1
|
Chan M, Chan JJ, Wright JM. Effect of amphetamines on blood pressure. Cochrane Database Syst Rev 2025; 3:CD007896. [PMID: 40152309 PMCID: PMC11951410 DOI: 10.1002/14651858.cd007896.pub4] [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: 03/29/2025]
Abstract
BACKGROUND Daily ingestion of amphetamines is common, as they are widely prescribed for attention-deficit hyperactivity disorder (ADHD) and other diagnoses. People also use amphetamines recreationally or in an attempt to boost cognitive or athletic performance. Amphetamines have the potential to increase blood pressure, and we do not know if the long-term benefits of daily amphetamine use outweigh the potential harms. OBJECTIVES Primary: to quantify the changes in systolic and diastolic blood pressure in children and adults taking amphetamines, compared to placebo. Secondary: to quantify the changes in heart rate in children and adults taking amphetamines, compared to placebo; to quantify the number of withdrawals due to adverse effects of amphetamine, compared to placebo. SEARCH METHODS We used the Cochrane Hypertension Specialized Register, CENTRAL, MEDLINE, Embase and two clinical trial registers, together with reference checking and contact with study authors to identify the studies included in the review. We imposed no restrictions on language, publication year or publication status. The latest search date was March 2023. SELECTION CRITERIA Randomized controlled trials (RCTs) that compared the effects of daily oral amphetamines versus placebo on blood pressure. There were no restrictions on participants' age or gender. DATA COLLECTION AND ANALYSIS We used standard methods expected by Cochrane. Primary outcomes were change in systolic and diastolic blood pressure (measured in millimeters of mercury (mmHg) above atmospheric pressure; continuous outcomes). Secondary outcomes were heart rate (measured as beats per minute; continuous outcome) and withdrawals due to adverse effects (dichotomous outcome). We calculated continuous outcomes as mean differences (MD) with 95% confidence intervals (CI). We expressed withdrawals due to adverse effects as a risk ratio with 95% CI. We used a fixed-effect model to pool effect sizes from all studies. MAIN RESULTS We included 56 RCTs with a total of 10,583 participants, both adults and children. Most studies were conducted in North America (mainly the USA), followed by Europe. A few studies took place in Asia (Japan) and Australia. The studies tested racemic amphetamine, dextroamphetamine, mixed amphetamine salts, lisdexamfetamine dimesylate, methylenedioxymethamphetamine (MDMA) and methylphenidate. The amphetamines were prescribed for ADHD, weight loss and other indications. In 48 RCTs, blood pressure was measured within 24 hours of the last dose. Based on data from all included studies, amphetamines increased systolic blood pressure (SBP) by 1.93 mmHg (95% CI 1.54 to 2.31) and diastolic blood pressure (DBP) by 1.84 mmHg (95% CI 1.51 to 2.16) (56 studies, 10,583 participants; high-certainty evidence for both). Amphetamines increased heart rate by 3.71 beats per minute (95% CI 3.27 to 4.14; 47 studies, 10,075 participants; high-certainty evidence). In a subgroup analysis limited to studies that gave participants amphetamines for at least eight weeks, the effects were similar, suggesting that these are sustained effects. These findings suggest that people taking daily oral amphetamines are at increased risk of adverse cardiovascular events. Participants in the amphetamine group were also more likely to withdraw from the study due to adverse effects compared to those given placebo (risk ratio 2.69, 95% CI 2.13 to 3.40; absolute risk increase of 4.3% over an average duration of 1 month; 42 studies, 8952 participants; high-certainty evidence). In general, the studies were well-executed, and the methodology was sound. We judged most studies to have a low risk of bias across most domains. Selection bias (random sequence generation and allocation concealment) was the domain most often rated as at unclear risk of bias, because the methods used were not reported. We judged 13 studies (23%) to have a high risk of bias in at least one of the seven domains, primarily due to high dropout rates, leading to a high risk of attrition bias. AUTHORS' CONCLUSIONS Daily oral amphetamines increase blood pressure, heart rate, and withdrawals due to adverse effects, with these effects observed across all time points, including shorter (≤ four weeks) and longer durations (> four weeks to < eight weeks; ≥ eight weeks) of use. Future trials should measure blood pressure using 24-hour ambulatory monitoring and assess the effect of long-term use.
