1
|
Allen MI, Rahimi O, Johnson BN, Cao J, Newman AH, Nader MA. Contrasting the reinforcing effects of the novel dopamine transport inhibitors JJC8-088 and JJC8-091 in monkeys: Potential translation to medication assisted treatment. J Pharmacol Exp Ther 2025; 392:100033. [PMID: 39892998 PMCID: PMC11822870 DOI: 10.1124/jpet.124.002356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Despite considerable efforts, there remains no Food and Drug Administration-approved medications for cocaine use disorder. One strategy to mitigate cocaine craving and relapse is to elevate dopamine. The dopamine transport inhibitor and releaser d-amphetamine has been shown to decrease cocaine self-administration (SA), although it has abuse liability. Recently, several modafinil analogs reduced cocaine SA in rats and monkeys, including JJC8-088, characterized as "cocaine-like" in rats, and JJC8-091, characterized as "atypical" and not SA by rats. The present study evaluated the reinforcing effects of both compounds in monkeys under several conditions. For experiment 1, 4 male cocaine-experienced rhesus monkeys self-administered cocaine (0.001-0.3 mg/kg per injection), JJC8-088 (0.001-0.3 mg/kg per injection), and JJC8-091 (0.1-3.0 mg/kg per injection) under a progressive-ratio schedule of reinforcement. Both JJC compounds functioned as reinforcers with equal reinforcing strength to cocaine. Although JJC8-091 was less potent than cocaine, JJC8-088 and cocaine had similar potencies. For experiment 2, 1 male and 2 female drug-naïve cynomolgus monkeys responded on a fixed-ratio schedule of food reinforcement. JJC8-091 was self-administered at rates higher than saline in all 3 monkeys. In experiment 3, monkeys from experiment 2 responded under a concurrent drug versus food choice paradigm and given access to cocaine or JJC8-091 under these conditions. At doses equal to or one-half log-units higher than doses used in experiment 2, cocaine, but not JJC8-091, was chosen over food. Together, these results demonstrate that although JJC8-091 may be reinforcing under some conditions, its reinforcing strength, in the presence of an alternative reinforcer, is substantially less than cocaine. SIGNIFICANCE STATEMENT: JJC8-088 and JJC8-091 have shown efficacy is reducing cocaine self-administration (SA) in rats and in nonhuman primates. This study found that both compounds maintained SA in monkeys responding under several conditions. However, when given access to an alternative reinforcer during the SA session, JJC8-091 was not reinforcing, suggesting that JJC8-091 may have lower abuse liability than cocaine and may be a viable candidate for cocaine use disorder because, in the human population, alternatives to drug use are often available.
Collapse
Affiliation(s)
- Mia I Allen
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Omeed Rahimi
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Bernard N Johnson
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Jianjing Cao
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| | - Michael A Nader
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
2
|
Xi ZX, Soler-Cedeño O, Galaj E, Klein B, Cao J, Bi GH, Newman A. RDS04-010: A novel atypical DAT inhibitor that inhibits cocaine taking and seeking and itself has low abuse potential in experimental animals. RESEARCH SQUARE 2024:rs.3.rs-5269973. [PMID: 39606448 PMCID: PMC11601829 DOI: 10.21203/rs.3.rs-5269973/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Cocaine use disorder (CUD) is a severe public health problem, and currently, there is no FDA-approved medication for its treatment. Atypical dopamine (DA) transporter (DAT) inhibitors display low addictive liability by themselves and may have therapeutic potential for treatment of psychostimulant use disorders. Here, we report that RDS04-010, a novel atypical DAT inhibitor that binds to an inward-facing conformation of DAT due to its sulfoxide moiety, displayed distinct pharmacological profiles in animal models of addiction from its sulfide analog, RDS03-094, a DAT inhibitor that binds to a more outward-facing conformation. Systemic administration of RDS04-010 dose-dependently inhibited cocaine self-administration (SA), shifted the cocaine SA dose-response curve downward, decreased motivation for cocaine seeking under progressive ratio reinforcement conditions, and inhibited cocaine-primed reinstatement of drug-seeking behavior. RDS04-010 alone neither altered optical brain-stimulation reward nor evoked reinstatement of drug-seeking behavior. RDS04-010 substitution for cocaine was not able to maintain self-administration in rats trained to self-administer cocaine. In contrast, RDS03-094 displayed more cocaine-like reinforcing effects. Its pretreatment upward-shifted both the cocaine self-administration dose-response and optical brain-stimulation reward curves. RDS03-094 alone was able to reinstate extinguished cocaine-seeking behavior and sustain self-administration during a substitution test. Collectively, these findings suggest that RDS04-010 is a novel atypical DAT inhibitor with favorable therapeutic potential in reducing cocaine-taking and -seeking behavior with low addictive liability. Moreover, this extensive behavioral evaluation further confirms the role DAT binding conformation plays in the distinctive profiles of atypical DAT inhibitors that prefer the inward facing conformation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Amy Newman
- National Institute on Drug Abuse (NIDA), NIH
| |
Collapse
|
3
|
Mojtabai R, Susukida R, Farokhnia M, Nguyen TQ, Dunn KE, Amin-Esmaeili M. Trajectories of craving in the course of pharmacotherapy trials for methamphetamine use disorder. Addiction 2024; 119:1803-1812. [PMID: 38984671 DOI: 10.1111/add.16610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 06/03/2024] [Indexed: 07/11/2024]
Abstract
AIMS The aim of this study was to measure trajectories of craving for methamphetamine during the course of pharmacotherapy trials for methamphetamine use disorder. DESIGN, SETTING AND PARTICIPANTS Craving trajectories were identified using Group-Based Trajectory Modeling. The association of craving trajectories with drug use trajectories was examined using a dual trajectory model. Association of craving trajectories with other health and social outcomes was also examined. The study used pooled data from five randomized controlled pharmacotherapy trials for methamphetamine use disorder. A total of 866 adults with methamphetamine use disorder participated in randomized controlled pharmacotherapy trials. MEASUREMENT Craving was assessed weekly using the Brief Substance Craving Scale. Drug use was assessed using urine toxicology. Alcohol- and drug-related problems, as well as psychiatric, medical, legal, employment and relationship problems, were measured using the Addiction Severity Index. FINDINGS A three-trajectory model with high, medium and low craving trajectories was selected as the most parsimonious model. Craving trajectories were associated with methamphetamine use trajectories in the course of trial; 88.4% of those in the high craving trajectory group had a consistently high frequency of methamphetamine use compared with 18.7% of those in the low craving group. High craving was also associated with less improvement in most other outcomes and higher rate of dropout from treatment. In turn, low craving was associated with a rapidly decreasing frequency of methamphetamine use, greater improvement in most other outcomes and a lower rate of dropout. Participants on modafinil daily and ondansetron 1 mg twice daily were less likely to be in the high craving group compared with those on placebo. CONCLUSIONS Trajectories of methamphetamine craving in the course of clinical trials for methamphetamine use disorder appear to be both highly variable and strongly associated with greater frequency of drug use, other drug-related outcomes and dropout from trials. Two medications, modafinil daily and ondansetron at a dose of 1 mg two times daily, appear to be associated with greater reduction in craving in the course of treatment compared with placebo. A decrease in methamphetamine craving shows promise as an early indicator of recovery from methamphetamine use disorder.
Collapse
Affiliation(s)
- Ramin Mojtabai
- Department of Psychiatry and Behavioral Sciences, Tulane Medical School, New Orleans, LA, USA
| | - Ryoko Susukida
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Mehdi Farokhnia
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Baltimore, MD, USA
| | - Trang Quynh Nguyen
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kelly E Dunn
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Masoumeh Amin-Esmaeili
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Donlon J, Kumari P, Varghese SP, Bai M, Florentin OD, Frost ED, Banks J, Vadlapatla N, Kam O, Shad MU, Rahman S, Abulseoud OA, Stone TW, Koola MM. Integrative Pharmacology in the Treatment of Substance Use Disorders. J Dual Diagn 2024; 20:132-177. [PMID: 38117676 DOI: 10.1080/15504263.2023.2293854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The detrimental physical, mental, and socioeconomic effects of substance use disorders (SUDs) have been apparent to the medical community for decades. However, it has become increasingly urgent in recent years to develop novel pharmacotherapies to treat SUDs. Currently, practitioners typically rely on monotherapy. Monotherapy has been shown to be superior to no treatment at all for most substance classes. However, many randomized controlled trials (RCTs) have revealed that monotherapy leads to poorer outcomes when compared with combination treatment in all specialties of medicine. The results of RCTs suggest that monotherapy frequently fails since multiple dysregulated pathways, enzymes, neurotransmitters, and receptors are involved in the pathophysiology of SUDs. As such, research is urgently needed to determine how various neurobiological mechanisms can be targeted by novel combination treatments to create increasingly specific yet exceedingly comprehensive approaches to SUD treatment. This article aims to review the neurobiology that integrates many pathophysiologic mechanisms and discuss integrative pharmacology developments that may ultimately improve clinical outcomes for patients with SUDs. Many neurobiological mechanisms are known to be involved in SUDs including dopaminergic, nicotinic, N-methyl-D-aspartate (NMDA), and kynurenic acid (KYNA) mechanisms. Emerging evidence indicates that KYNA, a tryptophan metabolite, modulates all these major pathophysiologic mechanisms. Therefore, achieving KYNA homeostasis by harmonizing integrative pathophysiology and pharmacology could prove to be a better therapeutic approach for SUDs. We propose KYNA-NMDA-α7nAChRcentric pathophysiology, the "conductor of the orchestra," as a novel approach to treat many SUDs concurrently. KYNA-NMDA-α7nAChR pathophysiology may be the "command center" of neuropsychiatry. To date, extant RCTs have shown equivocal findings across comparison conditions, possibly because investigators targeted single pathophysiologic mechanisms, hit wrong targets in underlying pathophysiologic mechanisms, and tested inadequate monotherapy treatment. We provide examples of potential combination treatments that simultaneously target multiple pathophysiologic mechanisms in addition to KYNA. Kynurenine pathway metabolism demonstrates the greatest potential as a target for neuropsychiatric diseases. The investigational medications with the most evidence include memantine, galantamine, and N-acetylcysteine. Future RCTs are warranted with novel combination treatments for SUDs. Multicenter RCTs with integrative pharmacology offer a promising, potentially fruitful avenue to develop novel therapeutics for the treatment of SUDs.
Collapse
Affiliation(s)
- Jack Donlon
- Cooper Medical School of Rowan University, Camden, New Jersey, USA
| | - Pooja Kumari
- Community Living Trent Highlands, Peterborough, Canada
| | - Sajoy P Varghese
- Addiction Recovery Treatment Services, Veterans Affairs Northern California Health Care System, University of California, Davis, Sacramento, California, USA
| | - Michael Bai
- Columbia University, New York, New York, USA
| | - Ori David Florentin
- Department of Psychiatry, Westchester Medical Center, Valhalla, New York, USA
| | - Emma D Frost
- Department of Neurology, Cooper University Health Care, Camden, New Jersey, USA
| | - John Banks
- Talkiatry Mental Health Clinic, New York, New York, USA
| | - Niyathi Vadlapatla
- Thomas Jefferson High School for Science and Technology, Alexandria, Virginia, USA
| | - Olivia Kam
- Stony Brook University Renaissance School of Medicine, Stony Brook, New York, USA
| | - Mujeeb U Shad
- Department of Psychiatry, University of Nevada Las Vegas, Las Vegas, Nevada, USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, South Dakota, USA
| | - Osama A Abulseoud
- Department of Psychiatry and Psychology, Alix School of Medicine at Mayo Clinic, Phoenix, Arizona, USA
| | - Trevor W Stone
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Maju Mathew Koola
- Department of Psychiatry and Behavioral Health, Cooper University Health Care, Cooper Medical School of Rowan University, Camden, New Jersey, USA
| |
Collapse
|
5
|
Schmitz JM, Stotts AL, Vujanovic AA, Yoon JH, Webber HE, Lane SD, Weaver MF, Vincent J, Suchting R, Green CE. Contingency management plus acceptance and commitment therapy for initial cocaine abstinence: Results of a sequential multiple assignment randomized trial (SMART). Drug Alcohol Depend 2024; 256:111078. [PMID: 38309089 DOI: 10.1016/j.drugalcdep.2023.111078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND This study tested an adaptive intervention for optimizing abstinence outcomes over phases of treatment for cocaine use disorder using a SMART design. Phase 1 assessed whether 4 weeks of contingency management (CM) improved response with the addition of Acceptance and Commitment Therapy (ACT). Phase 2 assessed pharmacological augmentation with modafinil (MOD) vs. placebo (PLA) for individuals not achieving abstinence during Phase 1. METHOD For Phase 1 of treatment, participants (N=118) were randomly allocated to ACT+CM or Drug Counseling (DC+CM), the comparison condition. At week 4, treatment response was defined as the submission of six consecutive cocaine-negative urine drug screens (UDS). Phase 1 non-responders were re-randomized to MOD or PLA as adjunct to their initial treatment. Phase 1 responders continued receiving their initial treatment. Primary outcomes included response rate and proportion of cocaine-negative UDS for Phase 1 and 2. Analyses used Bayesian inference with 80% pre-specified as the posterior probability (PP) threshold constituting moderate evidence that an effect exists. RESULTS Phase 1 response was higher in the ACT+CM group (24.5%) compared to the DC+CM group (17.5%; PP = 84.5%). In Phase 2, the proportion of cocaine-negative UDS among Phase 1 responders did not differ by initial treatment (PP = 61.8%) but remained higher overall compared to Phase 1 non-responders (PPs > 99%). No evidence of an effect favoring augmentation with MOD was observed. DISCUSSION Adding ACT to CM increased abstinence initiation. Initial responders were more likely to remain abstinent compared to initial non-responders, for whom modafinil was not an effective pharmacotherapy augmentation strategy.
Collapse
Affiliation(s)
- Joy M Schmitz
- Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States.
| | - Angela L Stotts
- Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States; Department of Family and Community Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Anka A Vujanovic
- Department of Psychological and Brain Sciences, Texas A&M University, United States
| | - Jin H Yoon
- Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Heather E Webber
- Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Scott D Lane
- Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Michael F Weaver
- Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Jessica Vincent
- Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Robert Suchting
- Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Charles E Green
- Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States; UTHealth Center for Clinical Research & Evidence-Based Medicine, United States
| |
Collapse
|
6
|
Carpenter KM, Choi CJ, Basaraba C, Pavlicova M, Brooks DJ, Brezing CA, Bisaga A, Nunes EV, Mariani JJ, Levin FR. Mixed amphetamine salts-extended release (MAS-ER) as a behavioral treatment augmentation strategy for cocaine use disorder: A randomized clinical trial. Exp Clin Psychopharmacol 2024; 32:112-127. [PMID: 37732961 PMCID: PMC10872820 DOI: 10.1037/pha0000676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Psychosocial interventions remain the primary strategy for addressing cocaine use disorder (CUD), although many individuals do not benefit from these approaches. Amphetamine-based interventions have shown significant promise and may improve outcomes among individuals continuing to use cocaine in the context of behavioral interventions. One hundred forty-five adults (122 males) who used cocaine a minimum of 4 days in the prior month and met the criteria for a CUD enrolled in a two-stage intervention. All participants received a computer-delivered skills intervention and contingency management for reinforcing abstinence for a 1-month period. Participants demonstrating less than 3 weeks of abstinence in the first month were randomized to receive mixed amphetamine salts-extended release (MAS-ER) or placebo (80 mg/day) for 10 weeks under double-blind conditions. All participants continued with the behavioral intervention. The primary outcome was the proportion of individuals who achieved 3 consecutive weeks of abstinence as measured by urine toxicology confirmed self-report at the study end. The proportion of participants demonstrating 3 consecutive weeks of abstinence at study end did not differ between the medication groups: MAS-ER = 15.6% (7/45) and placebo = 12.2% (5/41). Participants who received MAS-ER reported greater reductions in the magnitude of wanting cocaine, although no group differences were noted in either the perceived improvement or the frequency of wanting cocaine. Retention rates were greater for both medication groups compared to behavioral responders. Overall, augmenting a behavioral intervention with MAS-ER did not significantly increase the abstinence rate among individuals continuing to use cocaine following a month of behavioral therapy alone. (PsycInfo Database Record (c) 2024 APA, all rights reserved).
Collapse
Affiliation(s)
| | | | | | | | - Daniel J Brooks
- Division on Substance Use Disorders, New York State Psychiatric Institute
| | | | - Adam Bisaga
- Division on Substance Use Disorders, New York State Psychiatric Institute
| | - Edward V Nunes
- Division on Substance Use Disorders, New York State Psychiatric Institute
| | - John J Mariani
- Division on Substance Use Disorders, New York State Psychiatric Institute
| | - Frances R Levin
- Division on Substance Use Disorders, New York State Psychiatric Institute
| |
Collapse
|
7
|
Hersey M, Tanda G. Modafinil, an atypical CNS stimulant? ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 99:287-326. [PMID: 38467484 PMCID: PMC12004278 DOI: 10.1016/bs.apha.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Modafinil is a central nervous system stimulant approved for the treatment of narcolepsy and sleep disorders. Due to its wide range of biochemical actions, modafinil has been explored for other potential therapeutic uses. Indeed, it has shown promise as a therapy for cognitive disfunction resulting from neurologic disorders like ADHD, and as a smart drug in non-medical settings. The mechanism(s) of actions underlying the therapeutic efficacy of this agent remains largely elusive. Modafinil is known to inhibit the dopamine transporter, thus decreasing dopamine reuptake following neuronal release, an effect shared by addictive psychostimulants. However, modafinil is unique in that only a few cases of dependence on this drug have been reported, as compared to other psychostimulants. Moreover, modafinil has been tested, with some success, as a potential therapeutic agent to combat psychostimulant and other substance use disorders. Modafinil has additional, but less understood, actions on other neurotransmitter systems (GABA, glutamate, serotonin, norepinephrine, etc.). These interactions, together with its ability to activate selected brain regions, are likely one of the keys to understand its unique pharmacology and therapeutic activity as a CNS stimulant. In this chapter, we outline the pharmacokinetics and pharmacodynamics of modafinil that suggest it has an "atypical" CNS stimulant profile. We also highlight the current approved and off label uses of modafinil, including its beneficial effects as a treatment for sleep disorders, cognitive functions, and substance use disorders.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, NIDA-IRP, NIH, Baltimore, MD, United States
| | - Gianluigi Tanda
- Medication Development Program, NIDA-IRP, NIH, Baltimore, MD, United States.
| |
Collapse
|
8
|
Rahimi O, Cao J, Lam J, Childers SR, Rais R, Porrino LJ, Newman AH, Nader MA. The Effects of the Dopamine Transporter Ligands JJC8-088 and JJC8-091 on Cocaine versus Food Choice in Rhesus Monkeys. J Pharmacol Exp Ther 2023; 384:372-381. [PMID: 36507847 PMCID: PMC9976790 DOI: 10.1124/jpet.122.001363] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/26/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Although there are no Food and Drug Administration-approved treatments for cocaine use disorder, several modafinil analogs have demonstrated promise in reducing cocaine self-administration and reinstatement in rats. Furthermore, the range of dopamine transporter (DAT) compounds provides an opportunity to develop pharmacotherapeutics without abuse liability. This study extended the comparison of JJC8-088 and JJC8-091, the former compound having higher DAT affinity and predicted abuse liability, to rhesus monkeys using a concurrent cocaine versus food schedule of reinforcement. First, binding to striatal DAT was examined in cocaine-naïve monkey tissue. Next, intravenous pharmacokinetics of both JJC compounds were evaluated in cocaine-experienced male monkeys (n = 3/drug). In behavioral studies, acute and chronic administration of both compounds were evaluated in these same monkeys responding under a concurrent food versus cocaine (0 and 0.003-0.1 mg/kg per injection) schedule of reinforcement. In nonhuman primate striatum, JJC8-088 had higher DAT affinity compared with JJC8-091 (14.4 ± 9 versus 2730 ± 1270 nM, respectively). Both JJC compounds had favorable plasma pharmacokinetics for behavioral assessments, with half-lives of 1.1 hours and 3.5 hours for JJC8-088 (0.7 mg/kg, i.v.) and JJC8-091 (1.9 mg/kg, i.v.), respectively. Acute treatment with both compounds shifted the cocaine dose-response curve to the left. Chronic treatment with JJC8-088 decreased cocaine choice in two of the three monkeys, whereas JJC8-091 only modestly reduced cocaine allocation in one monkey. Differences in affinities of JJC8-091 DAT binding in monkeys compared with rats may account for the poor rodent-to-monkey translation. Future studies should evaluate atypical DAT blockers in combination with behavioral interventions that may further decrease cocaine choice. SIGNIFICANCE STATEMENT: Cocaine use disorder (CUD) remains a significant public health problem with no Food and Drug Administration-approved treatments. The ability of drugs that act in the brain in a similar manner to cocaine, but with lower abuse liability, has clinical implications for a treatment of CUD.
Collapse
Affiliation(s)
- Omeed Rahimi
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Jianjing Cao
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Jenny Lam
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Steven R Childers
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Rana Rais
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Linda J Porrino
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Amy Hauck Newman
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Michael A Nader
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| |
Collapse
|
9
|
Lassi DLS, Malbergier A, Negrão AB, Florio L, De Aquino JP, Castaldelli-Maia JM. Pharmacological Treatments for Cocaine Craving: What Is the Way Forward? A Systematic Review. Brain Sci 2022; 12:1546. [PMID: 36421870 PMCID: PMC9688748 DOI: 10.3390/brainsci12111546] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/01/2022] [Accepted: 11/10/2022] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND cocaine craving is a core feature of cocaine use disorder and remains a critical challenge for abstinence and relapse prevention. This review summarizes the anti-craving efficacy of pharmacotherapies tested for cocaine use disorder, in the context of randomized-controlled clinical trials. OBJECTIVES we assessed the databases of the U.S. National Library of Medicine, Google Scholar, and PsycINFO, without date restrictions up to August 2022, to identify relevant studies. STUDY ELIGIBILITY CRITERIA, PARTICIPANTS, AND INTERVENTIONS we included double-blinded randomized-controlled trials investigating pharmacotherapies for cocaine craving and/or cocaine use disorder whose outcomes included cocaine craving. STUDY APPRAISAL AND SYNTHESIS METHODS Two authors screened studies' titles and abstracts for inclusion, and both read all the included studies. We systematically gathered information on the following aspects of each study: title; author(s); year of publication; sample size; mean age; sample characteristics; study set-ting; whether participants were treatment-seeking; study design; craving measures; study interventions; drop-out rates; and other relevant outcomes. RESULTS Overall, we appraised 130 clinical trials, including 8137 participants. We further considered the drugs from the studies that scored equal to or greater than six points in the quality assessment. There was a correlation between craving and cocaine use outcomes (self-reports, timeline follow-back or urinary benzoylecgonine) in the vast majority of studies. In the short-term treatment, acute phenylalanine-tyrosine depletion, clonidine, fenfluramine, meta-chlorophenylpiperazine (m-CPP) and mecamylamine presented promising effects. In the long term, amphetamine, biperiden, carbamazepine, lisdexamfetamine, lorcaserin, methamphetamine, mirtazapine, pioglitazone, progesterone, guanfacine, levodopa, nefazodone presented promising anti-craving effects. Unfortunately, the highly tested medications were not successful in most of the trials, as follows: propranolol in the short term; amantadine, aripiprazole, bromocriptine, citicoline, ketamine, modafinil, olanzapine, topiramate in the long term. The remaining 52 medications had no positive anti-craving outcomes. LIMITATIONS Our review was limited by high heterogeneity of craving assessments across the studies and by a great range of pharmacotherapies. Further, the majority of the studies considered abstinence and retention in treatment as the main outcomes, whereas craving was a secondary outcome and some of the studies evaluated patients with cocaine use disorder with comorbidities such as opioid or alcohol use disorder, schizophrenia, bipolar disorder or attention deficit hyperactivity. Lastly, most of the studies also included non-pharmacological treatments, such as counseling or psychotherapy. CONCLUSIONS There is a direct association between craving and cocaine use, underscoring craving as an important treatment target for promoting abstinence among persons with cocaine use disorder. Clonidine, fenfluramine and m-CPP showed to be promising medications for cocaine craving in the short-term treatment, and amphetamine, biperiden, carbamazepine, lisdexamfetamine, lorcaserin, methamphetamine, mirtazapine, pioglitazone, progesterone, guanfacine, levodopa, nefazodone in the long-term treatment.
