Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Aug 1.
Published in final edited form as: Annu Rev Pharmacol Toxicol. 2021 Jan 6;61:609–628. doi: 10.1146/annurev-pharmtox-030220-124205

New Drugs, Old Targets: Tweaking the Dopamine System to Treat Psychostimulant Use Disorders

Amy Hauck Newman 1, Therese Ku 1, Chloe Jordan 1, Alessandro Bonifazi 1, Zheng-Xiong Xi 1
PMCID: PMC9341034  NIHMSID: NIHMS1825750  PMID: 33411583

Abstract

The abuse of illicit psychostimulants, such as cocaine and methamphetamine, continues to pose significant health and societal challenges. Despite considerable efforts to develop medications to treat psychostimulant use disorders (PUD), none have proven effective, leaving an underserved patient population and unanswered questions as to what mechanism(s) of action should be targeted for developing pharmacotherapies. As both cocaine and methamphetamine rapidly increase dopamine (DA) levels in mesolimbic brain regions, leading to euphoria that in some can lead to addiction, targets in which this increased dopaminergic tone may be mitigated have been explored. Further, understanding and targeting mechanisms underlying relapse is fundamental to the success of discovering medications that either reduce the reinforcing effects of the drug of abuse and/or decrease the negative reinforcement or withdrawal/negative affect that occurs during abstinence. A focus on atypical inhibitors of the DA transporter (DAT) and partial agonists/antagonists at DA D3 receptors (D3R) are described as two promising targets for future drug development.

Keywords: cocaine, methamphetamine, dopamine transporter, dopamine D3 receptor, modafinil, VK4-40, JJC8-091

I. Introduction to psychostimulant use disorders: cocaine and methamphetamine

a. The problem

While the national focus has been centered on the opioid epidemic, deaths related to psychostimulants, such as cocaine and especially methamphetamine, have surged dramatically. According to the Center for Disease Control (CDC), the National Institute on Drug Abuse (NIDA) and various news sources, overdose fatalities caused by psychostimulants have more than doubled over the last five years (15). One of the likely culprits of this increase is co-administration of the opioid, fentanyl. The Journal of the American Medical Association reported a close to eight-fold increase of unprescribed fentanyl detected in urine samples positive for methamphetamine and 18-fold increase for those tested positive for cocaine in 2019 (4, 6). The contamination of deadly fentanyl in non-regulated psychostimulants has thus contributed to the increase in numbers of overdose related deaths of these drugs (79). Another cause of this health and societal tragedy is, ironically, the increased public awareness of the dangers of using and abusing opioids, which has led chronic opioid users to shift to psychostimulants that are viewed as less harmful (10, 11). Alarmingly, the grim picture painted by the official statistics does not account for the total number of drug-related deaths. While the numbers of deaths due to overdose does serve as a good metric for the lethality of these addictive substances, there are many other means by which they can inflict long-term health damage that can shorten the users’ lifespan (12). For example, cocaine causes severe cardio- and lung-toxicity over time, and chronic users of methamphetamine not only show dramatic change in mood and behavior, but also exhibit psychotic symptoms (1216). Taking into consideration all the factors such as these, the numbers of deaths related to drug use could easily be double of what has been reported (12). These alarming trends resulting from increased illicit psychostimulant abuse and subsequent mortality emphasizes the need for effective treatment strategies, including FDA approved pharmacotherapies, of which none currently exists.

b. Potential pharmacotherapies under development

Clinically, a combination of an FDA approved medication with psychosocial treatment, termed medication-assisted treatment, has been shown to be more effective in treating substance use disorders than employing monotherapies exclusively (1720). However, unlike opioid use disorders for which options such as buprenorphine, methadone and naloxone exists, Psychostimulant Use Disorders (PUDs) lack FDA approved pharmacotherapies (20, 21). Efforts in overcoming this setback have yielded a variety of potential options. For example, an anti-cocaine vaccine is currently being evaluated in a Phase I clinical trial. The dAd5GNE vaccine is composed of a disrupted serotype adenovirus with a GNE hapten, a cocaine analogue, coupled to its capsid protein (22). The administered vaccine induces the immune system to generate anti-cocaine antibodies that have been shown to sequester cocaine molecules in the blood and subsequently prevent cocaine from reaching the brain as the antibodies cannot cross the blood brain barrier (BBB), thereby eliminating cocaine’s stimulant effects (22). Interestingly, the cocaine-antibody complex also protects other cocaine sensitive organs from damage, hence lowering the toxic effects of cocaine in the periphery as well (23). Through this strategy, cocaine self-administration behavior and cocaine-induced hyperactivity in mouse, rat and nonhuman primate pre-clinical models were effectively blocked (2224). While this vaccine appears to provide an effective solution to cocaine use disorder, a drawback is the necessity of a monthly vaccine regimen, with no endpoint provided, thus, a potentially lifelong treatment (25).

In terms of pharmacotherapeutics, disulfiram is a medication that shows promise in treating cocaine use disorder, although it is FDA approved only to treat alcoholism. For its label purpose, disulfiram inhibits aldehyde dehydrogenase, which induces an aversive reaction to alcohol and deters further intake (26). Clinically, disulfiram has been observed to be effective in decreasing cocaine consumption for dependent individuals, likely through inhibition of dopamine (DA) β-hydroxylase, when combined with psychotherapy, although outcomes are superior in males (2629). A recent report indicated that patients with genetically high dopamine transporter (DAT) expression levels due to the presence of two rs28363170 10-repeat allele polymorphisms in SLC6A3 (DAT1) may benefit even more from disulfiram treatment (30).

Topiramate, a glutamate antagonist and GABA agonist was able to prevent cocaine self-administration and reinstatement in rodents, however, clinical trials showed insignificant improvement in patients who were administered either topiramate or placebo (31). Bupropion, a DAT inhibitor, was not significantly efficacious in treating either cocaine or methamphetamine use disorder (32, 33). Mirtazapine, which affects monoamine transporters, was effective in decreasing methamphetamine use (34, 35). More recently, cholinergic medications such as galantamine, an acetylcholinesterase antagonist, have shown success in treating cocaine use disorder in a small and a larger (120 patient) clinical trial; further investigation is needed (31).

Perhaps more controversially, the use of DA agonists, or substitution therapy, has emerged as a highly promising pharmacological strategy for cocaine use disorder treatment (31, 36, 37). Opioid agonists such as methadone and buprenorphine have shown success in treating opioid use disorders, however, using the same strategy for PUDs has thus far been rejected. Long-acting amphetamines have been efficacious in treating cocaine use disorder in clinical trials, however, due to stringent criteria, the retention rate of participants was poor, thus studies with a larger population are needed (31, 37). Atypical DAT inhibitors and DA D3 receptor antagonist/partial agonists have also emerged as promising solutions to PUDs and will be discussed in detail in this review.

II. Emergence of atypical DAT inhibitors as pharmacotherapies for PUD

Cocaine and methamphetamine occupy a similar binding pocket in the DAT protein, which corresponds to the substrate (DA) binding site. Cocaine and its analogues are DAT inhibitors that typically stabilize an open outward-facing conformation of the DAT and prevent DA from being recycled (3841). Hence, the rewarding effects imparted by cocaine stem primarily from the elevated levels of DA resulting from DAT blockade. In contrast, although methamphetamine also serves as a DAT inhibitor, unlike cocaine, methamphetamine is also a DAT substrate. As such, it is taken up into the cell where it blocks the Vesicular Monoamine Transporter (VMAT) releasing DA and reversing DAT from inside the DA cell body, resulting in increased levels of mesolimbic DA that produce stimulation and euphoria that can lead to abuse (4248).

In the mid 1990’s, the Newman lab reported a series of benztropine analogues that like the parent drug had binding affinities at the DAT that were comparable or higher than cocaine, but these compounds did not have a cocaine-like behavioral profile, in rodents (49, 50). The term “atypical DAT inhibitor” was defined as compounds that did not display a cocaine-like behavioral profile, despite sharing the common mechanism of action as a DAT inhibitor. Benztropine was chosen as a lead molecule due to structural features that were shared with cocaine (tropane ring) and another DAT inhibitor, GBR12909 (diphenyl ether; Fig. 1). Benztropine was also a clinically used drug for the treatment of symptoms associated with Parkinson’s disease, and there were no reports of its abuse, suggesting it may be atypical.

Fig. 1.

Fig. 1.