Collapse
Affiliation(s)
- Magnus Chan
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Jocelyn Joy Chan
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - James M Wright
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
2
|
Raymond JS, Athanasopoulos AG, Badolato CJ, Doolan TJ, Scicluna RL, Everett NA, Bowen MT, James MH. Emerging medications and pharmacological treatment approaches for substance use disorders. Pharmacol Biochem Behav 2025; 248:173952. [PMID: 39719161 DOI: 10.1016/j.pbb.2024.173952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Medications to treat substance use disorders (SUDs) remain suboptimal or, in the case of stimulants and cannabis, non-existent. Many factors have contributed to this paucity, including the biological complexity of addiction, regulatory challenges, and a historical lack of enthusiasm among pharmaceutical companies to commit resources to this disease space. Despite these headwinds, the recent opioid crisis has highlighted the devastating consequences of SUDs for both individuals and society, stimulating urgent efforts to identify novel treatment approaches. In addition, several neurobiological systems have been recently implicated in unique aspects of drug reward, opening the door to candidate medications with novel mechanisms of action. Here, we provide an overview of efforts to target several of these new systems, with a focus on those that are the subject of ongoing clinical trials as well as being areas of interest among the authors' research groups (MHJ, MTB, NAE). Specifically, we discuss new classes of medications targeting the serotonin 2A receptor (i.e., psychedelics), glucagon-like peptide 1 receptor, cannabidiol, dynorphin/kappa opioid receptor, orexin/hypocretin, and oxytocin receptor systems, as well as emergent approaches for modulating the more canonical dopaminergic system via agonist therapies for stimulant use disorders. Collectively, innovations in this space give reason for optimism for an improved therapeutic landscape for substance use disorders in the near future.
Collapse
Affiliation(s)
- Joel S Raymond
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA; Rutgers Addiction Research Center, Brain Health Institute, Rutgers Health, Piscataway, NJ, USA
| | - Alexander G Athanasopoulos
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Connie J Badolato
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Tylah J Doolan
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Rhianne L Scicluna
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas A Everett
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Michael T Bowen
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA; Rutgers Addiction Research Center, Brain Health Institute, Rutgers Health, Piscataway, NJ, USA; School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
3
|
Amerio A, Baccino C, Breda GS, Cortesi D, Spiezio V, Magnani L, De Berardis D, Conio B, Costanza A, De Paola G, Rocca G, Arduino G, Aguglia A, Amore M, Serafini G. Effects of transcranial magnetic stimulation on cocaine addiction: A systematic review of randomized controlled trials. Psychiatry Res 2023; 329:115491. [PMID: 37783092 DOI: 10.1016/j.psychres.2023.115491] [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: 06/12/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023]
Abstract
OBJECTIVE While pharmacological strategies appear to be ineffective in treating long-term addiction, repetitive transcranial magnetic stimulation (rTMS) is emerging as a promising new tool for the attenuation of craving among multiple substance dependent populations. METHOD A systematic review of randomized controlled trials (RCTs) was conducted on the efficacy and tolerability of rTMS in treating cocaine use disorder (CUD). Relevant papers published in English through November 30th 2022 were identified, searching the electronic databases MEDLINE, Embase, PsycINFO and the Cochrane Library. RESULTS Eight studies matched inclusion criteria. The best findings were reported by the RCTs conducted at high-frequency (≥5 Hz) multiple sessions of rTMS delivered over the left dorsolateral prefrontal cortex (DLPFC): a significant decrease in self-reported cue-induced cocaine craving and lower cocaine craving scores and a considerable amelioration in the tendency to act rashly under extreme negative emotions (impulsivity) were found in the active group compared to controls. CONCLUSION Although still scant and heterogeneous, the strongest evidence so far on the use of rTMS on individuals with CUD support the high frequency stimulation over the left DLPFC as a well tolerated treatment of cocaine craving and impulsivity.