Collapse
Affiliation(s)
- Dângela Layne Silva Lassi
- Interdisciplinary Group of Alcohol and Drug Studies (GREA), Department and Institute of Psychiatry, Medical School, São Paulo University, São Paulo 05403-010, SP, Brazil
| | - André Malbergier
- Interdisciplinary Group of Alcohol and Drug Studies (GREA), Department and Institute of Psychiatry, Medical School, São Paulo University, São Paulo 05403-010, SP, Brazil
| | - André Brooking Negrão
- Interdisciplinary Group of Alcohol and Drug Studies (GREA), Department and Institute of Psychiatry, Medical School, São Paulo University, São Paulo 05403-010, SP, Brazil
| | - Lígia Florio
- Interdisciplinary Group of Alcohol and Drug Studies (GREA), Department and Institute of Psychiatry, Medical School, São Paulo University, São Paulo 05403-010, SP, Brazil
| | - João P. De Aquino
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - João Maurício Castaldelli-Maia
- Interdisciplinary Group of Alcohol and Drug Studies (GREA), Department and Institute of Psychiatry, Medical School, São Paulo University, São Paulo 05403-010, SP, Brazil
- Department of Neuroscience, Medical School, ABC Health University Center, Santo André 09060-870, SP, Brazil
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| |
Collapse
|
10
|
Jensen KL, Jensen SB, Madsen KL. A mechanistic overview of approaches for the treatment of psychostimulant dependence. Front Pharmacol 2022; 13:854176. [PMID: 36160447 PMCID: PMC9493975 DOI: 10.3389/fphar.2022.854176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Psychostimulant use disorder is a major health issue around the world with enormous individual, family-related and societal consequences, yet there are no effective pharmacological treatments available. In this review, a target-based overview of pharmacological treatments toward psychostimulant addiction will be presented. We will go through therapeutic approaches targeting different aspects of psychostimulant addiction with focus on three major areas; 1) drugs targeting signalling, and metabolism of the dopamine system, 2) drugs targeting either AMPA receptors or metabotropic glutamate receptors of the glutamate system and 3) drugs targeting the severe side-effects of quitting long-term psychostimulant use. For each of these major modes of intervention, findings from pre-clinical studies in rodents to clinical trials in humans will be listed, and future perspectives of the different treatment strategies as well as their potential side-effects will be discussed. Pharmaceuticals modulating the dopamine system, such as antipsychotics, DAT-inhibitors, and disulfiram, have shown some promising results. Cognitive enhancers have been found to increase aspects of behavioural control, and drugs targeting the glutamate system such as modulators of metabotropic glutamate receptors and AMPA receptors have provided interesting changes in relapse behaviour. Furthermore, CRF-antagonists directed toward alleviating the symptoms of the withdrawal stage have been examined with interesting resulting changes in behaviour. There are promising results investigating therapeutics for psychostimulant addiction, but further preclinical work and additional human studies with a more stratified patient selection are needed to prove sufficient evidence of efficacy and tolerability.
Collapse
|
11
|
Martins B, Rutland W, De Aquino JP, Kazer BL, Funaro M, Potenza MN, Angarita GA. Helpful or Harmful? The Therapeutic Potential of Medications with Varying Degrees of Abuse Liability in the Treatment of Substance Use Disorders. CURRENT ADDICTION REPORTS 2022; 9:647-659. [PMID: 35990796 PMCID: PMC9376579 DOI: 10.1007/s40429-022-00432-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 11/06/2022]
Abstract
Purpose of Review This review summarizes recent clinical trial research on pharmacological treatments for substance use disorders, with a specific focus on agents with potential abuse liability. Recent Findings Pharmacological treatments for substance use disorders may include gabapentinoids, baclofen, modafinil, ketamine, cannabinoids, gamma-hydroxybutyrate, and psychedelics. Gabapentinoids may decrease negative subjective effects of withdrawal in alcohol and cannabis use disorders. Cannabinoids similarly appear to decrease use and withdrawal symptoms in cannabis use disorder, while research shows stimulant medications may reduce cravings and increase abstinence in cocaine use disorder. Ketamine and psychedelics may help treat multiple substance use disorders. Ketamine may reduce withdrawal symptoms, promote abstinence, and diminish cravings in alcohol and cocaine use disorders and psychedelics may promote remission, decrease use, and reduce cravings in alcohol and opioid use disorders. Summary Regardless of current regulatory approval statuses and potentials for abuse, multiple agents should not be dismissed prematurely as possible treatments for substance use disorders. However, further clinical research is needed before effective implementation can begin in practice. Supplementary Information The online version contains supplementary material available at 10.1007/s40429-022-00432-9.
Collapse
Affiliation(s)
- Bradford Martins
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| | - Will Rutland
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| | - Joao P. De Aquino
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| | - Benjamin L. Kazer
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| | - Melissa Funaro
- Harvey Cushing/John Hay Whitney Medical Library, Yale University, New Haven, CT 06510 USA
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Child Study Center, Yale University School of Medicine, New Haven, CT 06510 USA
- Department of Neuroscience, Yale University, New Haven, CT 06510 USA
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
- Connecticut Council On Problem Gambling, Wethersfield, CT 06109 USA
- Wu Tsai Institute, Yale University, New Haven, CT 06510 USA
| | - Gustavo A. Angarita
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| |
Collapse
|
12
|
DeVito EE, Poling J, Babuscio T, Nich C, Carroll KM, Sofuoglu M. Modafinil Does Not Reduce Cocaine Use in Methadone-Maintained Individuals. DRUG AND ALCOHOL DEPENDENCE REPORTS 2022; 2:100032. [PMID: 36310662 PMCID: PMC9615926 DOI: 10.1016/j.dadr.2022.100032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Introduction There are no approved medications for the treatment of cocaine use disorder (CUD). Modafinil, a cognitive-enhancer with weak stimulant-like effects, has shown promise in initial studies as a treatment for CUD. Its potential efficacy has not been examined in individuals dually dependent on cocaine and opioids. Methods This study examined the efficacy of modafinil, in combination with contingency management (CM), for reducing cocaine and opioid use and improving cognitive function in methadone-stabilized individuals with opioid and cocaine dependence. We conducted a 17-week, double-blind, randomized controlled trial in which participants were randomized to one of four conditions: 1) modafinil + CM; 2) modafinil + yoked-control (YC); 3) placebo +CM; or 4) placebo + YC. Additionally, all subjects received platform treatments of cognitive behavioral therapy (CBT) and methadone. While the original planned sample size was N=160, a total of 91 participants were randomized. The two primary cocaine use outcomes were percentage of urine specimens positive for cocaine and percent of days of self-reported abstinence from cocaine during treatment. Cognitive function, opioid use, and secondary cocaine use outcomes were also considered. Results Modafinil was well-tolerated with minimal reports of adverse effects. Modafinil was no more effective than placebo in reducing cocaine or opioid use or improving cognitive performance. Conclusions In the context of a trial with robust control conditions and platform treatments, findings did not provide support for the efficacy of modafinil treatment for the treatment of CUD in methadone-stabilized individuals with dual opioid and cocaine dependence.
Collapse
Affiliation(s)
- Elise E. DeVito
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - James Poling
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Theresa Babuscio
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Charla Nich
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Kathleen M. Carroll
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Mehmet Sofuoglu
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States,VA Connecticut Healthcare System, West Haven, CT, United States,Corresponding author at: VA Connecticut Healthcare System, 950 Campbell Ave., Bldg. 36/116A4, West Haven, CT 06516.
| |
Collapse
|
13
|
Reinstatement of synaptic plasticity in the aging brain through specific dopamine transporter inhibition. Mol Psychiatry 2021; 26:7076-7090. [PMID: 34244620 DOI: 10.1038/s41380-021-01214-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging-related neurological deficits negatively impact mental health, productivity, and social interactions leading to a pronounced socioeconomic burden. Since declining brain dopamine signaling during aging is associated with the onset of neurological impairments, we produced a selective dopamine transporter (DAT) inhibitor to restore endogenous dopamine levels and improve cognitive function. We describe the synthesis and pharmacological profile of (S,S)-CE-158, a highly specific DAT inhibitor, which increases dopamine levels in brain regions associated with cognition. We find both a potentiation of neurotransmission and coincident restoration of dendritic spines in the dorsal hippocampus, indicative of reinstatement of dopamine-induced synaptic plasticity in aging rodents. Treatment with (S,S)-CE-158 significantly improved behavioral flexibility in scopolamine-compromised animals and increased the number of spontaneously active prefrontal cortical neurons, both in young and aging rodents. In addition, (S,S)-CE-158 restored learning and memory recall in aging rats comparable to their young performance in a hippocampus-dependent hole board test. In sum, we present a well-tolerated, highly selective DAT inhibitor that normalizes the age-related decline in cognitive function at a synaptic level through increased dopamine signaling.
Collapse
|
14
|
Hesse M, Thomsen KR, Thylstrup B, Andersen CU, Reitzel LA, Worm-Leonhard M, Lindholst C. Purity of street-level cocaine across Denmark from 2006 to 2019: Analysis of seized cocaine. Forensic Sci Int 2021; 329:111050. [PMID: 34736046 DOI: 10.1016/j.forsciint.2021.111050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022]
Abstract
Cocaine-related emergency department admissions are increasing, and cocaine seizures are at an all-time high in Europe. Our aim was to investigate the trends in purity and adulterants over time in cocaine available to cocaine users at street level in Denmark. We used a representative sample of cocaine seized at street level and analyzed by the national departments of forensic medicine between 2006 and 2019 (n = 1460). Latent profile analysis was used to classify the samples based on cocaine, levamisole, and phenacetin content. Low purity cocaine comprised most of the cocaine seizures in early years, but its share began to decline in 2013, and from 2016 to 2019, the high purity profile was dominant. While the total number of samples containing adulterants decreased, levamisole remained a common and dangerous adulterant. The findings underline the need to inform the public, medical doctors, and service providers for people with drug use disorders about the higher potency of street cocaine.
Collapse
Affiliation(s)
- Morten Hesse
- Aarhus University, Center for Alcohol and Drug Research, Bartholins Allé 10, 8000 Aarhus C, Denmark.
| | - Kristine Rømer Thomsen
- Aarhus University, Center for Alcohol and Drug Research, Bartholins Allé 10, 8000 Aarhus C, Denmark.
| | - Birgitte Thylstrup
- Aarhus University, Center for Alcohol and Drug Research, Bartholins Allé 10, 8000 Aarhus C, Denmark.
| | - Charlotte Uggerhøj Andersen
- Aarhus University, Department of Forensic Medicine, Palle Juul-Jensens Boulevard 99, Building Z, 3.017, 8200 Aarhus N, Denmark.
| | | | | | - Christian Lindholst
- Aarhus University, Department of Forensic Medicine, Palle Juul-Jensens Boulevard 99, Building Z, 3.017, 8200 Aarhus N, Denmark.
| |
Collapse
|
15
|
Suchting R, Green CE, de Dios C, Vincent J, Moeller FG, Lane SD, Schmitz JM. Citalopram for treatment of cocaine use disorder: A Bayesian drop-the-loser randomized clinical trial. Drug Alcohol Depend 2021; 228:109054. [PMID: 34600245 PMCID: PMC8595787 DOI: 10.1016/j.drugalcdep.2021.109054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Medication development research for cocaine use disorder (CUD) has been a longstanding goal in addiction research, but has not resulted in an FDA-approved treatment. Rising cocaine use rates underscore the need for efficient adaptive designs. This study compared differences between two doses of the selective serotonin reuptake inhibitor (SSRI) citalopram (versus placebo) on duration of cocaine abstinence and applied adaptive decision rules to select the 'best efficacy' dose. METHODS A double-blind, placebo-controlled, randomized Bayesian drop-the-loser (DTL) trial with three arms compared placebo to citalopram 20 mg and 40 mg. Adults (N = 107) with CUD attended thrice-weekly clinic visits for 9 weeks. The primary outcome was longest duration of abstinence (LDA), based on continuous cocaine-negative urine drug screens (UDS). The secondary outcome was probability of cocaine-negative UDS during treatment. A planned interim analysis performed at approximately 50% of recruitment dropped the least-effective active medication. Bayesian inference was used for all analyses with a pre-specified posterior probability (PP) threshold PP ≥ 95% considered statistically reliable evidence RESULTS: Citalopram 40 mg satisfied interim efficacy criteria and was retained for the second half of the trial. For LDA, analyses indicated PP = 82% and PP = 65% of benefit for 40 mg and 20 mg, respectively (each relative to placebo). The odds of having cocaine-negative UDS decreased in all groups over 9 weeks but remained higher for 40 mg (PP = 97.4%) CONCLUSIONS: Neither dose met the 95% PP threshold for the primary outcome; however, 40 mg provided moderate-to-strong evidence for positive effects on LDA and cocaine-negative UDS. The 40 mg dose was declared the "winner" in this DTL trial.
Collapse
Affiliation(s)
- Robert Suchting
- Faillace Department of Psychiatry and Behavioral Sciences, UTHealth McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Charles E. Green
- Department of Pediatrics – Center for Clinical Research and Evidence-Based Medicine, UTHealth McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA,MD Anderson – UTHealth Graduate School of Biomedical Sciences, Program in Neuroscience, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Constanza de Dios
- Faillace Department of Psychiatry and Behavioral Sciences, UTHealth McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jessica Vincent
- Faillace Department of Psychiatry and Behavioral Sciences, UTHealth McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Scott D. Lane
- Faillace Department of Psychiatry and Behavioral Sciences, UTHealth McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joy M. Schmitz
- Faillace Department of Psychiatry and Behavioral Sciences, UTHealth McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
16
|
Fluyau D, Cook SC, Chima A, Kailasam VK, Revadigar N. Pharmacological management of psychoactive substance withdrawal syndrome. DRUGS & THERAPY PERSPECTIVES 2021. [DOI: 10.1007/s40267-021-00874-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
17
|
Treatment of multiple sclerosis fatigue with the synthetic psychoactive drug modafinil. Exp Neurol 2021; 347:113906. [PMID: 34710403 DOI: 10.1016/j.expneurol.2021.113906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/15/2021] [Accepted: 10/23/2021] [Indexed: 12/17/2022]
Abstract
Multiple sclerosis (MS) is a complex disorder characterized by a broad spectrum of symptoms that evolve throughout the disease. Symptoms can be categorized as visible and invisible based on external sight recognition. However, although others poorly recognize it, invisible symptoms such as mood dysfunction, neuropathic pain, or fatigue can significantly affect activities of daily living and the quality of life of people with MS (PwMS). PwMS frequently complain of fatigue, which has physical or cognitive manifestations. Fatigue in MS does not improve or resolve with rest, and it is disproportionate with respect to the exerted effort. Fatigue management in MS is challenging, and a few pharmacological approaches have been successfully proposed. Among them, the drug modafinil has attracted attention because of its properties as a synthetic psychoactive drug. In this review, we focus on the evidence available to date, supporting the use of modafinil in MS fatigue. However, despite the availability of some trials evaluating the effects of modafinil on fatigue, their contrasting results failed to support its usefulness in fatigue management in MS.
Collapse
|
18
|
Hersey M, Bacon AK, Bailey LG, Coggiano MA, Newman AH, Leggio L, Tanda G. Psychostimulant Use Disorder, an Unmet Therapeutic Goal: Can Modafinil Narrow the Gap? Front Neurosci 2021; 15:656475. [PMID: 34121988 PMCID: PMC8187604 DOI: 10.3389/fnins.2021.656475] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
The number of individuals affected by psychostimulant use disorder (PSUD) has increased rapidly over the last few decades resulting in economic, emotional, and physical burdens on our society. Further compounding this issue is the current lack of clinically approved medications to treat this disorder. The dopamine transporter (DAT) is a common target of psychostimulant actions related to their use and dependence, and the recent availability of atypical DAT inhibitors as a potential therapeutic option has garnered popularity in this research field. Modafinil (MOD), which is approved for clinical use for the treatment of narcolepsy and sleep disorders, blocks DAT just like commonly abused psychostimulants. However, preclinical and clinical studies have shown that it lacks the addictive properties (in both behavioral and neurochemical studies) associated with other abused DAT inhibitors. Clinical availability of MOD has facilitated its off-label use for several psychiatric disorders related to alteration of brain dopamine (DA) systems, including PSUD. In this review, we highlight clinical and preclinical research on MOD and its R-enantiomer, R-MOD, as potential medications for PSUD. Given the complexity of PSUD, we have also reported the effects of MOD on psychostimulant-induced appearance of several symptoms that could intensify the severity of the disease (i.e., sleep disorders and impairment of cognitive functions), besides the potential therapeutic effects of MOD on PSUD.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Amanda K. Bacon
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lydia G. Bailey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Mark A. Coggiano
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Amy H. Newman
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lorenzo Leggio
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
- Clinical Psychoneuroendo- crinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
- National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Bethesda, MD, United States
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
19
|
Mariani JJ, Choi CJ, Pavlicova M, Mahony AL, Brooks DJ, Grabowski J, Levin FR. Open-label pilot study of lisdexamfetamine for cocaine use disorder. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2021; 47:402-409. [PMID: 33797985 DOI: 10.1080/00952990.2021.1885677] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Cocaine use disorder (CUD) is a substantial public health problem with no FDA-approved medication treatments. Psychostimulants have shown promise as pharmacotherapy for CUD. Lisdexamfetamine, a novel prodrug psychostimulant, is roughly 40-50% as potent as dextroamphetamine.Objectives: To evaluate the safety, tolerability, and optimal dosing of lisdexamfetamine for treating CUD.Methods: Open-label, 8-week trial of 17 CUD adults. Participants were titrated to the maximum tolerated dose of 140 mg over 2-week period and maintained for 4 weeks, followed by a two-week taper period. The primary outcome measures were the maximum daily dose achieved during the study period and tolerability as measured by medication-related study drop-out.Results: Among the 16 participants with post-enrollment data, the mean dose of lisdexamfetamine achieved was 118.1 mg (standard deviation (SD) = 40.4), mean retention was 6.5 weeks (SD = 2.0), and no participants discontinued study medication due to adverse effects. Four participants had dose reductions due to adverse effects and continued in the trial. Six participants (37.5%) were abstinent for the last 3 weeks of their study participation. Mean dollars of cocaine spent per day significantly decreased from $19.72 at baseline to $7.57 during the last 3 weeks of study participation (t15 = 3.60, p = .003). The mean percent of using days significantly decreased from 25% at baseline to 12% during the last 3 weeks of study participation (t15 = 3.33, p = .005).Conclusion: The use of lisdexamfetamine for CUD in doses ranging to 140 mg daily was safe and generally well tolerated.
Collapse
Affiliation(s)
- John J Mariani
- Division on Substance Use Disorders, New York State Psychiatric Institute, New York, NY, USA.,Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - C Jean Choi
- Mental Health Data Science, New York State Psychiatric Institute, New York, NY, USA
| | - Martina Pavlicova
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Amy L Mahony
- Division on Substance Use Disorders, New York State Psychiatric Institute, New York, NY, USA
| | - Daniel J Brooks
- Division on Substance Use Disorders, New York State Psychiatric Institute, New York, NY, USA
| | - John Grabowski
- Department of Psychiatry, Office of Faculty Affairs-Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Frances R Levin
- Division on Substance Use Disorders, New York State Psychiatric Institute, New York, NY, USA.,Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
20
|
Brandt L, Chao T, Comer SD, Levin FR. Pharmacotherapeutic strategies for treating cocaine use disorder-what do we have to offer? Addiction 2021; 116:694-710. [PMID: 32888245 PMCID: PMC7930140 DOI: 10.1111/add.15242] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 08/28/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND Cocaine use contines to be a significant public health problem world-wide. However, despite substantial research efforts, no pharmacotherapies are approved for the treatment of cocaine use disorder (CUD). ARGUMENT Studies have identified positive signals for a range of medications for treating CUD. These include long-acting amphetamine formulations, modafinil, topiramate, doxazosin and combined topiramate and mixed amphetamine salts extended-release (MAS-ER). However, valid conclusions about a medication's clinical efficacy require nuanced approaches that take into account behavioural phenotypes of the target population (frequency of use, co-abuse of cocaine and other substances, genetic subgroups, psychiatric comorbidity), variables related to the medication (dose, short-/long-acting formulations, titration speed, medication adherence) and other factors that may affect treatment outcomes. Meta-analyses frequently do not account for these co-varying factors, which contributes to a somewhat nihilistic view on pharmacotherapeutic options for CUD. In addition, the predominant focus on abstinence, which is difficult for most patients to achieve, may overshadow more nuanced therapeutic signals. CONCLUSION While there is an emphasis on finding new medications with novel mechanisms of action for treating CUD, currently available medications deserve further investigation based on the existing literature. Evaluating refined metrics of treatment success in well-defined subgroups of patients, and further exploring combination therapies and their synergy with behavioural/psychosocial interventions, are promising avenues to establishing effective therapies for CUD.
Collapse
Affiliation(s)
- Laura Brandt
- Division on Substance Use Disorders, New York State Psychiatric Institute and Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Thomas Chao
- Division on Substance Use Disorders, New York State Psychiatric Institute and Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Psychology, The New School for Social Research, New York, NY, USA
| | - Sandra D. Comer
- Division on Substance Use Disorders, New York State Psychiatric Institute and Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Frances R. Levin
- Division on Substance Use Disorders, New York State Psychiatric Institute and Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
21
|
Haney M, Rubin E, Denson RK, Foltin RW. Modafinil reduces smoked cocaine self-administration in humans: effects vary as a function of cocaine 'priming' and cost. Drug Alcohol Depend 2021; 221:108554. [PMID: 33610094 PMCID: PMC8026732 DOI: 10.1016/j.drugalcdep.2021.108554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND The absence of an FDA-approved medication for the treatment of cocaine use disorder (CUD) may, in part, reflect the varying conditions present when the decision to use cocaine is made, with one medication unlikely to work under all conditions. The objective of this double-blind, placebo-controlled, human laboratory study was to test the effects of modafinil, a medication with mixed efficacy for the treatment of CUD, using a novel self-administration procedure designed to model distinct clinical scenarios. METHODS During modafinil maintenance (0, 300 mg/day), participants chose to self-administer up to 7 doses of smoked cocaine (25 mg) under 9 conditions: immediately after exposure to: (a) cues associated with cocaine and a non-contingent cocaine administration, i.e. 'prime' (25 mg), (b) only cocaine cues, and (c) neither cues nor cocaine. Each condition was tested when self-administered cocaine cost $5, $10 and $15/dose. RESULTS Nontreatment-seeking cocaine smokers (3 F,13 M), spending $388 ± 218/week on cocaine and with no history of alcohol use disorder, completed the study. Relative to placebo, modafinil robustly attenuated self-administration when cocaine was expensive ($10,$15/dose) and when there was no 'prime.' Modafinil had no effect on self-administration when cocaine was inexpensive ($5/dose) or when participants received a 'prime.' CONCLUSIONS Modafinil's effects on cocaine-taking varied substantially as a function of recent cocaine exposure and cost, which may help explain the mixed clinical findings. Modafinil may be most effective for preventing relapse in abstinent patients, particularly under conditions in which cocaine is costly, rather than initiating abstinence for those continuing to use cocaine.
Collapse
Affiliation(s)
- Margaret Haney
- Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, USA.
| | - Eric Rubin
- Department of Psychiatry, Harlem Hospital Center, Columbia University College of Physicians and Surgeons, USA
| | - Rebecca K Denson
- Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, USA
| | - Richard W Foltin
- Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, USA
| |
Collapse
|
22
|
GIPSON CD, BIMONTE-NELSON HA. Interactions between reproductive transitions during aging and addiction: promoting translational crosstalk between different fields of research. Behav Pharmacol 2021; 32:112-122. [PMID: 32960852 PMCID: PMC7965232 DOI: 10.1097/fbp.0000000000000591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Discovery of neural mechanisms underlying neuropsychiatric disorders within the aging and addiction fields has been a main focus of the National Institutes of Health. However, there is a dearth of knowledge regarding the biological interactions of aging and addiction, which may have important influences on progression of disease and treatment outcomes in aging individuals with a history of chronic drug use. Thus, there is a large gap in these fields of research, which has slowed progress in understanding and treating substance use disorders (SUDs) as well as age-related diseases, specifically in women who experience precipitous reproductive cycle transitions during aging. The goal of this review is to highlight overlap of SUDs and age-related processes with a specific focus on menopause and smoking, and identify critical gaps. We have narrowed the focus of the review to smoking, as the majority of findings on hormonal and aging influences on drug use have come from this area of research. Further, we highlight female-specific issues such as transitional menopause and exogenous estrogen use. These issues may impact drug use cessation as well as outcomes with aging and age-related neurodegenerative diseases in women. We first review clinical studies for smoking, normal aging, and pathological aging, and discuss the few aging-related studies taking smoking history into account. Conversely, we highlight the dearth of clinical smoking studies taking age as a biological variable into account. Preclinical and clinical literature show that aging, age-related pathological brain disease, and addiction engage overlapping neural mechanisms. We hypothesize that these putative drivers interact in meaningful ways that may exacerbate disease and hinder successful treatment outcomes in such comorbid populations. We highlight areas where preclinical studies are needed to uncover neural mechanisms in aging and addiction processes. Collectively, this review highlights the need for crosstalk between different fields of research to address medical complexities of older adults, and specifically women, who smoke.
Collapse
Affiliation(s)
- Cassandra D. GIPSON
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY
- Arizona Alzheimer’s Consortium
| | | |
Collapse
|
23
|
Giancola JB, Bonifazi A, Cao J, Ku T, Haraczy AJ, Lam J, Rais R, Coggiano MA, Tanda G, Newman AH. Structure-activity relationships for a series of (Bis(4-fluorophenyl)methyl)sulfinylethyl-aminopiperidines and -piperidine amines at the dopamine transporter: Bioisosteric replacement of the piperazine improves metabolic stability. Eur J Med Chem 2020; 208:112674. [PMID: 32947229 PMCID: PMC7680422 DOI: 10.1016/j.ejmech.2020.112674] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 01/13/2023]
Abstract
Despite considerable efforts to develop medications to treat psychostimulant use disorders, 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. Atypical dopamine transporter (DAT) inhibitors, based on (±)modafinil, have shown therapeutic potential in preclinical models of psychostimulant abuse. However, metabolic instability among other limitations to piperazine analogues 1-3 have impeded further development. Herein, bioisosteric substitutions of the piperazine ring were explored with a series of aminopiperidines (A) and piperidine amines (B) wherein compounds with either a terminal tertiary amine or amide were synthesized. Several lead compounds showed high to moderate DAT affinities and metabolic stability in rat liver microsomes. Aminopiperidines 7 (DAT Ki = 50.6 nM), 21b (DAT Ki = 77.2 nM) and 33 (DAT Ki = 30.0 nM) produced only minimal stimulation of ambulatory activity in mice, compared to cocaine, suggesting an atypical DAT inhibitor profile.
Collapse
Affiliation(s)
- JoLynn B Giancola
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, United States
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, United States
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, United States
| | - Therese Ku
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, United States
| | - Alexandra J Haraczy
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, United States; Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, United States
| | - Jenny Lam
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, United States; Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, United States
| | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, United States
| | - Mark A Coggiano
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, United States
| | - Gianluigi Tanda
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, United States
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, United States.
| |
Collapse
|
24
|
Tanda G, Hersey M, Hempel B, Xi ZX, Newman AH. Modafinil and its structural analogs as atypical dopamine uptake inhibitors and potential medications for psychostimulant use disorder. Curr Opin Pharmacol 2020; 56:13-21. [PMID: 32927246 DOI: 10.1016/j.coph.2020.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/20/2022]
Abstract
Pharmacotherapeutics for treatment of psychostimulant use disorder are still an unmet medical goal. Recently, off label use of modafinil (MOD), an approved medication for treatment of sleep disturbances, has been tested as a therapeutic for cocaine and methamphetamine use disorder. Positive results have been found in subjects dependent on psychostimulants without concurrent abuse of other substances. Novel structural analogs of MOD have been synthesized in the search for compounds with potentially broader therapeutic efficacy than the parent drug. In the present report we review their potential efficacy as treatments for psychostimulant abuse and dependence assessed in preclinical tests. Results from these preclinical proof of concept studies reveal that some modafinil analogs do not possess typical cocaine-like neurochemical and behavioral effects. Further, they might blunt the reinforcing effects of psychostimulants in animal models, suggesting their potential efficacy as pharmacotherapeutics for treatment of psychostimulant use disorders.
Collapse
Affiliation(s)
- Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| | - Melinda Hersey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Briana Hempel
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Amy Hauck Newman
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| |
Collapse
|
25
|
Modafinil potentiates cocaine self-administration by a dopamine-independent mechanism: possible involvement of gap junctions. Neuropsychopharmacology 2020; 45:1518-1526. [PMID: 32340023 PMCID: PMC7360549 DOI: 10.1038/s41386-020-0680-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/15/2020] [Accepted: 04/09/2020] [Indexed: 12/21/2022]
Abstract
Modafinil and methylphenidate are medications that inhibit the neuronal reuptake of dopamine, a mechanism shared with cocaine. Their use as "smart drugs" by healthy subjects poses health concerns and requires investigation. We show that methylphenidate, but not modafinil, maintained intravenous self-administration in Sprague-Dawley rats similar to cocaine. Both modafinil and methylphenidate pretreatments potentiated cocaine self-administration. Cocaine, at self-administered doses, stimulated mesolimbic dopamine levels. This effect was potentiated by methylphenidate, but not by modafinil pretreatments, indicating dopamine-dependent actions for methylphenidate, but not modafinil. Modafinil is known to facilitate electrotonic neuronal coupling by actions on gap junctions. Carbenoxolone, a gap junction inhibitor, antagonized modafinil, but not methylphenidate potentiation of cocaine self-administration. Our results indicate that modafinil shares mechanisms with cocaine and methylphenidate but has a unique pharmacological profile that includes facilitation of electrotonic coupling and lower abuse liability, which may be exploited in future therapeutic drug design for cocaine use disorder.
Collapse
|
26
|
Prescription psychostimulants for the treatment of stimulant use disorder: a systematic review and meta-analysis. Psychopharmacology (Berl) 2020; 237:2233-2255. [PMID: 32601988 DOI: 10.1007/s00213-020-05563-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/18/2020] [Indexed: 01/09/2023]
Abstract
RATIONALE Agonist-based pharmacologic intervention is an accepted approach in treatment of opioid and tobacco use disorders. OBJECTIVES We conducted a systematic review and meta-analysis to evaluate usefulness of an agonist approach as treatment of (psycho)stimulant use disorder (PSUD). METHODS We reviewed PubMed/Medline, LILACS, and ClinicalTrials.gov databases searching for randomized, double-blind, placebo-controlled, parallel-design studies evaluating outcomes of individuals treated for cocaine- or amphetamine-type substance use disorder. We combined results of all trials that included the following prescription psychostimulants (PPs): modafinil, methylphenidate, or amphetamines (mixed amphetamine salts, lisdexamphetamine, and dextroamphetamine). The combined sample consisted of 2889 patients. Outcomes of interest included the following: drug abstinence (defined as 2-3 weeks of sustained abstinence and the average maximum days of consecutive abstinence), percentage of drug-negative urine tests across trial, and retention in treatment. We conducted random-effects meta-analyses and assessed quality of evidence using the GRADE system. RESULTS Thirty-eight trials were included. Treatment with PPs increases rates of sustained abstinence [risk ratio (RR) = 1.45, 95% confidence interval (CI) = (1.10, 1.92)] and duration of abstinence [mean difference (MD) = 3.34, 95% CI = (1.06, 5.62)] in patients with PSUD, particularly those with cocaine use disorder (very low-quality evidence). Prescription amphetamines were particularly efficacious in promoting sustained abstinence in patients with cocaine use disorder [RR = 2.44, 95% CI = (1.66, 3.58)], and higher doses of PPs were particularly efficacious for treatment of cocaine use disorder [RR = 1.95, 95% CI = (1.38, 2.77)] (moderate-quality evidence). Treatment with prescription amphetamines also yielded more cocaine-negative urines [MD = 8.37%, 95% CI = (3.75, 12.98)]. There was no effect of PPs on the retention in treatment. CONCLUSION Prescription psychostimulants, particularly prescription amphetamines given in robust doses, have a clinically significant beneficial effect to promote abstinence in the treatment of individuals with PSUD, specifically the population with cocaine use disorder.
Collapse
|
27
|
Abstract
In this targeted review, we summarize current knowledge on substance-use disorder
(SUD)-related cognitive deficits, the link between these deficits and clinical outcomes,
and the cognitive training, remediation, and pharmacological approaches that have the
potential to rescue cognition. We conclude that: (i) people with SUDs have moderate
deficits in memory, attention, executive functions, and decision-making (including
reward expectancy, valuation, and learning); (ii) deficits in higher-order executive
functions and decision-making are significant predictors of relapse; (iii) cognitive
training programs targeting reward-related appetitive biases, cognitive remediation
strategies targeting goal-based decision-making, and pharmacotherapies targeting memory,
attention, and impulsivity have potential to rescue SUD-related cognitive deficits. We
suggest avenues for future research, including developing brief, clinically oriented
harmonized cognitive testing suites to improve individualized prediction of treatment
outcomes; computational modeling that can achieve deep phenotyping of cognitive subtypes
likely to respond to different interventions; and phenotype-targeted cognitive,
pharmacological, and combined interventions. We conclude with a tentative model of
neuroscience-informed precision medicine.
Collapse
Affiliation(s)
| | - Gloria Garcia-Fernandez
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia; Department of Psychology, University of Oviedo, Spain
| | - Geert Dom
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Antwerp University (UA), Antwerp, Belgium
| |
Collapse
|
28
|
Sleep quality improves during treatment with repetitive transcranial magnetic stimulation (rTMS) in patients with cocaine use disorder: a retrospective observational study. BMC Psychiatry 2020; 20:153. [PMID: 32252720 PMCID: PMC7137315 DOI: 10.1186/s12888-020-02568-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Sleep disturbance is a prominent and common complaint in people with cocaine use disorder (CUD), either during intake or withdrawal. Repetitive transcranial magnetic stimulation (rTMS) has shown promise as a treatment for CUD. Thus, we evaluated the relationship between self-perceived sleep quality and cocaine use pattern variables in outpatients with CUD undergoing an rTMS protocol targeted at the left dorsolateral prefrontal cortex. METHODS This is a retrospective observational study including 87 patients diagnosed with CUD according to the DSM-5 criteria. Scores in Pittsburgh Sleep Quality Index (PSQI), Cocaine Craving Questionnaire (CCQ), Beck Depression Inventory-II (BDI-II), Self-rating Anxiety Scale (SAS), and Symptoms checklist 90-Revised (outcome used: Global Severity Index, GSI) were recorded at baseline, and after 5, 30, 60, and 90 days of rTMS treatment. Cocaine use was assessed by self-report and regular urine screens. RESULTS Sleep disturbances (PSQI scores > 5) were common in patients at baseline (mean ± SD; PSQI score baseline: 9.24 ± 3.89; PSQI > 5 in 88.5% of patients). PSQI scores significantly improved after rTMS treatment (PSQI score Day 90: 6.12 ± 3.32). Significant and consistent improvements were also seen in craving and in negative-affect symptoms compared to baseline. Considering the lack of a control group, in order to help the conceptualization of the outcomes, we compared the results to a wait-list group (n = 10). No significant improvements were observed in the wait-list group in any of the outcome measures. CONCLUSIONS The present findings support the therapeutic role of rTMS interventions for reducing cocaine use and accompanying symptoms such as sleep disturbance and negative-affect symptoms. TRIAL REGISTRATION ClinicalTrials.gov.NCT03733821.
Collapse
|
29
|
Sousa A, Dinis-Oliveira RJ. Pharmacokinetic and pharmacodynamic of the cognitive enhancer modafinil: Relevant clinical and forensic aspects. Subst Abus 2020; 41:155-173. [PMID: 31951804 DOI: 10.1080/08897077.2019.1700584] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Modafinil is a nonamphetamine nootropic drug with an increasingly therapeutic interest due to its different sites of action and behavioral effects in comparison to cocaine or amphetamine. A review of modafinil (and of its prodrug adrafinil and its R-enantiomer armodafinil) chemical, pharmacokinetic, pharmacodynamic, toxicological, clinical and forensic aspects was performed, aiming to better understand possible health problems associated to its unconscious and unruled use. Modafinil is a racemate metabolized mainly in the liver into its inactive acid and sulfone metabolites, which undergo primarily renal excretion. Although not fully clarified, major effects seem to be associated to inhibition of dopamine reuptake and modulation of several other neurochemical pathways, namely noradrenergic, serotoninergic, orexinergic, histaminergic, glutamatergic and GABAergic. Due its wake-promoting effects, modafinil is used for the treatment of daily sleepiness associated to narcolepsy, obstructive sleep apnea and shift work sleep disorder. Its psychotropic and cognitive effects are also attractive in several other pathologies and conditions that affect sleep structure, induce fatigue and lethargy, and impair cognitive abilities. Additionally, in health subjects, including students, modafinil is being used off-label to overcome sleepiness, increase concentration and improve cognitive potential. The most common adverse effects associated to modafinil intake are headache, insomnia, anxiety, diarrhea, dry mouth and raise in blood pressure and heart rate. Infrequently, severe dermatologic effects in children, including maculopapular and morbilliform rash, erythema multiforme and Stevens-Johnson Syndrome have been reported. Intoxication and dependence associated to modafinil are uncommon. Further research on effects and health implications of modafinil and its analogs is steel needed to create evidence-based policies.
Collapse
Affiliation(s)
- Ana Sousa
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal.,IINFACTS - Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal.,UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
30
|
Regier PS, Kampman KM, Childress AR. Clinical Trials for Stimulant Use Disorders: Addressing Heterogeneities That May Undermine Treatment Outcomes. Handb Exp Pharmacol 2020; 258:299-322. [PMID: 32193666 DOI: 10.1007/164_2019_303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In recent years, use of cocaine and amphetamines and deaths associated with stimulants have been on the rise, and there are still no FDA-approved medications for stimulant use disorders. One contributing factor may involve heterogeneity. At the neurobiological level, dual dopamine dysfunction may be undermining medication efficacy, suggesting a need for combination pharmacotherapies. At the population level, individual variability is expressed in a number of ways and, if left unaddressed, may interfere with medication efficacy. This chapter reviews studies investigating medications to address dopamine dysfunction, and it also identifies several prominent heterogeneities associated with stimulant (and other substance) use disorders. The chapter has implications for improving interventions to treat stimulant use disorders, and the theme of individual heterogeneity may have broader application across substance use disorders.
Collapse
Affiliation(s)
- Paul S Regier
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, Philadelphia, PA, USA.
| | - Kyle M Kampman
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna Rose Childress
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
31
|
Kampman KM. The treatment of cocaine use disorder. SCIENCE ADVANCES 2019; 5:eaax1532. [PMID: 31663022 PMCID: PMC6795516 DOI: 10.1126/sciadv.aax1532] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/25/2019] [Indexed: 06/01/2023]
Abstract
Cocaine use continues to be a serious worldwide public health problem. Cocaine abuse is associated with substantial morbidity and mortality. Cocaine overdose deaths are increasing in the United States and, in certain populations, outnumber heroin and opiate overdose deaths. Psychosocial treatments remain the treatments of choice for cocaine use disorder (CUD), with standard approaches including contingency management and cognitive behavioral therapy. However, the effect sizes of these treatments are not large, and they are not effective for most patients. Consequently, investigators have sought to develop pharmacological agents to augment the efficacy of psychosocial treatments. Despite these efforts, no medications have yet been proven to be safe and effective for the treatment of CUD. The most promising pharmacological strategies for CUD treatment thus far include the use of dopamine agonists, such as long-acting amphetamine and modafinil or glutamatergic and GABAergic agents such as topiramate. Combination drugs may be especially promising.
Collapse
Affiliation(s)
- Kyle M Kampman
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, 3535 Market Street, Suite 500, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
In search of optimal psychoactivation: stimulants as cognitive performance enhancers. Arh Hig Rada Toksikol 2019; 70:150-159. [PMID: 32597132 DOI: 10.2478/aiht-2019-70-3298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022] Open
Abstract
An increasing number of people, students in particular, seek substances that improve their cognitive functioning. The most popular group of pharmacological cognitive enhancers (PCEs) are stimulants. Available studies suggest a small beneficial effect of methylphenidate and amphetamine on memory, executive functions, and processing speed. However small, this effect can make the difference between success and failure. In recent years, research has focused on the additional beneficial effect on the emotional state, increased motivation, and placebo-induced cognitive enhancement. This paper briefly reviews the latest and most important research on the relationship between popular stimulants and cognitive enhancement. One cannot understand this relationship without understanding the Yerkes-Dodson law, which explains the relationship between the degree of arousal and performance. It suggests that the effect of stimulants is a dose-dependent continuum. This law has repeatedly been confirmed by studies in which an optimal level of psychoactivation for cognitive enhancement was obtained with low stimulant doses, whereas exceeding the effective dose resulted in cognitive deficits, psychomotor agitation, and addiction. A separate section has been devoted to modafinil, an increasingly popular stimulant that differs from the rest in neurochemical profile and behavioural effects.
Collapse
|
33
|
Keighron JD, Giancola JB, Shaffer RJ, DeMarco EM, Coggiano MA, Slack RD, Hauck Newman A, Tanda G. Distinct effects of (R)-modafinil and its (R)- and (S)-fluoro-analogs on mesolimbic extracellular dopamine assessed by voltammetry and microdialysis in rats. Eur J Neurosci 2019; 50:2045-2053. [PMID: 30402972 PMCID: PMC8294075 DOI: 10.1111/ejn.14256] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Abstract
Psychostimulant use disorders remain an unabated public health concern worldwide, but no FDA approved medications are currently available for treatment. Modafinil (MOD), like cocaine, is a dopamine reuptake inhibitor and one of the few drugs evaluated in clinical trials that has shown promise for the treatment of cocaine or methamphetamine use disorders in some patient subpopulations. Recent structure-activity relationship and preclinical studies on a series of MOD analogs have provided insight into modifications of its chemical structure that may lead to advancements in clinical efficacy. Here, we have tested the effects of the clinically available (R)-enantiomer of MOD on extracellular dopamine levels in the nucleus accumbens shell, a mesolimbic dopaminergic projection field that plays significant roles in various aspects of psychostimulant use disorders, measured in vivo by fast-scan cyclic voltammetry and by microdialysis in Sprague-Dawley rats. We have compared these results with those obtained under identical experimental conditions with two novel and enantiopure bis(F) analogs of MOD, JBG1-048 and JBG1-049. The results show that (R)-modafinil (R-MOD), JBG1-048, and JBG1-049, when administered intravenously with cumulative drug-doses, will block the dopamine transporter and reduce the clearance rate of dopamine, increasing its extracellular levels. Differences among the compounds in their maximum stimulation of dopamine levels, and in their time course of effects were also observed. These data highlight the mechanistic underpinnings of R-MOD and its bis(F) analogs as pharmacological tools to guide the discovery of novel medications to treat psychostimulant use disorders.
Collapse
Affiliation(s)
- Jacqueline D. Keighron
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services; 333 Cassell Drive, TRIAD Bldg., Baltimore, MD, USA, 21224
| | - JoLynn B. Giancola
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services; 333 Cassell Drive, TRIAD Bldg., Baltimore, MD, USA, 21224
| | - Rachel J. Shaffer
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services; 333 Cassell Drive, TRIAD Bldg., Baltimore, MD, USA, 21224
| | - Emily M. DeMarco
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services; 333 Cassell Drive, TRIAD Bldg., Baltimore, MD, USA, 21224
| | - Mark A. Coggiano
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services; 333 Cassell Drive, TRIAD Bldg., Baltimore, MD, USA, 21224
| | - Rachel D. Slack
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services; 333 Cassell Drive, TRIAD Bldg., Baltimore, MD, USA, 21224
| | - Amy Hauck Newman
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services; 333 Cassell Drive, TRIAD Bldg., Baltimore, MD, USA, 21224
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services; 333 Cassell Drive, TRIAD Bldg., Baltimore, MD, USA, 21224
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services; 333 Cassell Drive, TRIAD Bldg., Baltimore, MD, USA, 21224
| |
Collapse
|
34
|
Newman AH, Cao J, Keighron JD, Jordan CJ, Bi GH, Liang Y, Abramyan AM, Avelar AJ, Tschumi CW, Beckstead MJ, Shi L, Tanda G, Xi ZX. Translating the atypical dopamine uptake inhibitor hypothesis toward therapeutics for treatment of psychostimulant use disorders. Neuropsychopharmacology 2019; 44:1435-1444. [PMID: 30858517 PMCID: PMC6785152 DOI: 10.1038/s41386-019-0366-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 11/10/2022]
Abstract
Medication-assisted treatments are unavailable to patients with cocaine use disorders. Efforts to develop potential pharmacotherapies have led to the identification of a promising lead molecule, JJC8-091, that demonstrates a novel binding mode at the dopamine transporter (DAT). Here, JJC8-091 and a structural analogue, JJC8-088, were extensively and comparatively assessed to elucidate neurochemical correlates to their divergent behavioral profiles. Despite sharing significant structural similarity, JJC8-088 was more cocaine-like, increasing extracellular DA concentrations in the nucleus accumbens shell (NAS) efficaciously and more potently than JJC8-091. In contrast, JJC8-091 was not self-administered and was effective in blocking cocaine-induced reinstatement to drug seeking. Electrophysiology experiments confirmed that JJC8-091 was more effective than JJC8-088 at inhibiting cocaine-mediated enhancement of DA neurotransmission. Further, when VTA DA neurons in DAT-cre mice were optically stimulated, JJC8-088 produced a significant leftward shift in the stimulation-response curve, similar to cocaine, while JJC8-091 shifted the curve downward, suggesting attenuation of DA-mediated brain reward. Computational models predicted that JJC8-088 binds in an outward facing conformation of DAT, similar to cocaine. Conversely, JJC8-091 steers DAT towards a more occluded conformation. Collectively, these data reveal the underlying molecular mechanism at DAT that may be leveraged to rationally optimize leads for the treatment of cocaine use disorders, with JJC8-091 representing a compelling candidate for development.
Collapse
Affiliation(s)
- Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| | - Jianjing Cao
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Jacqueline D. Keighron
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Chloe J. Jordan
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Guo-Hua Bi
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Ying Liang
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Ara M. Abramyan
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Alicia J. Avelar
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX USA
| | - Christopher W. Tschumi
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX USA ,0000 0000 8527 6890grid.274264.1Aging & Metabolism Research Group, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Michael J. Beckstead
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX USA ,0000 0000 8527 6890grid.274264.1Aging & Metabolism Research Group, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Lei Shi
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Gianluigi Tanda
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Zheng-Xiong Xi
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| |
Collapse
|
35
|
Mahoney JJ. Cognitive dysfunction in individuals with cocaine use disorder: Potential moderating factors and pharmacological treatments. Exp Clin Psychopharmacol 2019; 27:203-214. [PMID: 30556731 PMCID: PMC6538444 DOI: 10.1037/pha0000245] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
It is well-documented in the literature that individuals repeatedly exposed to cocaine exhibit cognitive impairment and that cognitive dysfunction is a risk factor for poor treatment outcomes in those with cocaine use disorder (CUD). Specific deficits related to attention, episodic memory, working memory, and executive functioning are the most common deficits noted in this population. Given that cognitive impairment is a risk factor for poor treatment outcomes in those with CUD, identifying possible moderating factors contributing to and/or exacerbating cocaine-related cognitive deficits is of great importance. Some of these factors may include premorbid intellectual functioning, cocaine use patterns, polysubstance use, comorbid emotional symptoms, and sleep dysfunction. It is plausible that by identifying moderating factors impacting cognition, behavioral interventions can then be modified accordingly and/or treatment regimens can be augmented with pharmacological interventions (e.g., cognitive enhancing agents), leading to a reduction in treatment attrition and improved treatment outcomes. The currently available treatments for CUD are mainly behavioral with variable efficacy, and even though there have been great preclinical and clinical research efforts focused on medication development for CUD, there are currently no Food and Drug Administration-approved medications for CUD. A description of some of the several potential moderating factors, along with some pharmacological treatments which have been shown to ameliorate, at least to some extent, cognitive dysfunction in those with CUD are discussed. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
|
36
|
Keighron JD, Quarterman JC, Cao J, DeMarco EM, Coggiano MA, Gleaves A, Slack RD, Zanettini C, Newman AH, Tanda G. Effects of ( R)-Modafinil and Modafinil Analogues on Dopamine Dynamics Assessed by Voltammetry and Microdialysis in the Mouse Nucleus Accumbens Shell. ACS Chem Neurosci 2019; 10:2012-2021. [PMID: 30645944 DOI: 10.1021/acschemneuro.8b00340] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent discoveries have improved our understanding of the physiological and pathological roles of the dopamine transporter (DAT); however, only a few drugs are clinically available for DAT-implicated disorders. Among those drugs, modafinil (MOD) and its ( R)-enantiomer (R-MOD) have been used off-label as therapies for psychostimulant use disorders, but they have shown limited effectiveness in clinical trials. Recent preclinical studies on MOD and R-MOD have led to chemically modified structures aimed toward improving their neurobiological properties that might lead to more effective therapeutics for stimulant use disorders. This study examines three MOD analogues (JJC8-016, JJC8-088, and JJC8-091) with improved DAT affinities compared to their parent compound. These compounds were investigated for their effects on the neurochemistry (brain microdialysis and FSCV) and behavior (ambulatory activity) of male Swiss-Webster mice. Our data indicate that these compounds have dissimilar effects on tonic and phasic dopamine in the nucleus accumbens shell and variability in producing ambulatory activity. These results suggest that small changes in the chemical structure of a DAT inhibitor can cause compounds such as JJC8-088 to produce effects similar to abused psychostimulants like cocaine. In contrast, other compounds like JJC8-091 do not share cocaine-like effects and have a more atypical DAT-inhibitor profile, which may prove to be an advancement in the treatment of psychostimulant use disorders.
Collapse
|
37
|
Nicholson HL, Ford JA. Sociodemographic, neighborhood, psychosocial, and substance use correlates of cocaine use among Black adults: Findings from a pooled analysis of national data. Addict Behav 2019; 88:182-186. [PMID: 30218941 DOI: 10.1016/j.addbeh.2018.08.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 08/31/2018] [Accepted: 08/31/2018] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Cocaine use (CU) is serious a public health issue affecting U.S. adults, including Blacks. Cocaine-related overdose deaths have also trended upwards among this population. However, there remains a lack of research on correlates of CU among a nationally representative sample of Black adults. METHODS The current study examines the prevalence and correlates of past-year CU among Black adults aged 18 and older (N = 9,821). Data from the National Survey on Drug Use and Health (2015-2016) were used and a weighted logistic regression model was estimated. RESULTS Findings showed 2.4% of Black adults reported past-year CU-significantly higher than rates of CU among other assessed racial groups. Opioid use/misuse, encounters with drug dealers, easier access to cocaine, unemployment, and being 35 or older were associated with increased odds of CU. Greater risk perception of CU and religiosity were associated decreased odds of CU. CONCLUSION Results identified several correlates associated with past-year CU among Black adults. Findings suggest addressing CU among this population will likely require the development of multilevel prevention and intervention strategies and an increased focus on opioid use/misuse as opioids have been recently implicated in cocaine-related overdose deaths.