Chemical structures of cocaine, benztropine, JHW007, rimcazole, GBR12909, modafinil, JJC8-016, 8-088, 8-089, 8-091, RDS3-094 with binding data from the Newman lab; adata from reference (78).

a. Benztropine and rimcazole analogues

Modifications of the benztropine structure at the tropane N-, 2-, 3-, and 6,7-bridgehead were made over the following decade to advance structure activity relationships (SAR) across the monoamine transporters, as well as sigma1 receptors (5162). In addition, decreasing binding affinities at off target sites such as muscarinic M1 and histamine H1 receptors was an important and achieved objective to be sure that these actions were not playing a role or masking the psychostimulant behavioral effects of these novel analogues. Of note, benztropine has ~100-fold higher affinity for muscarinic M1 receptors than DAT, which may be related to its lack of abuse potential (63). Importantly, several interesting lead molecules were discovered, with the most studied, JHW007 (Fig. 1), showing no detectable psychostimulant-like behaviors and demonstrating antagonism of behaviors produced by cocaine or methamphetamine across numerous animal models and species (6469).

At the same time, a series of rimcazole analogues (63, 7073) was discovered that in addition to binding with moderately high affinity to DAT also interacted with sigma1 receptors. These compounds also exhibited an atypical DAT inhibitor behavioral profile (7476), suggesting that a dual DAT/sigma1 binding profile might be beneficial for a pharmacotherapeutic to treat PUD. This concept was further expanded with selected benztropine analogues (77). Of note, GBR12909 was reported to bind to sigma1 receptors (78) and thus it was posited that this dual mechanism may be shared by other compounds that had been or might be developed to treat PUDs.

In addition to the benztropine and rimcazole analogues, other tropane based atypical DAT inhibitors emerged during this time (79, 80) as well as numerous series of GBR12909 analogues, although extensive behavioral evaluation of these latter compounds was not reported. Moreover, both benztropine and GBR12909 were evaluated clinically as potential treatments for cocaine abuse. Benztropine failed to significantly affect responses to acute cocaine administration (81) and further evaluation was not reported. GBR12909 (vanoxerine) was advanced into Phase I clinical trials before failing, due to rate-dependent QTc elongation in healthy subjects (8286). However, a slow release formulation has recently been reported to have beneficial effects in a cocaine abusing patient population (87).

From the outset, we noted significantly different SAR at DAT with the benztropine analogues compared to cocaine and the many analogues that were synthesized by other labs during that time period (63, 88). We hypothesized that although these two classes of DAT inhibitors share the tropane ring structure, they were likely binding to the DAT differently. Furthermore, that difference in binding might, at least in part, result in a behavioral profile that was not cocaine-like but indeed, could block cocaine from the DAT and hence mitigate cocaine’s psychostimulant actions. Through extensive molecular pharmacology with DAT mutants that either had no effect on cocaine’s binding affinity (e.g., Y156F) or preferred an inward facing conformation (e.g., Y335A) that significantly reduced cocaine’s binding affinity, we discovered that in fact, the benztropines appeared to prefer a more occluded, inward facing conformation of the DAT, compared to cocaine and these data were supported with computational studies (89). Moreover, a recent crystal structure confirmed that cocaine binds to an outward facing conformation of Drosophila melanogaster DAT (dDAT) (39), as predicted empirically and through computational models. No crystal structure of DAT with a benztropine analogue has been reported.

In addition, through ex vivo autoradiography studies, it was shown that although these benztropine analogues penetrate the BBB, they are slow to occupy DAT (e.g., JHW 007 requires >4 h to fully occupy DAT), as compared to cocaine, which fully occupies DAT in <30 min (64, 90). These binding kinetic differences could also play a critical role in their behavioral profiles as it has been shown through extensive PET imaging that cocaine’s rapid BBB penetration and occupancy of DAT is related to feeling of “high” in human subjects (91). Unfortunately, none of the benztropine analogues were advanced to clinical trials.

More recently, inspired by its pharmacological profile and early promise in clinical studies of patients with cocaine or methamphetamine use disorders (9295), the Newman lab has focused on (±)-modafinil, its (R)-enantiomer (96) and analogues thereof. We originally noted that although not entirely atypical, as we have defined it (i.e. DAT inhibitor without a cocaine-like behavioral profile), R-modafinil had some striking pharmacological features that made it an interesting template to undertake SAR studies. Importantly, although it is a DA uptake inhibitor, it does not appear to have significant addictive liability in humans. Nevertheless, some people use R-modafinil for cognitive enhancement, instead of its FDA approved use for narcolepsy and other sleep disorders (97, 98). It is a mild stimulant in both rodents and humans, but is not self-administered in rodents and can block some cocaine-induced behaviors (97, 98).

b. Modafinil analogues

Armed with 1) SAR in the benztropine class of molecules and 2) a hypothesis that DA uptake inhibitors that prefer a more occluded inward facing conformation may have an atypical behavioral profile, we began a synthetic campaign to identify modafinil analogues that had higher DAT affinities than the parent molecule, with improved solubility and lower abuse potential. Our goal was to discover an atypical DAT inhibitor that had the appropriate drug-like properties as well as a behavioral profile that might be developed as a therapeutic for treatment of PUD.

In our first series of modafinil analogues, we discovered that reducing the terminal amide to an amine improved DAT binding affinity (99, 100) and the salt form of these analogues would impart improved solubility as compared to the parent drug. The first lead in this series, JJC8-016 (Fig. 1) demonstrated a promising behavioral profile in numerous rodent models of cocaine abuse. For example, pretreatment with JJC8-016 dose-dependently inhibited cocaine-enhanced locomotion, cocaine self-administration, and cocaine-induced reinstatement of drug-seeking behavior (101). However, subsequent analysis of JJC8-016 suggested that it may be bind to the human ether-à-go-go-related potassium channel (hERG; (102)), which might predict cardiotoxicity, as had already been reported for its analogue, GBR12909. hERG is involved in the repolarization of the cardiac action potential (83, 86, 103106). Inhibition of this channel with small molecule drugs can lead to delays in repolarization, which can lead to QT prolongation and the lethal cardiac arrhythmia torsade de pointes. The hERG channel is highly promiscuous and has high affinity for molecules with aromatic (e.g., phenyl rings) and positively charged groups (e.g., protonatable amines). Many FDA-approved drugs have been withdrawn from the market due to cardiotoxicity related to hERG inhibition and thus high affinity for hERG is a red flag for medication development (107). This also poses a significant challenge for designing drugs that are BBB penetrant and that also bind with high affinity to G-protein coupled receptors or monoamine transporters. Hence additional chemical modification of this template was undertaken to further improve the drug-like properties of this molecule, resulting in several new leads (108).

In the next series of modafinil analogues, a piperazine linker was incorporated and several interesting analogues emerged, including JJC8-088, JJC8-089 and JJC8-091 (Fig. 1). Although initial behavioral testing in a murine locomotor assay suggested that the highest DAT affinity analogue, JJC8-088, might be a lead candidate for development, subsequent evaluation showed that it was not as effective in rats exposed to long access (6h) methamphetamine as JJC8-091 (102). Further exploration revealed that JJC8-088 was more cocaine-like than originally predicted and appeared to prefer a more open conformation of the DAT than JJC8-091. Subsequently, JJC8-091 became the new lead molecule in this series (21) and continues to undergo development (http://encepheal.com/). In rodent studies, JJC8-091 reduced cocaine self-administration and cocaine-primed reinstatement of cocaine seeking (Fig. 2A, C; (21)).

Fig. 2.

Fig. 2.

Effects of the novel atypical DAT inhibitor JJC8-091 and the novel D3R partial agonist (±)-VK4-40 on cocaine self-administration (0.5 mg/kg, FR2) (A, B) and cocaine-primed reinstatement of drug-seeking behavior (C, D) in rats extinguished from previous cocaine self-administration. *p<0.05 compared to the vehicle control group.

More recently, in order to further explore SAR and improve pharmacokinetics and metabolic stability, a new series of 2,6-dimethylpiperazine analogues was reported (109). By introducing the 2,6-dimethyl substitution on the piperazine ring, some improvement in drug-like properties was realized. Nevertheless, the piperazine ring remains a metabolically labile functional group in this series of molecules and hence a new series of analogues in which the piperazine ring has been replaced with a piperidine-amine or amino-piperidine function (Giancola et al, unpublished data) are currently being evaluated in rodent models of PUD as well as for pharmacokinetics, metabolic stability and off target actions. In addition, Lubec and colleagues have recently reported novel heterocyclic-based modafinil analogues that may also be promising new leads for PUD therapeutics (110, 111).

III. Introduction to DA D3R partial agonists/antagonists as potential medications to treat PUD

a. A brief history of D3R partial agonists/antagonists

By blocking DA reuptake at DAT, psychostimulants such as cocaine and methamphetamine effectively prolong dopaminergic activity in the synapse at both pre- and post-synaptic DA receptors. There are five major DA receptor subtypes, classified as excitatory “D1-like” receptors (including D1R and D2R) or inhibitory “D2-like” receptors (D2R, D3R, and D4R). Relative to other DA receptor subtypes, the D3R is of particular interest as a medicinal target due to its restricted distribution in the mesolimbic reward system, including the nucleus accumbens (e.g., ventral striatum), which play an integral role in psychostimulant reward and abuse (112115). Shortly after cloning of the D3R in 1990 (115), preclinical studies revealed that D3R-preferring ligands, such as 7-hydroxy-N,N-di-n-propyl-2-aminotetralin (7-OH-DPAT), reduce cocaine intravenous self-administration in rats (116). The DA D3R received increasing interest as a medicinal target for PUDs following reports of upregulated D3R levels in cocaine overdose patients in key reward-related regions of the brain such as the nucleus accumbens, caudate/putamen, and the basolateral amygdala (117119). More recent PET studies using a D3R-preferring radioligand, [11C](+)PHNO, confirmed that cocaine users exhibit upregulated D3R binding in the substantia nigra, hypothalamus and amygdala (120, 121).