Collapse
Affiliation(s)
- A Amerio
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - C Baccino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - G S Breda
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - D Cortesi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - V Spiezio
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - L Magnani
- Department of Psychiatry, San Maurizio Hospital, Bolzano, Italy
| | - D De Berardis
- NHS, Department of Mental Health, Psychiatric Service for Diagnosis and Treatment, Hospital "G. Mazzini", ASL 4, Teramo, Italy.
| | - B Conio
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - A Costanza
- Department of Psychiatry, Faculty of Medicine, Geneva University (UNIGE), Geneva, Switzerland; Department of Psychiatry, Adult Psychiatry Service (SPA), University Hospitals of Geneva (HUG), Geneva, Switzerland; Department of Psychiatry, Faculty of Biomedical Sciences, University of Italian Switzerland (USI), Lugano, Switzerland.
| | - G De Paola
- Ospedale Maria Luigia, Monticelli Terme, Italy
| | - G Rocca
- R&R Neuromodulation Lab, Piacenza, Italy
| | - G Arduino
- Department of Mental Health and Pathological Addictions, Piacenza Local Health Authority, Piacenza, Italy
| | - A Aguglia
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - M Amore
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - G Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Heal DJ, Smith SL. Prospects for new drugs to treat binge-eating disorder: Insights from psychopathology and neuropharmacology. J Psychopharmacol 2022; 36:680-703. [PMID: 34318734 PMCID: PMC9150143 DOI: 10.1177/02698811211032475] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Binge-eating disorder (BED) is a common psychiatric condition with adverse psychological and metabolic consequences. Lisdexamfetamine (LDX) is the only approved BED drug treatment. New drugs to treat BED are urgently needed. METHODS A comprehensive review of published psychopathological, pharmacological and clinical findings. RESULTS The evidence supports the hypothesis that BED is an impulse control disorder with similarities to ADHD, including responsiveness to catecholaminergic drugs, for example LDX and dasotraline. The target product profile (TPP) of the ideal BED drug combines treating the psychopathological drivers of the disorder with an independent weight-loss effect. Drugs with proven efficacy in BED have a common pharmacology; they potentiate central noradrenergic and dopaminergic neurotransmission. Because of the overlap between pharmacotherapy in attention deficit hyperactivity disorder (ADHD) and BED, drug-candidates from diverse pharmacological classes, which have already failed in ADHD would also be predicted to fail if tested in BED. The failure in BED trials of drugs with diverse pharmacological mechanisms indicates many possible avenues for drug discovery can probably be discounted. CONCLUSIONS (1) The efficacy of drugs for BED is dependent on reducing its core psychopathologies of impulsivity, compulsivity and perseveration and by increasing cognitive control of eating. (2) The analysis revealed a large number of pharmacological mechanisms are unlikely to be productive in the search for effective new BED drugs. (3) The most promising areas for new treatments for BED are drugs, which augment noradrenergic and dopaminergic neurotransmission and/or those which are effective in ADHD.
Collapse
Affiliation(s)
- David J Heal
- David J Heal, DevelRx Ltd, BioCity, Nottingham, NG1 1GF, UK.
| | | |
Collapse
|
7
|
Schmitz JM, Suchting R, Green CE, Webber HE, Vincent J, Moeller FG, Lane SD. The effects of combination levodopa-ropinirole on cognitive improvement and treatment outcome in individuals with cocaine use disorder: A bayesian mediation analysis. Drug Alcohol Depend 2021; 225:108800. [PMID: 34102508 DOI: 10.1016/j.drugalcdep.2021.108800] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/08/2021] [Accepted: 04/03/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Chronic cocaine users show impairments in cognitive processes associated with dopamine (DA) circuitry. Medications aimed at bolstering cognitive functions via DA modulation might enhance treatment outcome. METHODS The trial used a double-blind, double-dummy, parallel-group design with four treatment arms comparing placebo (PLC) to levodopa/carbidopa 800 mg/200 mg alone (LR0), levodopa plus extended release (XR) ropinirole 2 mg (LR2) or XR ropinirole 4 mg (LR4). Adults (n = 110) with cocaine use disorder attended thrice weekly clinic visits for 10 weeks. Potential cognitive mediators assessed at week 5 consisted of measures of decision-making (Iowa Gambling Task, Risky Decision-Making Task), attention/impulsivity (Immediate Memory Task), motivation (Progressive Ratio task), and cognitive control (Cocaine Stoop task). The primary outcome measure was the treatment effectiveness score (TES) calculated as the number of cocaine-negative urines collected from weeks 6-10. RESULTS Bayesian mediation examined indirect and total effects of the relationships between each active treatment (compared to PLC) and TES. Total (direct) effects were supported for LR0 and LR2, but not for LR4. Indirect effects were tested for each mediator. Notably, 22.3 % and 35.4 % of the total effects of LR0 and LR2 on TES were mediated by changes in attention/impulsivity. CONCLUSIONS The hypothesized mediation effect was strongest for levodopa plus 2 mg ropinirole, indicating that this DA medication combination predicted change (improvement) in attention/impulsivity, which in turn predicted change (reduction) in cocaine use. This finding provides modest support for cognitive enhancement as a target for medications to treat cocaine use disorder.
