists block METH-induced increases in locomotor activity and disruptions in prepulse inhibition,468,469 and decrease METH self-administration and choice for METH over food.470 Likewise, blocking mGluR5 selectively decreases METH self-administration.471 A positive allosteric modulator of mGluR5 reverses METH-induced deficits in recognition memory.472 However, negative allosteric modulators of mGluR5, but not mGluR1, attenuate the expression of METH CPP.473

N-Acetylcysteine

Instead of targeting a specific receptor, some research has focused on restoring glutamate homeostasis with N-acetylcysteine, a medication used to reverse acetaminophen overdose.474,475 Loss of glutamate homeostasis is proposed to be a major driving force of addiction.476 In animals, N-acetylcysteine blocks the development of behavioral sensitization induced by METH, which corresponds to the ability of N-acetylcysteine to prevent METH-induced depletion of dopamine in striatum.477 However, N-acetylcysteine fails to alter METH self-administration and drug-induced reinstatement of METH seeking in rats.158 Also, N-acetylcysteine, whether administered alone or in conjunction with naltrexone, is ineffective at reducing METH craving, METH use, or severity of dependence in those that are dependent on METH;478,479 but see.480

Anticonvulsant Drugs

Anticonvulsant drugs (ie, antiepileptics) decrease excitatory neurotransmission through several mechanisms: blocking sodium or calcium channels, blocking glutamatergic receptors, and/or stimulating GABAergic receptors.481 One commonly prescribed anticonvulsant is gabapentin, which is also used as a nerve pain medication.482 Gabapentin prevents membrane trafficking of the ⍺2δ subunit of voltage-gated calcium channels as opposed to blocking these channels directly.483 Gabapentin attenuates locomotor activity, locomotor sensitization, and CPP following METH administration.245,434,484 However, this drug fails to reduce METH use in dependent individuals.431 An early study reported that combination treatment of oral gabapentin and intravenous infusion of flumazenil, a GABA receptor antagonist, decreases drug cravings and METH use,485 but a later study did not replicate this initial finding.486

Topiramate has multiple mechanisms of action, blocking voltage-gated sodium channels and glutamate AMPA receptors, potentiating GABA transmission, and modulating L-type calcium channels.487 Topiramate has shown some promise in treating individuals with cocaine dependence.488,489 Specific to METH, topiramate decreases the desire to use METH,490 and increases drug-free urine samples in METH-dependent individuals.491,492; but see490,493 Inconsistencies are reported concerning topiramate’s effects on cognition. Topiramate increases concentration but impairs perceptual-motor in a digit symbol substitution test.494 This finding is consistent with cognitive deficits observed in cocaine-dependent individuals receiving combination topiramate/methadone therapy.495

Gamma-vinyl GABA (vigabatrin) increases inhibition in the nervous system by inhibiting GABA transaminase, the enzyme that converts GABA into glutamate.496 As vigabatrin decreases METH-induced increases in dopamine,497 this drug has received some attention. Vigabatrin blocks reinstatement of METH CPP in rats.498 Although vigabatrin does not alter subjective effects of METH,499 it increases the rate of methamphetamine-negative urine samples in individuals that use stimulants, including METH.500,501 These results are promising and indicate that increasing GABAergic transmission is a viable target for treating MUD.

Calcium Channel Blockers

As discussed above, some anticonvulsants work by blocking voltage-gated calcium channels or by preventing membrane trafficking of these channels (eg, gabapentin). Calcium channel blockers are also used to treat hypertension.502 Acute METH administration inhibits L-type calcium channels, but chronic administration leads to an upregulation of such channels.503 Relatedly, L-type calcium channels are upregulated in regions such as frontal cortex and limbic forebrain following CPP of various drugs, including METH,504 with upregulation being dependent on activation of dopamine receptors.245 Voltage-gated calcium channel blockers reduce METH-induced neuronal damage,505 and they delay the onset of and reduce the severity of METH-induced stereotypies and self-injurious behavior.506–508 Voltage-gated calcium channel blockers also decrease METH CPP and attenuate cue-induced reinstatement.509–512

In humans, isradipine decreases positive subjective effects of METH and decreases craving for METH;513 however, isradipine’s effects are dependent on when it is administered. Specifically, isradipine alters the subjective effects of METH when it is administered after placebo, but not when it is administered first.514 Given the promising results obtained with animals, future studies examining other calcium channel blockers on the direct reinforcing effects of METH in dependent individuals are warranted.

Sigma Receptor Ligands

Sigma receptors were once proposed to be a type of opioid receptor, but are now known to be a type of chaperone protein located in the endoplasmic reticulum of cells.515 One important mechanism of sigma receptors is regulating calcium homeostasis.516 As calcium channel blockers show some efficacy in decreasing the reinforcing effects of METH (see above), some have investigated the role of sigma receptor ligands on METH-related behaviors. Sigma 1 receptor antagonists attenuate the locomotor stimulant, neurotoxic, and hyperthermic effects METH.517–519 A similar effect is observed with the sigma receptor ligand CM156.520 Low doses of sigma receptor agonists potentiate METH-induced hyperactivity, whereas higher doses blunt locomotor activity.521,522 More recent work shows that the sigma 1 receptor ligand PD144418 and the sigma 2 receptor ligand YUN-252 dose dependently decrease METH-induced hyperactivity.523 Work is needed to determine if sigma receptors regulate the direct reinforcing effects of METH.

Cannabinoid Receptor Ligands

The endocannabinoid system has two receptor subtypes: CB1 and CB2, which are both coupled to Gi proteins; the most well-known exogenous cannabinoids are delta-9-tetrahydrocannibinol (THC) and cannabidiol (CBD), found in cannabis plants.524 Similar to psychedelic hallucinogens, there is increased interest in testing the efficacy of cannabinoids for multiple conditions, including fibromyalgia and chronic pain,525,526 mental disorders,527 and SUDs.528

While THC prevents METH-induced neurotoxicity,529 pretreatment with THC significantly potentiates locomotor activity in mice and ataxia and stereotypies in rabbits.530–532 THC attenuates drug-induced reinstatement but potentiates cue-induced reinstatement; however, when THC is administered repeatedly during extinction or 24 hours before the reinstatement test, reinstatement is blunted.533 Similarly, a CB1 positive allosteric modulator decreases reinstatement of METH seeking.534 Like THC, the full CB1 agonist WIN55, 212–2 prevents METH-induced neurotoxicity, but it also blunts METH-induced hyperactivity.535; but see536 One concerning finding is that a CB1 agonist potentiates METH CPP.537

As CBD is non-intoxicating,538 there is growing interest in studying its efficacy in multiple conditions.539 CBD decreases METH-induced behavioral sensitization,540 the reinforcing efficacy of METH and reinstatement of METH-seeking behavior,541 and METH CPP.542,543 Currently, there are no published clinical trials testing CBD in METH-dependent individuals. However, CBD does not appear to be an effective treatment for cocaine use disorder.544,545

Cannabinoid receptor antagonists decrease METH-induced behavioral sensitization, METH self-administration, and reinstatement of METH seeking.533,534,546–548; but see549 Additionally, the CB1 antagonist rimonabant administered after each conditioning session prevents acquisition of METH CPP, and rimonabant prevents the expression and the reinstatement of METH CPP.550,551 Rimonabant also improves METH-induced deficits in novel object recognition memory.552 Unfortunately, CB1 antagonists like rimonabant are associated with increased suicidal ideation,553 which greatly limits their therapeutic use.

Endocannabinoids are metabolized by fatty acid amide hydrolase and monoacylglycerol lipase.554 The fatty acid amide hydrolase inhibitor URB597 blunts the anxiogenic and the depressive effects of METH withdrawal.555 Preventing the hydrolysis of the endocannabinoid 2-arachidonoylglycerol (2-AG) inhibits cue- and stress-induced reinstatement of METH seeking, an effect not observed following administration of URB597.556 As such, monoacylglycerol lipase inhibitors merit further examination as a treatment for MUD.

Angiotensin Receptor Antagonists and Angiotensin-Converting Enzyme (ACE) Inhibitors

Angiotensin is a hormone that primarily regulates blood pressure and promotes sodium retention in the kidneys; angiotensin I is converted to angiotensin II by angiotensin-converting enzyme (ACE).557 Angiotensin receptor antagonists and ACE inhibitors, like some calcium channel blockers, are used to treat hypertension.557 In the brain, there is evidence that the renin-angiotensin system influences dopamine release. Specifically, angiotensin II induces dopamine release while dopamine depletion leads to an upregulation of angiotensin receptors.558 Therefore, there is recent interest in using angiotensin receptor antagonists or angiotensin-converting enzyme inhibitors to blunt the effects of METH, and early reports are promising thus far. The angiotensin-converting enzyme inhibitor perindopril attenuates METH-induced hyperlocomotion in animals, with a similar effect observed following selective blockade of angiotensin II type 1 receptors.559,560 Blocking angiotensin II receptors also reverses METH-induced increases in body temperature, rescues METH-induced deficits in memory and cognition, and decreases METH self-administration and reinstatement in rats.560–562 Perindopril also decreases subjective effects of METH, but interestingly, these effects disappear at a higher dose of perindopril.563,564 As argued previously, the inability of a higher dose of perindopril to alter subjective effects of METH may be due to inhibition of ACE in more brain regions compared to lower doses of perindopril.564

Oxytocin

Oxytocin is a hormone involved in various physiological processes (eg, milk let-down reflex) and maternal/social behaviors.565 METH is known to affect oxytocin levels, as repeated METH administration increases blood plasma oxytocin levels,566 and causes upregulation of oxytocin receptors in amygdala and hypothalamus, but not in nucleus accumbens or dorsal striatum.567 However, compulsive METH seeking leads to upregulated oxytocin gene expression in nucleus accumbens.568 Oxytocin inhibits METH-induced hyperactivity, METH self-administration, and reinstatement of METH seeking.569 Adolescent treatment of oxytocin attenuates the reinforcing efficacy of METH and reduces drug-induced reinstatement of METH seeking during adulthood.570 Repeated administration of oxytocin during extinction blunts reinstatement of METH CPP.571 The ability of oxytocin to decrease the addictive-like properties of METH may be related to attenuated glutamate release in medial prefrontal cortex and potentiated glutamate release in hippocampus following co-administration of METH and oxytocin.572 While oxytocin decreases reinstatement of METH seeking and incubation of craving in a self-administration paradigm,275,573–576 decreased reinstatement of sucrose seeking is also observed.564

In a clinical setting, oxytocin increases group therapy attendance without significantly altering METH craving or METH use.577 However, there is evidence of decreased craving and anxiety in METH-dependent individuals receiving 4 weeks of oxytocin treatment.578

Phosphodiesterase (PDE) Inhibitors

Phosphodiesterase (PDE) inhibitors are used to treat several health conditions, such as chronic obstructive pulmonary disease, pulmonary arterial hypertension, psoriasis, and erectile dysfunction.579 PDE is an enzyme that degrades cyclic nucleotides, including cyclic adenosine monophosphate (cAMP), an important second messenger involved in intracellular signal transduction. Drugs are known to increase cAMP levels in brain regions associated with reinforcement,580,581 but the degradation of cAMP by PDE is important mediator of the addiction process. Upregulation of cAMP is proposed to lead to tolerance to a drug’s reinforcing effects whereas the breakdown of cAMP promotes increased drug seeking.580 As such, PDE inhibitors may be an effective treatment for reducing the reinforcing effects of METH and recurrence of METH use.

In rodents, PDE inhibitors attenuate METH-induced hyperactivity, stereotypies, and cognitive deficits;581–587 but see.588 The non-selective PDE inhibitor ibudilast attenuates METH self-administration and drug- and stress-induced reinstatement of METH seeking,589,590 and the PDE 4 inhibitor roflumilast reduces incubation of METH craving following forced abstinence.591 While the PDE 4B inhibitor A33 decreases METH self-administration, it fails to block relapse-like behavior.592 This result suggest that PDE 4A inhibitors may be more efficacious in preventing recurrence of substance use. So far, only ibudilast has been tested in dependent individuals. Ibudilast does not facilitate METH abstinence,593 but it decreases peripheral markers of inflammation associated with METH use.594 This latter finding may partially account for the cognitive improvements observed in animals following administration of PDE inhibitors. More work is needed to determine if selective PDE inhibitors are a viable treatment for ameliorating cognitive deficits associated with long-term METH use.

Discussion

Summary of Findings

As covered in the current review, numerous molecular targets have been examined as putative treatments for MUD. Multiple treatments that blunt dopaminergic/noradrenergic neurotransmission reduce hyperactivity stemming from METH use (eg, antagonists at dopamine, muscarinic, and sigma receptors; agonists at GABA receptors; calcium channel blockers). Numerous pharmacological manipulations reduce the rewarding/reinforcing effects of METH in animals, but have little efficacy in treating MUD (eg, modafinil, antipsychotic medications, baclofen). However, there is potential that these treatments, plus medications like atomoxetine and varenicline, can be used to ameliorate the psychotomimetic effects of METH and/or reverse METH-induced cognitive deficits. Promising pharmacotherapies that have been tested in animals only include VMAT-2 inhibitors, mGluR ligands, and trace amine-associated receptor agonists.

Future Directions

In addition to continuing research with promising pharmacotherapies described above, there are important considerations that need to be taken into account when screening potential treatments for SUDs. These future directions will be discussed first.

Preclinical Screening Considerations

One major limitation to many of the drug self-administration studies highlighted in this review is the use of a fixed ratio (FR) schedules to measure the reinforcing effects of METH. Dissociating a drug’s locomotor effects from its reinforcing effects is difficult when FR schedules, as well as progressive ratio schedules, are used because animals may show increased responding for a drug like METH as they become more hyperactive. Similarly, if a potential pharmacotherapy decreases responding for METH, this effect becomes more difficult to interpret if the pharmacological manipulation suppresses METH-induced hyperactivity. This caveat may account for why some interventions decrease the reinforcing effects of METH in animals but fail to improve treatment outcomes in METH-dependent individuals (eg, gabapentin). To avoid this limitation, schedules that control the rate of reinforcement, such as variable interval or second-order schedules, can be used.470,595

Another major weakness of preclinical models assessing METH-dependence-like behavior is the near exclusive use of male subjects as sex/gender are shown to modulate the effects of METH in an individual and modulate treatment responses for MUD.596 For example, adolescent male, but not female, rats exposed to repeated nicotine treatments self-administer more METH,344,346 and preadolescent exposure to nicotine enhances oral METH self-administration in female, but not male, adolescent rats.597 Also, oxytocin is more effective at reducing the reinforcing efficacy of METH and cue-induced reinstatement in females relative to males.573

Given the increased prevalence of METH use in pregnant women,598 increased work is needed to determine if pharmacotherapies are safe for these individuals and do not cause serious, long-term side effects in children. For example, mice prenatally exposed to modafinil display increased anxiety-like behavior and show greater METH-induced behavioral sensitization.599 These results suggest that in utero exposure to modafinil may put individuals at risk for future addictive-like behaviors. In addition, nicotine treatment may be problematic for pregnant women as rats exposed to nicotine in utero respond more for METH in both fixed ratio and progressive ratio schedules.600,601

Pharmacogenetic Approaches

Pharmacogenetics incorporates one’s genome when prescribing a medication. For example, some individuals prescribed amitriptyline may not respond well to treatment if they have specific alleles for cytochrome P450 enzymes.602 Additionally, individuals that have a cytochrome P450-2D6 phenotype characterized by more efficient METH metabolism show increased METH-induced neurocognitive impairment;603 conversely, individuals classified as low cytochrome P450-2D6 metabolizers are less likely to develop METH-related heart failure or to develop METH dependence.604,605 Specific to treatments for psychostimulant use disorders, individuals with cocaine use disorder that have a single nucleotide polymorphism of the serotonin 5-HT3 receptor show a greater percentage of cocaine-free urine samples during ondansetron treatment.606 Additionally, individuals with genetically higher levels of DAT respond better to disulfiram (Antabuse) for cocaine dependence,607 and individuals with genetically lower levels of the dopamine metabolizing enzyme dopamine beta-hydroxylase are less likely to use cocaine during levodopa treatment.608

Currently, the only study examining pharmacogenetic approaches to treating MUD found that a single nucleotide polymorphism of the mu opioid receptor (A118G) does not predict response to naltrexone.609 A recent meta-analysis reported that two genes that are strongly related to MUD are those that encode fatty acid amide hydrolase and brain derived neurotrophic factor, a protein that is critical for neuron growth and plasticity.610 Considering one’s genetic makeup can allow for more tailored treatment options that can help an individual better manage cravings and enable them to maintain abstinence long term.

Conclusion

Currently, there is no single pharmacotherapy that prevents continuation of METH use or recurrence of METH use following abstinence. Given the numerous factors that influence the addiction process,328 such medication may not exist, but there appears to be pharmacological options that can be utilized in conjunction with other therapeutic approaches, leading to positive outcomes for the individual (eg, oxytocin to increase group therapy attendance). With increasing MUD rates, continued research is necessary to find treatment options that can (a) reduce METH use, (b) ameliorate adverse events associated with METH use (eg, psychotomimetic-like effects, cognitive disturbances), and/or (c) reduce the likelihood of recurrence of METH use following therapy.

Acknowledgments

The current work was supported by NIH grant R15DA047610.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Checkley SA. A new distinction between the euphoric and the anti-depressant effects of methylamphetamine. Br J Psychiatry. 1978;133:416–423. doi:10.1192/bjp.133.5.416

2. 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(2):245–258. doi:10.1002/cpt1971122part1245

3. Mohs RC, Tinklenberg JR, Roth WT, Kopell BS. Methamphetamine and diphenhydramine effects on the rate of cognitive processing. Psychopharmacology. 1978;59(1):13–19. doi:10.1007/BF00428024

4. Buchanan JF, Brown CR. ‘Designer drugs’. A problem in clinical toxicology. Med Toxicol Adverse Drug Exp. 1988;3(1):1–17. doi:10.1007/BF03259928

5. Haning W, Goebert D. Electrocardiographic abnormalities in methamphetamine abusers. Addiction. 2007;102 Suppl 1:70–75. doi:10.1111/j.1360-0443.2006.01776.x

6. Chiang M, Lombardi D, Du J, et al. Methamphetamine-associated psychosis: clinical presentation, biological basis, and treatment options. Hum Psychopharmacol. 2019;34(5):e2710. doi:10.1002/hup.2710

7. Glasner-Edwards S, Mooney LJ. Methamphetamine psychosis: epidemiology and management. CNS Drugs. 2014;28(12):1115–1126. doi:10.1007/s40263-014-0209-8

8. Comer SD, Hart CL, Ward AS, Haney M, Foltin RW, Fischman MW. Effects of repeated oral methamphetamine administration in humans. Psychopharmacology. 2001;155(4):397–404. doi:10.1007/s002130100727

9. Kono J, Miyata H, Ushijima S, et al. Nicotine, alcohol, methamphetamine, and inhalant dependence: a comparison of clinical features with the use of a new clinical evaluation form. Alcohol. 2001;24(2):99–106. doi:10.1016/s0741-8329(01)00143-4

10. Perez-Reyes M, White WR, McDonald SA, et al. Clinical effects of daily methamphetamine administration. Clin Neuropharmacol. 1991;14(4):352–358. doi:10.1097/00002826-199108000-00007

11. Darke S, Kaye S, McKetin R, Duflou J. Major physical and psychological harms of methamphetamine use. Drug Alcohol Rev. 2008;27(3):253–262. doi:10.1080/09595230801923702

12. Clague J, Belin TR, Shetty V. Mechanisms underlying methamphetamine-related dental disease. J Am Dent Assoc. 2017;148(6):377–386. doi:10.1016/j.adaj.2017.02.054

13. Ho EL, Josephson SA, Lee HS, Smith WS. Cerebrovascular complications of methamphetamine abuse. Neurocrit Care. 2009;10(3):295–305. doi:10.1007/s12028-008-9177-5

14. Molitor F, Ruiz JD, Flynn N, Mikanda JN, Sun RK, Anderson R. Methamphetamine use and sexual and injection risk behaviors among out-of-treatment injection drug users. Am J Drug Alcohol Abuse. 1999;25(3):475–493. doi:10.1081/ada-100101874

15. Yu Q, Larson DF, Watson RR. Heart disease, methamphetamine and AIDS. Life Sci. 2003;73(2):129–140. doi:10.1016/s0024-3205(03)00260-1

16. Weisheit R. Making methamphetamine. J Rural Soc Sci. 2008;23(2):78–107.

17. Onoka I, Banyika AT, Banerjee PN, Makangara JJ, Dujourdy L. A review of the newly identified impurity profiles in methamphetamine seizures. Forensic Sci Int Synerg. 2020;2:194–205. doi:10.1016/j.fsisyn.2020.06.004

18. Cruickshank CC, Dyer KR. A review of the clinical pharmacology of methamphetamine. Addiction. 2009;104(7):1085–1099. doi:10.1111/j.1360-0443.2009.02564.x

19. Kevil CG, Goeders NE, Woolard MD, et al. Methamphetamine use and cardiovascular disease. Arterioscler Thromb Vasc Biol. 2019;39(9):1739–1746. doi:10.1161/ATVBAHA.119.312461

20. Volkow ND, Fowler JS, Wang GJ, et al. Distribution and pharmacokinetics of methamphetamine in the human body: clinical implications. PLoS One. 2010;5(12):e15269. doi:10.1371/journal.pone.0015269

21. Lin LY, Di Stefano EW, Schmitz DA, et al. Oxidation of methamphetamine and methylenedioxymethamphetamine by CYP2D6. Drug Metab Dispos. 1997;25(9):1059–1064.

22. Cho AK, Melega WP. Patterns of methamphetamine abuse and their consequences. J Addict Dis. 2002;21(1):21–34. doi:10.1300/j069v21n01_03

23. Li L, Everhart T, Jacob Iii P, Jones R, Mendelson J. Stereoselectivity in the human metabolism of methamphetamine. Br J Clin Pharmacol. 2010;69(2):187–192. doi:10.1111/j.1365-2125.2009.03576.x

24. Cook CE, Jeffcoat AR, Hill JM, et al. Pharmacokinetics of methamphetamine self-administered to human subjects by smoking S-(+)-methamphetamine hydrochloride. Drug Metab Dispos. 1993;21(4):717–723.

25. Cook CE, Jeffcoat AR, Sadler BM, et al. Pharmacokinetics of oral methamphetamine and effects of repeated daily dosing in humans. Drug Metab Dispos. 1992;20(6):856–862.

26. Harris DS, Boxenbaum H, Everhart ET, Sequeira G, Mendelson JE, Jones RT. The bioavailability of intranasal and smoked methamphetamine. Clin Pharmacol Ther. 2003;74(5):475–486. doi:10.1016/j.clpt.2003.08.002

27. Schepers RJ, Oyler JM, Joseph RE, Cone EJ, Moolchan ET, Huestis MA. Methamphetamine and amphetamine pharmacokinetics in oral fluid and plasma after controlled oral methamphetamine administration to human volunteers. Clin Chem. 2003;49(1):121–132. doi:10.1373/49.1.121

28. Nickell JR, Siripurapu KB, Vartak A, Crooks PA, Dwoskin LP. The vesicular monoamine transporter-2: an important pharmacological target for the discovery of novel therapeutics to treat methamphetamine abuse. Adv Pharmacol. 2014;69:71–106. doi:10.1016/B978-0-12-420118-7.00002-0

29. Johnson RA, Eshleman AJ, Meyers T, Neve KA, Janowsky A. [3H]substrate- and cell-specific effects of uptake inhibitors on human dopamine and serotonin transporter-mediated efflux. Synapse. 1998;30(1):97–106. doi:10.1002/(SICI)1098-2396(199809)30:1<97::AID-SYN12>3.0.CO;2-M

30. Egashira T, Yamamoto T, Yamanaka Y. Effects of d-methamphetamine on monkey brain monoamine oxidase, in vivo and in vitro. Jpn J Pharmacol. 1987;45(1):79–88. doi:10.1254/jjp.45.79

31. Volkow ND, Fowler JS, Wang GJ. Role of dopamine in drug reinforcement and addiction in humans: results from imaging studies. Behav Pharmacol. 2002;13(5–6):355–366. doi:10.1097/00008877-200209000-00008

32. NIDA. What are the long-term effects of methamphetamine misuse? National Institute on Drug Abuse website; 2022. Available from: https://nida.nih.gov/publications/research-reports/methamphetamine/what-are-long-term-effects-methamphetamine-misuse. Accessed April 20, 2024.

33. European Monitoring Centre for Drugs and Drug Addiction. Signals of spreading methamphetamine use in Europe. Available from: https://www.emcdda.europa.eu/publications/eu-drug-markets/methamphetamine/use-in-europe_en. Accessed April 10, 2024.

34. Han B, Compton WM, Jones CM, Einstein EB, Volkow ND. Methamphetamine use, methamphetamine use disorder, and associated overdose deaths among US adults. JAMA Psychiatry. 2021;78(12):1329–1342. doi:10.1001/jamapsychiatry.2021.2588

35. Papamihali K, Collins D, Karamouzian M, Purssell R, Graham B, Buxton J. Crystal methamphetamine use in British Columbia, Canada: a cross-sectional study of people who access harm reduction services. PLoS One. 2021;16(5):e0252090. doi:10.1371/journal.pone.0252090

36. Zhang B, Yan X, Li Y, Zhu H, Lu Z, Jia Z. Trends in methamphetamine use in the mainland of China, 2006-2015. Front Public Health. 2022;10:852837. doi:10.3389/fpubh.2022.852837

37. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. American Psychiatric Association; 2013.

38. World Health Organization. International statistical classification of diseases and related health problems; 2019. https://icd.who.int/. Accessed August 21, 2024.

39. NIDA. Drug misuse and addiction. National Institute on Drug Abuse website; 2024. https://nida.nih.gov/publications/drugs-brains-behavior-science-addiction/drug-misuse-addiction. Accessed April 21, 2024.

40. Nicosia N, Pacula RL, Kilmer B, Lundberg R, Chiesa J The economic cost of methamphetamine use in the United States, 2005. The RAND Corporation 2009. https://www.rand.org/content/dam/rand/pubs/monographs/2009/RAND_MG829.pdf. Accessed April 4, 2024.

41. Tait RJ, Whetton S, Shanahan M, et al. Quantifying the societal cost of methamphetamine use to Australia. Int J Drug Policy. 2018;62:30–36. doi:10.1016/j.drugpo.2018.08.015

42. Zhao SX, Deluna A, Kelsey K, et al. Socioeconomic burden of rising methamphetamine-associated heart failure hospitalizations in California from 2008 to 2018. Circ Cardiovasc Qual Outcomes. 2021;14(7):e007638. doi:10.1161/CIRCOUTCOMES.120.007638

43. Rusyniak DE. Neurologic manifestations of chronic methamphetamine abuse. Neurol Clin. 2011;29(3):641–655. doi:10.1016/j.ncl.2011.05.004

44. Rommel N, Rohleder NH, Koerdt S, et al. Sympathomimetic effects of chronic methamphetamine abuse on oral health: a cross-sectional study. BMC Oral Health. 2016;16(1):59. doi:10.1186/s12903-016-0218-8

45. Salamanca SA, Sorrentino EE, Nosanchuk JD, Martinez LR. Impact of methamphetamine on infection and immunity. Front Neurosci. 2015;8:445. doi:10.3389/fnins.2014.00445

46. Akiyama K, Ujike H, Sakai K, Shimizu Y, Kodama M, Kuroda S. Effect of 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo(f)quinoxaline on methamphetamine- and cocaine-induced behavioral sensitization. Pharmacol Biochem Behav. 1998;61(4):419–426. doi:10.1016/s0091-3057(98)00121-x

47. Fujiwara Y, Kazahaya Y, Nakashima M, Sato M, Otsuki S. Behavioral sensitization to methamphetamine in the rat: an ontogenic study. Psychopharmacology. 1987;91(3):316–319. doi:10.1007/BF00518183

48. Hashimoto T, Koda H, Paik IH, Kuriyama K. Possible involvement of supersensitivity in cerebral ?-adrenergic and dopaminergic receptors in the occurrence of sensitization to d-methamphetamine in mice. Neurochem Int. 1990;16(1):17–25. doi:10.1016/0197-0186(90)90119-e

49. Peachey E, Rogers B, Brien JF, Maclean A, Rogers D. Measurement of acute and chronic behavioural effects of methamphetamine in the mouse. Psychopharmacology. 1976;48(3):271–275. doi:10.1007/BF00496860

50. Tatsuta T, Kitanaka N, Kitanaka J, Morita Y, Takemura M. Lack of effect of anticonvulsant topiramate on methamphetamine-induced stereotypy and rewarding property in mice. Pharmacol Biochem Behav. 2007;87(1):48–55. doi:10.1016/j.pbb.2007.03.019

51. Piazza PV, Deminière JM, Le Moal M, Simon H. Factors that predict individual vulnerability to amphetamine self-administration. Science. 1989;245(4925):1511–1513. doi:10.1126/science.2781295

52. Piazza PV, Deminiere JM, le Moal M, Simon H. Stress- and pharmacologically-induced behavioral sensitization increases vulnerability to acquisition of amphetamine self-administration. Brain Res. 1990;514(1):22–26. doi:10.1016/0006-8993(90)90431-a

53. Yap JJ, Miczek KA. Social defeat stress, sensitization, and intravenous cocaine self-administration in mice. Psychopharmacology. 2007;192(2):261–273. doi:10.1007/s00213-007-0712-4

54. Bardo MT, Bevins RA. Conditioned place preference: what does it add to our preclinical understanding of drug reward? Psychopharmacology. 2000;153(1):31–43. doi:10.1007/s002130000569

55. Tzschentke TM. Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol. 2007;12(3–4):227–462. doi:10.1111/j.1369-1600.2007.00070.x

56. Childs E, de Wit H. Amphetamine-induced place preference in humans. Biol Psychiatry. 2009;65(10):900–904. doi:10.1016/j.biopsych.2008.11.016

57. Bardo MT, Horton DB, Yates JY. Conditioned place preference as a preclinical model for screening pharmacotherapies for drug abuse. In: Markgraf CG, Hudzik TJ, Compton DR, editors. Nonclinical Assessment of Abuse Potential for New Pharmaceuticals. Academic Press; 2015:151–196.

58. Panlilio LV, Goldberg SR. Self-administration of drugs in animals and humans as a model and an investigative tool. Addiction. 2007;102(12):1863–1870. doi:10.1111/j.1360-0443.2007.02011.x

59. Balster RL, Schuster CR. A comparison of d-amphetamine, l-amphetamine, and methamphetamine self-administration in rhesus monkeys. Pharmacol Biochem Behav. 1973;1(1):67–71. doi:10.1016/0091-3057(73)90057-9

60. Carney JM, Landrum RW, Cheng MS, Seale TW. Establishment of chronic intravenous drug self-administration in the C57BL/6J mouse. Neuroreport. 1991;2(8):477–480. doi:10.1097/00001756-199108000-00017

61. Glick SD, Maisonneuve IM, Dickinson HA. 18-MC reduces methamphetamine and nicotine self-administration in rats. Neuroreport. 2000;11(9):2013–2015. doi:10.1097/00001756-200006260-00041

62. Stefanski R, Ladenheim B, Lee SH, Cadet JL, Goldberg SR. Neuroadaptations in the dopaminergic system after active self-administration but not after passive administration of methamphetamine. Eur J Pharmacol. 1999;371(2–3):123–135. doi:10.1016/s0014-2999(99)00094-1

63. Kitamura O, Wee S, Specio SE, Koob GF, Pulvirenti L. Escalation of methamphetamine self-administration in rats: a dose-effect function. Psychopharmacology. 2006;186(1):48–53. doi:10.1007/s00213-006-0353-z

64. Rogers JL, De Santis S, See RE. Extended methamphetamine self-administration enhances reinstatement of drug seeking and impairs novel object recognition in rats. Psychopharmacology. 2008;199(4):615–624. doi:10.1007/s00213-008-1187-7

65. Schwendt M, Rocha A, See RE, Pacchioni AM, McGinty JF, Kalivas PW. Extended methamphetamine self-administration in rats results in a selective reduction of dopamine transporter levels in the prefrontal cortex and dorsal striatum not accompanied by marked monoaminergic depletion. J Pharmacol Exp Ther. 2009;331(2):555–562. doi:10.1124/jpet.109.155770

66. Edwards S, Koob GF. Escalation of drug self-administration as a hallmark of persistent addiction liability. Behav Pharmacol. 2013;24(5–6):356–362. doi:10.1097/FBP.0b013e3283644d15

67. Ranaldi R, Poeggel K. Baclofen decreases methamphetamine self-administration in rats. Neuroreport. 2002;13(9):1107–1110. doi:10.1097/00001756-200207020-00007

68. Roth ME, Carroll ME. Sex differences in the acquisition of IV methamphetamine self-administration and subsequent maintenance under a progressive ratio schedule in rats. Psychopharmacology. 2004;172(4):443–449. doi:10.1007/s00213-003-1670-0

69. Hodos W. Progressive ratio as a measure of reward strength. Science. 1961;134(3483):943–944. doi:10.1126/science.134.3483.943

70. Haney M, Comer SD, Ward AS, Foltin RW, Fischman MW. Factors influencing marijuana self-administration by humans. Behav Pharmacol. 1997;8(2–3):101–112.

71. Marks KR, Lile JA, Stoops WW, Glaser PE, Hays LR, Rush CR. Separate and combined effects of naltrexone and extended-release alprazolam on the reinforcing, subject-rated, and cardiovascular effects of methamphetamine. J Clin Psychopharmacol. 2016;36(3):213–221. doi:10.1097/JCP.0000000000000488

72. Griffiths RR, Wurster RM, Brady JV. Discrete-trial choice procedure: effects of naloxone and methadone on choice between food and heroin. Pharmacol Rev. 1975;27(3):357–365.

73. Thomsen M, Fink-Jensen A, Woldbye DP, et al. Effects of acute and chronic aripiprazole treatment on choice between cocaine self-administration and food under a concurrent schedule of reinforcement in rats. Psychopharmacology. 2008;201(1):43–53. doi:10.1007/s00213-008-1245-1

74. Cole-Harding S, de Wit H. Self-administration of pentobarbital in light and moderate alcohol drinkers. Pharmacol Biochem Behav. 1992;43(2):563–569. doi:10.1016/0091-3057(92)90191-h

75. Foltin RW, Fischman MW. Effects of methadone or buprenorphine maintenance on the subjective and reinforcing effects of intravenous cocaine in humans. J Pharmacol Exp Ther. 1996;278(3):1153–1164.

76. Foltin RW, Fischman MW. Self-administration of cocaine by humans: choice between smoked and intravenous cocaine. J Pharmacol Exp Ther. 1992;261(3):841–849.

77. Rush CR, Stoops WW, Lile JA, Glaser PE, Hays LR. Subjective and physiological effects of acute intranasal methamphetamine during d-amphetamine maintenance. Psychopharmacology. 2011;214(3):665–674. doi:10.1007/s00213-010-2067-5

78. Trivedi MH, Walker R, Ling W, et al. Bupropion and naltrexone in methamphetamine use disorder. N Engl J Med. 2021;384(2):140–153. doi:10.1056/NEJMoa2020214

79. Wang B, Luo F, Zhang WT, Han JS. Stress or drug priming induces reinstatement of extinguished conditioned place preference. Neuroreport. 2000;11(12):2781–2784. doi:10.1097/00001756-200008210-00034

80. Shaham Y, Rajabi H, Stewart J. Relapse to heroin-seeking in rats under opioid maintenance: the effects of stress, heroin priming, and withdrawal. J Neurosci. 1996;16(5):1957–1963. doi:10.1523/JNEUROSCI.16-05-01957.1996

81. Stretch R, Gerber GJ. Drug-induced reinstatement of amphetamine self-administration behaviour in monkeys. Can J Psychol. 1973;27(2):168–177. doi:10.1037/h0082466

82. Grimm JW, Shaham Y, Hope BT. Effect of cocaine and sucrose withdrawal period on extinction behavior, cue-induced reinstatement, and protein levels of the dopamine transporter and tyrosine hydroxylase in limbic and cortical areas in rats. Behav Pharmacol. 2002;13(5–6):379–388. doi:10.1097/00008877-200209000-00011

83. Grimm JW, Hope BT, Wise RA, Shaham Y. Neuroadaptation. Incubation of cocaine craving after withdrawal. Nature. 2001;412:6843):141–142. doi:10.1038/35084134

84. Mehta TR, Monegro A, Nene Y, Fayyaz M, Bollu PC. Neurobiology of ADHD: a review. Curr Dev Disord Rep. 2019;6:235–240. doi:10.1007/s40474-019-00182-w

85. Faraone SV. The pharmacology of amphetamine and methylphenidate: relevance to the neurobiology of attention-deficit/hyperactivity disorder and other psychiatric comorbidities. Neurosci Biobehav Rev. 2018;87:255–270. doi:10.1016/j.neubiorev.2018.02.001

86. Stoops WW, Rush CR. Agonist replacement for stimulant dependence: a review of clinical research. Curr Pharm Des. 2013;19(40):7026–7035. doi:10.2174/138161281940131209142843

87. Jordan CJ, Cao J, Newman AH, Xi ZX. Progress in agonist therapy for substance use disorders: lessons learned from methadone and buprenorphine. Neuropharmacology. 2019;158:107609. doi:10.1016/j.neuropharm.2019.04.015

88. Logan DE, Marlatt GA. Harm reduction therapy: a practice-friendly review of research. J Clin Psychol. 2010;66(2):201–214. doi:10.1002/jclp.20669

89. Clemow DB, Walker DJ. The potential for misuse and abuse of medications in ADHD: a review. Postgrad Med. 2014;126(5):64–81. doi:10.3810/pgm.2014.09.2801

90. Sembower MA, Ertischek MD, Buchholtz C, Dasgupta N, Schnoll SH. Surveillance of diversion and nonmedical use of extended-release prescription amphetamine and oral methylphenidate in the United States. J Addict Dis. 2013;32(1):26–38. doi:10.1080/10550887.2012.759880

91. Schindler CW, Gilman JP, Panlilio LV, McCann DJ, Goldberg SR. Comparison of the effects of methamphetamine, bupropion, and methylphenidate on the self-administration of methamphetamine by rhesus monkeys. Exp Clin Psychopharmacol. 2011;19(1):1–10. doi:10.1037/a0022432

92. Schwienteck KL, Banks ML. Effects of 7-day continuous D-amphetamine, methylphenidate, and cocaine treatment on choice between methamphetamine and food in male rhesus monkeys. Drug Alcohol Depend. 2015;155:16–23. doi:10.1016/j.drugalcdep.2015.08.022

93. Baladi MG, Nielsen SM, Umpierre A, Hanson GR, Fleckenstein AE. Prior methylphenidate self-administration alters the subsequent reinforcing effects of methamphetamine in rats. Behav Pharmacol. 2014;25(8):758–765. doi:10.1097/FBP.0000000000000094

94. Rush CR, Stoops WW, Lile JA, Glaser PE, Hays LR. Physiological and subjective effects of acute intranasal methamphetamine during atomoxetine maintenance. Pharmacol Biochem Behav. 2011;100(1):40–47. doi:10.1016/j.pbb.2011.06.024

95. Rabiey A, Hassani-Abharian P, Farhad M, Moravveji AR, Akasheh G, Banafshe HR. Atomoxetine efficacy in methamphetamine dependence during methadone maintenance therapy. Arch Iran Med. 2019;22(12):692–698.

96. Broos N, Loonstra R, van Mourik Y, et al. Subchronic administration of atomoxetine causes an enduring reduction in context-induced relapse to cocaine seeking without affecting impulsive decision making. Addict Biol. 2015;20(4):714–723. doi:10.1111/adb.12168

97. Economidou D, Dalley JW, Everitt BJ. Selective norepinephrine reuptake inhibition by atomoxetine prevents cue-induced heroin and cocaine seeking. Biol Psychiatry. 2011;69(3):266–274. doi:10.1016/j.biopsych.2010.09.040

98. Janak PH, Bowers MS, Corbit LH. Compound stimulus presentation and the norepinephrine reuptake inhibitor atomoxetine enhance long-term extinction of cocaine-seeking behavior. Neuropsychopharmacology. 2012;37(4):975–985. doi:10.1038/npp.2011.281

99. Gerrard P, Malcolm R. Mechanisms of modafinil: a review of current research. Neuropsychiatr Dis Treat. 2007;3(3):349–364.

100. Tunstall BJ, Ho CP, Cao J, et al. Atypical dopamine transporter inhibitors attenuate compulsive-like methamphetamine self-administration in rats. Neuropharmacology. 2018;131:96–103. doi:10.1016/j.neuropharm.2017.12.006

101. Holtz NA, Lozama A, Prisinzano TE, Carroll ME. Reinstatement of methamphetamine seeking in male and female rats treated with modafinil and allopregnanolone. Drug Alcohol Depend. 2012;120(1–3):233–237. doi:10.1016/j.drugalcdep.2011.07.010

102. Reichel CM, See RE. Modafinil effects on reinstatement of methamphetamine seeking in a rat model of relapse. Psychopharmacology. 2010;210(3):337–346. doi:10.1007/s00213-010-1828-5

103. Reichel CM, See RE. Chronic modafinil effects on drug-seeking following methamphetamine self-administration in rats. Int J Neuropsychopharmacol. 2012;15(7):919–929. doi:10.1017/S1461145711000988

104. Hirate K, Kuribara H. Characteristics of the ambulation-increasing effect of GBR-12909, a selective dopamine uptake inhibitor, in mice. Jpn J Pharmacol. 1991;55(4):501–511. doi:10.1254/jjp.55.501

105. Carati C, Schenk S. Role of dopamine D1- and D2-like receptor mechanisms in drug-seeking following methamphetamine self-administration in rats. Pharmacol Biochem Behav. 2011;98(3):449–454. doi:10.1016/j.pbb.2011.02.010

106. Minzenberg MJ, Carter CS. Modafinil: a review of neurochemical actions and effects on cognition. Neuropsychopharmacology. 2008;33(7):1477–1502. doi:10.1038/sj.npp.1301534

107. De La Garza R, Zorick T, London ED, Newton TF. Evaluation of modafinil effects on cardiovascular, subjective, and reinforcing effects of methamphetamine in methamphetamine-dependent volunteers. Drug Alcohol Depend. 2010;106(2–3):173–180. doi:10.1016/j.drugalcdep.2009.08.013

108. Shearer J, Darke S, Rodgers C, et al. A double-blind, placebo-controlled trial of modafinil (200 mg/day) for methamphetamine dependence. Addiction. 2009;104(2):224–233. doi:10.1111/j.1360-0443.2008.02437.x

109. McGaugh J, Mancino MJ, Feldman Z, et al. Open-label pilot study of modafinil for methamphetamine dependence. J Clin Psychopharmacol. 2009;29(5):488–491. doi:10.1097/JCP.0b013e3181b591e0

110. Anderson AL, Li SH, Biswas K, et al. Modafinil for the treatment of methamphetamine dependence. Drug Alcohol Depend. 2012;120(1–3):135–141. doi:10.1016/j.drugalcdep.2011.07.007

111. Heinzerling KG, Swanson AN, Kim S, et al. Randomized, double-blind, placebo-controlled trial of modafinil for the treatment of methamphetamine dependence. Drug Alcohol Depend. 2010;109(1–3):20–29. doi:10.1016/j.drugalcdep.2009.11.023

112. Hester R, Lee N, Pennay A, Nielsen S, Ferris J. The effects of modafinil treatment on neuropsychological and attentional bias performance during 7-day inpatient withdrawal from methamphetamine dependence. Exp Clin Psychopharmacol. 2010;18(6):489–497. doi:10.1037/a0021791

113. Lee N, Pennay A, Hester R, McKetin R, Nielsen S, Ferris J. A pilot randomised controlled trial of modafinil during acute methamphetamine withdrawal: feasibility, tolerability and clinical outcomes. Drug Alcohol Rev. 2013;32(1):88–95. doi:10.1111/j.1465-3362.2012.00473.x

114. McElhiney MC, Rabkin JG, Rabkin R, Nunes EV. Provigil (modafinil) plus cognitive behavioral therapy for methamphetamine use in HIV+ gay men: a pilot study. Am J Drug Alcohol Abuse. 2009;35(1):34–37. doi:10.1080/00952990802342907

115. Raineri M, Gonzalez B, Goitia B, et al. Modafinil abrogates methamphetamine-induced neuroinflammation and apoptotic effects in the mouse striatum. PLoS One. 2012;7(10):e46599. doi:10.1371/journal.pone.0046599

116. Baumann MH, Phillips JM, Ayestas MA, Ali SF, Rice KC, Rothman RB. Preclinical evaluation of GBR12909 decanoate as a long-acting medication for methamphetamine dependence. Ann N Y Acad Sci. 2002;965:92–108. doi:10.1111/j.1749-6632.2002.tb04154.x

117. Stephans SE, Yamamoto BK. Methamphetamine-induced neurotoxicity: roles for glutamate and dopamine efflux. Synapse. 1994;17(3):203–209. doi:10.1002/syn.890170310

118. Kalechstein AD, De La Garza R, Newton TF. Modafinil administration improves working memory in methamphetamine-dependent individuals who demonstrate baseline impairment. Am J Addict. 2010;19(4):340–344. doi:10.1111/j.1521-0391.2010.00052.x

119. González B, Raineri M, Cadet JL, García-Rill E, Urbano FJ, Bisagno V. Modafinil improves methamphetamine-induced object recognition deficits and restores prefrontal cortex ERK signaling in mice. Neuropharmacology. 2014;87:188–197. doi:10.1016/j.neuropharm.2014.02.002

120. Reichel CM, Gilstrap MG, Ramsey LA, See RE. Modafinil restores methamphetamine induced object-in-place memory deficits in rats independent of glutamate N-methyl-D-aspartate receptor expression. Drug Alcohol Depend. 2014;134:115–122. doi:10.1016/j.drugalcdep.2013.09.018

121. Ghahremani DG, Tabibnia G, Monterosso J, Hellemann G, Poldrack RA, London ED. Effect of modafinil on learning and task-related brain activity in methamphetamine-dependent and healthy individuals. Neuropsychopharmacology. 2011;36(5):950–959. doi:10.1038/npp.2010.233

122. Dean AC, Sevak RJ, Monterosso JR, Hellemann G, Sugar CA, London ED. Acute modafinil effects on attention and inhibitory control in methamphetamine-dependent humans. J Stud Alcohol Drugs. 2011;72(6):943–953. doi:10.15288/jsad.2011.72.943

123. Howard P, Twycross R, Shuster J, Mihalyo M, Wilcock A. Antidepressant drugs. J Pain Symptom Manage. 2012;44(5):763–783. doi:10.1016/j.jpainsymman.2012.09.001

124. Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract. 2004;10(4):239–248. doi:10.1097/00131746-200407000-00005

125. Newton TF, De La Garza R, Fong T, et al. A comprehensive assessment of the safety of intravenous methamphetamine administration during treatment with selegiline. Pharmacol Biochem Behav. 2005;82(4):704–711. doi:10.1016/j.pbb.2005.11.012

126. Galloway GP, Newmeyer J, Knapp T, Stalcup SA, Smith D. Imipramine for the treatment of cocaine and methamphetamine dependence. J Addict Dis. 1994;13(4):201–216. doi:10.1300/j069v13n04_08

127. Galloway GP, Newmeyer J, Knapp T, Stalcup SA, Smith D. A controlled trial of imipramine for the treatment of methamphetamine dependence. J Subst Abuse Treat. 1996;13(6):493–497. doi:10.1016/s0740-5472(96)00154-7

128. Shoblock JR, Maisonneuve IM, Glick SD. Differential interactions of desipramine with amphetamine and methamphetamine: evidence that amphetamine releases dopamine from noradrenergic neurons in the medial prefrontal cortex. Neurochem Res. 2004;29(7):1437–1442. doi:10.1023/b:nere.0000026409.76261.f3

129. Balsara JJ, Chandorkar AG. Experimental evaluation of the possible neuroleptic activity of clomipramine. Indian J Physiol Pharmacol. 1978;22(3):263–269.

130. Balsara JJ, Jadhav JH, Muley MP, Chandorkar AG. Effect of drugs influencing central serotonergic mechanisms on methamphetamine-induced stereotyped behavior in the rat. Psychopharmacology. 1979;64(3):303–307. doi:10.1007/BF00427514

131. Ginawi OT, Al-Majed AA, Al-Suwailem AK, El-Hadiyah TM. Involvement of some 5-HT receptors in methamphetamine-induced locomotor activity in mice. J Physiol Pharmacol. 2004;55(2):357–369.

132. McDaid J, Tedford CE, Mackie AR, et al. Nullifying drug-induced sensitization: behavioral and electrophysiological evaluations of dopaminergic and serotonergic ligands in methamphetamine-sensitized rats. Drug Alcohol Depend. 2007;86(1):55–66. doi:10.1016/j.drugalcdep.2006.05.014

133. Balsara JJ, Nandal NV, Mane VR, Chandorkar AG. Experimental evaluation of the antidepressant and neuroleptic activity of maprotiline. Indian J Physiol Pharmacol. 1982;26(3):183–195.

134. Herrold AA, Shen F, Graham MP, et al. Mirtazapine treatment after conditioning with methamphetamine alters subsequent expression of place preference. Drug Alcohol Depend. 2009;99(1–3):231–239. doi:10.1016/j.drugalcdep.2008.08.005

135. Graves SM, Napier TC. Mirtazapine alters cue-associated methamphetamine seeking in rats. Biol Psychiatry. 2011;69(3):275–281. doi:10.1016/j.biopsych.2010.09.032

136. Coffin PO, Santos GM, Hern J, et al. Effects of mirtazapine for methamphetamine use disorder among cisgender men and transgender women who have sex with men: a placebo-controlled randomized clinical trial. JAMA Psychiatry. 2020;77(3):246–255. doi:10.1001/jamapsychiatry.2019.3655

137. Colfax GN, Santos GM, Das M, et al. Mirtazapine to reduce methamphetamine use: a randomized controlled trial. Arch Gen Psychiatry. 2011;68(11):1168–1175. doi:10.1001/archgenpsychiatry.2011.124

138. Cruickshank CC, Montebello ME, Dyer KR, et al. A placebo-controlled trial of mirtazapine for the management of methamphetamine withdrawal. Drug Alcohol Rev. 2008;27(3):326–333. doi:10.1080/09595230801935672

139. Kessler DS, MacNeill SJ, Tallon D, et al. Mirtazapine added to SSRIs or SNRIs for treatment resistant depression in primary care: Phase III randomised placebo controlled trial (MIR. BMJ. 2018:363:k4218. doi:10.1136/bmj.k4218

140. Kameda K, Tanaka T, Miura J, Kusumi I, Koyama T. Effects of acute citalopram treatment on the methamphetamine-induced locomotor activity. Prog Neuropsychopharmacol Biol Psychiatry. 2001;25(8):1583–1595. doi:10.1016/s0278-5846(01)00206-8

141. Ricaurte GA, Fuller RW, Perry KW, Seiden LS, Schuster CR. Fluoxetine increases long-lasting neostriatal dopamine depletion after administration of d-methamphetamine and d-amphetamine. Neuropharmacology. 1983;22(10):1165–1169. doi:10.1016/0028-3908(83)90075-8

142. Kaneko Y, Kashiwa A, Ito T, Ishii S, Umino A, Nishikawa T. Selective serotonin reuptake inhibitors, fluoxetine and paroxetine, attenuate the expression of the established behavioral sensitization induced by methamphetamine. Neuropsychopharmacology. 2007;32(3):658–664. doi:10.1038/sj.npp.1301111

143. Takamatsu Y, Yamamoto H, Ogai Y, Hagino Y, Markou A, Ikeda K. Fluoxetine as a potential pharmacotherapy for methamphetamine dependence: studies in mice. Ann N Y Acad Sci. 2006;1074:295–302. doi:10.1196/annals.1369.026

144. Takamatsu Y, Yamamoto H, Hagino Y, Markou A, Ikeda K. The selective serotonin reuptake inhibitor paroxetine, but not fluvoxamine, decreases methamphetamine conditioned place preference in mice. Curr Neuropharmacol. 2011;9(1):68–72. doi:10.2174/157015911795017236

145. Shoptaw S, Huber A, Peck J, et al. Randomized, placebo-controlled trial of sertraline and contingency management for the treatment of methamphetamine dependence. Drug Alcohol Depend. 2006;85(1):12–18. doi:10.1016/j.drugalcdep.2006.03.005

146. Zorick T, Sugar CA, Hellemann G, Shoptaw S, London ED. Poor response to sertraline in methamphetamine dependence is associated with sustained craving for methamphetamine. Drug Alcohol Depend. 2011;118(2–3):500–503. doi:10.1016/j.drugalcdep.2011.04.015

147. Rahimi Borumand M, Motaghinejad M, Motevalian M, Gholami M. Duloxetine by modulating the Akt/GSK3 signaling pathways has neuroprotective effects against methamphetamine-induced neurodegeneration and cognition impairment in rats. Iran J Med Sci. 2019;44(2):146–154.

148. Ramezany Yasuj S, Nourhashemi M, Keshavarzi S, Motaghinejad M, Motevalian M. Possible role of cyclic AMP response element binding/brain-derived neurotrophic factor signaling pathway in mediating the pharmacological effects of duloxetine against methamphetamine use-induced cognitive impairment and withdrawal-induced anxiety and depression in rats. Adv Biomed Res. 2019;8:11. doi:10.4103/abr.abr_34_18

149. Althobaiti YS. Role of venlafaxine in relapse to methamphetamine seeking. Potential treatment option for drug dependence. Saudi Med J. 2019;40(4):339–346. doi:10.15537/smj.2019.4.23718

150. Ciraulo DA, Sarid-Segal O, Knapp CM, et al. Efficacy screening trials of paroxetine, pentoxifylline, riluzole, pramipexole and venlafaxine in cocaine dependence. Addiction. 2005;100 Suppl 1:12–22. doi:10.1111/j.1360-0443.2005.00985.x

151. Passos SR, Camacho LA, Lopes CS, Dos Santos MA. Nefazodone in out-patient treatment of inhaled cocaine dependence: a randomized double-blind placebo-controlled trial. Addiction. 2005;100(4):489–494. doi:10.1111/j.1360-0443.2005.01041.x

152. Raby WN, Rubin EA, Garawi F, et al. A randomized, double-blind, placebo-controlled trial of venlafaxine for the treatment of depressed cocaine-dependent patients. Am J Addict. 2014;23(1):68–75. doi:10.1111/j.1521-0391.2013.12065.x

153. Ciraulo DA, Knapp C, Rotrosen J, et al. Nefazodone treatment of cocaine dependence with comorbid depressive symptoms. Addiction. 2005;100 Suppl 1:23–31. doi:10.1111/j.1360-0443.2005.00984.x

154. McDowell DM, Levin FR, Seracini AM, Nunes EV. Venlafaxine treatment of cocaine abusers with depressive disorders. Am J Drug Alcohol Abuse. 2000;26(1):25–31. doi:10.1081/ada-100100588

155. Foley KF, DeSanty KP, Kast RE. Bupropion: pharmacology and therapeutic applications. Expert Rev Neurother. 2006;6(9):1249–1265. doi:10.1586/14737175.6.9.1249

156. Newton TF, Roache JD, De La Garza R, et al. Bupropion reduces methamphetamine-induced subjective effects and cue-induced craving. Neuropsychopharmacology. 2006;31(7):1537–1544. doi:10.1038/sj.npp.1300979

157. Banks ML, Blough BE. Effects of environmental manipulations and treatment with bupropion and risperidone on choice between methamphetamine and food in rhesus monkeys. Neuropsychopharmacology. 2015;40(9):2198–2206. doi:10.1038/npp.2015.63

158. Charntikov S, Pittenger ST, Pudiak CM, Bevins RA. The effect of N-acetylcysteine or bupropion on methamphetamine self-administration and methamphetamine-triggered reinstatement of female rats. Neuropharmacology. 2018;135:487–495. doi:10.1016/j.neuropharm.2018.03.021

159. Reichel CM, Linkugel JD, Bevins RA. Bupropion differentially impacts acquisition of methamphetamine self-administration and sucrose-maintained behavior. Pharmacol Biochem Behav. 2008;89(3):463–472. doi:10.1016/j.pbb.2008.02.002

160. Reichel CM, Murray JE, Grant KM, Bevins RA. Bupropion attenuates methamphetamine self-administration in adult male rats. Drug Alcohol Depend. 2009;100(1–2):54–62. doi:10.1016/j.drugalcdep.2008.09.006

161. Walsh AEL, Huneke NTM, Brown R, Browning M, Cowen P, Harmer CJ. A Dissociation of the acute effects of bupropion on positive emotional processing and reward processing in healthy volunteers. Front Psychiatry. 2018;9:482. doi:10.3389/fpsyt.2018.00482

162. Beyens MN, Guy C, Mounier G, Laporte S, Ollagnier M. Serious adverse reactions of bupropion for smoking cessation: analysis of the French Pharmacovigilance Database from 2001 to 2004. Drug Saf. 2008;31(11):1017–1026. doi:10.2165/00002018-200831110-00006

163. Simpson M, Troger A, Feng C, Whitledge JD, Monuteaux M, Burns MM. Clinical and electrocardiographic factors associated with adverse cardiovascular events in bupropion exposures. Clin Toxicol. 2023;61(7):529–535. doi:10.1080/15563650.2023.2227997

164. Van Wyck Fleet J, Manberg PJ, Miller LL, et al. Overview of clinically significant adverse reactions to bupropion. J Clin Psychiatry. 1983;44(5):191–196.

165. Harrod SB, Dwoskin LP, Crooks PA, Klebaur JE, Bardo MT. Lobeline attenuates d-methamphetamine self-administration in rats. J Pharmacol Exp Ther. 2001;298(1):172–179.

166. Tatsuta T, Kitanaka N, Kitanaka J, Morita Y, Takemura M. Lobeline attenuates methamphetamine-induced stereotypy in adolescent mice. Neurochem Res. 2006;31(11):1359–1369. doi:10.1007/s11064-006-9180-1

167. Miller DK, Crooks PA, Dwoskin LP. Lobeline inhibits nicotine-evoked [(3)H]dopamine overflow from rat striatal slices and nicotine-evoked (86)Rb(+) efflux from thalamic synaptosomes. Neuropharmacology. 2000;39(13):2654–2662. doi:10.1016/s0028-3908(00)00140-4

168. Miller DK, Crooks PA, Zheng G, Grinevich VP, Norrholm SD, Dwoskin LP. Lobeline analogs with enhanced affinity and selectivity for plasmalemma and vesicular monoamine transporters. J Pharmacol Exp Ther. 2004;310(3):1035–1045. doi:10.1124/jpet.104.068098

169. Nickell JR, Krishnamurthy S, Norrholm S, et al. Lobelane inhibits methamphetamine-evoked dopamine release via inhibition of the vesicular monoamine transporter-2. J Pharmacol Exp Ther. 2010;332(2):612–621. doi:10.1124/jpet.109.160275

170. Neugebauer NM, Harrod SB, Stairs DJ, Crooks PA, Dwoskin LP, Bardo MT. Lobelane decreases methamphetamine self-administration in rats. Eur J Pharmacol. 2007;571(1):33–38. doi:10.1016/j.ejphar.2007.06.003

171. Beckmann JS, Siripurapu KB, Nickell JR, et al. The novel pyrrolidine nor-lobelane analog UKCP-110 [cis-2,5-di-(2-phenethyl)-pyrrolidine hydrochloride] inhibits VMAT2 function, methamphetamine-evoked dopamine release, and methamphetamine self-administration in rats. J Pharmacol Exp Ther. 2010;335(3):841–851. doi:10.1124/jpet.110.172742

172. Alvers KM, Beckmann JS, Zheng G, Crooks PA, Dwoskin LP, Bardo MT. The effect of VMAT2 inhibitor GZ-793A on the reinstatement of methamphetamine-seeking in rats. Psychopharmacology. 2012;224(2):255–262. doi:10.1007/s00213-012-2748-3

173. Beckmann JS, Denehy ED, Zheng G, Crooks PA, Dwoskin LP, Bardo MT. The effect of a novel VMAT2 inhibitor, GZ-793A, on methamphetamine reward in rats. Psychopharmacology. 2012;220(2):395–403. doi:10.1007/s00213-011-2488-9

174. Kangiser MM, Dwoskin LP, Zheng G, Crooks PA, Stairs DJ. Varenicline and GZ-793A differentially decrease methamphetamine self-administration under a multiple schedule of reinforcement in rats. Behav Pharmacol. 2018;29(1):87–97. doi:10.1097/FBP.0000000000000340

175. Wilmouth CE, Zheng G, Crooks PA, Dwoskin LP, Bardo MT. Oral administration of GZ-793A, a VMAT2 inhibitor, decreases methamphetamine self-administration in rats. Pharmacol Biochem Behav. 2013;112:29–33. doi:10.1016/j.pbb.2013.09.006

176. Chandler CM, Nickell JR, George Wilson A, et al. Vesicular monoamine transporter-2 inhibitor JPC-141 prevents methamphetamine-induced dopamine toxicity and blocks methamphetamine self-administration in rats. Biochem Pharmacol. 2024:116189. doi:10.1016/j.bcp.2024.116189

177. Lee NR, Zheng G, Leggas M, et al. GZ-11608, a vesicular monoamine transporter-2 inhibitor, decreases the neurochemical and behavioral effects of methamphetamine. J Pharmacol Exp Ther. 2019;371(2):526–543. doi:10.1124/jpet.119.258699

178. Thomas DM, Francescutti-Verbeem DM, Kuhn DM. The newly synthesized pool of dopamine determines the severity of methamphetamine-induced neurotoxicity. J Neurochem. 2008;105(3):605–616. doi:10.1111/j.1471-4159.2007.05155.x

179. Albers DS, Sonsalla PK. Methamphetamine-induced hyperthermia and dopaminergic neurotoxicity in mice: pharmacological profile of protective and nonprotective agents. J Pharmacol Exp Ther. 1995;275(3):1104–1114.

180. Axt KJ, Commins DL, Vosmer G, Seiden LS. alpha-Methyl-p-tyrosine pretreatment partially prevents methamphetamine-induced endogenous neurotoxin formation. Brain Res. 1990;515(1–2):269–276. doi:10.1016/0006-8993(90)90606-c

181. Commins DL, Seiden LS. alpha-Methyltyrosine blocks methylamphetamine-induced degeneration in the rat somatosensory cortex. Brain Res. 1986;365(1):15–20. doi:10.1016/0006-8993(86)90717-1

182. Metzger RR, Haughey HM, Wilkins DG, Gibb JW, Hanson GR, Fleckenstein AE. Methamphetamine-induced rapid decrease in dopamine transporter function: role of dopamine and hyperthermia. J Pharmacol Exp Ther. 2000;295(3):1077–1085.

183. Schmidt CJ, Ritter JK, Sonsalla PK, Hanson GR, Gibb JW. Role of dopamine in the neurotoxic effects of methamphetamine. J Pharmacol Exp Ther. 1985;233(3):539–544.

184. Wagner GC, Lucot JB, Schuster CR, Seiden LS. Alpha-methyltyrosine attenuates and reserpine increases methamphetamine-induced neuronal changes. Brain Res. 1983;270(2):285–288. doi:10.1016/0006-8993(83)90602-9

185. McTavish SF, McPherson MH, Harmer CJ, et al. Antidopaminergic effects of dietary tyrosine depletion in healthy subjects and patients with manic illness. Br J Psychiatry. 2001;179:356–360. doi:10.1192/bjp.179.4.356

186. Leyton M, Casey KF, Delaney JS, Kolivakis T, Benkelfat C. Cocaine craving, euphoria, and self-administration: a preliminary study of the effect of catecholamine precursor depletion. Behav Neurosci. 2005;119(6):1619–1627. doi:10.1037/0735-7044.119.6.1619

187. McLean A, Rubinsztein JS, Robbins TW, Sahakian BJ. The effects of tyrosine depletion in normal healthy volunteers: implications for unipolar depression. Psychopharmacology. 2004;171(3):286–297. doi:10.1007/s00213-003-1586-8

188. Krulich L, Giachetti A, Marchlewska-Koj A, Hefco E, Jameson HE. On the role of the central noradrenergic and dopaminergic systems in the regulation of TSH secretion in the rat. Endocrinology. 1977;100(2):496–505. doi:10.1210/endo-100-2-496

189. Kaufman S, Friedman S. Dopamine-beta-hydroxylase. Pharmacol Rev. 1965;17:71–100.

190. Berridge CW, Waterhouse BD. The locus coeruleus-noradrenergic system: modulation of behavioral state and state-dependent cognitive processes. Brain Res Brain Res Rev. 2003;42(1):33–84. doi:10.1016/s0165-0173(03)00143-7

191. Martel JC, Gatti McArthur S. Dopamine receptor subtypes, physiology and pharmacology: new ligands and concepts in schizophrenia. Front Pharmacol. 2020;11:1003. doi:10.3389/fphar.2020.01003

192. Nichols DE, Nichols CD. Serotonin receptors. Chem Rev. 2008;108(5):1614–1641. doi:10.1021/cr078224o

193. Du Y, Li Q, Dou Y, et al. Side effects and cognitive benefits of buspirone: a systematic review and meta-analysis. Heliyon. 2024;10(7):e28918. doi:10.1016/j.heliyon.2024.e28918

194. Shelton KL, Hendrick ES, Beardsley PM. Efficacy of buspirone for attenuating cocaine and methamphetamine reinstatement in rats. Drug Alcohol Depend. 2013;129(3):210–216. doi:10.1016/j.drugalcdep.2013.01.003

195. John WS, Banala AK, Newman AH, Nader MA. Effects of buspirone and the dopamine D3 receptor compound PG619 on cocaine and methamphetamine self-administration in rhesus monkeys using a food-drug choice paradigm. Psychopharmacology. 2015;232(7):1279–1289. doi:10.1007/s00213-014-3760-6

196. Reynolds AR, Strickland JC, Stoops WW, Lile JA, Rush CR. Buspirone maintenance does not alter the reinforcing, subjective, and cardiovascular effects of intranasal methamphetamine. Drug Alcohol Depend. 2017;181:25–29. doi:10.1016/j.drugalcdep.2017.08.038

197. Pike E, Stoops WW, Rush CR. Acute buspirone dosing enhances abuse-related subjective effects of oral methamphetamine. Pharmacol Biochem Behav. 2016;150-151:87–93. doi:10.1016/j.pbb.2016.09.009

198. Grinchii D, Dremencov E. Mechanism of action of atypical antipsychotic drugs in mood disorders. Int J Mol Sci. 2020;21(24):9532. doi:10.3390/ijms21249532

199. Burris KD, Molski TF, Xu C, et al. Aripiprazole, a novel antipsychotic, is a high-affinity partial agonist at human dopamine D2 receptors. J Pharmacol Exp Ther. 2002;302(1):381–389. doi:10.1124/jpet.102.033175

200. Funakoshi T, Chaki S, Kawashima N, et al. In vitro and in vivo pharmacological profile of 5-[2-[4-(6-fluoro-1H-indole-3-yl)piperidin-1-yl]ethyl]-4-(4-fluorophenyl)thiazole-2-carboxylic acid amide (NRA0562), a novel and putative atypical antipsychotic. Life Sci. 2002;71(12):1371–1384. doi:10.1016/s0024-3205(02)01863-5

201. Futamura T, Akiyama S, Sugino H, Forbes A, McQuade RD, Kikuchi T. Aripiprazole attenuates established behavioral sensitization induced by methamphetamine. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34(6):1115–1119. doi:10.1016/j.pnpbp.2010.06.006

202. Ishibashi T, Horisawa T, Tokuda K, et al. Pharmacological profile of lurasidone, a novel antipsychotic agent with potent 5-hydroxytryptamine 7 (5-HT7) and 5-HT1A receptor activity. J Pharmacol Exp Ther. 2010;334(1):171–181. doi:10.1124/jpet.110.167346

203. Jing L, Liu B, Zhang M, Liang JH. Involvement of dopamine D2 receptor in a single methamphetamine-induced behavioral sensitization in C57BL/6J mice. Neurosci Lett. 2018;681:87–92. doi:10.1016/j.neulet.2018.02.067

204. Kuribara H. Early post-treatment with haloperidol retards induction of methamphetamine sensitization in mice. Eur J Pharmacol. 1994;256(3):295–299. doi:10.1016/0014-2999(94)90555-x

205. Kuribara H, Uchihashi Y. Effects of haloperidol on the methamphetamine sensitization: assessment by ambulatory activity in mice. Jpn J Psychiatry Neurol. 1993;47(3):661–668. doi:10.1111/j.1440-1819.1993.tb01815.x

206. Lucot JB, Wagner GC, Schuster CR, Seiden LS. The effects of dopaminergic agents on the locomotor activity of rats after high doses of methylamphetamine. Pharmacol Biochem Behav. 1980;13(3):409–413. doi:10.1016/0091-3057(80)90247-6

207. Maehara S, Hikichi H, Satow A, Okuda S, Ohta H. Antipsychotic property of a muscarinic receptor agonist in animal models for schizophrenia. Pharmacol Biochem Behav. 2008;91(1):140–149. doi:10.1016/j.pbb.2008.06.023

208. Shintomi K. Effects of psychotropic drugs on methamphetamine-induced behavioral excitation in grouped mice. Eur J Pharmacol. 1975;31(2):195–206. doi:10.1016/0014-2999(75)90040-0

209. Steed E, Jones CA, McCreary AC. Serotonergic involvement in methamphetamine-induced locomotor activity: a detailed pharmacological study. Behav Brain Res. 2011;220(1):9–19. doi:10.1016/j.bbr.2011.01.026

210. Tada M, Shirakawa K, Matsuoka N, Mutoh S. Combined treatment of quetiapine with haloperidol in animal models of antipsychotic effect and extrapyramidal side effects: comparison with risperidone and chlorpromazine. Psychopharmacology. 2004;176(1):94–100. doi:10.1007/s00213-004-1866-y

211. Zhang L, Zeng Z, Lu X, et al. CNTN1 in the nucleus accumbens is involved in methamphetamine-induced conditioned place preference in mice. Neurotox Res. 2023;41(4):324–337. doi:10.1007/s12640-023-00640-9

212. Cheng MC, Hsu SH, Chen CH. Repetitive administration of aripiprazole enhances locomotor response to methamphetamine in mice. Behav Brain Res. 2011;216(2):621–625. doi:10.1016/j.bbr.2010.09.002

213. Wee S, Wang Z, Woolverton WL, Pulvirenti L, Koob GF. Effect of aripiprazole, a partial dopamine D2 receptor agonist, on increased rate of methamphetamine self-administration in rats with prolonged session duration. Neuropsychopharmacology. 2007;32(10):2238–2247. doi:10.1038/sj.npp.1301353

214. Wachtel SR, Ortengren A, de Wit H. The effects of acute haloperidol or risperidone on subjective responses to methamphetamine in healthy volunteers. Drug Alcohol Depend. 2002;68(1):23–33. doi:10.1016/s0376-8716(02)00104-7

215. Meredith CW, Jaffe C, Yanasak E, Cherrier M, Saxon AJ. An open-label pilot study of risperidone in the treatment of methamphetamine dependence. J Psychoactive Drugs. 2007;39(2):167–172. doi:10.1080/02791072.2007.10399875

216. Meredith CW, Jaffe C, Cherrier M, et al. Open trial of injectable risperidone for methamphetamine dependence. J Addict Med. 2009;3(2):55–65. doi:10.1097/ADM.0b013e31818e2185

217. Newton TF, Reid MS, De La Garza R, et al. Evaluation of subjective effects of aripiprazole and methamphetamine in methamphetamine-dependent volunteers. Int J Neuropsychopharmacol. 2008;11(8):1037–1045. doi:10.1017/S1461145708009097

218. Sevak RJ, Vansickel AR, Stoops WW, Glaser PE, Hays LR, Rush CR. Discriminative-stimulus, subject-rated, and physiological effects of methamphetamine in humans pretreated with aripiprazole. J Clin Psychopharmacol. 2011;31(4):470–480. doi:10.1097/JCP.0b013e318221b2db

219. Stoops WW, Bennett JA, Lile JA, Sevak RJ, Rush CR. Influence of aripiprazole pretreatment on the reinforcing effects of methamphetamine in humans. Prog Neuropsychopharmacol Biol Psychiatry. 2013;47:111–117. doi:10.1016/j.pnpbp.2013.08.007

220. Coffin PO, Santos GM, Das M, et al. Aripiprazole for the treatment of methamphetamine dependence: a randomized, double-blind, placebo-controlled trial. Addiction. 2013;108(4):751–761. doi:10.1111/add.12073

221. Sulaiman AH, Gill JS, Said MA, Zainal NZ, Hussein HM, Guan NC. A randomized, placebo-controlled trial of aripiprazole for the treatment of methamphetamine dependence and associated psychosis. Int J Psychiatry Clin Pract. 2013;17(2):131–138. doi:10.3109/13651501.2012.667116

222. Amarasekera R, Wood E. Worsening stimulant use disorder outcomes coinciding with off-label antipsychotic prescribing: a commonly unrecognised side effect? BMJ Case Rep. 2023;16(10):e255129. doi:10.1136/bcr-2023-255129

223. Abekawa T, Ito K, Nakagawa S, Nakato Y, Koyama T. Olanzapine and risperidone block a high dose of methamphetamine-induced schizophrenia-like behavioral abnormalities and accompanied apoptosis in the medial prefrontal cortex. Schizophr Res. 2008;101(1–3):84–94. doi:10.1016/j.schres.2007.12.488

224. Abekawa T, Ito K, Nakagawa S, Nakato Y, Koyama T. Effects of aripiprazole and haloperidol on progression to schizophrenia-like behavioural abnormalities and apoptosis in rodents. Schizophr Res. 2011;125(1):77–87. doi:10.1016/j.schres.2010.08.011

225. Mori T, Ito S, Kita T, Sawaguchi T. Effects of dopamine- and serotonin-related compounds on methamphetamine-induced self-injurious behavior in mice. J Pharmacol Sci. 2004;96(4):459–464. doi:10.1254/jphs.fpj04040x

226. Farnia V, Shakeri J, Tatari F, et al. Randomized controlled trial of aripiprazole versus risperidone for the treatment of amphetamine-induced psychosis. Am J Drug Alcohol Abuse. 2014;40(1):10–15. doi:10.3109/00952990.2013.861843

227. Sato M, Chen CC, Akiyama K, Otsuki S. Acute exacerbation of paranoid psychotic state after long-term abstinence in patients with previous methamphetamine psychosis. Biol Psychiatry. 1983;18(4):429–440.

228. Verachai V, Rukngan W, Chawanakrasaesin K, et al. Treatment of methamphetamine-induced psychosis: a double-blind randomized controlled trial comparing haloperidol and quetiapine. Psychopharmacology. 2014;231(16):3099–3108. doi:10.1007/s00213-014-3485-6

229. Wang G, Zhang Y, Zhang S, et al. Aripiprazole and risperidone for treatment of methamphetamine-associated psychosis in Chinese patients. J Subst Abuse Treat. 2016;62:84–88. doi:10.1016/j.jsat.2015.11.009

230. Wang G, Ding F, Chawarski MC, et al. Randomized controlled trial of paliperidone extended release versus risperidone for the treatment of methamphetamine-associated psychosis in Chinese patients. Front Psychiatry. 2020;11:237. doi:10.3389/fpsyt.2020.00237

231. Richards JR, Derlet RW, Duncan DR. Methamphetamine toxicity: treatment with a benzodiazepine versus a butyrophenone. Eur J Emerg Med. 1997;4(3):130–135. doi:10.1097/00063110-199709000-00003

232. Shioda K, Nisijima K, Yoshino T, Kato S. Effect of risperidone on acute methamphetamine-induced hyperthermia in rats. Drug Alcohol Depend. 2010;111(3):241–249. doi:10.1016/j.drugalcdep.2010.05.001

233. Adinoff B. Neurobiologic processes in drug reward and addiction. Harv Rev Psychiatry. 2004;12(6):305–320. doi:10.1080/10673220490910844

234. Gong X, Yue K, Ma B, et al. Levo-tetrahydropalmatine, a natural, mixed dopamine receptor antagonist, inhibits methamphetamine self-administration and methamphetamine-induced reinstatement. Pharmacol Biochem Behav. 2016;144:67–72. doi:10.1016/j.pbb.2016.01.010

235. Brennan KA, Carati C, Lea RA, Fitzmaurice PS, Schenk S. Effect of D1-like and D2-like receptor antagonists on methamphetamine and 3,4-methylenedioxymethamphetamine self-administration in rats. Behav Pharmacol. 2009;20(8):688–694. doi:10.1097/FBP.0b013e328333a28d

236. Shahveisi K, Abdoli N, Khazaie H, Farnia V, Khodamoradi M. Maternal sleep deprivation affects extinction and reinstatement of methamphetamine reward memory in male offspring: role of the D1-like and D2-like dopamine receptors. Brain Res. 2022;1792:148033. doi:10.1016/j.brainres.2022.148033

237. Hamamura T, Akiyama K, Akimoto K, et al. Co-administration of either a selective D1 or D2 dopamine antagonist with methamphetamine prevents methamphetamine-induced behavioral sensitization and neurochemical change, studied by in vivo intracerebral dialysis. Brain Res. 1991;546(1):40–46. doi:10.1016/0006-8993(91)91156-u

238. Kuribara H. Effects of sulpiride and nemonapride, benzamide derivatives having distinct potencies of antagonistic action on dopamine D2 receptors, on sensitization to methamphetamine in mice. J Pharm Pharmacol. 1996;48(3):292–296. doi:10.1111/j.2042-7158.1996.tb05919.x

239. Kuribara H, Uchihashi Y. Dopamine antagonists can inhibit methamphetamine sensitization, but not cocaine sensitization, when assessed by ambulatory activity in mice. J Pharm Pharmacol. 1993;45(12):1042–1045. doi:10.1111/j.2042-7158.1993.tb07177.x

240. Ujike H, Onoue T, Akiyama K, Hamamura T, Otsuki S. Effects of selective D-1 and D-2 dopamine antagonists on development of methamphetamine-induced behavioral sensitization. Psychopharmacology. 1989;98(1):89–92. doi:10.1007/BF00442011

241. Yoshida H, Ohno M, Watanabe S. Roles of dopamine D1 receptors in striatal fos protein induction associated with methamphetamine behavioral sensitization in rats. Brain Res Bull. 1995;38(4):393–397. doi:10.1016/0361-9230(95)02005-c

242. Avchalumov Y, Trenet W, Piña-Crespo J, Mandyam C. SCH23390 Reduces Methamphetamine self-administration and prevents methamphetamine-induced striatal LTD. Int J Mol Sci. 2020;21(18):6491. doi:10.3390/ijms21186491

243. Gu SM, Cha HJ, Seo SW, Hong JT, Yun J. Dopamine D1 receptor antagonist reduces stimulant-induced conditioned place preferences and dopamine receptor supersensitivity. Naunyn Schmiedebergs Arch Pharmacol. 2020;393(1):131–138. doi:10.1007/s00210-019-01694-3

244. Kurokawa K, Mizuno K, Shibasaki M, Ohkuma S. Regulation of ryanodine receptors by dopamine D1 receptors during methamphetamine-induced place conditioning. J Neurochem. 2010;115(5):1206–1214. doi:10.1111/j.1471-4159.2010.07010.x

245. Kurokawa K, Shibasaki M, Ohkuma S. Methamphetamine-induced up-regulation of α2/δ subunit of voltage-gated calcium channels is regulated by DA receptors. Synapse. 2010;64(11):822–828. doi:10.1002/syn.20797

246. Liu M, Mu S, Han W, et al. Dopamine D1 receptor in orbitofrontal cortex to dorsal striatum pathway modulates methamphetamine addiction. Biochem Biophys Res Commun. 2023;671:96–104. doi:10.1016/j.bbrc.2023.06.005

247. Mizoguchi H, Yamada K, Mizuno M, et al. Regulations of methamphetamine reward by extracellular signal-regulated kinase 1/2/ets-like gene-1 signaling pathway via the activation of dopamine receptors. Mol Pharmacol. 2004;65(5):1293–1301. doi:10.1124/mol.65.5.1293

248. Saika F, Kiguchi N, Wakida N, et al. Upregulation of CCL7 and CCL2 in reward system mediated through dopamine D1 receptor signaling underlies methamphetamine-induced place preference in mice. Neurosci Lett. 2018;665:33–37. doi:10.1016/j.neulet.2017.11.042

249. Larson TA, Winkler MC, Stafford J, Levis SC, O’Neill CE, Bachtell RK. Role of dopamine D2-like receptors and their modulation by adenosine receptor stimulation in the reinstatement of methamphetamine seeking. Psychopharmacology. 2019;236(4):1207–1218. doi:10.1007/s00213-018-5126-y

250. Taverna S, Ilijic E, Surmeier DJ. Recurrent collateral connections of striatal medium spiny neurons are disrupted in models of Parkinson’s disease. J Neurosci. 2008;28(21):5504–5512. doi:10.1523/JNEUROSCI.5493-07.2008

251. Gallo EF, Meszaros J, Sherman JD, et al. Accumbens dopamine D2 receptors increase motivation by decreasing inhibitory transmission to the ventral pallidum. Nat Commun. 2018;9(1):1086. doi:10.1038/s41467-018-03272-2

252. Carr KD, Cabeza de Vaca S, Sun Y, Chau LS. Reward-potentiating effects of D-1 dopamine receptor agonist and AMPAR GluR1 antagonist in nucleus accumbens shell and their modulation by food restriction. Psychopharmacology. 2009;202(4):731–743. doi:10.1007/s00213-008-1355-9

253. Paterson NE, Vocci F, Sevak RJ, Wagreich E, London ED. Dopamine D3 receptors as a therapeutic target for methamphetamine dependence. Am J Drug Alcohol Abuse. 2014;40(1):1–9. doi:10.3109/00952990.2013.858723

254. Sun L, Song R, Chen Y, et al. A selective D3 receptor antagonist YQA14 attenuates methamphetamine-induced behavioral sensitization and conditioned place preference in mice. Acta Pharmacol Sin. 2016;37(2):157–165. doi:10.1038/aps.2015.96

255. Sun X, Gou HY, Li F, et al. Y-QA31, a novel dopamine D3 receptor antagonist, exhibits antipsychotic-like properties in preclinical animal models of schizophrenia. Acta Pharmacol Sin. 2016;37(3):322–333. doi:10.1038/aps.2015.105

256. Baladi MG, Newman AH, Nielsen SM, Hanson GR, Fleckenstein AE. Dopamine D(3) receptors contribute to methamphetamine-induced alterations in dopaminergic neuronal function: role of hyperthermia. Eur J Pharmacol. 2014;732:105–110. doi:10.1016/j.ejphar.2014.03.023

257. Higley AE, Kiefer SW, Li X, Gaál J, Xi ZX, Gardner EL. Dopamine D(3) receptor antagonist SB-277011A inhibits methamphetamine self-administration and methamphetamine-induced reinstatement of drug-seeking in rats. Eur J Pharmacol. 2011;659(2–3):187–192. doi:10.1016/j.ejphar.2011.02.046

258. Spiller K, Xi ZX, Peng XQ, et al. The selective dopamine D3 receptor antagonists SB-277011A and NGB 2904 and the putative partial D3 receptor agonist BP-897 attenuate methamphetamine-enhanced brain stimulation reward in rats. Psychopharmacology. 2008;196(4):533–542. doi:10.1007/s00213-007-0986-6

259. Chen Y, Song R, Yang RF, Wu N, Li J. A novel dopamine D3 receptor antagonist YQA14 inhibits methamphetamine self-administration and relapse to drug-seeking behaviour in rats. Eur J Pharmacol. 2014;743:126–132. doi:10.1016/j.ejphar.2014.09.026

260. Higley AE, Spiller K, Grundt P, et al. PG01037, a novel dopamine D3 receptor antagonist, inhibits the effects of methamphetamine in rats. J Psychopharmacol. 2011;25(2):263–273. doi:10.1177/0269881109358201

261. Orio L, Wee S, Newman AH, Pulvirenti L, Koob GF. The dopamine D3 receptor partial agonist CJB090 and antagonist PG01037 decrease progressive ratio responding for methamphetamine in rats with extended-access. Addict Biol. 2010;15(3):312–323. doi:10.1111/j.1369-1600.2010.00211.x

262. Ranaldi R, Timken P, Parasram D, et al. The D3 receptor antagonist SR 21502 reduces cue-induced reinstatement of methamphetamine-seeking in rats. Neurosci Lett. 2023;806:137237. doi:10.1016/j.neulet.2023.137237

263. Di Ciano P, Grandy DK, Le Foll B. Dopamine D4 receptors in psychostimulant addiction. Adv Pharmacol. 2014;69:301–321. doi:10.1016/B978-0-12-420118-7.00008-1

264. Okuyama S, Kawashima N, Chaki S, et al. A selective dopamine D4 receptor antagonist, NRA0160: a preclinical neuropharmacological profile. Life Sci. 1999;65(20):2109–2125. doi:10.1016/s0024-3205(99)00476-2

265. Liao IM, Chen JC. Lack of dopamine D4 receptor participation in mouse hyperdopaminergic locomotor response. Behav Brain Res. 2021;396:112925. doi:10.1016/j.bbr.2020.112925

266. Boateng CA, Nilson AN, Placide R, et al. Pharmacology and therapeutic potential of benzothiazole analogues for cocaine use disorder. J Med Chem. 2023;66(17):12141–12162. doi:10.1021/acs.jmedchem.3c00734

267. Johnson DA, Hricik JG. The pharmacology of alpha-adrenergic decongestants. Pharmacotherapy. 1993;13(6):110S–146S.

268. Kee VR. Hemodynamic pharmacology of intravenous vasopressors. Crit Care Nurse. 2003;23(4):79–82.

269. Burkes RM, Panos RJ. Ultra long-acting β-agonists in chronic obstructive pulmonary disease. J Exp Pharmacol. 2020;12:589–602. doi:10.2147/JEP.S259328

270. Graham RM. Selective alpha 1-adrenergic antagonists: therapeutically relevant antihypertensive agents. Am J Cardiol. 1984;53(3):16A–20A. doi:10.1016/0002-9149(84)90829-4

271. Shanks RG. Clinical pharmacology of vasodilatory beta-blocking drugs. Am Heart J. 1991;121(3):1006–1011. doi:10.1016/0002-8703(91)90612-l

272. Lemke KA. Perioperative use of selective alpha-2 agonists and antagonists in small animals. Can Vet J. 2004;45(6):475–480.

273. Venables M, Ntzani E, Hsia Y, Gillies D. Alpha2 adrenergic agonists for attention deficit hyperactivity disorder (ADHD). Cochrane Database Syst Rev. 2017;2017(1):CD010016. doi:10.1002/14651858.CD010016.pub2

274. Jarvis N, England GC. Reversal of xylazine sedation in dogs. Vet Rec. 1991;128(14):323–325. doi:10.1136/vr.128.14.323

275. Baracz SJ, Robinson KJ, Wright AL, et al. Oxytocin as an adolescent treatment for methamphetamine addiction after early life stress in male and female rats. Neuropsychopharmacology. 2022;47(8):1561–1573. doi:10.1038/s41386-022-01336-y

276. Shepard JD, Bossert JM, Liu SY, Shaham Y. The anxiogenic drug yohimbine reinstates methamphetamine seeking in a rat model of drug relapse. Biol Psychiatry. 2004;55(11):1082–1089. doi:10.1016/j.biopsych.2004.02.032

277. Harrell R, Speaker HA, Mitchell SL, Sabol KE. The effects of the β1 antagonist, metoprolol, on methamphetamine-induced changes in core temperature in the rat. Neurosci Lett. 2015;609:81–86. doi:10.1016/j.neulet.2015.09.018

278. Kikuchi-Utsumi K, Ishizaka M, Matsumura N, et al. Involvement of the α(1D)-adrenergic receptor in methamphetamine-induced hyperthermia and neurotoxicity in rats. Neurotox Res. 2013;24(2):130–138. doi:10.1007/s12640-012-9369-9

279. Rauhut AS. Propranolol blocks the unconditioned and conditioned hyperactive effects of methamphetamine in CD-1 mice. Behav Pharmacol. 2023;34(6):375–379. doi:10.1097/FBP.0000000000000742

280. White AO, Rauhut AS. Time-dependent effects of prazosin on the development of methamphetamine conditioned hyperactivity and context-specific sensitization in mice. Behav Brain Res. 2014;263:80–89. doi:10.1016/j.bbr.2014.01.032

281. Perry AN, Westenbroek C, Jagannathan L, Becker JB. The roles of dopamine and α1-adrenergic receptors in cocaine preferences in female and male rats. Neuropsychopharmacology. 2015;40(12):2696–2704. doi:10.1038/npp.2015.116

282. Wee S, Mandyam CD, Lekic DM, Koob GF. Alpha 1-noradrenergic system role in increased motivation for cocaine intake in rats with prolonged access. Eur Neuropsychopharmacol. 2008;18(4):303–311. doi:10.1016/j.euroneuro.2007.08.003

283. Zhang XY, Kosten TA. Previous exposure to cocaine enhances cocaine self-administration in an alpha 1-adrenergic receptor dependent manner. Neuropsychopharmacology. 2007;32(3):638–645. doi:10.1038/sj.npp.1301120

284. Woolverton WL. Evaluation of the role of norepinephrine in the reinforcing effects of psychomotor stimulants in rhesus monkeys. Pharmacol Biochem Behav. 1987;26(4):835–839. doi:10.1016/0091-3057(87)90618-6

285. Beldjoud H, Avelar A, de Guglielmo G, et al. Chronic administration of a norepinephrine antagonist prevents and partially reverses escalation of cocaine self-administration. Addict Biol. 2023;28(9):e13316. doi:10.1111/adb.13316

286. Harris GC, Hedaya MA, Pan WJ, Kalivas P. beta-adrenergic antagonism alters the behavioral and neurochemical responses to cocaine. Neuropsychopharmacology. 1996;14(3):195–204. doi:10.1016/0893-133X(95)00089-V

287. Smith RJ, Aston-Jones G. α(2) Adrenergic and imidazoline receptor agonists prevent cue-induced cocaine seeking. Biol Psychiatry. 2011;70(8):712–719. doi:10.1016/j.biopsych.2011.06.010

288. Kitanaka N, Kitanaka J, Hall FS, et al. Agmatine attenuates methamphetamine-induced hyperlocomotion and stereotyped behavior in mice. Behav Pharmacol. 2014;25(2):158–165. doi:10.1097/FBP.0000000000000030

289. Thorn DA, Li J, Qiu Y, Li JX. Agmatine attenuates the discriminative stimulus and hyperthermic effects of methamphetamine in male rats. Behav Pharmacol. 2016;27(6):542–548. doi:10.1097/FBP.0000000000000244

290. Thorn DA, Winter JC, Li JX. Agmatine attenuates methamphetamine-induced conditioned place preference in rats. Eur J Pharmacol. 2012;680(1–3):69–72. doi:10.1016/j.ejphar.2012.01.037

291. Kohut SJ, Fivel PA, Mello NK. Differential effects of acute and chronic treatment with the α2-adrenergic agonist, lofexidine, on cocaine self-administration in rhesus monkeys. Drug Alcohol Depend. 2013;133(2):593–599. doi:10.1016/j.drugalcdep.2013.07.032

292. Czoty PW, Nader MA. Effects of the α-2 adrenergic receptor agonists lofexidine and guanfacine on food-cocaine choice in socially housed cynomolgus monkeys. J Pharmacol Exp Ther. 2020;375(1):193–201. doi:10.1124/jpet.120.266007

293. Freund N, Jordan CJ, Lukkes JL, Norman KJ, Andersen SL. Juvenile exposure to methylphenidate and guanfacine in rats: effects on early delay discounting and later cocaine-taking behavior. Psychopharmacology. 2019;236(2):685–698. doi:10.1007/s00213-018-5096-0

294. Buffalari DM, Baldwin CK, See RE. Treatment of cocaine withdrawal anxiety with guanfacine: relationships to cocaine intake and reinstatement of cocaine seeking in rats. Psychopharmacology. 2012;223(2):179–190. doi:10.1007/s00213-012-2705-1

295. Erb S, Hitchcott PK, Rajabi H, Mueller D, Shaham Y, Stewart J. Alpha-2 adrenergic receptor agonists block stress-induced reinstatement of cocaine seeking. Neuropsychopharmacology. 2000;23(2):138–150. doi:10.1016/S0893-133X(99)00158-X

296. Platt DM, Rowlett JK, Spealman RD. Noradrenergic mechanisms in cocaine-induced reinstatement of drug seeking in squirrel monkeys. J Pharmacol Exp Ther. 2007;322(2):894–902. doi:10.1124/jpet.107.121806

297. Ago Y, Nakamura S, Uda M, et al. Attenuation by the 5-HT1A receptor agonist osemozotan of the behavioral effects of single and repeated methamphetamine in mice. Neuropharmacology. 2006;51(4):914–922. doi:10.1016/j.neuropharm.2006.06.001

298. Picard M, Morisset S, Cloix JF, et al. Pharmacological, neurochemical, and behavioral profile of JB-788, a new 5-HT1A agonist. Neuroscience. 2010;169(3):1337–1346. doi:10.1016/j.neuroscience.2010.05.040

299. Yamazaki M, Harada K, Yamamoto N, et al. ASP5736, a novel 5-HT5A receptor antagonist, ameliorates positive symptoms and cognitive impairment in animal models of schizophrenia. Eur Neuropsychopharmacol. 2014;24(10):1698–1708. doi:10.1016/j.euroneuro.2014.07.009

300. Der-Ghazarian TS, Charmchi D, Noudali SN, et al. Neural circuits associated with 5-HT1B receptor agonist inhibition of methamphetamine seeking in the conditioned place preference model. ACS Chem Neurosci. 2019;10(7):3271–3283. doi:10.1021/acschemneuro.8b00709

301. Suzuki T, Shiozaki Y, Masukawa Y, Misawa M. 5-HT3 receptor antagonists block cocaine- and methamphetamine-induced place preference. Yakubutsu Seishin Kodo. 1992;12(1):33–38.

302. Sicignano D, Hernandez AV, Schiff B, Elmahy N, White CM. The impact of psychedelics on patients with alcohol use disorder: a systematic review with meta-analysis. Curr Med Res Opin. 2024;40(2):293–302. doi:10.1080/03007995.2023.2296968

303. Zaretsky TG, Jagodnik KM, Barsic R, et al. The psychedelic future of post-traumatic stress disorder treatment. Curr Neuropharmacol. 2024;22(4):636–735. doi:10.2174/1570159X22666231027111147

304. Kwan AC, Olson DE, Preller KH, Roth BL. The neural basis of psychedelic action. Nat Neurosci. 2022;25(11):1407–1419. doi:10.1038/s41593-022-01177-4

305. Sakloth F, Leggett E, Moerke MJ, Townsend EA, Banks ML, Negus SS. Effects of acute and repeated treatment with serotonin 5-HT2A receptor agonist hallucinogens on intracranial self-stimulation in rats. Exp Clin Psychopharmacol. 2019;27(3):215–226. doi:10.1037/pha0000253

306. Wang J, Liang M, Shang Q, et al. Psilocin suppresses methamphetamine-induced hyperlocomotion and acquisition of conditioned place preference via D2R-mediated ERK signaling. CNS Neurosci Ther. 2023;29(3):831–841. doi:10.1111/cns.14054

307. Banks ML. Effects of 7-day repeated treatment with the 5-HT2A inverse agonist/antagonist pimavanserin on methamphetamine vs. food choice in male rhesus monkeys. Drug Alcohol Depend. 2016;165:260–264. doi:10.1016/j.drugalcdep.2016.05.014

308. Khera R, Murad MH, Chandar AK, et al. Association of pharmacological treatments for obesity with weight loss and adverse events: a systematic review and meta-analysis. JAMA. 2016;315(22):2424–2434. doi:10.1001/jama.2016.7602

309. Campbell EJ, Bonomo Y, Pastor A, et al. The 5-HT2C receptor as a therapeutic target for alcohol and methamphetamine use disorders: a pilot study in treatment-seeking individuals. Pharmacol Res Perspect. 2021;9(3):e00767. doi:10.1002/prp2.767

310. Johns SE, Keyser-Marcus L, Abbate A, et al. Safety and preliminary efficacy of lorcaserin for cocaine use disorder: a Phase I randomized clinical trial. Front Psychiatry. 2021;12:666945. doi:10.3389/fpsyt.2021.666945

311. McCann DJ, Chen HH, Devine EG, Gyaw S, Ramey T. Results of a randomized, double-blind, placebo-controlled trial of lorcaserin in cocaine use disorder. Drug Alcohol Depend. 2024;255:111063. doi:10.1016/j.drugalcdep.2023.111063

312. de Andrade Mesquita L, Fagundes Piccoli G, Richter da Natividade G, Frison Spiazzi B, Colpani V, Gerchman F. Is lorcaserin really associated with increased risk of cancer? A systematic review and meta-analysis. Obes Rev. 2021;22(3):e13170. doi:10.1111/obr.13170

313. Graves SM, Napier TC. SB 206553, a putative 5-HT2C inverse agonist, attenuates methamphetamine-seeking in rats. BMC Neurosci. 2012;13:65. doi:10.1186/1471-2202-13-65

314. Milne RJ, Heel RC. Ondansetron. Therapeutic use as an antiemetic. Drugs. 1991;41(4):574–595. doi:10.2165/00003495-199141040-00006

315. Yoo JH, Cho JH, Yu HS, et al. Involvement of 5-HT receptors in the development and expression of methamphetamine-induced behavioral sensitization: 5-HT receptor channel and binding study. J Neurochem. 2006;99(3):976–988. doi:10.1111/j.1471-4159.2006.04137.x

316. Johnson BA, Ait-Daoud N, Elkashef AM, et al. A preliminary randomized, double-blind, placebo-controlled study of the safety and efficacy of ondansetron in the treatment of methamphetamine dependence. Int J Neuropsychopharmacol. 2008;11(1):1–14. doi:10.1017/S1461145707007778

317. Ginawi OT, Al-Majed AA, Al-Suwailem AK. Ondansetron, a selective 5-HT3 antagonist, antagonizes methamphetamine-induced anorexia in mice. Pharmacol Res. 2005;51(3):255–259. doi:10.1016/j.phrs.2004.09.002

318. Gainetdinov RR, Hoener MC, Berry MD. Trace amines and their receptors. Pharmacol Rev. 2018;70(3):549–620. doi:10.1124/pr.117.015305

319. Xie Z, Miller GM. Trace amine-associated receptor 1 is a modulator of the dopamine transporter. J Pharmacol Exp Ther. 2007;321(1):128–136. doi:10.1124/jpet.106.117382

320. Xie Z, Miller GM. Beta-phenylethylamine alters monoamine transporter function via trace amine-associated receptor 1: implication for modulatory roles of trace amines in brain. J Pharmacol Exp Ther. 2008;325(2):617–628. doi:10.1124/jpet.107.134247

321. Dave S, Chen L, Yu C, et al. Methamphetamine decreases K+ channel function in human fetal astrocytes by activating the trace amine-associated receptor type-1. J Neurochem. 2019;148(1):29–45. doi:10.1111/jnc.14606

322. Liu H, Zheng Y, Wang Y, et al. Recognition of methamphetamine and other amines by trace amine receptor TAAR1. Nature. 2023;624:7992):663–671. doi:10.1038/s41586-023-06775-1

323. Reese EA, Bunzow JR, Arttamangkul S, Sonders MS, Grandy DK. Trace amine-associated receptor 1 displays species-dependent stereoselectivity for isomers of methamphetamine, amphetamine, and para-hydroxyamphetamine. J Pharmacol Exp Ther. 2007;321(1):178–186. doi:10.1124/jpet.106.115402

324. Cotter R, Pei Y, Mus L, et al. The trace amine-associated receptor 1 modulates methamphetamine’s neurochemical and behavioral effects. Front Neurosci. 2015;9:39. doi:10.3389/fnins.2015.00039

325. Jing L, Zhang Y, Li JX. Effects of the trace amine associated receptor 1 agonist RO5263397 on abuse-related behavioral indices of methamphetamine in rats. Int J Neuropsychopharmacol. 2014;18(4):pyu060. doi:10.1093/ijnp/pyu060

326. Pei Y, Asif-Malik A, Hoener M, Canales JJ. A partial trace amine-associated receptor 1 agonist exhibits properties consistent with a methamphetamine substitution treatment. Addict Biol. 2017;22(5):1246–1256. doi:10.1111/adb.12410

327. Xue Z, Siemian JN, Johnson BN, Zhang Y, Li JX. Methamphetamine-induced impulsivity during chronic methamphetamine treatment in rats: effects of the TAAR 1 agonist RO5263397. Neuropharmacology. 2018;129:36–46. doi:10.1016/j.neuropharm.2017.11.012

328. Yates JR. Determinants of Addiction: Neurobiological, Behavioral, Cognitive, and Sociocultural Factors. London: Academic Press; 2023.

329. Picciotto MR, Higley MJ, Mineur YS. Acetylcholine as a neuromodulator: cholinergic signaling shapes nervous system function and behavior. Neuron. 2012;76(1):116–129. doi:10.1016/j.neuron.2012.08.036

330. Tintignac LA, Brenner HR, Rüegg MA. Mechanisms regulating neuromuscular junction development and function and causes of muscle wasting. Physiol Rev. 2015;95(3):809–852. doi:10.1152/physrev.00033.2014

331. Dobbs LK, Mark GP. Comparison of systemic and local methamphetamine treatment on acetylcholine and dopamine levels in the ventral tegmental area in the mouse. Neuroscience. 2008;156(3):700–711. doi:10.1016/j.neuroscience.2008.07.052

332. Taguchi K, Atobe J, Kato M, et al. The effect of methamphetamine on the release of acetylcholine in the rat striatum. Eur J Pharmacol. 1998;360(2–3):131–137. doi:10.1016/s0014-2999(98)00653-0

333. Koob GF, Volkow ND. Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry. 2016;3(8):760–773. doi:10.1016/S2215-0366(16)00104-8

334. Collins AL, Aitken TJ, Greenfield VY, Ostlund SB, Wassum KM. Nucleus accumbens acetylcholine receptors modulate dopamine and motivation. Neuropsychopharmacology. 2016;41(12):2830–2838. doi:10.1038/npp.2016.81

335. Tiwari P, Dwivedi S, Singh MP, Mishra R, Chandy A. Basic and modern concepts on cholinergic receptor: a review. Asian Pac J Trop Dis. 2013;3(5):413–420. doi:10.1016/S2222-1808(13)60094-8

336. Magee JC, Winhusen T. The coupling of nicotine and stimulant craving during treatment for stimulant dependence. J Consult Clin Psychol. 2016;84(3):230–237. doi:10.1037/ccp0000054

337. Gatch MB, Flores E, Forster MJ. Nicotine and methamphetamine share discriminative stimulus effects. Drug Alcohol Depend. 2008;93(1–2):63–71. doi:10.1016/j.drugalcdep.2007.08.020

338. Hiranita T, Anggadiredja K, Fujisaki C, Watanabe S, Yamamoto T. Nicotine attenuates relapse to methamphetamine-seeking behavior (craving) in rats. Ann N Y Acad Sci. 2004;1025:504–507. doi:10.1196/annals.1316.062

339. Berry JN, Neugebauer NM, Bardo MT. Reinstatement of methamphetamine conditioned place preference in nicotine-sensitized rats. Behav Brain Res. 2012;235(2):158–165. doi:10.1016/j.bbr.2012.07.043

340. Camarasa J, Ratés SG, Pubill D, Escubedo E. The involvement of nicotinic receptor subtypes in the locomotor activity and analgesia induced by methamphetamine in mice. Behav Pharmacol. 2009;20(7):623–630. doi:10.1097/FBP.0b013e328331ba5b

341. Suemaru K, Gomita Y, Furuno K, Araki Y. Chronic nicotine treatment potentiates behavioral responses to dopaminergic drugs in rats. Pharmacol Biochem Behav. 1993;46(1):135–139. doi:10.1016/0091-3057(93)90329-r

342. Kuribara H. Does nicotine modify the psychotoxic effect of methamphetamine? Assessment in terms of locomotor sensitization in mice. J Toxicol Sci. 1999;24(1):55–62. doi:10.2131/jts.24.55

343. Neugebauer NM, Harrod SB, Bardo MT. Nicotine elicits methamphetamine-seeking in rats previously administered nicotine. Drug Alcohol Depend. 2010;106(1):72–78. doi:10.1016/j.drugalcdep.2009.07.018

344. Cardenas A, Lotfipour S. Age- and sex-dependent nicotine pretreatment effects on the enhancement of methamphetamine self-administration in Sprague-Dawley rats. Nicotine Tob Res. 2022;24(8):1186–1192. doi:10.1093/ntr/ntab218

345. Pittenger ST, Chou S, Barrett ST, Catalano I, Lydiatt M, Bevins RA. Nicotine- and cocaine-triggered methamphetamine reinstatement in female and male Sprague-Dawley rats. Pharmacol Biochem Behav. 2017;159:69–75. doi:10.1016/j.pbb.2017.07.003

346. Pipkin JA, Kaplan GJ, Plant CP, et al. Nicotine exposure beginning in adolescence enhances the acquisition of methamphetamine self-administration, but not methamphetamine-primed reinstatement in male rats. Drug Alcohol Depend. 2014;142:341–344. doi:10.1016/j.drugalcdep.2014.06.029

347. Mizoguchi H, Arai S, Koike H, et al. Therapeutic potential of nicotine for methamphetamine-induced impairment of sensorimotor gating: involvement of pallidotegmental neurons. Psychopharmacology. 2009;207(2):235–243. doi:10.1007/s00213-009-1651-z

348. Mizoguchi H, Ibi D, Takase F, et al. Nicotine ameliorates impairment of working memory in methamphetamine-treated rats. Behav Brain Res. 2011;220(1):159–163. doi:10.1016/j.bbr.2011.01.036

349. Vieira-Brock PL, McFadden LM, Nielsen SM, Smith MD, Hanson GR, Fleckenstein AE. Nicotine administration attenuates methamphetamine-induced novel object recognition deficits. Int J Neuropsychopharmacol. 2015;18(12):pyv073. doi:10.1093/ijnp/pyv073

350. Mizoguchi H, Wang T, Kusaba M, Fukumoto K, Yamada K. Nicotine and varenicline ameliorate changes in reward-based choice strategy and altered decision-making in methamphetamine-treated rats. Behav Brain Res. 2019;359:935–941. doi:10.1016/j.bbr.2018.06.016

351. Burke MV, Hays JT, Ebbert JO. Varenicline for smoking cessation: a narrative review of efficacy, adverse effects, use in at-risk populations, and adherence. Patient Prefer Adherence. 2016;10:435–441. doi:10.2147/PPA.S83469

352. Pittenger ST, Barrett ST, Chou S, Bevins RA. The effects of varenicline on methamphetamine self-administration and drug-primed reinstatement in female rats. Behav Brain Res. 2016;300:150–159. doi:10.1016/j.bbr.2015.11.033

353. Pittenger ST, Barrett ST, Chou S, Bevins RA. The effects of varenicline on methamphetamine self-administration and drug-primed reinstatement in male rats. Behav Brain Res. 2017;320:195–199. doi:10.1016/j.bbr.2016.12.005

354. Verrico CD, Mahoney JJ, Thompson-Lake DG, Bennett RS, Newton TF, De La Garza R. Safety and efficacy of varenicline to reduce positive subjective effects produced by methamphetamine in methamphetamine-dependent volunteers. Int J Neuropsychopharmacol. 2014;17(2):223–233. doi:10.1017/S146114571300134X

355. Briones M, Shoptaw S, Cook R, et al. Varenicline treatment for methamphetamine dependence: a randomized, double-blind Phase II clinical trial. Drug Alcohol Depend. 2018;189:30–36. doi:10.1016/j.drugalcdep.2018.04.023

356. Levin ED, Rezvani AH, Wells C, et al. α4β2 Nicotinic receptor desensitizing compounds can decrease self-administration of cocaine and methamphetamine in rats. Eur J Pharmacol. 2019;845:1–7. doi:10.1016/j.ejphar.2018.12.010

357. Köck P, Froelich K, Walter M, Lang U, Dürsteler KM. A systematic literature review of clinical trials and therapeutic applications of ibogaine. J Subst Abuse Treat. 2022;138:108717. doi:10.1016/j.jsat.2021.108717

358. Yu X, Imam SZ, Newport GD, Slikker WJ, Ali SF. Ibogaine blocked methamphetamine-induced hyperthermia and induction of heat shock protein in mice. Brain Res. 1999;823(1–2):213–216. doi:10.1016/s0006-8993(99)01154-3

359. Szumlinski KK, Balogun MY, Maisonneuve IM, Glick SD. Interactions between iboga agents and methamphetamine sensitization: studies of locomotion and stereotypy in rats. Psychopharmacology. 2000;151(2–3):234–241. doi:10.1007/s002130000478

360. Glick SD, Maisonneuve IM, Kitchen BA, Fleck MW. Antagonism of alpha 3 beta 4 nicotinic receptors as a strategy to reduce opioid and stimulant self-administration. Eur J Pharmacol. 2002;438(1–2):99–105. doi:10.1016/s0014-2999(02)01284-0

361. Maisonneuve IM, Glick SD. Anti-addictive actions of an iboga alkaloid congener: a novel mechanism for a novel treatment. Pharmacol Biochem Behav. 2003;75(3):607–618. doi:10.1016/s0091-3057(03)00119-9

362. Friedman JH, Koller WC, Lannon MC, Busenbark K, Swanson-Hyland E, Smith D. Benztropine versus clozapine for the treatment of tremor in Parkinson’s disease. Neurology. 1997;48(4):1077–1081. doi:10.1212/wnl.48.4.1077

363. Dassanayake AF, Canales JJ. Replacement treatment during extinction training with the atypical dopamine uptake inhibitor, JHW-007, reduces relapse to methamphetamine seeking. Neurosci Lett. 2018;671:88–92. doi:10.1016/j.neulet.2018.02.025

364. Ferragud A, Velázquez-Sánchez C, Canales JJ. Modulation of methamphetamine’s locomotor stimulation and self-administration by JHW 007, an atypical dopamine reuptake blocker. Eur J Pharmacol. 2014;731:73–79. doi:10.1016/j.ejphar.2014.03.015

365. Hiranita T, Hong WC, Kopajtic T, Katz JL. σ receptor effects of n-substituted benztropine analogs: implications for antagonism of cocaine self-administration. J Pharmacol Exp Ther. 2017;362(1):2–13. doi:10.1124/jpet.117.241109

366. Spinks A, Wasiak J. Scopolamine (hyoscine) for preventing and treating motion sickness. Cochrane Database Syst Rev. 2011;2011(6):CD002851. doi:10.1002/14651858.CD002851.pub4

367. Ohmori T, Abekawa T, Koyama T. Scopolamine prevents the development of sensitization to methamphetamine. Life Sci. 1995;56(14):1223–1229. doi:10.1016/0024-3205(95)00062-b

368. Ohmori T, Abekawa T, Koyama T. Scopolamine prevents augmentation of stereotypy induced by chronic methamphetamine treatment. Psychopharmacology. 1995;121(2):158–163. doi:10.1007/BF02245625

369. Shimosato K, Watanabe S, Kitayama S. Differential effects of trihexyphenidyl on place preference conditioning and locomotor stimulant activity of cocaine and methamphetamine. Naunyn Schmiedebergs Arch Pharmacol. 2001;364(1):74–80. doi:10.1007/s002100100433

370. Yui K, Miura T. Behavioral responses induced by repeated treatment with methamphetamine alone and in combination with scopolamine in rats. Neuropsychobiology. 1996;33(1):21–27. doi:10.1159/000119244

371. Yui K, Miura T, Sugiyama K, Ono M, Nagase M. Effects of repeated treatment with methamphetamine plus scopolamine and methamphetamine on behavioral sensitization and conditioning. Behav Brain Res. 1996;80(1–2):169–175. doi:10.1016/0166-4328(96)00032-0

372. Lee KW, Tian YH, You IJ, et al. Blockade of M1 muscarinic acetylcholine receptors modulates the methamphetamine-induced psychomotor stimulant effect. Neuroscience. 2008;153(4):1235–1244. doi:10.1016/j.neuroscience.2008.02.021

373. Mandai T, Kasahara M, Kurimoto E, et al. In vivo pharmacological comparison of TAK-071, a positive allosteric modulator of muscarinic M1 receptor, and xanomeline, an agonist of muscarinic M1/M4 Receptor, in rodents. Neuroscience. 2019;414:60–76. doi:10.1016/j.neuroscience.2019.07.003

374. Decima EE, Rand RW. oxotremorine induced tremor in the decorticated cat. Int J Neuropharmacol. 1965;4:139–148. doi:10.1016/0028-3908(65)90003-1

375. Miwa H, Hama K, Kajimoto Y, Kondo T. Effects of zonisamide on experimental tremors in rats. Parkinsonism Relat Disord. 2008;14(1):33–36. doi:10.1016/j.parkreldis.2007.05.008

376. Sur C, Mallorga PJ, Wittmann M, et al. N-desmethylclozapine, an allosteric agonist at muscarinic 1 receptor, potentiates N-methyl-D-aspartate receptor activity. Proc Natl Acad Sci U S A. 2003;100(23):13674–13679. doi:10.1073/pnas.1835612100

377. Maehara S, Satow A, Hikichi H, Ohta H. Antipsychotic effects of N-desmethylclozapine on sensorimotor gating function in rats--possible involvement of activation of M(1) muscarinic receptors. Eur J Pharmacol. 2011;667(1–3):242–249. doi:10.1016/j.ejphar.2011.05.063

378. Soreq H, Seidman S. Acetylcholinesterase--new roles for an old actor. Nat Rev Neurosci. 2001;2(4):294–302. doi:10.1038/35067589

379. Marucci G, Buccioni M, Ben DD, Lambertucci C, Volpini R, Amenta F. Efficacy of acetylcholinesterase inhibitors in Alzheimer’s disease. Neuropharmacology. 2021;190:108352. doi:10.1016/j.neuropharm.2020.108352

380. Takamatsu Y, Yamanishi Y, Hagino Y, Yamamoto H, Ikeda K. Differential effects of donepezil on methamphetamine and cocaine dependencies. Ann N Y Acad Sci. 2006;1074:418–426. doi:10.1196/annals.1369.042

381. Noda Y, Mouri A, Ando Y, et al. Galantamine ameliorates the impairment of recognition memory in mice repeatedly treated with methamphetamine: involvement of allosteric potentiation of nicotinic acetylcholine receptors and dopaminergic-ERK1/2 systems. Int J Neuropsychopharmacol. 2010;13(10):1343–1354. doi:10.1017/S1461145710000222

382. Hiranita T, Nawata Y, Sakimura K, Anggadiredja K, Yamamoto T. Suppression of methamphetamine-seeking behavior by nicotinic agonists. Proc Natl Acad Sci U S A. 2006;103(22):8523–8527. doi:10.1073/pnas.0600347103

383. Koseki T, Mouri A, Suzuki S, et al. Galantamine attenuates reinstatement of cue-induced methamphetamine-seeking behavior in mice. Addict Biol. 2014;19(1):1–4. doi:10.1111/j.1369-1600.2011.00425.x

384. De La Garza R, Mahoney JJ, Culbertson C, Shoptaw S, Newton TF. The acetylcholinesterase inhibitor rivastigmine does not alter total choices for methamphetamine, but may reduce positive subjective effects, in a laboratory model of intravenous self-administration in human volunteers. Pharmacol Biochem Behav. 2008;89(2):200–208. doi:10.1016/j.pbb.2007.12.010

385. De La Garza R, Shoptaw S, Newton TF. Evaluation of the cardiovascular and subjective effects of rivastigmine in combination with methamphetamine in methamphetamine-dependent human volunteers. Int J Neuropsychopharmacol. 2008;11(6):729–741. doi:10.1017/S1461145708008456

386. De La Garza R, Newton TF, Haile CN, et al. Rivastigmine reduces ”Likely to use methamphetamine” in methamphetamine-dependent volunteers. Prog Neuropsychopharmacol Biol Psychiatry. 2012;37(1):141–146. doi:10.1016/j.pnpbp.2011.12.014

387. Kalechstein AD, Yoon JH, Croft DE, Jaeggi S, Mahoney JJ, De La Garza R. Low dose, short-term rivastigmine administration does not affect neurocognition in methamphetamine dependent individuals. Pharmacol Biochem Behav. 2011;99(3):423–427. doi:10.1016/j.pbb.2011.05.013

388. Pathan H, Williams J. Basic opioid pharmacology: an update. Br J Pain. 2012;6(1):11–16. doi:10.1177/2049463712438493

389. Darcq E, Kieffer BL. Opioid receptors: drivers to addiction? Nat Rev Neurosci. 2018;19(8):499–514. doi:10.1038/s41583-018-0028-x

390. Roozen HG, de Waart R, van der Windt DA, van den Brink W, de Jong CA, Kerkhof AJ. A systematic review of the effectiveness of naltrexone in the maintenance treatment of opioid and alcohol dependence. Eur Neuropsychopharmacol. 2006;16(5):311–323. doi:10.1016/j.euroneuro.2005.11.001

391. Wermeling DP. Review of naloxone safety for opioid overdose: practical considerations for new technology and expanded public access. Ther Adv Drug Saf. 2015;6(1):20–31. doi:10.1177/2042098614564776

392. Kitanaka J, Kitanaka N, Hall FS, et al. The selective μ opioid receptor antagonist β-funaltrexamine attenuates methamphetamine-induced stereotypical biting in mice. Brain Res. 2013;1522:88–98. doi:10.1016/j.brainres.2013.05.027

393. Balsara JJ, Nandal NV, Burte NP, Jadhav JH, Chandorkar AG. Effects of naloxone on methamphetamine and apomorphine stereotypy and on haloperidol catalepsy in rats. Psychopharmacology. 1984;82(3):237–240. doi:10.1007/BF00427781

394. Anggadiredja K, Sakimura K, Hiranita T, Yamamoto T. Naltrexone attenuates cue- but not drug-induced methamphetamine seeking: a possible mechanism for the dissociation of primary and secondary reward. Brain Res. 2004;1021(2):272–276. doi:10.1016/j.brainres.2004.06.051

395. Chen LC, Chan MH, Chen HH. Extinction and reinstatement of methamphetamine-induced conditioned place preference in zebrafish. Addict Biol. 2023;28(12):e13351. doi:10.1111/adb.13351

396. Chiu CT, Ma T, Ho IK. Attenuation of methamphetamine-induced behavioral sensitization in mice by systemic administration of naltrexone. Brain Res Bull. 2005;67(1–2):100–109. doi:10.1016/j.brainresbull.2005.05.028

397. Guo LK, Wang ZY, Lu GY, et al. Inhibition of naltrexone on relapse in methamphetamine self-administration and conditioned place preference in rats. Eur J Pharmacol. 2019;865:172671. doi:10.1016/j.ejphar.2019.172671

398. Wang ZY, Guo LK, Han X, et al. Naltrexone attenuates methamphetamine-induced behavioral sensitization and conditioned place preference in mice. Behav Brain Res. 2021;399:112971. doi:10.1016/j.bbr.2020.112971

399. Nawata Y, Ooishi R, Nishioku T, Yamaguchi T. Nalmefene attenuates reinstatement of methamphetamine-seeking behavior in rats through group II metabotropic glutamate receptors (mGluR2/3). Behav Brain Res. 2024;456:114708. doi:10.1016/j.bbr.2023.114708

400. Jones JD, Mumtaz M, Vadhan NP, et al. The effects of acute oral naltrexone pretreatment on the abuse potential of intranasal methamphetamine, and the relationship between reward/punishment sensitivity and methamphetamine’s effects. Behav Pharmacol. 2022;33(4):255–265. doi:10.1097/FBP.0000000000000671

401. Santos GM, Coffin P, Santos D, et al. Feasibility, acceptability, and tolerability of targeted naltrexone for nondependent methamphetamine-using and binge-drinking men who have sex with men. J Acquir Immune Defic Syndr. 2016;72(1):21–30. doi:10.1097/QAI.0000000000000922

402. Kohno M, Dennis LE, McCready H, Schwartz DL, Hoffman WF, Korthuis PT. A preliminary randomized clinical trial of naltrexone reduces striatal resting state functional connectivity in people with methamphetamine use disorder. Drug Alcohol Depend. 2018;192:186–192. doi:10.1016/j.drugalcdep.2018.07.045

403. Ray LA, Bujarski S, Courtney KE, et al. The effects of naltrexone on subjective response to methamphetamine in a clinical sample: a double-blind, placebo-controlled laboratory study. Neuropsychopharmacology. 2015;40(10):2347–2356. doi:10.1038/npp.2015.83

404. Roche DJO, Worley MJ, Courtney KE, et al. Naltrexone moderates the relationship between cue-induced craving and subjective response to methamphetamine in individuals with methamphetamine use disorder. Psychopharmacology. 2017;234(13):1997–2007. doi:10.1007/s00213-017-4607-8

405. Lim AC, Grodin EN, Green R, et al. Executive function moderates naltrexone effects on methamphetamine-induced craving and subjective responses. Am J Drug Alcohol Abuse. 2020;46(5):565–576. doi:10.1080/00952990.2020.1741002

406. Courtney KE, Ghahremani DG, Ray LA. The effects of pharmacological opioid blockade on neural measures of drug cue-reactivity in humans. Neuropsychopharmacology. 2016;41(12):2872–2881. doi:10.1038/npp.2016.99

407. Stoops WW, Pike E, Hays LR, Glaser PE, Rush CR. Naltrexone and bupropion, alone or combined, do not alter the reinforcing effects of intranasal methamphetamine. Pharmacol Biochem Behav. 2015;129:45–50. doi:10.1016/j.pbb.2014.11.018

408. Kidd JD, Smiley SL, Coffin PO, et al. Sexual orientation differences among men in a randomized clinical trial of extended-release naltrexone and bupropion for methamphetamine use disorder. Drug Alcohol Depend. 2023;250:110899. doi:10.1016/j.drugalcdep.2023.110899

409. Coffin PO, Santos GM, Hern J, et al. Extended-release naltrexone for methamphetamine dependence among men who have sex with men: a randomized placebo-controlled trial. Addiction. 2018;113(2):268–278. doi:10.1111/add.13950

410. Philbin MM, Greene ER, LaBossier NJ, Martins SS, McCrimmon T, Mauro PM. Age-related patterns of cocaine and methamphetamine use across the life course in the United States: disparities by gender and sexual identity among adults. Addict Behav. 2023;137:107539. doi:10.1016/j.addbeh.2022.107539

411. Whitfield TW, Schlosburg JE, Wee S, et al. κ Opioid receptors in the nucleus accumbens shell mediate escalation of methamphetamine intake. J Neurosci. 2015;35(10):4296–4305. doi:10.1523/JNEUROSCI.1978-13.2015

412. Suzuki T, Mori T, Tsuji M, Misawa M, Nagase H. The role of delta-opioid receptor subtypes in cocaine- and methamphetamine-induced place preferences. Life Sci. 1994;55(17):PL339–PL344. doi:10.1016/0024-3205(94)00774-8

413. Shan Y, Zhao J, Zheng Y, Guo S, Schrodi SJ, He D. Understanding the function of the GABAergic system and its potential role in rheumatoid arthritis. Front Immunol. 2023;14:1114350. doi:10.3389/fimmu.2023.1114350

414. Nutt DJ, Nestor LJ. The GABA System and Addiction. Oxford: Oxford University Press; 2013. Addiction.

415. Padgett CL, Lalive AL, Tan KR, et al. Methamphetamine-evoked depression of GABA(B) receptor signaling in GABA neurons of the VTA. Neuron. 2012;73(5):978–989. doi:10.1016/j.neuron.2011.12.031

416. Robinson KJ, Everett NA, Baracz SJ, Cornish JL. The effect of self-administered methamphetamine on GABAergic interneuron populations and functional connectivity of the nucleus accumbens and prefrontal cortex. Psychopharmacology. 2022;239(9):2903–2919. doi:10.1007/s00213-022-06175-9

417. Study RE, Barker JL. Cellular mechanisms of benzodiazepine action. JAMA. 1982;247(15):2147–2151.

418. Ito K, Ohmori T, Abekawa T, Koyama T. Clonazepam prevents the development of sensitization to methamphetamine. Pharmacol Biochem Behav. 1997;58(4):875–879. doi:10.1016/s0091-3057(97)00049-x

419. Ito K, Ohmori T, Abekawa T, Koyama T. The role of benzodiazepine receptors in the acquisition and expression of behavioral sensitization to methamphetamine. Pharmacol Biochem Behav. 2000;65(4):705–710. doi:10.1016/s0091-3057(99)00263-4

420. Goeders JE, Goeders NE. Effects of oxazepam on methamphetamine-induced conditioned place preference. Pharmacol Biochem Behav. 2004;78(1):185–188. doi:10.1016/j.pbb.2004.03.008

421. Keller CM, Cornett EM, Guerin GF, Goeders NE. Combinations of oxazepam and metyrapone attenuate cocaine and methamphetamine cue reactivity. Drug Alcohol Depend. 2013;133(2):405–412. doi:10.1016/j.drugalcdep.2013.06.025

422. Spence AL, Guerin GF, Goeders NE. The differential effects of alprazolam and oxazepam on methamphetamine self-administration in rats. Drug Alcohol Depend. 2016;166:209–217. doi:10.1016/j.drugalcdep.2016.07.015

423. Tracy ME, Banks ML, Shelton KL. Negative allosteric modulation of GABAA receptors inhibits facilitation of brain stimulation reward by drugs of abuse in C57BL6/J mice. Psychopharmacology. 2016;233(4):715–725. doi:10.1007/s00213-015-4155-z

424. Lopes EF, Roberts BM, Siddorn RE, Clements MA, Cragg SJ. Inhibition of nigrostriatal dopamine release by striatal GABAA and GABAB receptors. J Neurosci. 2019;39(6):1058–1065. doi:10.1523/JNEUROSCI.2028-18.2018

425. Balsara JJ, Muley MP, Vaidya AS, Chandorkar AG. Effects of baclofen on dopamine-dependent behaviors in mice. Psychopharmacology. 1981;75(4):396–399. doi:10.1007/BF00435861

426. Li SM, Yin LL, Ren YH, Pan LS, Zheng JW. GABA(B) receptor agonist baclofen attenuates the development and expression of d-methamphetamine-induced place preference in rats. Life Sci. 2001;70(3):349–356. doi:10.1016/s0024-3205(01)01397-2

427. Voigt RM, Herrold AA, Napier TC. Baclofen facilitates the extinction of methamphetamine-induced conditioned place preference in rats. Behav Neurosci. 2011;125(2):261–267. doi:10.1037/a0022893

428. Arai S, Takuma K, Mizoguchi H, et al. Involvement of pallidotegmental neurons in methamphetamine- and MK-801-induced impairment of prepulse inhibition of the acoustic startle reflex in mice: reversal by GABAB receptor agonist baclofen. Neuropsychopharmacology. 2008;33(13):3164–3175. doi:10.1038/npp.2008.41

429. Arai S, Takuma K, Mizoguchi H, et al. GABAB receptor agonist baclofen improves methamphetamine-induced cognitive deficit in mice. Eur J Pharmacol. 2009;602(1):101–104. doi:10.1016/j.ejphar.2008.10.065

430. Mizoguchi H, Yamada K. Pharmacologic treatment with GABA(B) receptor agonist of methamphetamine-induced cognitive impairment in mice. Curr Neuropharmacol. 2011;9(1):109–112. doi:10.2174/157015911795016976

431. Heinzerling KG, Shoptaw S, Peck JA, et al. Randomized, placebo-controlled trial of baclofen and gabapentin for the treatment of methamphetamine dependence. Drug Alcohol Depend. 2006;85(3):177–184. doi:10.1016/j.drugalcdep.2006.03.019

432. Ozawa S, Kamiya H, Tsuzuki K. Glutamate receptors in the mammalian central nervous system. Prog Neurobiol. 1998;54(5):581–618. doi:10.1016/s0301-0082(97)00085-3

433. Debono MW, Le Guern J, Canton T, Doble A, Pradier L. Inhibition by riluzole of electrophysiological responses mediated by rat kainate and NMDA receptors expressed in Xenopus oocytes. Eur J Pharmacol. 1993;235(2–3):283–289. doi:10.1016/0014-2999(93)90147-a

434. Itzhak Y, Martin JL. Effect of riluzole and gabapentin on cocaine- and methamphetamine-induced behavioral sensitization in mice. Psychopharmacology. 2000;151(2–3):226–233. doi:10.1007/s002130000394

435. Hrickova M, Amchova P, Ruda-Kucerova J. The effect of CNQX on self-administration: present in nicotine, absent in methamphetamine model. Front Behav Neurosci. 2024;17:1305412. doi:10.3389/fnbeh.2023.1305412

436. Ruda-Kucerova J, Amchova P, Siska F, Tizabi Y. NBQX attenuates relapse of nicotine seeking but not nicotine and methamphetamine self-administration in rats. World J Biol Psychiatry. 2021;22(10):733–743. doi:10.1080/15622975.2021.1907714

437. Johnson JW, Kotermanski SE. Mechanism of action of memantine. Curr Opin Pharmacol. 2006;6(1):61–67. doi:10.1016/j.coph.2005.09.007

438. Camarasa J, Rodrigo T, Pubill D, Escubedo E. Memantine is a useful drug to prevent the spatial and non-spatial memory deficits induced by methamphetamine in rats. Pharmacol Res. 2010;62(5):450–456. doi:10.1016/j.phrs.2010.05.004

439. Long JD, Liu Y, Jiao DL, et al. The neuroprotective effect of memantine on methamphetamine-induced cognitive deficits. Behav Brain Res. 2017;323:133–140. doi:10.1016/j.bbr.2017.01.042

440. Kitanaka N, Kitanaka J, Hall FS, et al. Psychotomimetic-like behavioral effects of memantine in the mouse. Biomed Pharmacother. 2018;100:116–123. doi:10.1016/j.biopha.2018.01.160

441. Hiranita T, Kohut SJ, Soto PL, Tanda G, Kopajtic TA, Katz JL. Preclinical efficacy of N-substituted benztropine analogs as antagonists of methamphetamine self-administration in rats. J Pharmacol Exp Ther. 2014;348(1):174–191. doi:10.1124/jpet.113.208264

442. Hart CL, Haney M, Foltin RW, Fischman MW. Effects of the NMDA antagonist memantine on human methamphetamine discrimination. Psychopharmacology. 2002;164(4):376–384. doi:10.1007/s00213-002-1225-9

443. Kurdi MS, Theerth KA, Deva RS. Ketamine: current applications in anesthesia, pain, and critical care. Anesth Essays Res. 2014;8(3):283–290. doi:10.4103/0259-1162.143110

444. Matveychuk D, Thomas RK, Swainson J, et al. Ketamine as an antidepressant: overview of its mechanisms of action and potential predictive biomarkers. Ther Adv Psychopharmacol. 2020;10:2045125320916657. doi:10.1177/2045125320916657

445. Ruda-Kucerova J, Babinska Z, Stark T, Micale V. Suppression of methamphetamine self-administration by ketamine pre-treatment is absent in the methylazoxymethanol (MAM) rat model of schizophrenia. Neurotox Res. 2017;32(1):121–133. doi:10.1007/s12640-017-9718-9

446. Uzbay IT, Wallis CJ, Lal H, Forster MJ. Effects of NMDA receptor blockers on cocaine-stimulated locomotor activity in mice. Behav Brain Res. 2000;108(1):57–61. doi:10.1016/s0166-4328(99)00129-1

447. Dakwar E, Hart CL, Levin FR, Nunes EV, Foltin RW. Cocaine self-administration disrupted by the N-methyl-D-aspartate receptor antagonist ketamine: a randomized, crossover trial. Mol Psychiatry. 2017;22(1):76–81. doi:10.1038/mp.2016.39

448. Zanettini C, Wilkinson DS, Katz JL. Behavioral economic analysis of the effects of N-substituted benztropine analogs on cocaine self-administration in rats. Psychopharmacology. 2018;235(1):47–58. doi:10.1007/s00213-017-4739-x

449. Maltbie EA, Gopinath KS, Howell LL. Effects of ketamine treatment on cocaine-induced reinstatement and disruption of functional connectivity in unanesthetized rhesus monkeys. Psychopharmacology. 2019;236(7):2105–2118. doi:10.1007/s00213-019-05204-4

450. Dakwar E, Nunes EV, Hart CL, et al. A single ketamine infusion combined with mindfulness-based behavioral modification to treat cocaine dependence: a randomized clinical trial. Am J Psychiatry. 2019;176(11):923–930. doi:10.1176/appi.ajp.2019.18101123

451. Vrajová M, Schutová B, Klaschka J, Stěpánková H, Rípová D, Šlamberová R. Age-related differences in NMDA receptor subunits of prenatally methamphetamine-exposed male rats. Neurochem Res. 2014;39(11):2040–2046. doi:10.1007/s11064-014-1381-4

452. Furlong TM, Corbit LH, Brown RA, Balleine BW. Methamphetamine promotes habitual action and alters the density of striatal glutamate receptor and vesicular proteins in dorsal striatum. Addict Biol. 2018;23(3):857–867. doi:10.1111/adb.12534

453. Li J, Liu W, Peng Q, et al. Effect of rhynchophylline on conditioned place preference on expression of NR2B in methamphetamine-dependent mice. Biochem Biophys Res Commun. 2014;452(3):695–700. doi:10.1016/j.bbrc.2014.08.127

454. Mishra D, Pena-Bravo JI, Leong KC, Lavin A, Reichel CM. Methamphetamine self-administration modulates glutamate neurophysiology. Brain Struct Funct. 2017;222(5):2031–2039. doi:10.1007/s00429-016-1322-x

455. Gipson CD, Reissner KJ, Kupchik YM, et al. Reinstatement of nicotine seeking is mediated by glutamatergic plasticity. Proc Natl Acad Sci U S A. 2013;110(22):9124–9129. doi:10.1073/pnas.1220591110

456. Shen H, Moussawi K, Zhou W, Toda S, Kalivas PW. Heroin relapse requires long-term potentiation-like plasticity mediated by NMDA2b-containing receptors. Proc Natl Acad Sci U S A. 2011;108(48):19407–19412. doi:10.1073/pnas.1112052108

457. Westbrook SR, Gulley JM. Effects of the GluN2B antagonist, Ro 25-6981, on extinction consolidation following adolescent- or adult-onset methamphetamine self-administration in male and female rats. Behav Pharmacol. 2020;31(8):748–758. doi:10.1097/FBP.0000000000000586

458. Li L, Qiao C, Chen G, et al. Ifenprodil attenuates the acquisition and expression of methamphetamine-induced behavioral sensitization and activation of Ras-ERK1/2 cascade in the caudate putamen. Neuroscience. 2016;335:20–29. doi:10.1016/j.neuroscience.2016.08.022

459. Li MH, Underhill SM, Reed C, Phillips TJ, Amara SG, Ingram SL. Amphetamine and methamphetamine increase NMDAR-GluN2B synaptic currents in midbrain dopamine neurons. Neuropsychopharmacology. 2017;42(7):1539–1547. doi:10.1038/npp.2016.278

460. Yates JR, Campbell HL, Hawley LL, Horchar MJ, Kappesser JL, Wright MR. Effects of the GluN2B-selective antagonist Ro 63-1908 on acquisition and expression of methamphetamine conditioned place preference in male and female rats. Drug Alcohol Depend. 2021;225:108785. doi:10.1016/j.drugalcdep.2021.108785

461. Schwendt M, Reichel CM, See RE. Extinction-dependent alterations in corticostriatal mGluR2/3 and mGluR7 receptors following chronic methamphetamine self-administration in rats. PLoS One. 2012;7(3):e34299. doi:10.1371/journal.pone.0034299

462. Ago Y, Araki R, Yano K, et al. Activation of metabotropic glutamate 2/3 receptors attenuates methamphetamine-induced hyperlocomotion and increase in prefrontal serotonergic neurotransmission. Psychopharmacology. 2011;217(3):443–452. doi:10.1007/s00213-011-2295-3

463. Hiyoshi T, Marumo T, Hikichi H, et al. Neurophysiologic and antipsychotic profiles of TASP0433864, a novel positive allosteric modulator of metabotropic glutamate 2 receptor. J Pharmacol Exp Ther. 2014;351(3):642–653. doi:10.1124/jpet.114.218651

464. Crawford JT, Roberts DC, Beveridge TJ. The group II metabotropic glutamate receptor agonist, LY379268, decreases methamphetamine self-administration in rats. Drug Alcohol Depend. 2013;132(3):414–419. doi:10.1016/j.drugalcdep.2013.07.024

465. Kufahl PR, Watterson LR, Nemirovsky NE, et al. Attenuation of methamphetamine seeking by the mGluR2/3 agonist LY379268 in rats with histories of restricted and escalated self-administration. Neuropharmacology. 2013;66:290–301. doi:10.1016/j.neuropharm.2012.05.037

466. Iijima M, Koike H, Chaki S. Effect of an mGlu2/3 receptor antagonist on depressive behavior induced by withdrawal from chronic treatment with methamphetamine. Behav Brain Res. 2013;246:24–28. doi:10.1016/j.bbr.2013.02.039

467. Murray CH, Loweth JA, Milovanovic M, et al. AMPA receptor and metabotropic glutamate receptor 1 adaptations in the nucleus accumbens core during incubation of methamphetamine craving. Neuropsychopharmacology. 2019;44(9):1534–1541. doi:10.1038/s41386-019-0425-5

468. Satow A, Maehara S, Ise S, et al. Pharmacological effects of the metabotropic glutamate receptor 1 antagonist compared with those of the metabotropic glutamate receptor 5 antagonist and metabotropic glutamate receptor 2/3 agonist in rodents: detailed investigations with a selective allosteric metabotropic glutamate receptor 1 antagonist, FTIDC [4-[1-(2-fluoropyridine-3-yl)-5-methyl-1H-1,2,3-triazol-4-yl]-N-isopropyl-N-methyl-3,6-dihydropyridine-1(2H)-carboxamide]. J Pharmacol Exp Ther. 2008;326(2):577–586. doi:10.1124/jpet.108.138107

469. Satow A, Suzuki G, Maehara S, et al. Unique antipsychotic activities of the selective metabotropic glutamate receptor 1 allosteric antagonist 2-cyclopropyl-5-[1-(2-fluoro-3-pyridinyl)-5-methyl-1H-1,2,3-triazol-4-yl]-2,3-dihydro-1H-isoindol-1-one. J Pharmacol Exp Ther. 2009;330(1):179–190. doi:10.1124/jpet.109.151118

470. Achat-Mendes C, Platt DM, Spealman RD. Antagonism of metabotropic glutamate 1 receptors attenuates behavioral effects of cocaine and methamphetamine in squirrel monkeys. J Pharmacol Exp Ther. 2012;343(1):214–224. doi:10.1124/jpet.112.196295

471. Osborne MP, Olive MF. A role for mGluR5 receptors in intravenous methamphetamine self-administration. Ann N Y Acad Sci. 2008;1139:206–211. doi:10.1196/annals.1432.034

472. Reichel CM, Schwendt M, McGinty JF, Olive MF, See RE. Loss of object recognition memory produced by extended access to methamphetamine self-administration is reversed by positive allosteric modulation of metabotropic glutamate receptor 5. Neuropsychopharmacology. 2011;36(4):782–792. doi:10.1038/npp.2010.212

473. Herrold AA, Voigt RM, Napier TC. mGluR5 is necessary for maintenance of methamphetamine-induced associative learning. Eur Neuropsychopharmacol. 2013;23(7):691–696. doi:10.1016/j.euroneuro.2012.05.014

474. Pedre B, Barayeu U, Ezeriņa D, Dick TP. The mechanism of action of N-acetylcysteine (NAC): the emerging role of H2S and sulfane sulfur species. Pharmacol Ther. 2021;228:107916. doi:10.1016/j.pharmthera.2021.107916

475. Smaga I, Frankowska M, Filip M. N-acetylcysteine in substance use disorder: a lesson from preclinical and clinical research. Pharmacol Rep. 2021;73(5):1205–1219. doi:10.1007/s43440-021-00283-7

476. Kalivas PW. The glutamate homeostasis hypothesis of addiction. Nat Rev Neurosci. 2009;10(8):561–572. doi:10.1038/nrn2515

477. Fukami G, Hashimoto K, Koike K, Okamura N, Shimizu E, Iyo M. Effect of antioxidant N-acetyl-L-cysteine on behavioral changes and neurotoxicity in rats after administration of methamphetamine. Brain Res. 2004;1016(1):90–95. doi:10.1016/j.brainres.2004.04.072

478. Grant JE, Odlaug BL, Kim SW. A double-blind, placebo-controlled study of N-acetyl cysteine plus naltrexone for methamphetamine dependence. Eur Neuropsychopharmacol. 2010;20(11):823–828. doi:10.1016/j.euroneuro.2010.06.018

479. McKetin R, Dean OM, Turner A, et al. N-acetylcysteine (NAC) for methamphetamine dependence: a randomised controlled trial. EClinicalMedicine. 2021;38:101005. doi:10.1016/j.eclinm.2021.101005

480. Mousavi SG, Sharbafchi MR, Salehi M, Peykanpour M, Karimian Sichani N, Maracy M. The efficacy of N-acetylcysteine in the treatment of methamphetamine dependence: a double-blind controlled, crossover study. Arch Iran Med. 2015;18(1):28–33.

481. Rogawski MA, Löscher W, Rho JM. Mechanisms of action of antiseizure drugs and the ketogenic diet. Cold Spring Harb Perspect Med. 2016;6(5):a022780. doi:10.1101/cshperspect.a022780

482. Wiffen PJ, Derry S, Bell RF, et al. Gabapentin for chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2017;6(6):CD007938. doi:10.1002/14651858.CD007938.pub4

483. Kukkar A, Bali A, Singh N, Jaggi AS. Implications and mechanism of action of gabapentin in neuropathic pain. Arch Pharm Res. 2013;36(3):237–251. doi:10.1007/s12272-013-0057-y

484. Kurokawa K, Shibasaki M, Mizuno K, Ohkuma S. Gabapentin blocks methamphetamine-induced sensitization and conditioned place preference via inhibition of α₂/δ-1 subunits of the voltage-gated calcium channels. Neuroscience. 2011;176:328–335. doi:10.1016/j.neuroscience.2010.11.062

485. Urschel HC, Hanselka LL, Gromov I, White L, Baron M. Open-label study of a proprietary treatment program targeting type A gamma-aminobutyric acid receptor dysregulation in methamphetamine dependence. Mayo Clin Proc. 2007;82(10):1170–1178. doi:10.4065/82.10.1170

486. Ling W, Shoptaw S, Hillhouse M, et al. Double-blind placebo-controlled evaluation of the PROMETA™ protocol for methamphetamine dependence. Addiction. 2012;107(2):361–369. doi:10.1111/j.1360-0443.2011.03619.x

487. Mula M, Cavanna AE, Monaco F. Psychopharmacology of topiramate: from epilepsy to bipolar disorder. Neuropsychiatr Dis Treat. 2006;2(4):475–488. doi:10.2147/nedt.2006.2.4.475

488. Johnson BA, Ait-Daoud N, Wang XQ, et al. Topiramate for the treatment of cocaine addiction: a randomized clinical trial. JAMA Psychiatry. 2013;70(12):1338–1346. doi:10.1001/jamapsychiatry.2013.2295

489. Kampman KM, Pettinati HM, Lynch KG, Spratt K, Wierzbicki MR, O’Brien CP. A double-blind, placebo-controlled trial of topiramate for the treatment of comorbid cocaine and alcohol dependence. Drug Alcohol Depend. 2013;133(1):94–99. doi:10.1016/j.drugalcdep.2013.05.026

490. Moghaddam Sadegh A, Nazarinasab M, Behrouzian F, Rostami H, Mehrabi M. The effectiveness of topiramate in the treatment of amphetamine and methamphetamine use disorder: a randomized controlled trial. Iran J Psychiatry. 2023;18(4):371–379. doi:10.18502/ijps.v18i4.13624

491. Elkashef A, Kahn R, Yu E, et al. Topiramate for the treatment of methamphetamine addiction: a multi-center placebo-controlled trial. Addiction. 2012;107(7):1297–1306. doi:10.1111/j.1360-0443.2011.03771.x

492. Rezaei F, Ghaderi E, Mardani R, Hamidi S, Hassanzadeh K. Topiramate for the management of methamphetamine dependence: a pilot randomized, double-blind, placebo-controlled trial. Fundam Clin Pharmacol. 2016;30(3):282–289. doi:10.1111/fcp.12178

493. Ma JZ, Johnson BA, Yu E, et al. Fine-grain analysis of the treatment effect of topiramate on methamphetamine addiction with latent variable analysis. Drug Alcohol Depend. 2013;130(1–3):45–51. doi:10.1016/j.drugalcdep.2012.10.009

494. Johnson BA, Roache JD, Ait-Daoud N, et al. Effects of topiramate on methamphetamine-induced changes in attentional and perceptual-motor skills of cognition in recently abstinent methamphetamine-dependent individuals. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31(1):123–130. doi:10.1016/j.pnpbp.2006.08.002

495. Rass O, Umbricht A, Bigelow GE, Strain EC, Johnson MW, Mintzer MZ. Topiramate impairs cognitive function in methadone-maintained individuals with concurrent cocaine dependence. Psychol Addict Behav. 2015;29(1):237–246. doi:10.1037/adb0000027

496. Gatson TN, Freemont OA, Coleman RL, et al. Vigabatrin (Sabril) for the treatment of refractory complex focal seizures in adults: pharmacologic and clinical considerations. Cureus. 2023;15(10):e46414. doi:10.7759/cureus.46414

497. Gerasimov MR, Ashby CR, Gardner EL, Mills MJ, Brodie JD, Dewey SL. Gamma-vinyl GABA inhibits methamphetamine, heroin, or ethanol-induced increases in nucleus accumbens dopamine. Synapse. 1999;34(1):11–19. doi:10.1002/(SICI)1098-2396(199910)34:1<11::AID-SYN2>3.0.CO;2-5

498. DeMarco A, Dalal RM, Pai J, et al. Racemic gamma vinyl-GABA (R,S-GVG) blocks methamphetamine-triggered reinstatement of conditioned place preference. Synapse. 2009;63(2):87–94. doi:10.1002/syn.20582

499. De La Garza R, Zorick T, Heinzerling KG, et al. The cardiovascular and subjective effects of methamphetamine combined with gamma-vinyl-gamma-aminobutyric acid (GVG) in non-treatment seeking methamphetamine-dependent volunteers. Pharmacol Biochem Behav. 2009;94(1):186–193. doi:10.1016/j.pbb.2009.08.007

500. Brodie JD, Figueroa E, Laska EM, Dewey SL. Safety and efficacy of gamma-vinyl GABA (GVG) for the treatment of methamphetamine and/or cocaine addiction. Synapse. 2005;55(2):122–125. doi:10.1002/syn.20097

501. Fechtner RD, Khouri AS, Figueroa E, et al. Short-term treatment of cocaine and/or methamphetamine abuse with vigabatrin: ocular safety pilot results. Arch Ophthalmol. 2006;124(9):1257–1262. doi:10.1001/archopht.124.9.1257

502. Elliott WJ, Ram CV. Calcium channel blockers. J Clin Hypertens. 2011;13(9):687–689. doi:10.1111/j.1751-7176.2011.00513.x

503. Andres MA, Cooke IM, Bellinger FP, et al. Methamphetamine acutely inhibits voltage-gated calcium channels but chronically up-regulates L-type channels. J Neurochem. 2015;134(1):56–65. doi:10.1111/jnc.13104

504. Shibasaki M, Kurokawa K, Ohkuma S. Upregulation of L-type Ca(v)1 channels in the development of psychological dependence. Synapse. 2010;64(6):440–444. doi:10.1002/syn.20745

505. Chen X, Xing J, Jiang L, et al. Involvement of calcium/calmodulin-dependent protein kinase II in methamphetamine-induced neural damage. J Appl Toxicol. 2016;36(11):1460–1467. doi:10.1002/jat.3301

506. Khanzode SD, Mahakalkar SM, Belorkar NR, Kharkar VT, Manekar MS. Effect of pre-treatment of some calcium channel blockers on catalepsy and stereotypic behaviour in rats. Indian J Physiol Pharmacol. 1996;40(2):159–162.

507. Moorthy NS, Balsara JJ. Effects of flunarizine on dopamine dependent behaviours in rats. Indian J Med Sci. 1999;53(2):43–48.

508. Blake BL, Muehlmann AM, Egami K, Breese GR, Devine DP, Jinnah HA. Nifedipine suppresses self-injurious behaviors in animals. Dev Neurosci. 2007;29(3):241–250. doi:10.1159/000096414

509. Shibasaki M, Mizuno K, Kurokawa K, Ohkuma S. L-type voltage-dependent calcium channels facilitate acetylation of histone H3 through PKCγ phosphorylation in mice with methamphetamine-induced place preference. J Neurochem. 2011;118(6):1056–1066. doi:10.1111/j.1471-4159.2011.07387.x

510. Suzuki T, Shiozaki Y, Masukawa Y, Misawa M. Effects of calcium antagonists on the cocaine- and methamphetamine-induced conditioned place preference. Arukoru Kenkyuto Yakubutsu Ison. 1992;27(1):81–90.

511. Voigt RM, Riddle JL, Napier TC. Effect of fendiline on the maintenance and expression of methamphetamine-induced conditioned place preference in Sprague-Dawley rats. Psychopharmacology. 2014;231(9):2019–2029. doi:10.1007/s00213-013-3347-7

512. Choi S, Methiwala HN, Graves SM. Isradipine, an L-type calcium channel inhibitor, attenuates cue-associated methamphetamine-seeking in mice. Brain Res. 2023;1818:148528. doi:10.1016/j.brainres.2023.148528

513. Johnson BA, Roache JD, Bordnick PS, Ait-Daoud N. Isradipine, a dihydropyridine-class calcium channel antagonist, attenuates some of d-methamphetamine’s positive subjective effects: a preliminary study. Psychopharmacology. 1999;144(3):295–300. doi:10.1007/s002130051007

514. Johnson BA, Roache JD, Ait-Daoud N, et al. Effects of isradipine, a dihydropyridine-class calcium-channel antagonist, on d-methamphetamine’s subjective and reinforcing effects. Int J Neuropsychopharmacol. 2005;8(2):203–213. doi:10.1017/S1461145704005036

515. Pergolizzi J, Varrassi G, Coleman M, et al. The sigma enigma: a narrative review of sigma receptors. Cureus. 2023;15(3):e35756. doi:10.7759/cureus.35756

516. Tchedre KT, Huang RQ, Dibas A, Krishnamoorthy RR, Dillon GH, Yorio T. Sigma-1 receptor regulation of voltage-gated calcium channels involves a direct interaction. Invest Ophthalmol Vis Sci. 2008;49(11):4993–5002. doi:10.1167/iovs.08-1867

517. Matsumoto RR, Shaikh J, Wilson LL, Vedam S, Coop A. Attenuation of methamphetamine-induced effects through the antagonism of sigma (sigma) receptors: evidence from in vivo and in vitro studies. Eur Neuropsychopharmacol. 2008;18(12):871–881. doi:10.1016/j.euroneuro.2008.07.006

518. Nguyen EC, McCracken KA, Liu Y, Pouw B, Matsumoto RR. Involvement of sigma (sigma) receptors in the acute actions of methamphetamine: receptor binding and behavioral studies. Neuropharmacology. 2005;49(5):638–645. doi:10.1016/j.neuropharm.2005.04.016

519. Seminerio MJ, Kaushal N, Shaikh J, Huber JD, Coop A, Matsumoto RR. Sigma (σ) receptor ligand, AC927 (N-phenethylpiperidine oxalate), attenuates methamphetamine-induced hyperthermia and serotonin damage in mice. Pharmacol Biochem Behav. 2011;98(1):12–20. doi:10.1016/j.pbb.2010.11.023

520. Kaushal N, Seminerio MJ, Shaikh J, et al. CM156, a high affinity sigma ligand, attenuates the stimulant and neurotoxic effects of methamphetamine in mice. Neuropharmacology. 2011;61(5–6):992–1000. doi:10.1016/j.neuropharm.2011.06.028

521. Miller DK, Park ES, Lever SZ, Lever JR. N-phenylpropyl-N’-substituted piperazines occupy sigma receptors and alter methamphetamine-induced hyperactivity in mice. Pharmacol Biochem Behav. 2016;150-151:198–206. doi:10.1016/j.pbb.2016.11.003

522. Rodvelt KR, Oelrichs CE, Blount LR, et al. The sigma receptor agonist SA4503 both attenuates and enhances the effects of methamphetamine. Drug Alcohol Depend. 2011;116(1–3):203–210. doi:10.1016/j.drugalcdep.2010.12.018

523. Tapia MA, Lever JR, Lever SZ, Will MJ, Park ES, Miller DK. Sigma-1 receptor ligand PD144418 and sigma-2 receptor ligand YUN-252 attenuate the stimulant effects of methamphetamine in mice. Psychopharmacology. 2019;236(11):3147–3158. doi:10.1007/s00213-019-05268-2

524. Battista N, Di Tommaso M, Bari M, Maccarrone M. The endocannabinoid system: an overview. Front Behav Neurosci. 2012;6:9. doi:10.3389/fnbeh.2012.00009

525. Berger AA, Keefe J, Winnick A, et al. Cannabis and cannabidiol (CBD) for the treatment of fibromyalgia. Best Pract Res Clin Anaesthesiol. 2020;34(3):617–631. doi:10.1016/j.bpa.2020.08.010

526. Mücke M, Phillips T, Radbruch L, Petzke F, Häuser W. Cannabis-based medicines for chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2018;3(3):CD012182. doi:10.1002/14651858.CD012182.pub2

527. Black N, Stockings E, Campbell G, et al. Cannabinoids for the treatment of mental disorders and symptoms of mental disorders: a systematic review and meta-analysis. Lancet Psychiatry. 2019;6(12):995–1010. doi:10.1016/S2215-0366(19)30401-8

528. Spanagel R. Cannabinoids and the endocannabinoid system in reward processing and addiction: from mechanisms to interventions. Dialogues Clin Neurosci. 2020;22(3):241–250. doi:10.31887/DCNS.2020.22.3/rspanagel

529. Castelli MP, Madeddu C, Casti A, et al. Δ9-tetrahydrocannabinol prevents methamphetamine-induced neurotoxicity. PLoS One. 2014;9(5):e98079. doi:10.1371/journal.pone.0098079

530. Evans MA, Harbison RD, Brown DJ, Forney RB. Stimulant actions of delta9-tetrahydrocannabinol in mice. Psychopharmacology. 1976;50(3):245–250. doi:10.1007/BF00426840

531. Consroe PF, Jones BC, Akins F. Delta9-tetrahydrocannabinol methamphetamine interaction in the rabbit. Neuropharmacology. 1975;14(5–6):377–383. doi:10.1016/0028-3908(75)90020-9

532. Consroe P, Jones B, Laird H. EEG and behavioral effects of delta9-tetrahydrocannabinol in combination with stimulant drugs in rabbits. Psychopharmacology. 1976;50(1):47–52. doi:10.1007/BF00634153

533. Anggadiredja K, Nakamichi M, Hiranita T, et al. Endocannabinoid system modulates relapse to methamphetamine seeking: possible mediation by the arachidonic acid cascade. Neuropsychopharmacology. 2004;29(8):1470–1478. doi:10.1038/sj.npp.1300454

534. Jing L, Qiu Y, Zhang Y, Li JX. Effects of the cannabinoid CB₁ receptor allosteric modulator ORG 27569 on reinstatement of cocaine- and methamphetamine-seeking behavior in rats. Drug Alcohol Depend. 2014;143:251–256. doi:10.1016/j.drugalcdep.2014.08.004

535. Ramshini E, Dabiri S, Arjmand S, et al. Attenuation effect of cannabinoid type 1 receptor activation on methamphetamine-induced neurodegeneration and locomotion impairments among male rats. Addict Health. 2017;9(4):206–213.

536. Ramshini E, Sepehri G, Ahmadi-Zeidabadi M, Arjmand S, Khaksari M, Shabani M. Contribution of CB1Rs in anxiety-related behaviors but not locomotor deficits induced by methamphetamine. Neurosci Lett. 2018;665:240–245. doi:10.1016/j.neulet.2017.12.021

537. Gu SM, Seo S, Park D, et al. Cannabinoid receptor type 1 regulates drug reward behavior via glutamate decarboxylase 67 transcription. Int J Mol Sci. 2021;22(19):10486. doi:10.3390/ijms221910486

538. Kicman A, Toczek M. The effects of cannabidiol, a non-intoxicating compound of cannabis, on the cardiovascular system in health and disease. Int J Mol Sci. 2020;21(18):6740. doi:10.3390/ijms21186740

539. Legare CA, Raup-Konsavage WM, Vrana KE. Therapeutic potential of cannabis, cannabidiol, and cannabinoid-based pharmaceuticals. Pharmacology. 2022;107(3–4):131–149. doi:10.1159/000521683

540. Umpierrez LS, Costa PA, Michelutti EA, et al. Cannabidiol but not cannabidiolic acid reduces behavioural sensitisation to methamphetamine in rats, at pharmacologically effective doses. Psychopharmacology. 2022;239(5):1593–1603. doi:10.1007/s00213-022-06119-3

541. Hay GL, Baracz SJ, Everett NA, et al. Cannabidiol treatment reduces the motivation to self-administer methamphetamine and methamphetamine-primed relapse in rats. J Psychopharmacol. 2018;32(12):1369–1378. doi:10.1177/0269881118799954

542. Liu L, Li J, Wang C, et al. Cannabidiol attenuates methamphetamine-induced conditioned place preference in male rats and viability in PC12 cells through the Sigma1R/AKT/GSK3β/CREB signaling pathway. Am J Drug Alcohol Abuse. 2022;48(5):548–561. doi:10.1080/00952990.2022.2073450

543. Yang G, Liu L, Zhang R, et al. Cannabidiol attenuates methamphetamine-induced conditioned place preference via the Sigma1R/AKT/GSK-3β/CREB signaling pathway in rats. Toxicol Res. 2020;9(3):202–211. doi:10.1093/toxres/tfaa021

544. de Meneses-Gaya C, Crippa JA, Hallak JE, et al. Cannabidiol for the treatment of crack-cocaine craving: an exploratory double-blind study. Braz J Psychiatry. 2021;43(5):467–476. doi:10.1590/1516-4446-2020-1416

545. Mongeau-Pérusse V, Brissette S, Bruneau J, et al. Cannabidiol as a treatment for craving and relapse in individuals with cocaine use disorder: a randomized placebo-controlled trial. Addiction. 2021;116(9):2431–2442. doi:10.1111/add.15417

546. Landa L, Sulcova A, Slais K. Involvement of cannabinoid CB1 and CB2 receptor activity in the development of behavioural sensitization to methamphetamine effects in mice. Neuro Endocrinol Lett. 2006;27(1–2):63–69.

547. Schindler CW, Panlilio LV, Gilman JP, et al. Effects of cannabinoid receptor antagonists on maintenance and reinstatement of methamphetamine self-administration in rhesus monkeys. Eur J Pharmacol. 2010;633(1–3):44–49. doi:10.1016/j.ejphar.2010.02.005

548. Vinklerová J, Nováková J, Sulcová A. Inhibition of methamphetamine self-administration in rats by cannabinoid receptor antagonist AM 251. J Psychopharmacol. 2002;16(2):139–143. doi:10.1177/026988110201600204

549. Boctor SY, Martinez JL, Koek W, France CP. The cannabinoid CB1 receptor antagonist AM251 does not modify methamphetamine reinstatement of responding. Eur J Pharmacol. 2007;571(1):39–43. doi:10.1016/j.ejphar.2007.06.004

550. Yu LL, Wang XY, Zhao M, et al. Effects of cannabinoid CB1 receptor antagonist rimonabant in consolidation and reconsolidation of methamphetamine reward memory in mice. Psychopharmacology. 2009;204(2):203–211. doi:10.1007/s00213-008-1450-y

551. Yu LL, Zhou SJ, Wang XY, et al. Effects of cannabinoid CB₁ receptor antagonist rimonabant on acquisition and reinstatement of psychostimulant reward memory in mice. Behav Brain Res. 2011;217(1):111–116. doi:10.1016/j.bbr.2010.10.008

552. Khodamoradi M, Tirgar F, Ghazvini H, et al. Role of the cannabinoid CB1 receptor in methamphetamine-induced social and recognition memory impairment. Neurosci Lett. 2022;779:136634. doi:10.1016/j.neulet.2022.136634

553. Moreira FA, Crippa JA. The psychiatric side-effects of rimonabant. Braz J Psychiatry. 2009;31(2):145–153. doi:10.1590/s1516-44462009000200012

554. Lu HC, Mackie K. An introduction to the endogenous cannabinoid system. Biol Psychiatry. 2016;79(7):516–525. doi:10.1016/j.biopsych.2015.07.028

555. Ebrahimi-Ghiri M, Khakpai F, Zarrindast MR. URB597 abrogates anxiogenic and depressive behaviors in the methamphetamine-withdrawal mice: role of the cannabinoid receptor type 1, cannabinoid receptor type 2, and transient receptor potential vanilloid 1 channels. J Psychopharmacol. 2021;35(7):875–884. doi:10.1177/0269881120965934

556. Nawata Y, Yamaguchi T, Fukumori R, Yamamoto T. Inhibition of monoacylglycerol lipase reduces the reinstatement of methamphetamine-seeking and anxiety-like behaviors in methamphetamine self-administered rats. Int J Neuropsychopharmacol. 2019;22(2):165–172. doi:10.1093/ijnp/pyy086

557. Benigni A, Cassis P, Remuzzi G. Angiotensin II revisited: new roles in inflammation, immunology and aging. EMBO Mol Med. 2010;2(7):247–257. doi:10.1002/emmm.201000080

558. Kobiec T, Otero-Losada M, Chevalier G, et al. The renin-angiotensin system modulates dopaminergic neurotransmission: a new player on the scene. Front Synaptic Neurosci. 2021;13:638519. doi:10.3389/fnsyn.2021.638519

559. Jiang L, Zhu R, Bu Q, et al. Brain renin-angiotensin system blockade attenuates methamphetamine-induced hyperlocomotion and neurotoxicity. Neurotherapeutics. 2018;15(2):500–510. doi:10.1007/s13311-018-0613-8

560. Xu X, Fan R, Ruan Y, et al. Inhibition of PLCβ1 signaling pathway regulates methamphetamine self-administration and neurotoxicity in rats. Food Chem Toxicol. 2021;149:111970. doi:10.1016/j.fct.2021.111970

561. Qian L, Ruan Y, Gong X, et al. The neuroprotective effect of LCZ696 on methamphetamine-induced cognitive impairment in mice. Neurosci Lett. 2024;823:137630. doi:10.1016/j.neulet.2024.137630

562. Xu X, Pan J, Li X, et al. Inhibition of methamphetamine self-administration and reinstatement by central blockade of angiotensin II receptor in rats. J Pharmacol Exp Ther. 2019;369(2):244–258. doi:10.1124/jpet.118.255729

563. Newton TF, De La Garza R, Grasing K. The angiotensin-converting enzyme inhibitor perindopril treatment alters cardiovascular and subjective effects of methamphetamine in humans. Psychiatry Res. 2010;179(1):96–100. doi:10.1016/j.psychres.2009.11.011

564. Verrico CD, Haile CN, De La Garza R, Grasing K, Kosten TR, Newton TF. Subjective and cardiovascular effects of intravenous methamphetamine during perindopril maintenance: a randomized, double-blind, placebo-controlled human laboratory study. Int J Neuropsychopharmacol. 2016;19(7):pyw029. doi:10.1093/ijnp/pyw029

565. Cochran DM, Fallon D, Hill M, Frazier JA. The role of oxytocin in psychiatric disorders: a review of biological and therapeutic research findings. Harv Rev Psychiatry. 2013;21(5):219–247. doi:10.1097/HRP.0b013e3182a75b7d

566. Baracz SJ, Parker LM, Suraev AS, et al. Chronic methamphetamine self-administration dysregulates oxytocin plasma levels and oxytocin receptor fibre density in the nucleus accumbens core and subthalamic nucleus of the rat. J Neuroendocrinol. 2016;28(4):10.1111/jne.12337. doi:10.1111/jne.12337

567. Zanos P, Wright SR, Georgiou P, et al. Chronic methamphetamine treatment induces oxytocin receptor up-regulation in the amygdala and hypothalamus via an adenosine A2A receptor-independent mechanism. Pharmacol Biochem Behav. 2014;119:72–79. doi:10.1016/j.pbb.2013.05.009

568. Krasnova IN, Gerra MC, Walther D, et al. Compulsive methamphetamine taking in the presence of punishment is associated with increased oxytocin expression in the nucleus accumbens of rats. Sci Rep. 2017;7(1):8331. doi:10.1038/s41598-017-08898-8

569. Carson DS, Cornish JL, Guastella AJ, Hunt GE, McGregor IS. Oxytocin decreases methamphetamine self-administration, methamphetamine hyperactivity, and relapse to methamphetamine-seeking behaviour in rats. Neuropharmacology. 2010;58(1):38–43. doi:10.1016/j.neuropharm.2009.06.018

570. Hicks C, Cornish JL, Baracz SJ, Suraev A, McGregor IS. Adolescent pre-treatment with oxytocin protects against adult methamphetamine-seeking behavior in female rats. Addict Biol. 2016;21(2):304–315. doi:10.1111/adb.12197

571. Subiah CO, Mabandla MV, Phulukdaree A, Chuturgoon AA, Daniels WM. The effects of vasopressin and oxytocin on methamphetamine-induced place preference behaviour in rats. Metab Brain Dis. 2012;27(3):341–350. doi:10.1007/s11011-012-9297-7

572. Qi J, Han WY, Yang JY, et al. Oxytocin regulates changes of extracellular glutamate and GABA levels induced by methamphetamine in the mouse brain. Addict Biol. 2012;17(4):758–769. doi:10.1111/j.1369-1600.2012.00439.x

573. Cox BM, Young AB, See RE, Reichel CM. Sex differences in methamphetamine seeking in rats: impact of oxytocin. Psychoneuroendocrinology. 2013;38(10):2343–2353. doi:10.1016/j.psyneuen.2013.05.005

574. Everett NA, McGregor IS, Baracz SJ, Cornish JL. The role of the vasopressin V1A receptor in oxytocin modulation of methamphetamine primed reinstatement. Neuropharmacology. 2018;133:1–11. doi:10.1016/j.neuropharm.2017.12.036

575. Everett NA, Baracz SJ, Cornish JL. The effect of chronic oxytocin treatment during abstinence from methamphetamine self-administration on incubation of craving, reinstatement, and anxiety. Neuropsychopharmacology. 2020;45(4):597–605. doi:10.1038/s41386-019-0566-6

576. Ferland CL, Reichel CM, McGinty JF. Effects of oxytocin on methamphetamine-seeking exacerbated by predator odor pre-exposure in rats. Psychopharmacology. 2016;233(6):1015–1024. doi:10.1007/s00213-015-4184-7

577. Stauffer CS, Moschetto JM, McKernan S, et al. Oxytocin-enhanced group therapy for methamphetamine use disorder: randomized controlled trial. J Subst Abuse Treat. 2020;116:108059. doi:10.1016/j.jsat.2020.108059

578. Azadbakht A, Salehi M, Maracy MR, Banafshe HR. The effects of oxytocin on craving, mental health parameters, and stress hormones in methamphetamine-dependent patients undergoing matrix treatment model: a randomized, double-blind clinical trial. Eur Addict Res. 2022;28(5):340–349. doi:10.1159/000525443

579. Bondarev AD, Attwood MM, Jonsson J, et al. Recent developments of phosphodiesterase inhibitors: clinical trials, emerging indications and novel molecules. Front Pharmacol. 2022;13:1057083. doi:10.3389/fphar.2022.1057083

580. Self DW, Genova LM, Hope BT, Barnhart WJ, Spencer JJ, Nestler EJ. Involvement of cAMP-dependent protein kinase in the nucleus accumbens in cocaine self-administration and relapse of cocaine-seeking behavior. J Neurosci. 1998;18(5):1848–1859. doi:10.1523/JNEUROSCI.18-05-01848.1998

581. Enomoto T, Tatara A, Goda M, et al. A novel phosphodiesterase 1 inhibitor DSR-141562 exhibits efficacies in animal models for positive, negative, and cognitive symptoms associated with schizophrenia. J Pharmacol Exp Ther. 2019;371(3):692–702. doi:10.1124/jpet.119.260869

582. Harada A, Kaushal N, Suzuki K, et al. Balanced activation of striatal output pathways by faster off-rate PDE10A inhibitors elicits not only antipsychotic-like effects but also procognitive effects in rodents. Int J Neuropsychopharmacol. 2020;23(2):96–107. doi:10.1093/ijnp/pyz056

583. Iyo M, Maeda Y, Inada T, Kitao Y, Sasaki H, Fukui S. The effects of a selective cAMP phosphodiesterase inhibitor, rolipram, on methamphetamine-induced behavior. Neuropsychopharmacology. 1995;13(1):33–39. doi:10.1016/0893-133X(94)00133-K

584. Iyo M, Bi Y, Hashimoto K, Inada T, Fukui S. Prevention of methamphetamine-induced behavioral sensitization in rats by a cyclic AMP phosphodiesterase inhibitor, rolipram. Eur J Pharmacol. 1996;312(2):163–170. doi:10.1016/0014-2999(96)00479-7

585. Mori T, Baba J, Ichimaru Y, Suzuki T. Effects of rolipram, a selective inhibitor of phosphodiesterase 4, on hyperlocomotion induced by several abused drugs in mice. Jpn J Pharmacol. 2000;83(2):113–118. doi:10.1254/jjp.83.113

586. Snider SE, Vunck SA, van den Oord EJ, Adkins DE, McClay JL, Beardsley PM. The glial cell modulators, ibudilast and its amino analog, AV1013, attenuate methamphetamine locomotor activity and its sensitization in mice. Eur J Pharmacol. 2012;679(1–3):75–80. doi:10.1016/j.ejphar.2012.01.013

587. Suzuki K, Harada A, Suzuki H, Miyamoto M, Kimura H. TAK-063, a PDE10A Inhibitor with balanced activation of direct and indirect pathways, provides potent antipsychotic-like effects in multiple paradigms. Neuropsychopharmacology. 2016;41(9):2252–2262. doi:10.1038/npp.2016.20

588. Nakashima M, Imada H, Shiraishi E, et al. Phosphodiesterase 2A inhibitor TAK-915 ameliorates cognitive impairments and social withdrawal in N-methyl-d-aspartate receptor antagonist-induced rat models of schizophrenia. J Pharmacol Exp Ther. 2018;365(1):179–188. doi:10.1124/jpet.117.245506

589. Beardsley PM, Shelton KL, Hendrick E, Johnson KW. The glial cell modulator and phosphodiesterase inhibitor, AV411 (ibudilast), attenuates prime- and stress-induced methamphetamine relapse. Eur J Pharmacol. 2010;637(1–3):102–108. doi:10.1016/j.ejphar.2010.04.010

590. Snider SE, Hendrick ES, Beardsley PM. Glial cell modulators attenuate methamphetamine self-administration in the rat. Eur J Pharmacol. 2013;701(1–3):124–130. doi:10.1016/j.ejphar.2013.01.016

591. Baek JJ, Kline H, Deveau CM, Yamamoto BK. Roflumilast treatment during forced abstinence reduces relapse to methamphetamine seeking and taking. Addict Biol. 2022;27(1):e13082. doi:10.1111/adb.13082

592. Honeywell KM, Doren EV, Szumlinski KK. Selective inhibition of PDE4B reduces methamphetamine reinforcement in two C57BL/6 substrains. Int J Mol Sci. 2022;23(9):4872. doi:10.3390/ijms23094872

593. Heinzerling KG, Briones M, Thames AD, et al. Randomized, placebo-controlled trial of targeting neuroinflammation with ibudilast to treat methamphetamine use disorder. J Neuroimmune Pharmacol. 2020;15(2):238–248. doi:10.1007/s11481-019-09883-w

594. Li MJ, Briones MS, Heinzerling KG, Kalmin MM, Shoptaw SJ. Ibudilast attenuates peripheral inflammatory effects of methamphetamine in patients with methamphetamine use disorder. Drug Alcohol Depend. 2020;206:107776. doi:10.1016/j.drugalcdep.2019.107776

595. Powell GL, Bonadonna JP, Vannan A, et al. Dopamine D3 receptor partial agonist LS-3-134 attenuates cocaine-motivated behaviors. Pharmacol Biochem Behav. 2018;175:123–129. doi:10.1016/j.pbb.2018.10.002

596. Daiwile AP, Jayanthi S, Cadet JL. Sex differences in methamphetamine use disorder perused from pre-clinical and clinical studies: potential therapeutic impacts. Neurosci Biobehav Rev. 2022;137:104674. doi:10.1016/j.neubiorev.2022.104674

597. Harmony ZR, Alderson EM, Garcia-Carachure I, Bituin LD, Crawford CA. Effects of nicotine exposure on oral methamphetamine self-administration, extinction, and drug-primed reinstatement in adolescent male and female rats. Drug Alcohol Depend. 2020;209:107927. doi:10.1016/j.drugalcdep.2020.107927

598. Admon LK, Bart G, Kozhimannil KB, Richardson CR, Dalton VK, Winkelman TNA. Amphetamine- and opioid-affected births: incidence, outcomes, and costs, United States, 2004-2015. Am J Public Health. 2019;109(1):148–154. doi:10.2105/AJPH.2018.304771

599. Ruda-Kucerova J, Pistovcakova J, Amchova P, Sulcova A, Machalova A. Prenatal exposure to modafinil alters behavioural response to methamphetamine in adult male mice. Int J Dev Neurosci. 2018;67:37–45. doi:10.1016/j.ijdevneu.2018.03.005

600. Harrod SB, Lacy RT, Morgan AJ. Offspring of prenatal IV nicotine exposure exhibit increased sensitivity to the reinforcing effects of methamphetamine. Front Pharmacol. 2012;3:116. doi:10.3389/fphar.2012.00116

601. Lacy RT, Morgan AJ, Harrod SB. IV prenatal nicotine exposure increases the reinforcing efficacy of methamphetamine in adult rat offspring. Drug Alcohol Depend. 2014;141:92–98. doi:10.1016/j.drugalcdep.2014.05.010

602. Steimer W, Zöpf K, von Amelunxen S, et al. Amitriptyline or not, that is the question: pharmacogenetic testing of CYP2D6 and CYP2C19 identifies patients with low or high risk for side effects in amitriptyline therapy. Clin Chem. 2005;51(2):376–385. doi:10.1373/clinchem.2004.041327

603. Cherner M, Bousman C, Everall I, et al. Cytochrome P450-2D6 extensive metabolizers are more vulnerable to methamphetamine-associated neurocognitive impairment: preliminary findings. J Int Neuropsychol Soc. 2010;16(5):890–901. doi:10.1017/S1355617710000779

604. Otani K, Ujike H, Sakai A, et al. Reduced CYP2D6 activity is a negative risk factor for methamphetamine dependence. Neurosci Lett. 2008;434(1):88–92. doi:10.1016/j.neulet.2008.01.033

605. Sutter ME, Gaedigk A, Albertson TE, et al. Polymorphisms in CYP2D6 may predict methamphetamine related heart failure. Clin Toxicol. 2013;51(7):540–544. doi:10.3109/15563650.2013.818684

606. Blevins D, Seneviratne C, Wang XQ, Johnson BA, Ait-Daoud N. A randomized, double-blind, placebo-controlled trial of ondansetron for the treatment of cocaine use disorder with post hoc pharmacogenetic analysis. Drug Alcohol Depend. 2021;228:109074. doi:10.1016/j.drugalcdep.2021.109074

607. Kampangkaew JP, Spellicy CJ, Nielsen EM, et al. Pharmacogenetic role of dopamine transporter (SLC6A3) variation on response to disulfiram treatment for cocaine addiction. Am J Addict. 2019;28(4):311–317. doi:10.1111/ajad.12891

608. Liu S, Green CE, Lane SD, et al. The influence of dopamine β-hydroxylase gene polymorphism rs1611115 on levodopa/carbidopa treatment for cocaine dependence: a preliminary study. Pharmacogenet Genomics. 2014;24(7):370–373. doi:10.1097/FPC.0000000000000055

609. Pal R, Mendelson JE, Flower K, et al. Impact of prospectively determined A118G polymorphism on treatment response to injectable naltrexone among methamphetamine-dependent patients: an open-label, pilot study. J Addict Med. 2015;9(2):130–135. doi:10.1097/ADM.0000000000000107

610. Guerin AA, Nestler EJ, Berk M, Lawrence AJ, Rossell SL, Kim JH. Genetics of methamphetamine use disorder: a systematic review and meta-analyses of gene association studies. Neurosci Biobehav Rev. 2021;120:48–74. doi:10.1016/j.neubiorev.2020.11.001

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.