Collapse
Affiliation(s)
| | - Jason A Ford
- University of Central Florida, Department of Sociology, United States
| |
Collapse
|
38
|
Zanettini C, Scaglione A, Keighron JD, Giancola JB, Lin SC, Newman AH, Tanda G. Pharmacological classification of centrally acting drugs using EEG in freely moving rats: an old tool to identify new atypical dopamine uptake inhibitors. Neuropharmacology 2018; 161:107446. [PMID: 30481526 DOI: 10.1016/j.neuropharm.2018.11.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/05/2018] [Accepted: 11/23/2018] [Indexed: 10/27/2022]
Abstract
Atypical dopamine uptake inhibitors (DUIs) bind to the dopamine transporter and inhibit the reuptake of dopamine but have lower abuse potential than psychostimulants. Several atypical DUIs can block abuse-related effects of cocaine and methamphetamine, thus making them potential medication candidates for psychostimulant use disorders. The aim of the current study is to establish an in-vivo assay using EEG for the rapid identification of atypical DUIs with potential for medication development. The typical DUIs cocaine and methylphenidate dose-dependently decreased the power of the alpha, beta, and gamma bands. The atypical DUI modafinil and its F-analog, JBG1-049, decreased the power of beta, but in contrast to cocaine, none of the other frequency bands, while JHW007 did not significantly alter the EEG spectrum. The mu-opioid receptor agonists heroin and morphine dose-dependently decreased the power of gamma and increased power of the other bands. The effect of morphine on EEG power bands was antagonized by naltrexone. The NMDA receptor antagonist ketamine increased the power of all frequency bands. Therefore, typical and atypical DUIs and drugs of other classes differentially affected EEG spectra, showing distinctive features in the magnitude and direction of their effects on EEG. Comparative analysis of the effects of test drugs on EEG indicates a potential atypical profile of JBG1-049 with similar potency and effectiveness to its parent compound modafinil. These data suggest that EEG can be used to rapidly screen compounds for potential activity at specific pharmacological targets and provide valuable information for guiding the early stages of drug development. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- Claudio Zanettini
- Medication Development Program, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA.
| | - Alessandro Scaglione
- Neural Circuits and Cognition Unit, Laboratory of Behavioral Neuroscience, NIA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - Jacqueline D Keighron
- Medication Development Program, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - JoLynn B Giancola
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - Shih-Chieh Lin
- Neural Circuits and Cognition Unit, Laboratory of Behavioral Neuroscience, NIA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - Amy H Newman
- Medication Development Program, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA; Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA
| |
Collapse
|
39
|
Penna LMDC, Düsman E, Vicentini VEP. The mutagenic effect of psychostimulant modafinil in Wistar rats in vivo. Drug Chem Toxicol 2018; 43:373-377. [PMID: 30293447 DOI: 10.1080/01480545.2018.1517771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The demands imposed by today's world require a fast and efficient society, then, significant section of the population looks for support from psychotropic medicine. Modafinil is a psychostimulant that promotes wakefulness, it being recommended for treatment of narcolepsy, obstructive sleep apnea, and shift-work sleep disorder, besides being a cognitive function potentiator. However, chemical components of drugs can alter genetic material. Thus, the present study evaluated the cytotoxic and clastogenic/mutagenic potential of 0.25, 0.50, and 0.75 mg of Modafinil/mL of corn oil/100g body weight in acute treatments and subacute treatments, 15 days, to Rattus norvegicus, treated via gavage in a single daily dose. The drug was not cytotoxic at any of the evaluated doses in either of the treatments. However, the medicine showed clastogenic/mutagenic activity in the acute treatment group at the standard dose and at double dose. Data from the present study indicates that there should be greater caution as to the use of this psychostimulant by human beings.
Collapse
Affiliation(s)
| | - Elisângela Düsman
- Academic Department of Chemistry and Biology, Federal Technological University of Paraná, Francisco Beltrão, Paraná, Brazil
| | | |
Collapse
|
40
|
Lacoste J, Michel G, Rollier S, Charles-Nicolas A. Cocaïne et cocaïnomanie. Enseignements récents et stratégies actuelles. ANNALES MEDICO-PSYCHOLOGIQUES 2018. [DOI: 10.1016/j.amp.2018.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
41
|
Walker LC, Lawrence AJ. Investigational drug therapies in phase I and phase II clinical trials for alcohol use disorders. Expert Opin Investig Drugs 2018; 27:1-14. [PMID: 30019949 DOI: 10.1080/13543784.2018.1502269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Alcohol use disorder (AUD) is a complex psychiatric condition characterized by craving, compulsive seeking, loss of control of alcohol consumption as well as the emergence of negative emotional states during withdrawal. Despite the large socioeconomic burden of AUD, therapeutic treatment options lag behind. AREAS COVERED This review covers pharmacotherapies currently in phase I/II clinical trials for the treatment of AUDs listed on clinicaltrials.gov. We discuss drug therapies that modulate monoamine, GABA/Glutamate, neuropeptide and neuroimmune systems. We examine in depth preclinical and clinical evidence of a select range of these compounds and consider their utility in treating AUDs. EXPERT OPINION Current therapeutic options to treat AUD are inadequate at a population level. Currently there are 30 different compounds and one compound combination in phase I/II clinical trials for AUD. These compounds target various aspects of neurotransmitter signaling, neuroimmune modulation, and alcohol metabolism. Almost 75% of these compounds under trial are Food and Drug Administration (FDA) approved for other indications, which may save time and costs in treatment development. Further, development of therapeutics focused on genetic biomarkers and behavioral screening may improve how treatment decisions are made in the future on a case-by-case basis.
Collapse
Affiliation(s)
- Leigh C Walker
- a Florey Department of Neuroscience and Mental Health , University of Melbourne , Parkville , VIC , Australia
| | - Andrew J Lawrence
- a Florey Department of Neuroscience and Mental Health , University of Melbourne , Parkville , VIC , Australia
| |
Collapse
|
42
|
Lai S, Heaphy CM, Rizzo AJ, Celentano DD, Gerstenblith G, Li J, Moore RD, Treisman G, Chen S, Foster P, Kickler T, Lai H. Cocaine use may induce telomere shortening in individuals with HIV infection. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:11-17. [PMID: 29410247 PMCID: PMC5880737 DOI: 10.1016/j.pnpbp.2018.01.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/14/2018] [Accepted: 01/22/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Although cocaine use may induce/accelerate HIV-associated comorbidities in HIV-infected individuals on antiretroviral therapy (ART), and that HIV itself may accelerate aging, the issue of whether cocaine use plays a role in HIV-associated aging in HIV-infected cocaine users has not been reported. The goals of this study were (1) to explore factor(s) associated with peripheral blood leukocyte telomere length, a marker of cellular replicative history, and telomere shortening in HIV-infected individuals, and (2) to assess whether cocaine use plays a role in accelerating telomere shortening in cocaine users with HIV infection. METHODS Between June 2010 and December 2016, 147 HIV-infected participants in Baltimore, Maryland, were enrolled in a cross-sectional study investigating factor(s) associated with telomere length. Of these 147, 93 participated in a follow-up study to examine factor(s) associated with telomere shortening. Robust regression model was used to analyze cross-sectional data and the generalized estimating equation approach was used to analyze follow-up data. RESULTS Cross-sectional analyses demonstrated that (1) both daily alcohol consumption and use of non-nucleoside reverse transcriptase inhibitors (NNRTIs) were independently associated with telomere length, and cocaine use modified the associations of daily alcohol use and NNRTI use with telomere length. Longitudinal analyses suggested that both daily alcohol consumption and duration of NNRTI use were independently associated with telomere shortening, and (2) cocaine use induced/accelerated telomere shortening in HIV-infected individuals. CONCLUSIONS Our findings suggest that cocaine use may promote premature aging in HIV-infected individuals who are on ART. Our results emphasize the importance of cocaine abstinence/reduced use, which may retard HIV-associated premature aging.
Collapse
Affiliation(s)
- Shenghan Lai
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | | | - Anthony J. Rizzo
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - David D. Celentano
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Gary Gerstenblith
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ji Li
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Richard D. Moore
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Glenn Treisman
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shaoguang Chen
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Parker Foster
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thomas Kickler
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hong Lai
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
43
|
Schmitz JM, Stotts AL, Vujanovic AA, Weaver MF, Yoon JH, Vincent J, Green CE. A sequential multiple assignment randomized trial for cocaine cessation and relapse prevention: Tailoring treatment to the individual. Contemp Clin Trials 2018; 65:109-115. [PMID: 29287664 PMCID: PMC5803345 DOI: 10.1016/j.cct.2017.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/23/2017] [Accepted: 12/24/2017] [Indexed: 10/18/2022]
Abstract
Drug addiction is a chronic, devastating, but treatable disorder. A core principle of drug addiction treatment states that no single treatment is appropriate for everyone (NIDA, 2012); treatments need to adjust based on patient characteristics and response in order to be maximally effective. For cocaine use disorders (CUD), specifically, the most potent intervention currently available for initiating abstinence is behavior therapy using contingency management (CM) procedures, with early cessation being a robust predictor of future abstinence. This raises two key questions for treatment development research: First, can we significantly improve initial CM response rates with targeted adjunctive interventions? Second, for individuals who fail to achieve initial abstinence with CM, is pharmacotherapy an effective augmentation strategy? This paper describes how a sequential, multiple assignment, randomized trial (SMART) design has advantages over a fixed-intervention approach when it comes to collecting data needed to answer both questions. The first aim will examine whether Acceptance and Commitment Therapy (ACT) in combination with CM increases initial abstinence response rates (i.e., 2 consecutive weeks of cocaine-negative urine screens). The second aim will examine whether ACT+CM in combination with modafinil promotes abstinence achievement in initial non-responders. Results are expected to inform how we tailor treatment of CUD to maximize outcomes.
Collapse
Affiliation(s)
- Joy M Schmitz
- Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States.
| | - Angela L Stotts
- Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States; Department of Family and Community Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | | | - Michael F Weaver
- Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Jin H Yoon
- Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Jessica Vincent
- Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Charles E Green
- Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States; UT-Houston Center for Clinical Research & Evidence-Based Medicine, United States
| |
Collapse
|
44
|
Tunstall BJ, Ho CP, Cao J, Vendruscolo JCM, Schmeichel BE, Slack RD, Tanda G, Gadiano AJ, Rais R, Slusher BS, Koob GF, Newman AH, Vendruscolo LF. Atypical dopamine transporter inhibitors attenuate compulsive-like methamphetamine self-administration in rats. Neuropharmacology 2017; 131:96-103. [PMID: 29217282 DOI: 10.1016/j.neuropharm.2017.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 11/30/2022]
Abstract
Methamphetamine (METH) is a highly addictive drug, but no pharmacological treatment is yet available for METH use disorders. Similar to METH, the wake-promoting drug (R)-modafinil (R-MOD) binds to the dopamine transporter (DAT). Unlike METH, R-MOD is not a substrate for transport by DAT and has low abuse potential. We tested the hypothesis that the atypical DAT inhibitor R-MOD and compounds that are derived from modafinil would decrease METH intake by reducing the actions of METH at the DAT. We tested the effects of systemic injections of R-MOD and four novel modafinil-derived ligands with increased DAT affinity (JJC8-016, JJC8-088, JJC8-089, and JJC8-091) on intravenous (i.v.) METH self-administration in rats that were allowed short access (ShA; 1 h) or long access (LgA; 6 h) to the drug. ShA rats exhibited stable METH intake over sessions, whereas LgA rats exhibited an escalation of drug intake. R-MOD decreased METH self-administration in ShA and LgA rats (in the 1st hour only). JJC8-091 and JJC8-016 decreased METH self-administration in both ShA and LgA rats. JJC8-089 decreased METH self-administration in LgA rats only, whereas JJC8-088 had no effect on METH self-administration in either ShA or LgA rats. These findings support the potential of atypical DAT inhibitors for the treatment of METH use disorders and suggest several novel compounds as candidate drugs.
Collapse
Affiliation(s)
- Brendan J Tunstall
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Chelsea P Ho
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Janaína C M Vendruscolo
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Brooke E Schmeichel
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Rachel D Slack
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Gianluigi Tanda
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Alexandra J Gadiano
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA; Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara S Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Amy H Newman
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA.
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
45
|
Trends and correlates of cocaine use and cocaine use disorder in the United States from 2011 to 2015. Drug Alcohol Depend 2017; 180:376-384. [PMID: 28961544 PMCID: PMC5828499 DOI: 10.1016/j.drugalcdep.2017.08.031] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 08/25/2017] [Accepted: 08/25/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND Recent epidemiological data suggest a resurgence in cocaine use (CU) and cocaine-related problems in the United States. Demographic trends and correlates of problem CU are needed to determine potential factors that may be influencing the increased trend and to inform targeted prevention and intervention strategies. METHODS Trends in any past-year CU, weekly CU, and cocaine use disorder (CUD) were examined among persons aged ≥12 years using the National Survey on Drug Use and Health from 2011 to 2015. Logistic regression analyses were used to determine correlates of past-year and weekly CU and CUD among adolescents and adults. RESULTS The prevalence of past-year CU from 2011 to 2015 increased among females, ages 18-25, ages ≥50, non-Hispanic Blacks, and persons reporting low income, past-year tobacco use, past-year alcohol use, and past-month binge and heavy alcohol use. The prevalence of weekly CU increased among persons aged ≥50 years and persons reporting past-month heavy alcohol use. A significant increase in the prevalence of CUD was only found among persons aged ≥50 years. Adjusted logistic regression showed that older age, large metropolitan residence, past-year tobacco, alcohol, cannabis, and heroin use, and major depressive episode were associated with increased odds of CU or CUD among both adolescents and adults; however, sex and race/ethnicity correlates differed among adolescents and adults. CONCLUSIONS Findings have implications for increased monitoring of CU-related indicators among some high-risk groups, such as females, older adults, Blacks, and polysubstance users. Targeted screening and intervention strategies among these population subgroups may be needed.
Collapse
|
46
|
Avelar AJ, Cao J, Newman AH, Beckstead MJ. Atypical dopamine transporter inhibitors R-modafinil and JHW 007 differentially affect D2 autoreceptor neurotransmission and the firing rate of midbrain dopamine neurons. Neuropharmacology 2017; 123:410-419. [PMID: 28625719 PMCID: PMC5546153 DOI: 10.1016/j.neuropharm.2017.06.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/17/2017] [Accepted: 06/14/2017] [Indexed: 12/12/2022]
Abstract
Abuse of psychostimulants like cocaine that inhibit dopamine (DA) reuptake through the dopamine transporter (DAT) represents a major public health issue, however FDA-approved pharmacotherapies have yet to be developed. Recently a class of ligands termed "atypical DAT inhibitors" has gained attention due to their range of effectiveness in increasing extracellular DA levels without demonstrating significant abuse liability. These compounds not only hold promise as therapeutic agents to treat stimulant use disorders but also as experimental tools to improve our understanding of DAT function. Here we used patch clamp electrophysiology in mouse brain slices to explore the effects of two atypical DAT inhibitors (R-modafinil and JHW 007) on the physiology of single DA neurons in the substantia nigra and ventral tegmental area. Despite their commonalities of being DAT inhibitors that lack cocaine-like behavioral profiles, these compounds exhibited surprisingly divergent cellular effects. Similar to cocaine, R-modafinil slowed DA neuron firing in a D2 receptor-dependent manner and rapidly enhanced the amplitude and duration of D2 receptor-mediated currents in the midbrain. In contrast, JHW 007 exhibited little effect on firing, slow DAT blockade, and an unexpected inhibition of D2 receptor-mediated currents that may be due to direct D2 receptor antagonism. Furthermore, pretreatment with JHW 007 blunted the cellular effects of cocaine, suggesting that it may be valuable to investigate similar DAT inhibitors as potential therapeutic agents. Further exploration of these and other atypical DAT inhibitors may reveal important cellular effects of compounds that will have potential as pharmacotherapies for treating cocaine use disorders.
Collapse
Affiliation(s)
- Alicia J Avelar
- Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX, 78229, USA.
| | - Jianjing Cao
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Michael J Beckstead
- Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX, 78229, USA.
| |
Collapse
|
47
|
Sangroula D, Motiwala F, Wagle B, Shah VC, Hagi K, Lippmann S. Modafinil Treatment of Cocaine Dependence: A Systematic Review and Meta-Analysis. Subst Use Misuse 2017; 52:1292-1306. [PMID: 28350194 DOI: 10.1080/10826084.2016.1276597] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Currently, there is none FDA-approved medication to treat cocaine dependency. Studies conducted with various medications, including antipsychotics, antidepressants, anticonvulsants, and others, revealed inconsistent results. OBJECTIVES To meta-analytically investigate the efficacy and safety of modafinil in the treatment of cocaine-dependent patients. METHODS Randomized controlled trials with ≥20 subjects comparing the numerical therapeutic outcomes of modafinil with placebo were identified in databases, such as PUBMED, psycINFO, EMBASE, and Clinicaltrials.gov. Relevant data on efficacy and safety were extracted. Relative risk (RR) and standardized mean difference were applied for reporting dichotomous and continuous outcomes respectively. Random effects, subgroup, and meta-regression analyses were conducted to further explore the results and evaluate for any moderators. RESULTS In total, 11 studies (participants = 896, duration = 6.7 ± 1.9 weeks) comparing modafinil with placebo were systematically analyzed, which indicated that modafinil was not superior to placebo in improving the treatment retention rate (studies = 11, participants = 891, RR = 1.030, 95% CI = 0.918-1.156, p = .613). Similarly, data from 7/11 studies did not evidence superiority of modafinil in achieving cocaine abstinence (participants = 696, RR = 1.259, 95% CI = 0.813-1.949, p = .302). However, subgroup analysis of six studies conducted in the United States demonstrated superiority of modafinil in cocaine abstinence rate (studies = 6, participants = 669, 95% CI = 1.027-2.020, p = 0.035). In addition, no evidence suggested modafinil-related discontinuation or specific adverse events than placebo. CONCLUSIONS Overall, there is no evidence to conclude superiority of modafinil in increasing cocaine abstinence and treatment retention rate. However, promising result in subgroup analysis of cocaine abstinence, secondary outcomes, and good safety profile urged the need of larger studies to derive more conclusive results.
Collapse
Affiliation(s)
- Dinesh Sangroula
- a Department of Psychiatry , Jamaica Hospital Medical Center , New York , New York , USA
| | - Fatima Motiwala
- b Department of Psychiatric Research , Columbia University Medical Center , New York , New York , USA
| | - Bivek Wagle
- c Department of Biology , California State University , Hayward , California , USA
| | - Vivek C Shah
- d Department of Psychiatry , Nassau University Medical Center , East Meadow , New York , USA
| | | | - Steven Lippmann
- f Department of Psychiatry , University of Louisville School of Medicine , Louisville , Kentucky , USA
| |
Collapse
|
48
|
The Novel Modafinil Analog, JJC8-016, as a Potential Cocaine Abuse Pharmacotherapeutic. Neuropsychopharmacology 2017; 42:1871-1883. [PMID: 28266501 PMCID: PMC5564383 DOI: 10.1038/npp.2017.41] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/15/2017] [Accepted: 02/20/2017] [Indexed: 12/22/2022]
Abstract
(±)Modafinil ((±)MOD) and its R-enantiomer (R-modafinil; R-MOD) have been investigated for their potential as treatments for psychostimulant addiction. We recently reported a series of (±)MOD analogs, of which JJC8-016 (N-(2-((bis(4-fluorophenyl)methyl)thio)ethyl)-3-phenylpropan-1-amine) was selected for further development. JJC8-016 and R-MOD were evaluated for binding across ~70 receptors, transporters, and enzymes. Although at a concentration of 10 μM, there were many hits for JJC8-016, binding affinities in the range of its DAT affinity were only observed at the serotonin transporter (SERT), dopamine D2-like, and sigma1 receptors. R-MOD was more selective, but had much lower affinity at the DAT (Ki=3 μM) than JJC8-016 (Ki=116 nM). In rats, systemic administration of R-MOD alone (10-30 mg/kg i.p.) dose-dependently increased locomotor activity and electrical brain-stimulation reward, whereas JJC8-016 (10-30 mg/kg i.p.) did not produce these effects. Strikingly, pretreatment with JJC8-016 dose-dependently inhibited cocaine-enhanced locomotion, cocaine self-administration, and cocaine-induced reinstatement of drug-seeking behavior, whereas R-MOD inhibited cocaine-induced reinstatement only at the high dose of 100 mg/kg. Notably, JJC8-016 alone neither altered extracellular dopamine in the nucleus accumbens nor maintained self-administration. It also failed to induce reinstatement of drug-seeking behavior. These findings suggest that JJC8-016 is a unique DAT inhibitor that has no cocaine-like abuse potential by itself. Moreover, pretreatment with JJC8-016 significantly inhibits cocaine-taking and cocaine-seeking behavior likely by interfering with cocaine binding to DAT. In addition, off-target actions may also contribute to its potential therapeutic utility in the treatment of cocaine abuse.
Collapse
|
49
|
Dowling G, Kavanagh PV, Talbot B, O’Brien J, Hessman G, McLaughlin G, Twamley B, Brandt SD. Outsmarted by nootropics? An investigation into the thermal degradation of modafinil, modafinic acid, adrafinil, CRL-40,940 and CRL-40,941 in the GC injector: formation of 1,1,2,2-tetraphenylethane and its tetra fluoro analog. Drug Test Anal 2016; 9:518-528. [DOI: 10.1002/dta.2142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/26/2016] [Accepted: 11/26/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Geraldine Dowling
- School of Chemical and Pharmaceutical Sciences; Dublin Institute of Technology; Kevin Street Dublin D08 NF82 Ireland
- Department of Pharmacology and Therapeutics; School of Medicine, Trinity Centre for Health Sciences, St. James's Hospital; James's Street Dublin D08 W9RT Ireland
| | - Pierce V. Kavanagh
- Department of Pharmacology and Therapeutics; School of Medicine, Trinity Centre for Health Sciences, St. James's Hospital; James's Street Dublin D08 W9RT Ireland
| | - Brian Talbot
- School of Pharmacy and Pharmaceutical Sciences; Trinity College Dublin, College Green; Dublin D02 EV57 Ireland
| | - John O’Brien
- School of Chemistry, Trinity College Dublin, College Green; Dublin D02 EV57 Ireland
| | - Gary Hessman
- School of Chemistry, Trinity College Dublin, College Green; Dublin D02 EV57 Ireland
| | - Gavin McLaughlin
- Department of Pharmacology and Therapeutics; School of Medicine, Trinity Centre for Health Sciences, St. James's Hospital; James's Street Dublin D08 W9RT Ireland
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, College Green; Dublin D02 EV57 Ireland
| | - Simon D. Brandt
- School of Pharmacy and Biomolecular Sciences; Liverpool John Moores University; Byrom Street Liverpool L3 3AF UK
| |
Collapse
|
50
|
Cao J, Slack RD, Bakare OM, Burzynski C, Rais R, Slusher BS, Kopajtic T, Bonifazi A, Ellenberger MP, Yano H, He Y, Bi GH, Xi ZX, Loland CJ, Newman AH. Novel and High Affinity 2-[(Diphenylmethyl)sulfinyl]acetamide (Modafinil) Analogues as Atypical Dopamine Transporter Inhibitors. J Med Chem 2016; 59:10676-10691. [PMID: 27933960 DOI: 10.1021/acs.jmedchem.6b01373] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The development of pharmacotherapeutic treatments of psychostimulant abuse has remained a challenge, despite significant efforts made toward relevant mechanistic targets, such as the dopamine transporter (DAT). The atypical DAT inhibitors have received attention due to their promising pharmacological profiles in animal models of cocaine and methamphetamine abuse. Herein, we report a series of modafinil analogues that have an atypical DAT inhibitor profile. We extended SAR by chemically manipulating the oxidation states of the sulfoxide and the amide functional groups, halogenating the phenyl rings, and/or functionalizing the terminal nitrogen with substituted piperazines, resulting in several novel leads such as 11b, which demonstrated high DAT affinity (Ki = 2.5 nM) and selectivity without producing concomitant locomotor stimulation in mice, as compared to cocaine. These results are consistent with an atypical DAT inhibitor profile and suggest that 11b may be a potential lead for development as a psychostimulant abuse medication.
Collapse
Affiliation(s)
- Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Rachel D Slack
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Oluyomi M Bakare
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Caitlin Burzynski
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States.,Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine , 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine , 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Barbara S Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine , 855 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Theresa Kopajtic
- Psychobiology Section, Molecular Neuropsychiatry Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 251 Bayview Boulevard, Baltimore, Maryland 21224, United States
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Michael P Ellenberger
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Hideaki Yano
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Yi He
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Guo-Hua Bi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Claus J Loland
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen , DK-2200 Copenhagen, Denmark
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| |
Collapse
|