Collapse
Affiliation(s)
- 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.
| | - 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
| | - Heather E Webber
- 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
| |
Collapse
|
8
|
Bentzley BS, Han SS, Neuner S, Humphreys K, Kampman KM, Halpern CH. Comparison of Treatments for Cocaine Use Disorder Among Adults: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e218049. [PMID: 33961037 PMCID: PMC8105751 DOI: 10.1001/jamanetworkopen.2021.8049] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPORTANCE In the US and the United Kingdom, cocaine use is the second leading cause of illicit drug overdose death. Psychosocial treatments for cocaine use disorder are limited, and no pharmacotherapy is approved for use in the US or Europe. OBJECTIVE To compare treatments for active cocaine use among adults. DATA SOURCES PubMed and the Cochrane Database of Systematic Reviews were searched for clinical trials published between December 31, 1995, and December 31, 2017. STUDY SELECTION This meta-analysis was registered on Covidence.org (study 8731) on December 31, 2015. Clinical trials were included if they (1) had the term cocaine in the article title; (2) were published between December 31, 1995, and December 31, 2017; (3) were written in English; (4) enrolled outpatients 18 years or older with active cocaine use at baseline; and (5) reported treatment group size, treatment duration, retention rates, and urinalysis results for the presence of cocaine metabolites. A study was excluded if (1) more than 25% of participants were not active cocaine users or more than 80% of participants had negative test results for the presence of cocaine metabolites at baseline and (2) it reported only pooled urinalysis results indicating the presence of multiple substances and did not report the specific proportion of positive test results for cocaine metabolites. Multiple reviewers reached criteria consensus. Of 831 records screened, 157 studies (18.9%) met selection criteria and were included in the analysis. DATA EXTRACTION AND SYNTHESIS This study followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guideline. Search results were imported from PubMed XML into Covidence.org then Microsoft Excel. Data extraction was completed in 2 iterations to ensure fidelity. Analyses included a multilevel random-effects model, a multilevel mixed-effects meta-regression model, and sensitivity analyses. Treatments were clustered into 11 categories (psychotherapy, contingency management programs, placebo, opioids, psychostimulants, anticonvulsants, dopamine agonists, antidepressants, antipsychotics, miscellaneous medications, and other therapies). Missing data were imputed using multiple imputation by chained equations. The significance threshold for all analyses was P = .05. Data were analyzed using the metafor and mice packages in R software, version 3.3.2 (R Foundation for Statistical Computing). Data were analyzed from January 1, 2018, to February 28, 2021. MAIN OUTCOMES AND MEASURES The primary outcome was the intention-to-treat logarithm of the odds ratio (OR) of having a negative urinalysis result for the presence of cocaine metabolites at the end of each treatment period compared with baseline. The hypothesis, which was formulated after data collection, was that no treatment category would have a significant association with objective reductions in cocaine use. RESULTS A total of 157 studies comprising 402 treatment groups and 15 842 participants were included. Excluding other therapies, the largest treatment groups across all studies were psychotherapy (mean [SD] number of participants, 40.04 [36.88]) and contingency management programs (mean [SD] number of participants, 37.51 [25.51]). Only contingency management programs were significantly associated with an increased likelihood of having a negative test result for the presence of cocaine (OR, 2.13; 95% CI, 1.62-2.80), and this association remained significant in all sensitivity analyses. CONCLUSIONS AND RELEVANCE In this meta-analysis, contingency management programs were associated with reductions in cocaine use among adults. Research efforts and policies that align with this treatment modality may benefit those who actively use cocaine and attenuate societal burdens.
Collapse
Affiliation(s)
- Brandon S. Bentzley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Summer S. Han
- Department of Neurosurgery, Stanford University, Stanford, California
| | - Sophie Neuner
- Department of Neurosurgery, Stanford University, Stanford, California
| | - Keith Humphreys
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Kyle M. Kampman
- