1
|
Martin CE, Bjork JM, Keyser-Marcus L, Sabo RT, Pignatello T, Simmons K, La Rosa C, Arias AJ, Ramey T, Moeller FG. Phase 1b/2a safety study of lemborexant as an adjunctive treatment for insomnia to buprenorphine-naloxone for opioid use disorder: A randomized controlled trial. DRUG AND ALCOHOL DEPENDENCE REPORTS 2025; 14:100304. [PMID: 39807348 PMCID: PMC11728975 DOI: 10.1016/j.dadr.2024.100304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/07/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025]
Abstract
Background Evidence supports the common incidence of sleep disturbance in opioid use disorder (OUD) as a potential marker of disrupted orexin system functioning. This study evaluated the initial safety and tolerability of a challenge dose of lemborexant, a dual orexin antagonist, as an adjunct to buprenorphine/naloxone. Methods Patients (18-65 years old) with OUD receiving sublingual buprenorphine/naloxone, with a Pittsburgh Sleep Quality Index total score of 6 or higher, were recruited from outpatient clinics. After randomization, while being monitored on an inpatient research unit over two 10-hour daytime periods, participants received a placebo or lemborexant (5 mg on day one and 10 mg on day two) along with buprenorphine/naloxone. Primary outcomes included safety and tolerability: adverse events, physiologic measures, sedation level assessments. Generalized linear mixed model analysis assessed the effect of study drug and time on outcomes. Results N=18 (14=male, 4=female) were randomized to lemborexant (n=12) or placebo (n=6). No unanticipated problems occurred; five adverse events occurred in the lemborexant group and two in the placebo group with no serious adverse events. None of the physiologic measures showed a significant interaction of time and placebo vs. lemborexant (5 or 10 mg): Pulse oximetry (F=0.6; p=0.84), End-tidal CO2 (F=0.5; p=0.91), Heart rate (F=0.6; p=0.82), Systolic blood pressure (F=0.7; p=0.73), Diastolic blood pressure (F=2.0; p=0.06). At 9 hours after study drug administration, all participants returned to baseline sedation levels and were discharged. Conclusions Findings support the initial safety and tolerability of lemborexant as an adjunctive treatment for insomnia in humans receiving buprenorphine for OUD. Future longitudinal work is warranted with larger samples.
Collapse
Affiliation(s)
- Caitlin E. Martin
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA 23219, USA
| | - James M. Bjork
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA 23219, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Lori Keyser-Marcus
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA 23219, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Roy T. Sabo
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Tiffany Pignatello
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA 23219, USA
| | - Kameron Simmons
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA 23219, USA
| | - Christina La Rosa
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA 23219, USA
| | - Albert J. Arias
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA 23219, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Tatiana Ramey
- National Institute on Drug Abuse, 9000 Rockville Pike, Baltimore, MD 20892, USA
| | - F. Gerard Moeller
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA 23219, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23219, USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
2
|
Ubhayarathna M, Langmead CJ, Diepenhorst NA, Stewart GD. Molecular and structural insights into the 5-HT 2C receptor as a therapeutic target for substance use disorders. Br J Pharmacol 2024; 181:4414-4429. [PMID: 37679998 DOI: 10.1111/bph.16233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Substance use disorder (SUD) is a chronic condition, with maintained abuse of a substance leading to physiological and psychological alterations and often changes in cognitive and social behaviours. Current therapies include psychotherapy coupled with medication; however, high relapse rates reveal the shortcomings of these therapies. The signalling, expression profile, and neurological function of the serotonin 2C receptor (5-HT2C receptor) make it a candidate of interest for the treatment of SUD. Recently, psychedelics, which broadly act at 5-HT2 receptors, have indicated potential for the treatment of SUD, implicating the 5-HT2C receptor. The modern psychedelic movement has rekindled interest in the 5-HT2C receptor, resulting in many new studies, especially structural analyses. This review explores the structural, molecular and cellular mechanisms governing 5-HT2C receptor function in the context of SUD. This provides the basis of the preclinical and clinical evidence for their role in SUD and highlights the potential for future exploration.