Early studies on putative D3R medications for PUDs were limited by poor selectivity and specificity for the D3R over the D2R. Recent advances in medicinal chemistry have led to significant improvements in selectivity and specificity for the D3R (Fig. 3). For example, the D3R partial agonist, BP897, is ~70 fold more selective for the D3R > D2R. BP897 reduced conditioned locomotor activity to both amphetamine and cocaine and modestly decreased lever responding to cocaine-paired cues in rats (122125), but was ineffective when co-administered with amphetamine or when cocaine was available for self-administration (122, 126). Similarly, SB277,011A, a D3R antagonist with approximately 100-fold selectivity for the D3R over the D2R (127), dose-dependently reduced cocaine-enhanced brain stimulation reward, suppressed cocaine conditioned place preference, and attenuated cocaine-primed reinstatement behaviors in rats (128130). However, like BP897, SB277,011A could not reduce cocaine intake under an “easy” fixed ratio 1 (FR1) reinforcement schedule (130).

Fig. 3.

Fig. 3.

Chemical structures of D3R antagonists and partial agonists studied as potential medications for PUD, including highly selective bitopic D3R antagonists and partial agonists (CJB090, PG01037, VK4-116 and VK4-40) developed in the Newman lab (138140). Binding data from Newman lab; adata from reference (141).

In addition to limited efficacy, SB277,011A had poor translational utility due to low bioavailability (<2%) and a short half-life (<20 min) in non-human primates (131, 132). Subsequently, PG01037 and its parent molecule, NGB2904 (Fig. 3) were studied as potentially more viable D3R antagonists with >100-fold selectivity for D3R over D2R. In rats, NGB 2904 reduced progressive ratio breakpoints for cocaine, inhibited both cocaine- and cue-primed reinstatement of cocaine seeking, and reduced cocaine-enhanced brain stimulation reward (133). However, the reward-attenuating effects of NGB 2904 could be overcome by higher doses of cocaine, and NGB 2904 was found to enhance amphetamine-induced locomotor activity in wild type (but not D3R knockout) mice, suggested possible abuse liability (133, 134). Similar to SB277,011A, PG01037 reduced progressive ratio breakpoints for methamphetamine, suppressed cue-primed reinstatement of methamphetamine seeking, and attenuated methamphetamine-enhanced brain stimulation reward in rats, but failed to alter methamphetamine self-administration under an FR2 reinforcement schedule in rats (135). In non-human primates, PG01037 reduced cocaine choice over food, but tolerance to these effects developed after 5 days of administration (136). Moreover, PG01037 was not effective in reducing methamphetamine choice or intake in the same self-administering rhesus monkeys (136, 137).

b. Cardiovascular implications: Not a D3R class effect

Despite limited efficacy in reducing psychostimulant intake when drugs are available under low-cost (FR1, FR2) reinforcement conditions, the D3R antagonists described above significantly attenuated psychostimulant seeking under high motivation (progressive ratio) or reinstatement conditions, suggesting that D3R antagonists have value in attenuating relapse behaviors. However, translational efforts towards moving D3R antagonists to the clinic were essentially halted following reports of cardiovascular side effects by GSK598,809 (142), a D3R antagonist previously evaluated in clinical trials for smoking cessation (ClinicalTrials.gov Identifier: NCT01188967). Briefly, GSK598,809 was found to increase blood pressure in dogs fitted with telemetry transmitters, particularly when combined with cocaine. Because the D3R is expressed in the kidney, GSK598,809’s blood pressor effects were predicted to apply to all D3R ligands.

Subsequent investigations following the GSK598,809 telemetry study confirmed that SB277,011A similarly increased blood pressure and heart rate in rats (143). However, two structurally unique D3R antagonists, R-VK4-116 and R-VK4-40, which exhibit higher D3R affinity and selectivity than previous D3R ligands (1735-fold and 305-fold more selective for D3R over D2R, respectively), did not potentiate the adverse cardiovascular effects caused by oxycodone or cocaine (140, 143). Instead, R-VK4-116 reduced cocaine-enhanced heart rate and blood pressure. R-VK4-40 similarly attenuated cocaine-induced increases in heart rate, and further reduced blood pressure and heart rate when administered alone. The reasons for which R-VK4-116 and R-VK4-40 do not exert the same adverse cardiovascular effects as previous generations of D3R antagonists are unclear. Differences in ligand affinity, selectivity, unknown off-target effects, or biased activation of different intracellular D3R signaling pathways in the kidney may all play a role (143). Regardless of the reasons why, these R-VK4-116 and R-VK4-40 results renewed interest in the D3R as a medication target for PUD, and provided fresh impetus for the continued development of D3R ligands in the treatment of psychostimulant and other drug use disorders, including opioid use disorder (139, 140, 144).

c. Emerging evidence supporting D3R partial agonist/antagonist treatments for PUD

Although D3R antagonists effectively attenuate the motivation to earn psychostimulants and relapse-related behaviors in rodent models, these compounds have been relatively ineffective in reducing cocaine intake when response requirements are low and/or the reinforcing value of cocaine is high, such as under low FR schedules. As a result, additional efforts have focused instead on partial D3R agonists as treatments for PUDs. Partial agonists can functionally block the effects of drugs of abuse (due to occupation of their receptor target) but elicit partial activation of their receptor targets under abstinence conditions, thereby potentially mitigating withdrawal effects. As such, partial agonists have historically been more effective at maintaining abstinence and reducing relapse rates than antagonist therapies, particularly for opioid and nicotine use disorders (145, 146).

Prior generations of partial D3R agonists have shown promise in attenuating psychostimulant seeking in rodent models. The D3R preferring partial agonist, RGH-188 (Cariprazine, Vraylar®), reduced cocaine intake under an FR1 schedule and suppressed cue-induced reinstatement of cocaine seeking in rats (147). However, while the D3R partial agonists BP897 and RGH-237 also suppress cue-driven cocaine seeking, these compounds were not effective when cocaine or methamphetamine were available for self-administration under FR schedules (122, 126). CJB090, another partial D3R agonist from the Newman lab, decreased methamphetamine intake under both FR and progressive ratio schedules and was more effective at attenuating psychostimulant reward than the D3R antagonist PG01037 in rats (148) but showed mixed efficacy in non-human primates (149).

As with D3R antagonists, prior generations of partial D3R agonists have suffered from poor selectivity (BP897 and CJB090 are ~60-70 fold more selective for D3R > D2R (150152)) or poor pharmacokinetic profiles (RGH-237 does not readily penetrate the BBB and is eliminated within ~5 hours of administration (141)). However, a recently developed series of new D3R ligands may circumvent these limitations (139, 140). As described above, R-VK4-40, which was found to attenuate blood pressor effects of cocaine as described above (143), functions as a D3R antagonist. Interestingly, the racemic (±)-VK4-40 compound, and its S-VK4-40 enantiomer, both function as D3R partial agonists. (±)-VK4-40 is ~300 fold selective for the D3R>D2R and remains at significant levels in the brain up to 8 hours following oral administration in the rat (Jordan et al., under review). Preliminary results indicate that (±)-VK4-40 not only attenuates cocaine self-administration and cocaine-primed reinstatement (Fig. 2B, D). In optical brain-stimulation reward model, in which adenosine-associated virus-mediated channelrhodopsin 2 (AAV-ChR2-eYFP) expression is driven by the DAT gene promotor selectively in VTA DA neurons of the midbrain (Fig. 4A, B), (±)-VK4-40, R-VK4-40, or S-VK4-40 (alone significantly inhibits optical brain-stimulation reward maintained by selective stimulation of VTA DA neurons in transgenic DAT-cre mice (Fig. 4C, D, E), and pretreatment with either of them significantly blocks cocaine-induced increase in brain-stimulation reward (Fig. 4F, G, H). These early findings support the efficacy of partial D3R agonists or antagonists as putative treatments for PUDs, and critically extend the translational utility of this medication class. As such, the VK compounds are currently undergoing further development as new investigational drugs for treatment of substance use disorders.

Fig. 4.

Fig. 4.