Collapse
Affiliation(s)
- Maleesha Ubhayarathna
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Christopher J Langmead
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Australia
| | - Natalie A Diepenhorst
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Gregory D Stewart
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Australia
| |
Collapse
|
3
|
Yates JR. Pharmacological Treatments for Methamphetamine Use Disorder: Current Status and Future Targets. Subst Abuse Rehabil 2024; 15:125-161. [PMID: 39228432 PMCID: PMC11370775 DOI: 10.2147/sar.s431273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024] Open
Abstract
The illicit use of the psychostimulant methamphetamine (METH) is a major concern, with overdose deaths increasing substantially since the mid-2010s. One challenge to treating METH use disorder (MUD), as with other psychostimulant use disorders, is that there are no available pharmacotherapies that can reduce cravings and help individuals achieve abstinence. The purpose of the current review is to discuss the molecular targets that have been tested in assays measuring the physiological, the cognitive, and the reinforcing effects of METH in both animals and humans. Several drugs show promise as potential pharmacotherapies for MUD when tested in animals, but fail to produce long-term changes in METH use in dependent individuals (eg, modafinil, antipsychotic medications, baclofen). However, these drugs, plus medications like atomoxetine and varenicline, may be better served as treatments to ameliorate the psychotomimetic effects of METH or to reverse METH-induced cognitive deficits. Preclinical studies show that vesicular monoamine transporter 2 inhibitors, metabotropic glutamate receptor ligands, and trace amine-associated receptor agonists are efficacious in attenuating the reinforcing effects of METH; however, clinical studies are needed to determine if these drugs effectively treat MUD. In addition to screening these compounds in individuals with MUD, potential future directions include increased emphasis on sex differences in preclinical studies and utilization of pharmacogenetic approaches to determine if genetic variances are predictive of treatment outcomes. These future directions can help lead to better interventions for treating MUD.
Collapse
Affiliation(s)
- Justin R Yates
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA
| |
Collapse
|
4
|
Castle ME, Flanigan ME. The role of brain serotonin signaling in excessive alcohol consumption and withdrawal: A call for more research in females. Neurobiol Stress 2024; 30:100618. [PMID: 38433994 PMCID: PMC10907856 DOI: 10.1016/j.ynstr.2024.100618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/05/2024] Open
Abstract
Alcohol Use Disorder (AUD) is a leading cause of death and disability worldwide, but current treatments are insufficient in fully addressing the symptoms that often lead to relapses in alcohol consumption. The brain's serotonin system has been implicated in AUD for decades and is a major regulator of stress-related behaviors associated with increased alcohol consumption. This review will discuss the current literature on the association between neurobiological adaptations in serotonin systems and AUD in humans as well as the effectiveness of serotonin receptor manipulations on alcohol-related behaviors like consumption and withdrawal. We will further discuss how these findings in humans relate to findings in animal models, including a comparison of systemic pharmacological manipulations modulating alcohol consumption. We next provide a detailed overview of brain region-specific roles for serotonin and serotonin receptor signaling in alcohol-related behaviors in preclinical animal models, highlighting the complexity of forming a cohesive model of serotonin function in AUD and providing possible avenues for more effective therapeutic intervention. Throughout the review, we discuss what is known about sex differences in the sequelae of AUD and the role of serotonin in these sequelae. We stress a critical need for additional studies in women and female animals so that we may build a clearer path to elucidating sex-specific serotonergic mechanisms and develop better treatments.
Collapse
Affiliation(s)
- Megan E. Castle
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Meghan E. Flanigan
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
5
|
McCann DJ, Chen HH, Devine EG, Gyaw S, Ramey T. Results of a randomized, double-blind, placebo-controlled trial of lorcaserin in cocaine use disorder. Drug Alcohol Depend 2024; 255:111063. [PMID: 38163425 PMCID: PMC10872513 DOI: 10.1016/j.drugalcdep.2023.111063] [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: 05/04/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Cocaine use disorder (CUD) is a major public health problem for which there is no approved pharmacotherapy. The primary purpose of this study was to evaluate the ability of lorcaserin, a 5-hydroxytryptamine2 C (5-HT2 C) receptor agonist, to facilitate abstinence in individuals seeking treatment for CUD. METHODS This was a 12-site, randomized, parallel arm study with a 13-week Treatment Phase that included a 1-week, single-blind run-in period when all participants received twice daily 15mg acetazolamide capsules (a medication adherence marker), followed by randomization to either twice daily 10mg lorcaserin or placebo capsules for the remaining 12 weeks. Pre-randomization data were utilized in an enrichment strategy aimed at achieving high levels of medication adherence and low placebo response rates in a subgroup of participants that qualified for the "efficacy population." For lorcaserin vs. placebo, the primary efficacy endpoint was the proportion of participants in the efficacy population achieving abstinence during the last three weeks of treatment, as evidenced by self-report of no cocaine use, confirmed by urine testing. RESULTS Within the efficacy population, 1.1% of 91 participants receiving lorcaserin and 4.3% of 92 receiving placebo achieved abstinence during the last 3 weeks of treatment. Among all randomized participants, 2.5% of 118 receiving lorcaserin and 5.6% of 124 receiving placebo achieved similar abstinence. Study participants receiving lorcaserin exhibited significantly greater reductions in body weight and BMI, indicating that medication adherence was sufficient to produce a pharmacological effect. CONCLUSIONS Twice daily 10mg lorcaserin failed to demonstrate efficacy in the treatment of CUD.
Collapse
Affiliation(s)
- David J McCann
- Division of Therapeutics and Medical Consequences, National Institute on Drug Abuse, Bethesda, MD, USA.
| | - Hegang H Chen
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Eric G Devine
- Department of Psychiatry, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston, MA, USA.
| | - Shwe Gyaw
- Division of Therapeutics and Medical Consequences, National Institute on Drug Abuse, Bethesda, MD, USA.
| | - Tatiana Ramey
- Division of Therapeutics and Medical Consequences, National Institute on Drug Abuse, Bethesda, MD, USA.
| |
Collapse
|
6
|
Mazza M, Kotzalidis GD, Marano G, De Berardis D, Martinotti G, Romagnoli E, Biondi-Zoccai G, Abbate A, Sani G. Lorcaserin: Worthy of Further Insights? Results from Recent Research. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:278-283. [PMID: 37005521 DOI: 10.2174/1871527322666230330124137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/20/2023] [Accepted: 02/20/2023] [Indexed: 04/04/2023]
Abstract
Lorcaserin is a 3-benzazepine that binds 5-HT2C serotonin receptors in the hypothalamus, where it mediates lack of hunger and/or satiety, and in the ventral tegmental area, the site of origin of the mesolimbic and mesocortical dopaminergic projections, which mediate pleasure and reward. The drug has been first developed for the treatment of obesity, where it has shown efficacy, and subsequently trialed to counter substance use (mostly cocaine, cannabis, opioids, and nicotine) and craving, but showed inconsistent effects. Since 2020, the US Food and Drug Administration obtained that the drug was voluntarily withdrawn from the US market on the grounds that its long-term use was found to be associated with a greater incidence of some types of cancer. Provided it can show to be free from cancerogenic effects, ongoing research suggests that lorcaserin may have therapeutic potential for a variety of disorders and conditions beyond obesity. Since 5-HT2C receptors are involved in many diversified physiological functions (mood, feeding, reproductive behavior, neuronal processes related to impulsiveness, and modulating reward-related mechanisms) this drug has the potential to treat different central nervous system conditions, such as depression and schizophrenia.
Collapse
Affiliation(s)
- Marianna Mazza
- Department of Geriatrics, Institute of Psychiatry and Psychology, Neuroscience and Orthopedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Psychiatry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Georgios D Kotzalidis
- Department of Geriatrics, Institute of Psychiatry and Psychology, Neuroscience and Orthopedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Neurosciences, Mental Health, and Sensory Organs (NESMOS), Sapienza University of Rome, 00189 Rome, Italy
| | - Giuseppe Marano
- Department of Geriatrics, Institute of Psychiatry and Psychology, Neuroscience and Orthopedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Psychiatry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Giovanni Martinotti
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
- Department of Pharmacy, Pharmacology, Clinical Science, University of Hertfordshire, AL 10 9AB Herts, UK
| | - Enrico Romagnoli
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
- Mediterranea Cardiocentro, 80122 Napoli, Italy
| | - Antonio Abbate
- Berne Cardiovascular Research Center, University of Virginia, 22908 Charlottesville, Virginia, USA
| | - Gabriele Sani
- Department of Geriatrics, Institute of Psychiatry and Psychology, Neuroscience and Orthopedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Psychiatry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
7
|
Chao YS, Parrilla-Carrero J, Eid M, Culver OP, Jackson TB, Lipat R, Taniguchi M, Jhou TC. Innate cocaine-seeking vulnerability arising from loss of serotonin-mediated aversive effects of cocaine in rats. Cell Rep 2023; 42:112404. [PMID: 37083325 PMCID: PMC12035767 DOI: 10.1016/j.celrep.2023.112404] [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: 08/12/2022] [Revised: 01/11/2023] [Accepted: 04/02/2023] [Indexed: 04/22/2023] Open
Abstract
Cocaine blocks dopamine reuptake, thereby producing rewarding effects that are widely studied. However, cocaine also blocks serotonin uptake, which we show drives, in rats, individually variable aversive effects that depend on serotonin 2C receptors (5-HT2CRs) in the rostromedial tegmental nucleus (RMTg), a major GABAergic afferent to midbrain dopamine neurons. 5-HT2CRs produce depolarizing effects in RMTg neurons that are particularly strong in some rats, leading to aversive effects that reduce acquisition of and relapse to cocaine seeking. In contrast, 5-HT2CR signaling is largely lost after cocaine exposure in other rats, leading to reduced aversive effects and increased cocaine seeking. These results suggest a serotonergic biological marker of cocaine-seeking vulnerability that can be targeted to modulate drug seeking.
Collapse
Affiliation(s)
- Ying S Chao
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - Maya Eid
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Oliver P Culver
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Tyler B Jackson
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Rachel Lipat
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Thomas C Jhou
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
8
|
Chang VN, Peters J. Neural circuits controlling choice behavior in opioid addiction. Neuropharmacology 2023; 226:109407. [PMID: 36592884 PMCID: PMC9898219 DOI: 10.1016/j.neuropharm.2022.109407] [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: 08/01/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022]
Abstract
As the opioid epidemic presents an ever-expanding public health threat, there is a growing need to identify effective new treatments for opioid use disorder (OUD). OUD is characterized by a behavioral misallocation in choice behavior between opioids and other rewards, as opioid use leads to negative consequences, such as job loss, family neglect, and potential overdose. Preclinical models of addiction that incorporate choice behavior, as opposed to self-administration of a single drug reward, are needed to understand the neural circuits governing opioid choice. These choice models recapitulate scenarios that humans suffering from OUD encounter in their daily lives. Indeed, patients with substance use disorders (SUDs) exhibit a propensity to choose drug under certain conditions. While most preclinical addiction models have focused on relapse as the outcome measure, our data suggest that choice is an independent metric of addiction severity, perhaps relating to loss of cognitive control over choice, as opposed to excessive motivational drive to seek drugs during relapse. In this review, we examine both preclinical and clinical literature on choice behavior for drugs, with a focus on opioids, and the neural circuits that mediate drug choice versus relapse. We argue that preclinical models of opioid choice are needed to identify promising new avenues for OUD therapy that are translationally relevant. Both forward and reverse translation will be necessary to identify novel treatment interventions. This article is part of the Special Issue on "Opioid-induced changes in addiction and pain circuits".
Collapse
Affiliation(s)
- Victoria N Chang
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jamie Peters
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, 80045, USA; Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
9
|
De A, Grasing KW. The antidepressant agomelatine attenuates morphine-induced reinstatement but not self-administration or precipitated withdrawal. Pharmacol Biochem Behav 2023; 223:173525. [PMID: 36758685 PMCID: PMC11895839 DOI: 10.1016/j.pbb.2023.173525] [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: 09/25/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Exogenous melatonin appears to have anti-addictive properties and was recently shown to improve mental health and metabolic measures in patients receiving chronic opioid maintenance therapy. Agomelatine is a marketed antidepressant which acts as a melatonin agonist. We evaluated its effects using a rat model of morphine-reinforced behavior. METHODS After pretreatment with noncontingent morphine, male Wistar rats were trained to self-administer intravenous morphine (1.0 mg/kg-injection) under a progressive-ratio schedule. Rats were pretreated with vehicle or agomelatine during extinction, reinstatement, and reacquisition of morphine-reinforced behavior. RESULTS Daily treatment with 10 mg/kg-day of agomelatine decreased the number of ratios completed and prolonged latency during morphine-induced reinstatement. There were no significant effects on cue-induced reinstatement, morphine self-administration, or naloxone-precipitated withdrawal. Treatment with 32 mg/kg-day of agomelatine caused postural changes. That dose prolonged withdrawal-induced loss of body weight and caused delayed reductions in food reinforcement. SUMMARY In addition to postural effects, high-dose agomelatine worsened the course of spontaneous withdrawal and produced nonspecific effects on food-reinforced behavior. When administered at a selective dose, agomelatine did not modify morphine self-administration or precipitated withdrawal, but decreased morphine-induced reinstatement. Our findings show potential detrimental effects of high-dose agomelatine, with reductions in opioid-seeking behavior after a lower, more selective dose.
Collapse
Affiliation(s)
- Alok De
- Substance Use Research Laboratory, Kansas City Veterans Affairs Medical Center, 4801 Linwood Boulevard, Kansas City, MO 64128, United States of America
| | - Ken W Grasing
- Substance Use Research Laboratory, Kansas City Veterans Affairs Medical Center, 4801 Linwood Boulevard, Kansas City, MO 64128, United States of America; Division of Clinical Pharmacology, Department of Medicine, University of Kansas School of Medicine, Kansas City, KS 66160, United States of America.
| |
Collapse
|
10
|
Merritt CR, Smith AE, Khanipov K, Golovko G, Dineley KT, Anastasio NC, Cunningham KA. Heightened cocaine-seeking in male rats associates with a distinct transcriptomic profile in the medial prefrontal cortex. Front Pharmacol 2022; 13:1022863. [PMID: 36588704 PMCID: PMC9797046 DOI: 10.3389/fphar.2022.1022863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
Drug overdose deaths involving cocaine have skyrocketed, an outcome attributable in part to the lack of FDA-approved medications for the treatment of cocaine use disorder (CUD), highlighting the need to identify new pharmacotherapeutic targets. Vulnerability to cocaine-associated environmental contexts and stimuli serves as a risk factor for relapse in CUD recovery, with individual differences evident in the motivational aspects of these cues. The medial prefrontal cortex (mPFC) provides top-down control of striatal circuitry to regulate the incentive-motivational properties of cocaine-associated stimuli. Clinical and preclinical studies have identified genetic variations that impact the degree of executive restraint over drug-motivated behaviors, and we designed the present study to employ next-generation sequencing to identify specific genes associated with heightened cue-evoked cocaine-seeking in the mPFC of male, outbred rats. Rats were trained to stably self-administer cocaine, and baseline cue-reinforced cocaine-seeking was established. Rats were phenotyped as either high cue (HC) or low cue (LC) responders based upon lever pressing for previously associated cocaine cues and allowed 10 days of abstinence in their home cages prior to mPFC collection for RNA-sequencing. The expression of 309 genes in the mPFC was significantly different in HC vs. LC rats. Functional gene enrichment analyses identified ten biological processes that were overrepresented in the mPFC of HC vs. LC rats. The present study identifies distinctions in mPFC mRNA transcripts that characterizes individual differences in relapse-like behavior and provides prioritized candidates for future pharmacotherapeutics aimed to help maintain abstinence in CUD. In particular the Htr2c gene, which encodes the serotonin 5-HT2C receptor (5-HT2CR), is expressed to a lower extent in HC rats, relative to LC rats. These findings build on a plethora of previous studies that also point to the 5-HT2CR as an attractive target for the treatment of CUD.
Collapse
Affiliation(s)
- Christina R. Merritt
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ashley E. Smith
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kamil Khanipov
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - George Golovko
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kelly T. Dineley
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, United States
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, United States
| | - Noelle C. Anastasio
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kathryn A. Cunningham
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
11
|
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
|
12
|
Grasing KW, Burnell K, De A. Biphasic reward effects are characteristic of both lorcaserin and drugs of abuse: implications for treatment of substance use disorders. Behav Pharmacol 2022; 33:238-248. [PMID: 35324488 PMCID: PMC9149059 DOI: 10.1097/fbp.0000000000000672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Lorcaserin is a modestly selective agonist for 2C serotonin receptors (5-HT2CR). Despite early promising data, it recently failed to facilitate cocaine abstinence in patients and has been compared with dopamine antagonist medications (antipsychotics). Here, we review the effects of both classes on drug reinforcement. In addition to not being effective treatments for cocaine use disorder, both dopamine antagonists and lorcaserin can have biphasic effects on dopamine and reward behavior. Lower doses can cause enhanced drug taking with higher doses causing reductions. This biphasic pattern is shared with certain stimulants, opioids, and sedative-hypnotics, as well as compounds without abuse potential that include agonists for muscarinic and melatonin receptors. Additional factors associated with decreased drug taking include intermittent dosing for dopamine antagonists and use of progressive-ratio schedules for lorcaserin. Clinically relevant doses of lorcaserin were much lower than those that inhibited cocaine-reinforced behavior and can also augment this same behavior in different species. Diminished drug-reinforced behavior only occurred in animals after higher doses that are not suitable for use in patients. In conclusion, drugs of abuse and related compounds often act as biphasic modifiers of reward behavior, especially when evaluated over a broad range of doses. This property may reflect the underlying physiology of the reward system, allowing homeostatic influences on behavior.
Collapse
Affiliation(s)
- Ken W Grasing
- Substance Use Research Laboratory, Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
- Division of Clinical Pharmacology, Department of Medicine, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Kim Burnell
- Substance Use Research Laboratory, Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Alok De
- Substance Use Research Laboratory, Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| |
Collapse
|
13
|
Fletcher PJ, Li Z, Ji X, Higgins GA, Funk D, Lê A. Effects of pimavanserin and lorcaserin on alcohol self-administration and reinstatement in male and female rats. Neuropharmacology 2022; 215:109150. [DOI: 10.1016/j.neuropharm.2022.109150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/25/2022] [Accepted: 05/23/2022] [Indexed: 11/27/2022]
|
14
|
Hadizadeh H, Flores J, Nunes E, Mayerson T, Potenza MN, Angarita GA. Novel Pharmacological Agents for the Treatment of Cocaine Use Disorder. Curr Behav Neurosci Rep 2022. [DOI: 10.1007/s40473-022-00246-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
15
|
Negus SS, Banks ML. Confronting the challenge of failed translation in medications development for substance use disorders. Pharmacol Biochem Behav 2021; 210:173264. [PMID: 34461148 PMCID: PMC8418188 DOI: 10.1016/j.pbb.2021.173264] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/20/2021] [Indexed: 11/25/2022]
Affiliation(s)
- S S Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America.
| | - M L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America
| |
Collapse
|