Effects of the racemic (±)-VK4-40 (a D3R partial agonist) and its enantiomers R-VK4-40 (a D3R antagonist) and S-VK4-40 (a D3R partial agonist) on lever responding for brain-stimulation reward maintained by optogenetic stimulation of VTA DA neurons in DAT-Cre mice. A: Schematic of the experimental model, illustrating AAV-ChR2-EGFP was microinjected into the VTA and an optical fiber was implanted into the VTA to stimulate DA neurons in DAT-Cre mice; B: Representative images, showing that AAV-ChR2-EGFP is selectively expressed in VTA tyrosine hydroxylase (TH)-positive DA neurons. C, D, E: Systemic administration of (±)-VK4-40 (C), R-VK4-40 (D), or S-VK4-40 (E) alone inhibits optical brain-stimulation reward and dose-dependently shifts the stimulation frequency-active lever responding curve downward. F, G, H: Pretreatment with (±)-VK4-40 (F), R-VK4-40 (G), or S-VK4-40 (H) dose-dependently attenuated cocaine-enhanced brain-stimulation reward as assessed by the upward or leftward shift of the stimulation-response curve after cocaine administration. * p < 0.05, ** p < 0.01, *** p < 0.001, compared to vehicle control group.

IV. Summary

There has never been a time when the development of novel strategies and pharmacotherapeutics to treat substance use disorders was more urgent. The loss of lives and livelihood due to health challenges associated with addiction, incarceration and stigma is unprecedented and the resurgence of cocaine and methamphetamine use and abuse is staggering. There is no doubt that targeting multiple mechanisms for medication development is required (153155) and that a single medication will never be a “cure all” for everyone who suffers from PUD. Targeted medications, focused clinical trials, recognition of other mental health co-morbidities and a plethora of other challenges have made it extremely difficult to identify and ultimately provide medications to be a part of a successful treatment strategy for those who suffer from PUD. Identifying subpopulations that are positively affected by experimental treatments is critical, instead of only determining that a large clinical trial failed. Undoubtedly there are at least subpopulations of people who would benefit from the medications that have failed to receive FDA approval for PUD (e.g., modafinil), but we can’t stop there.

Accelerating research and engaging pharmaceutical industry in this challenging area is critical to our success. Academic labs can do formidable basic and preclinical research, identifying new leads and moving them forward through animal models of PUD. But, that is not enough. There is an enormous chasm between preclinical research and actually getting a drug into Phase 1 clinical trials, much less beyond, and this is going to require more than NIH funded research. Partnerships and ongoing communication that build bridges between academia, government, and private industry are required. We have made slow but steady strides toward identifying the two sets of lead molecules described in this review: atypical DAT inhibitors and dopamine D3R partial agonists/antagonists. But frankly, reducing rats’ psychostimulant seeking is not the point. Until we can get our lead molecules into the clinic and until we can determine which of our animal models is translational to humans, we are spinning our wheels. Our data support moving forward with either or both of these mechanistic targets and doing further medicinal chemistry to find drug candidates that not only have the desired pharmacological profile, but also appropriate pharmacokinetics, metabolic stability and lack of off target actions that may preclude further development due to toxicity (e.g., hERG activity). These are challenging tasks, but not impossible. Indeed, the NIH Helping to End Addiction Long-term (HEAL) initiative has been developed and funded to address the Opioid Crisis and this is enormously important to ultimately curtail the unprecedented morbidity related to prescription opioid abuse that has led to increased heroin and fentanyl addiction. Lessons learned and critical research in this area will undoubtedly feed into other substance use disorders. And, although one medication or a single targeted mechanism of action is not going to mitigate all substance use disorders, there will be overlap. Indeed, the D3R partial agonists/antagonists are an excellent example and are currently being developed toward the prevention and treatment of opioid use disorder. However, as we have discussed herein, preclinical data support their efficacy for treatment of PUD. More work and resources are required. Certainly, people who suffer from PUD and indeed all substance use disorders are depending on us.

Acknowledgement.

The authors would like to thank the many collaborators, post-doctoral and post-baccalaureate fellows, and graduate students that have worked tirelessly on these targets, over the years. And a special thanks to J. Cao for >20 years of technical support to the Medicinal Chemistry Section. This work was funded by Z1A DA000389 and Z1A DA000424. The authors declare no conflicts of interest.

References

  • 1.NIDA. 2019. Overdose Death Rates. https://www.drugabuse.gov/related-topics/trends-statistics/overdose-death-rates
  • 2.DEA. 2017. 2017 National Drug Threat Assessment. https://www.dea.gov/documents/2017/10/01/2017-national-drug-threat-assessment
  • 3.News KH. 2019. Meth Vs. Opioids: America Has Two Drug Epidemics, But Focuses On One. [Google Scholar]
  • 4.Twillman RK, Dawson E, LaRue L, Guevara MG, Whitley P, Huskey A. 2020. Evaluation of Trends of Near-Real-Time Urine Drug Test Results for Methamphetamine, Cocaine, Heroin, and Fentanyl. JAMA Netw Open 3: e1918514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.CDC. 2020. Provisional Drug Overdose Death Counts. [Google Scholar]
  • 6.LaRue L, Twillman RK, Dawson E, Whitley P, Frasco MA, et al. 2019. Rate of Fentanyl Positivity Among Urine Drug Test Results Positive for Cocaine or Methamphetamine. JAMA Netw Open 2: e192851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.DEA. 2018. Cocaine laced with fentanyl leads to multiple deaths, overdoses.
  • 8.Nolan ML, Shamasunder S, Colon-Berezin C, Kunins HV, Paone D. 2019. Increased Presence of Fentanyl in Cocaine-Involved Fatal Overdoses: Implications for Prevention. J Urban Health 96: 49–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.McCall Jones C, Baldwin GT, Compton WM. 2017. Recent Increases in Cocaine-Related Overdose Deaths and the Role of Opioids. Am J Public Health 107: 430–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lopez G 2020. The rise in meth and cocaine overdoses, explained: America’s drug overdose crisis could be reaching a new phase. Vox.com: Vox media [Google Scholar]
  • 11.Ellis MS, Kasper ZA, Cicero TJ. 2018. Twin epidemics: The surging rise of methamphetamine use in chronic opioid users. Drug Alcohol Depend 193: 14–20 [DOI] [PubMed] [Google Scholar]
  • 12.Glei DA, Preston SH. 2020. Estimating the impact of drug use on US mortality, 1999-2016. PLOS ONE 15: e0226732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.AHA. 2018. Illegal Drugs and Heart Disease. American Heart Association [Google Scholar]
  • 14.NIH-NIDA. 2017. Health Consequences of Drug Misuse. [Google Scholar]
  • 15.Rusyniak DE. 2013. Neurologic manifestations of chronic methamphetamine abuse. Psychiatr Clin North Am 36: 261–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Akindipe T, Wilson D, Stein DJ. 2014. Psychiatric disorders in individuals with methamphetamine dependence: prevalence and risk factors. Metab Brain Dis 29: 351–7 [DOI] [PubMed] [Google Scholar]
  • 17.Mattick RP, Breen C, Kimber J, Davoli M. 2009. Methadone maintenance therapy versus no opioid replacement therapy for opioid dependence. Cochrane Db. Syst. Rev [Google Scholar]
  • 18.Mattick RP, Breen C, Kimber J, Davoli M. 2014. Buprenorphine maintenance versus placebo or methadone maintenance for opioid dependence. Cochrane Db. Syst. Rev [Google Scholar]
  • 19.Schwartz RP, Gryczynski J, O’Grady KE, Sharfstein JM, Warren G, et al. 2013. Opioid agonist treatments and heroin overdose deaths in Baltimore, Maryland, 1995-2009. Am. J. Public Health 103: 917–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jordan CJ, Cao J, Newman AH, Xi ZX. 2019. Progress in agonist therapy for substance use disorders: Lessons learned from methadone and buprenorphine. Neuropharmacology [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Newman AH, Cao J, Keighron JD, Jordan CJ, Bi GH, et al. 2019. Translating the atypical dopamine uptake inhibitor hypothesis toward therapeutics for treatment of psychostimulant use disorders. Neuropsychopharmacology 44: 1435–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Koob G, Hicks MJ, Wee S, Rosenberg JB, De BP, et al. 2011. Anti-cocaine vaccine based on coupling a cocaine analog to a disrupted adenovirus. CNS Neurol Disord Drug Targets 10: 899–904 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hicks MJ, Kaminsky SM, De BP, Rosenberg JB, Evans SM, et al. 2014. Fate of systemically administered cocaine in nonhuman primates treated with the dAd5GNE anticocaine vaccine. Hum Gene Ther Clin Dev 25: 40–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Evans SM, Foltin RW, Hicks MJ, Rosenberg JB, De BP, et al. 2016. Efficacy of an adenovirus-based anti-cocaine vaccine to reduce cocaine self-administration and reacqusition using a choice procedure in rhesus macaques. Pharmacol Biochem Behav 150-151: 76–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Havlicek DF, De B, Rosenberg J, Pagovich O, Sondhi D, et al. 2016. Translation of an Adenovirus-Based Cocaine Vaccine dAd5GNE to a Clinical Trial. Molecular Therapy 24: S16–S16 [Google Scholar]
  • 26.Gaval-Cruz M, Weinshenker D. 2009. mechanisms of disulfiram-induced cocaine abstinence: antabuse and cocaine relapse. Mol Interv 9: 175–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Carroll KM, Fenton LR, Ball SA, Nich C, Frankforter TL, et al. 2004. Efficacy of disulfiram and cognitive behavior therapy in cocaine-dependent outpatients: a randomized placebo-controlled trial. Arch Gen Psychiatry 61: 264–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.DeVito EE, Babuscio TA, Nich C, Ball SA, Carroll KM. 2014. Gender differences in clinical outcomes for cocaine dependence: randomized clinical trials of behavioral therapy and disulfiram. Drug Alcohol Depend 145: 156–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Carroll KM, Nich C, Ball SA, McCance E, Rounsavile BJ. 1998. Treatment of cocaine and alcohol dependence with psychotherapy and disulfiram. Addiction 93: 713–27 [DOI] [PubMed] [Google Scholar]
  • 30.Kampangkaew JP, Spellicy CJ, Nielsen EM, Harding MJ, Ye A, et al. 2019. Pharmacogenetic role of dopamine transporter (SLC6A3) variation on response to disulfiram treatment for cocaine addiction. Am J Addict 28: 311–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kampman KM. 2019. The treatment of cocaine use disorder. Science Advances 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Margolin A, Kosten TR, Avants SK, Wilkins J, Ling W, et al. 1995. A multicenter trial of bupropion for cocaine dependence in methadone-maintained patients. Drug Alcohol Depend 40: 125–31 [DOI] [PubMed] [Google Scholar]
  • 33.Elkashef AM, Rawson RA, Anderson AL, Li SH, Holmes T, et al. 2008. Bupropion for the treatment of methamphetamine dependence. Neuropsychopharmacology 33: 1162–70 [DOI] [PubMed] [Google Scholar]
  • 34.Colfax GN, Santos GM, Das M, Santos DM, Matheson T, et al. 2011. Mirtazapine to reduce methamphetamine use: a randomized controlled trial. Arch Gen Psychiatry 68: 1168–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Coffin PO, Santos GM, Hern J, Vittinghoff E, Walker JE, et al. 2019. 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mariani JJ, Levin FR. 2012. Psychostimulant treatment of cocaine dependence. Psychiatr Clin North Am 35: 425–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Negus SS, Henningfield J. 2015. Agonist Medications for the Treatment of Cocaine Use Disorder. Neuropsychopharmacology 40: 1815–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Beuming T, Kniazeff J, Bergmann ML, Shi L, Gracia L, et al. 2008. The binding sites for cocaine and dopamine in the dopamine transporter overlap. Nat. Neurosci 11: 780–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wang KH, Penmatsa A, Gouaux E. 2015. Neurotransmitter and psychostimulant recognition by the dopamine transporter. Nature 521: 322–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Razavi AM, Khelashvili G, Weinstein H. 2017. A Markov state-based quantitative kinetic model of sodium release from the dopamine transporter. Sci. Rep 7: 40076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cheng MH, Kaya C, Bahar I. 2018. Quantitative assessment of the energetics of dopamine translocation by human dopamine transporter. J. Phys. Chem. B 122: 5336–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Balster RL, Kuhar MJ, Schuster CR. 1996. Pharmacological Aspects of Drug Dependence : Toward an Integrated Neurobehavioral Approach. Berlin ; New York: Springer. xxv, 658 p. pp. [Google Scholar]
  • 43.Schmitt KC, Zhen J, Kharkar P, Mishra M, Chen N, et al. 2010. Interaction of cocaine-, benztropine-, and GBR12909-like compounds with wildtype and mutant human dopamine transporters: molecular features that differentially determine antagonist binding properties. J. Neurochem 115: 296–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Schmitt KC, Reith MEA. 2010. Regulation of the dopamine transporter Aspects relevant to psychostimulant drugs of abuse. Ann. Ny. Acad. Sci 1187: 316–40 [DOI] [PubMed] [Google Scholar]
  • 45.Fleckenstein AE, Volz TJ, Riddle EL, Gibb JW, Hanson GR. 2007. New insights into the mechanism of action of amphetamines. Annu. Rev. Pharmacol 47: 681–98 [DOI] [PubMed] [Google Scholar]
  • 46.Wood S, Sage JR, Shuman T, Anagnostaras SG. 2014. Psychostimulants and cognition: A continuum of behavioral and cognitive activation. Pharmacol. Rev 66: 193–221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Madras B, Kuhar MJ. 2014. The Effects of Drug Abuse on the Human Nervous System. Amsterdam: Elsevier. xiii, 609 pages pp. [Google Scholar]
  • 48.Wise RA, Robble MA. 2020. Dopamine and Addiction. Annu Rev Psychol 71: 79–106 [DOI] [PubMed] [Google Scholar]
  • 49.Newman AH, Allen AC, Izenwasser S, Katz JL. 1994. Novel 3 alpha-(diphenylmethoxy)tropane analogs: potent dopamine uptake inhibitors without cocaine-like behavioral profiles. J Med Chem 37: 2258–61 [DOI] [PubMed] [Google Scholar]
  • 50.Newman AH, Kline RH, Allen AC, Izenwasser S, George C, Katz JL. 1995. Novel 4′-substituted and 4′,4″-disubstituted 3 alpha-(diphenylmethoxy)tropane analogs as potent and selective dopamine uptake inhibitors. J Med Chem 38: 3933–40 [DOI] [PubMed] [Google Scholar]
  • 51.Kline RH, Izenwasser S, Katz JL, Joseph DB, Bowen WD, Newman AH. 1997. 3′-Chloro-3 alpha-(diphenylmethoxy)tropane but not 4′-chloro-3 alpha-(diphenylmethoxy)tropane produces a cocaine-like behavioral profile. J Med Chem 40: 851–7 [DOI] [PubMed] [Google Scholar]
  • 52.Agoston GE, Wu JH, Izenwasser S, George C, Katz J, et al. 1997. Novel N-substituted 3 alpha-[bis(4′-fluorophenyl)methoxy]tropane analogues: selective ligands for the dopamine transporter. J Med Chem 40: 4329–39 [DOI] [PubMed] [Google Scholar]
  • 53.Katz JL, Izenwasser S, Kline RH, Allen AC, Newman AH. 1999. Novel 3alpha-diphenylmethoxytropane analogs: selective dopamine uptake inhibitors with behavioral effects distinct from those of cocaine. J Pharmacol Exp Ther 288: 302–15 [PubMed] [Google Scholar]
  • 54.Robarge MJ, Agoston GE, Izenwasser S, Kopajtic T, George C, et al. 2000. Highly selective chiral N-substituted 3alpha-[bis(4′-fluorophenyl)methoxy]tropane analogues for the dopamine transporter: synthesis and comparative molecular field analysis. J Med Chem 43: 1085–93 [DOI] [PubMed] [Google Scholar]
  • 55.Newman AH, Robarge MJ, Howard IM, Wittkopp SL, George C, et al. 2001. Structure-activity relationships at monoamine transporters and muscarinic receptors for N-substituted-3alpha-(3′-chloro-, 4′-chloro-, and 4′,4″-dichloro-substituted-diphenyl)methoxytropanes. J Med Chem 44: 633–40 [DOI] [PubMed] [Google Scholar]
  • 56.Katz JL, Agoston GE, Alling KL, Kline RH, Forster MJ, et al. 2001. Dopamine transporter binding without cocaine-like behavioral effects: synthesis and evaluation of benztropine analogs alone and in combination with cocaine in rodents. Psychopharmacology (Berl) 154: 362–74 [DOI] [PubMed] [Google Scholar]
  • 57.Zou MF, Kopajtic T, Katz JL, Newman AH. 2003. Structure-activity relationship comparison of (S)-2beta-substituted 3alpha-(bis[4-fluorophenyl]methoxy)tropanes and (R)-2beta-substituted 3beta-(3,4-dichlorophenyl)tropanes at the dopamine transporter. J Med Chem 46: 2908–16 [DOI] [PubMed] [Google Scholar]
  • 58.Katz JL, Kopajtic TA, Agoston GE, Newman AH. 2004. Effects of N-substituted analogs of benztropine: diminished cocaine-like effects in dopamine transporter ligands. J Pharmacol Exp Ther 309: 650–60 [DOI] [PubMed] [Google Scholar]
  • 59.Grundt P, Kopajtic TA, Katz JL, Newman AH. 2004. The effect of 6-substituted-4′,4″-difluorobenztropines on monoamine transporters and the muscarinic M1 receptor. Bioorg Med Chem Lett 14: 3295–8 [DOI] [PubMed] [Google Scholar]
  • 60.Kulkarni SS, Grundt P, Kopajtic T, Katz JL, Newman AH. 2004. Structure-activity relationships at monoamine transporters for a series of N-substituted 3alpha-(bis[4-fluorophenyl]methoxy)tropanes: comparative molecular field analysis, synthesis, and pharmacological evaluation. J Med Chem 47: 3388–98 [DOI] [PubMed] [Google Scholar]
  • 61.Grundt P, Kopajtic TA, Katz JL, Newman AH. 2005. N-8-Substituted benztropinamine analogs as selective dopamine transporter ligands. Bioorg Med Chem Lett 15: 5419–23 [DOI] [PubMed] [Google Scholar]
  • 62.Zou MF, Cao J, Kopajtic T, Desai RI, Katz JL, Newman AH. 2006. Structure-activity relationship studies on a novel series of (S)-2beta-substituted 3alpha-[bis(4-fluoro- or 4-chlorophenyl)methoxy]tropane analogues for in vivo investigation. J Med Chem 49: 6391–9 [DOI] [PubMed] [Google Scholar]
  • 63.Newman AH, Kulkarni S. 2002. Probes for the dopamine transporter: new leads toward a cocaine-abuse therapeutic--A focus on analogues of benztropine and rimcazole. Med Res Rev 22: 429–64 [DOI] [PubMed] [Google Scholar]
  • 64.Desai RI, Kopajtic TA, Koffarnus M, Newman AH, Katz JL. 2005. Identification of a dopamine transporter ligand that blocks the stimulant effects of cocaine. J Neurosci 25: 1889–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Velazquez-Sanchez C, Garcia-Verdugo JM, Murga J, Canales JJ. 2013. The atypical dopamine transport inhibitor, JHW 007, prevents amphetamine-induced sensitization and synaptic reorganization within the nucleus accumbens. Prog Neuropsychopharmacol Biol Psychiatry 44: 73–80 [DOI] [PubMed] [Google Scholar]
  • 66.Hiranita T, Wilkinson DS, Hong WC, Zou MF, Kopajtic TA, et al. 2014. 2-isoxazol-3-phenyltropane derivatives of cocaine: molecular and atypical system effects at the dopamine transporter. J Pharmacol Exp Ther 349: 297–309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Desai RI, Grandy DK, Lupica CR, Katz JL. 2014. Pharmacological characterization of a dopamine transporter ligand that functions as a cocaine antagonist. J Pharmacol Exp Ther 348: 106–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kohut SJ, Hiranita T, Hong SK, Ebbs AL, Tronci V, et al. 2014. Preference for distinct functional conformations of the dopamine transporter alters the relationship between subjective effects of cocaine and stimulation of mesolimbic dopamine. Biol Psychiatry 76: 802–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Avelar AJ, Cao J, Newman AH, Beckstead MJ. 2017. Atypical dopamine transporter inhibitors R-modafinil and JHW 007 differentially affect D2 autoreceptor neurotransmission and the firing rate of midbrain dopamine neurons. Neuropharmacology 123: 410–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Husbands SM, Izenwasser S, Kopajtic T, Bowen WD, Vilner BJ, et al. 1999. Structure-activity relationships at the monoamine transporters and sigma receptors for a novel series of 9-[3-(cis-3, 5-dimethyl-1-piperazinyl)propyl]carbazole (rimcazole) analogues. J Med Chem 42: 4446–55 [DOI] [PubMed] [Google Scholar]
  • 71.Cao J, Husbands SM, Kopajtic T, Katz JL, Newman AH. 2001. [3-cis-3,5-Dimethyl-(1-piperazinyl)alkyl]-bis-(4′-fluorophenyl)amine analogues as novel probes for the dopamine transporter. Bioorg Med Chem Lett 11: 3169–73 [DOI] [PubMed] [Google Scholar]
  • 72.Cao J, Kulkarni SS, Husbands SM, Bowen WD, Williams W, et al. 2003. Dual probes for the dopamine transporter and sigma1 receptors: novel piperazinyl alkyl-bis(4′-fluorophenyl)amine analogues as potential cocaine-abuse therapeutic agents. J Med Chem 46: 2589–98 [DOI] [PubMed] [Google Scholar]
  • 73.Cao J, Kopajtic T, Katz JL, Newman AH. 2008. Dual DAT/sigma1 receptor ligands based on 3-(4-(3-(bis(4-fluorophenyl)amino)propyl)piperazin-1-yl)-1-phenylpropan-1-ol. Bioorg Med Chem Lett 18: 5238–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Matsumoto RR, Hewett KL, Pouw B, Bowen WD, Husbands SM, et al. 2001. Rimcazole analogs attenuate the convulsive effects of cocaine: correlation with binding to sigma receptors rather than dopamine transporters. Neuropharmacology 41: 878–86 [DOI] [PubMed] [Google Scholar]
  • 75.Katz JL, Libby TA, Kopajtic T, Husbands SM, Newman AH. 2003. Behavioral effects of rimcazole analogues alone and in combination with cocaine. Eur J Pharmacol 468: 109–19 [DOI] [PubMed] [Google Scholar]
  • 76.Hiranita T, Soto PL, Kohut SJ, Kopajtic T, Cao J, et al. 2011. Decreases in cocaine self-administration with dual inhibition of the dopamine transporter and sigma receptors. J Pharmacol Exp Ther 339: 662–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hiranita T, Hong WC, Kopajtic T, Katz JL. 2017. sigma Receptor Effects of N-Substituted Benztropine Analogs: Implications for Antagonism of Cocaine Self-Administration. J Pharmacol Exp Ther 362: 2–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Contreras PC, Bremer ME, Rao TS. 1990. GBR-12909 and fluspirilene potently inhibited binding of [3H] (+)3-PPP to sigma receptors in rat brain. Life Sci 47: PL133–7 [DOI] [PubMed] [Google Scholar]
  • 79.Reith ME, Blough BE, Hong WC, Jones KT, Schmitt KC, et al. 2015. Behavioral, biological, and chemical perspectives on atypical agents targeting the dopamine transporter. Drug Alcohol Depend 147: 1–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hong WC, Kopajtic TA, Xu L, Lomenzo SA, Jean B, et al. 2016. 2-Substituted 3beta-Aryltropane Cocaine Analogs Produce Atypical Effects without Inducing Inward-Facing Dopamine Transporter Conformations. J Pharmacol Exp Ther 356: 624–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Penetar DM, Looby AR, Su Z, Lundahl LH, Eros-Sarnyai M, et al. 2006. Benztropine pretreatment does not affect responses to acute cocaine administration in human volunteers. Hum Psychopharmacol 21: 549–59 [DOI] [PubMed] [Google Scholar]
  • 82.Sambo DO, Lin M, Owens A, Lebowitz JJ, Richardson B, et al. 2017. The sigma-1 receptor modulates methamphetamine dysregulation of dopamine neurotransmission. Nat Commun 8: 2228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Jamieson C, Moir EM, Rankovic Z, Wishart G. 2006. Medicinal chemistry of hERG optimizations: Highlights and hang-ups. J Med Chem 49: 5029–46 [DOI] [PubMed] [Google Scholar]
  • 84.Perrin MJ, Kuchel PW, Campbell TJ, Vandenberg JI. 2008. Drug binding to the inactivated state is necessary but not sufficient for high-affinity binding to human ether-a-go-go-related gene channels. Mol Pharmacol 74: 1443–52 [DOI] [PubMed] [Google Scholar]
  • 85.Gintant G, Sager PT, Stockbridge N. 2016. Evolution of strategies to improve preclinical cardiac safety testing. Nat Rev Drug Discov 15: 457–71 [DOI] [PubMed] [Google Scholar]
  • 86.Kalyaanamoorthy S, Barakat KH. 2018. Binding modes of hERG blockers: an unsolved mystery in the drug design arena. Expert Opin Drug Discov 13: 207–10 [DOI] [PubMed] [Google Scholar]
  • 87.Kadric S, Mohler H, Kallioniemi O, Altmann KH. 2019. A Multicenter, Randomized, Placebo-Controlled Study to Evaluate the Efficacy and Safety of Long-Acting Injectable Formulation of Vanoxerine (Vanoxerine Consta 394.2 mg) for Cocaine Relapse Prevention. World Journal of Neuroscience 9: 113–37 [Google Scholar]
  • 88.Tanda G, Newman AH, Katz JL. 2009. Discovery of drugs to treat cocaine dependence: behavioral and neurochemical effects of atypical dopamine transport inhibitors. Adv Pharmacol 57: 253–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Beuming T, Kniazeff J, Bergmann ML, Shi L, Gracia L, et al. 2008. The binding sites for cocaine and dopamine in the dopamine transporter overlap. Nat Neurosci 11: 780–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Desai RI, Kopajtic TA, French D, Newman AH, Katz JL. 2005. Relationship between in vivo occupancy at the dopamine transporter and behavioral effects of cocaine, GBR 12909 [1-{2-[bis-(4-fluorophenyl)methoxy]ethyl}−4-(3-phenylpropyl)piperazine], and benztropine analogs. J Pharmacol Exp Ther 315: 397–404 [DOI] [PubMed] [Google Scholar]
  • 91.Volkow ND, Ding YS, Fowler JS, Wang GJ. 1996. Cocaine addiction: hypothesis derived from imaging studies with PET. J Addict Dis 15: 55–71 [DOI] [PubMed] [Google Scholar]
  • 92.Dackis CA, Kampman KM, Lynch KG, Pettinati HM, O’Brien CP. 2005. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology 30: 205–11 [DOI] [PubMed] [Google Scholar]
  • 93.Ballon JS, Feifel D. 2006. A systematic review of modafinil: Potential clinical uses and mechanisms of action. J Clin Psychiatry 67: 554–66 [DOI] [PubMed] [Google Scholar]
  • 94.Hart CL, Haney M, Vosburg SK, Rubin E, Foltin RW. 2008. Smoked cocaine self-administration is decreased by modafinil. Neuropsychopharmacology 33: 761–8 [DOI] [PubMed] [Google Scholar]
  • 95.Anderson AL, Reid MS, Li SH, Holmes T, Shemanski L, et al. 2009. Modafinil for the treatment of cocaine dependence. Drug Alcohol Depend 104: 133–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Loland CJ, Mereu M, Okunola OM, Cao J, Prisinzano TE, et al. 2012. R-modafinil (armodafinil): a unique dopamine uptake inhibitor and potential medication for psychostimulant abuse. Biol Psychiatry 72: 405–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Mereu M, Bonci A, Newman AH, Tanda G. 2013. The neurobiology of modafinil as an enhancer of cognitive performance and a potential treatment for substance use disorders. Psychopharmacology (Berl) 229: 415–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Mereu M, Chun LE, Prisinzano TE, Newman AH, Katz JL, Tanda G. 2017. The unique psychostimulant profile of (+/−)-modafinil: investigation of behavioral and neurochemical effects in mice. Eur J Neurosci 45: 167–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Cao J, Prisinzano TE, Okunola OM, Kopajtic T, Shook M, et al. 2010. Structure-Activity Relationships at the Monoamine Transporters for a Novel Series of Modafinil (2-[(diphenylmethyl)sulfinyl]acetamide) Analogues. ACS Med Chem Lett 2: 48–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Okunola-Bakare OM, Cao J, Kopajtic T, Katz JL, Loland CJ, et al. 2014. Elucidation of structural elements for selectivity across monoamine transporters: novel 2-[(diphenylmethyl)sulfinyl]acetamide (modafinil) analogues. J Med Chem 57: 1000–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Zhang HY, Bi GH, Yang HJ, He Y, Xue G, et al. 2017. The Novel Modafinil Analog, JJC8-016, as a Potential Cocaine Abuse Pharmacotherapeutic. Neuropsychopharmacology 42: 1871–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Tunstall BJ, Ho CP, Cao J, Vendruscolo JCM, Schmeichel BE, et al. 2018. Atypical dopamine transporter inhibitors attenuate compulsive-like methamphetamine self-administration in rats. Neuropharmacology 131: 96–103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Fant AD, Wacker S, Jung J, Guo J, Abramyan AM, et al. 2019. Toward reducing hERG affinities for DAT inhibitors with a combined machine learning and molecular modeling approach. Biophys. J 116: 562a–62a [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Johannesen L, Vicente J, Mason JW, Erato C, Sanabria C, et al. 2016. Late sodium current block for drug-induced long QT syndrome: Results from a prospective clinical trial. Clin Pharmacol Ther 99: 214–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Bolleddula J, DeMent K, Driscoll JP, Worboys P, Brassil PJ, Bourdet DL. 2014. Biotransformation and bioactivation reactions of alicyclic amines in drug molecules. Drug Metab Rev 46: 379–419 [DOI] [PubMed] [Google Scholar]
  • 106.Reilly SW, Griffin S, Taylor M, Sahlholm K, Weng CC, et al. 2017. Highly Selective Dopamine D3 Receptor Antagonists with Arylated Diazaspiro Alkane Cores. J Med Chem 60: 9905–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Aronov AM. 2005. Predictive in silico modeling for hERG channel blockers. Drug Discov Today 10: 149–55 [DOI] [PubMed] [Google Scholar]
  • 108.Cao J, Slack RD, Bakare OM, Burzynski C, Rais R, et al. 2016. Novel and High Affinity 2-[(Diphenylmethyl)sulfinyl]acetamide (Modafinil) Analogues as Atypical Dopamine Transporter Inhibitors. J Med Chem 59: 10676–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Slack RD, Ku TC, Cao J, Giancola JB, Bonifazi A, et al. 2019. Structure-Activity Relationships for a Series of (Bis(4-fluorophenyl)methyl)sulfinyl Alkyl Alicyclic Amines at the Dopamine Transporter: Functionalizing the Terminal Nitrogen Affects Affinity, Selectivity, and Metabolic Stability. J Med Chem [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kalaba P, Aher NY, Ilic M, Dragacevic V, Wieder M, et al. 2017. Heterocyclic Analogues of Modafinil as Novel, Atypical Dopamine Transporter Inhibitors. J Med Chem 60: 9330–48 [DOI] [PubMed] [Google Scholar]
  • 111.Kalaba P, Ilic M, Aher NY, Dragacevic V, Wieder M, et al. 2020. Structure-Activity Relationships of Novel Thiazole-Based Modafinil Analogues Acting at Monoamine Transporters. J Med Chem 63: 391–417 [DOI] [PubMed] [Google Scholar]
  • 112.Di Chiara G, Acquas E, Tanda G, Cadoni C. 1993. Drugs of abuse: biochemical surrogates of specific aspects of natural reward? Biochem Soc Symp 59: 65–81 [PubMed] [Google Scholar]
  • 113.Landwehrmeyer B, Mengod G, Palacios JM. 1993. Dopamine D3 receptor mRNA and binding sites in human brain. Brain Res Mol Brain Res 18: 187–92 [DOI] [PubMed] [Google Scholar]
  • 114.Landwehrmeyer B, Mengod G, Palacios JM. 1993. Differential visualization of dopamine D2 and D3 receptor sites in rat brain. A comparative study using in situ hybridization histochemistry and ligand binding autoradiography. Eur J Neurosci 5: 145–53 [DOI] [PubMed] [Google Scholar]
  • 115.Sokoloff P, Giros B, Martres MP, Bouthenet ML, Schwartz JC. 1990. Molecular cloning and characterization of a novel dopamine receptor (D3) as a target for neuroleptics. Nature 347: 146–51 [DOI] [PubMed] [Google Scholar]
  • 116.Caine SB, Koob GF. 1993. Modulation of cocaine self-administration in the rat through D-3 dopamine receptors. Science 260: 1814–6 [DOI] [PubMed] [Google Scholar]
  • 117.Mash DC, Staley JK. 1999. D3 dopamine and kappa opioid receptor alterations in human brain of cocaine-overdose victims. Ann N Y Acad Sci 877: 507–22 [DOI] [PubMed] [Google Scholar]
  • 118.Segal DM, Moraes CT, Mash DC. 1997. Up-regulation of D3 dopamine receptor mRNA in the nucleus accumbens of human cocaine fatalities. Brain Res Mol Brain Res 45: 335–9 [DOI] [PubMed] [Google Scholar]
  • 119.Staley JK, Mash DC. 1996. Adaptive increase in D3 dopamine receptors in the brain reward circuits of human cocaine fatalities. J Neurosci 16: 6100–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Matuskey D, Gallezot JD, Pittman B, Williams W, Wanyiri J, et al. 2014. Dopamine D(3) receptor alterations in cocaine-dependent humans imaged with [(1)(1)C](+)PHNO. Drug Alcohol Depend 139: 100–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Payer D, Balasubramaniam G, Boileau I. 2014. What is the role of the D3 receptor in addiction? A mini review of PET studies with [(11)C]-(+)-PHNO. Prog Neuropsychopharmacol Biol Psychiatry 52: 4–8 [DOI] [PubMed] [Google Scholar]
  • 122.Aujla H, Sokoloff P, Beninger RJ. 2002. A dopamine D3 receptor partial agonist blocks the expression of conditioned activity. Neuroreport 13: 173–6 [DOI] [PubMed] [Google Scholar]
  • 123.Cervo L, Carnovali F, Stark JA, Mennini T. 2003. Cocaine-seeking behavior in response to drug-associated stimuli in rats: involvement of D3 and D2 dopamine receptors. Neuropsychopharmacology 28: 1150–9 [DOI] [PubMed] [Google Scholar]
  • 124.Gal K, Gyertyan I. 2006. Dopamine D3 as well as D2 receptor ligands attenuate the cue-induced cocaine-seeking in a relapse model in rats. Drug Alcohol Depend 81: 63–70 [DOI] [PubMed] [Google Scholar]
  • 125.Le Foll B, Frances H, Diaz J, Schwartz JC, Sokoloff P. 2002. Role of the dopamine D3 receptor in reactivity to cocaine-associated cues in mice. Eur J Neurosci 15: 2016–26 [DOI] [PubMed] [Google Scholar]
  • 126.Pilla M, Perachon S, Sautel F, Garrido F, Mann A, et al. 1999. Selective inhibition of cocaine-seeking behaviour by a partial dopamine D3 receptor agonist. Nature 400: 371–5 [DOI] [PubMed] [Google Scholar]
  • 127.Reavill C, Taylor SG, Wood MD, Ashmeade T, Austin NE, et al. 2000. Pharmacological actions of a novel, high-affinity, and selective human dopamine D(3) receptor antagonist, SB-277011-A. J Pharmacol Exp Ther 294: 1154–65 [PubMed] [Google Scholar]
  • 128.Guerrero-Bautista R, Do Couto BR, Hidalgo JM, Carceles-Moreno FJ, Molina G, et al. 2019. Modulation of stress- and cocaine prime-induced reinstatement of conditioned place preference after memory extinction through dopamine D3 receptor. Prog Neuropsychopharmacol Biol Psychiatry 92: 308–20 [DOI] [PubMed] [Google Scholar]
  • 129.Vorel SR, Ashby CR Jr., Paul M, Liu X, Hayes R, et al. 2002. Dopamine D3 receptor antagonism inhibits cocaine-seeking and cocaine-enhanced brain reward in rats. J Neurosci 22: 9595–603 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Di Ciano P, Underwood RJ, Hagan JJ, Everitt BJ. 2003. Attenuation of cue-controlled cocaine-seeking by a selective D3 dopamine receptor antagonist SB-277011-A. Neuropsychopharmacology 28: 329–38 [DOI] [PubMed] [Google Scholar]
  • 131.Austin NE, Baldwin SJ, Cutler L, Deeks N, Kelly PJ, et al. 2001. Pharmacokinetics of the novel, high-affinity and selective dopamine D3 receptor antagonist SB-277011 in rat, dog and monkey: in vitro/in vivo correlation and the role of aldehyde oxidase. Xenobiotica 31: 677–86 [DOI] [PubMed] [Google Scholar]
  • 132.Remington G, Kapur S. 2001. SB-277011 GlaxoSmithKline. Curr Opin Investig Drugs 2: 946–9 [PubMed] [Google Scholar]
  • 133.Xi ZX, Gardner EL. 2007. Pharmacological actions of NGB 2904, a selective dopamine D3 receptor antagonist, in animal models of drug addiction. CNS Drug Rev 13: 240–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Pritchard LM, Newman AH, McNamara RK, Logue AD, Taylor B, et al. 2007. The dopamine D3 receptor antagonist NGB 2904 increases spontaneous and amphetamine-stimulated locomotion. Pharmacol Biochem Behav 86: 718–26 [DOI] [PubMed] [Google Scholar]
  • 135.Higley AE, Spiller K, Grundt P, Newman AH, Kiefer SW, et al. 2011. PG01037, a novel dopamine D3 receptor antagonist, inhibits the effects of methamphetamine in rats. J Psychopharmacol 25: 263–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.John WS, Newman AH, Nader MA. 2015. Differential effects of the dopamine D3 receptor antagonist PG01037 on cocaine and methamphetamine self-administration in rhesus monkeys. Neuropharmacology 92: 34–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Keck TM, John WS, Czoty PW, Nader MA, Newman AH. 2015. Identifying Medication Targets for Psychostimulant Addiction: Unraveling the Dopamine D3 Receptor Hypothesis. J Med Chem 58: 5361–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Newman AH, Battiti FO, Bonifazi A. 2019. 2016 Philip S. Portoghese Medicinal Chemistry Lectureship: Designing Bivalent or Bitopic Molecules for G-Protein Coupled Receptors. The Whole Is Greater Than the Sum of Its Parts. J Med Chem [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Kumar V, Bonifazi A, Ellenberger MP, Keck TM, Pommier E, et al. 2016. Highly Selective Dopamine D3 Receptor (D3R) Antagonists and Partial Agonists Based on Eticlopride and the D3R Crystal Structure: New Leads for Opioid Dependence Treatment. J Med Chem 59: 7634–50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Shaik AB, Kumar V, Bonifazi A, Guerrero AM, Cemaj SL, et al. 2019. Investigation of Novel Primary and Secondary Pharmacophores and 3-Substitution in the Linking Chain of a Series of Highly Selective and Bitopic Dopamine D3 Receptor Antagonists and Partial Agonists. J Med Chem 62: 9061–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Gyertyan I, Kiss B, Gal K, Laszlovszky I, Horvath A, et al. 2007. Effects of RGH-237 [N-{4-[4-(3-aminocarbonyl-phenyl)-piperazin-1-yl]-butyl}−4-bromo-benzamide], an orally active, selective dopamine D(3) receptor partial agonist in animal models of cocaine abuse. J Pharmacol Exp Ther 320: 1268–78 [DOI] [PubMed] [Google Scholar]
  • 142.Appel NM, Li SH, Holmes TH, Acri JB. 2015. Dopamine D3 Receptor Antagonist (GSK598809) Potentiates the Hypertensive Effects of Cocaine in Conscious, Freely-Moving Dogs. J Pharmacol Exp Ther 354: 484–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Jordan CJ, Humburg BA, Thorndike EB, Shaik AB, Xi ZX, et al. 2019. Newly Developed Dopamine D3 Receptor Antagonists, R-VK4-40 and R-VK4-116, Do Not Potentiate Cardiovascular Effects of Cocaine or Oxycodone in Rats. J Pharmacol Exp Ther 371: 602–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.You ZB, Bi GH, Galaj E, Kumar V, Cao J, et al. 2019. Dopamine D3R antagonist VK4-116 attenuates oxycodone self-administration and reinstatement without compromising its antinociceptive effects. Neuropsychopharmacology 44: 1415–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Jordan CJ, Cao J, Newman AH, Xi ZX. 2019. Progress in agonist therapy for substance use disorders: Lessons learned from methadone and buprenorphine. Neuropharmacology 158: 107609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Jordan CJ, Xi ZX. 2018. Discovery and development of varenicline for smoking cessation. Expert Opin Drug Discov 13: 671–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Roman V, Gyertyan I, Saghy K, Kiss B, Szombathelyi Z. 2013. Cariprazine (RGH-188), a D(3)-preferring dopamine D(3)/D(2) receptor partial agonist antipsychotic candidate demonstrates anti-abuse potential in rats. Psychopharmacology (Berl) 226: 285–93 [DOI] [PubMed] [Google Scholar]
  • 148.Orio L, Wee S, Newman AH, Pulvirenti L, Koob GF. 2010. The dopamine D3 receptor partial agonist CJB090 and antagonist PG01037 decrease progressive ratio responding for methamphetamine in rats with extended-access. Addict Biol 15: 312–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Martelle JL, Claytor R, Ross JT, Reboussin BA, Newman AH, Nader MA. 2007. Effects of two novel D3-selective compounds, NGB 2904 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butyl)-9H-fluorene-2-carboxamide] and CJB 090 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butyl)-4-(pyridin-2-yl)benzamide], on the reinforcing and discriminative stimulus effects of cocaine in rhesus monkeys. J Pharmacol Exp Ther 321: 573–82 [DOI] [PubMed] [Google Scholar]
  • 150.Garcia-Ladona FJ, Cox BF. 2003. BP 897, a selective dopamine D3 receptor ligand with therapeutic potential for the treatment of cocaine-addiction. CNS Drug Rev 9: 141–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Achat-Mendes C, Platt DM, Newman AH, Spealman RD. 2009. The dopamine D3 receptor partial agonist CJB 090 inhibits the discriminative stimulus but not the reinforcing or priming effects of cocaine in squirrel monkeys. Psychopharmacology (Berl) 206: 73–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Hachimine P, Seepersad N, Ananthan S, Ranaldi R. 2014. The novel dopamine D3 receptor antagonist, SR 21502, reduces cocaine conditioned place preference in rats. Neurosci Lett 569: 137–41 [DOI] [PubMed] [Google Scholar]
  • 153.Czoty PW, Stoops WW, Rush CR. 2016. Evaluation of the “Pipeline” for Development of Medications for Cocaine Use Disorder: A Review of Translational Preclinical, Human Laboratory, and Clinical Trial Research. Pharmacol Rev 68: 533–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Kampman KM. 2019. The treatment of cocaine use disorder. Sci Adv 5: eaax1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Jordan CJ, Xi ZX. 2019. Progress in brain cannabinoid CB2 receptor research: From genes to behavior. Neurosci Biobehav Rev 98: 208–20 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES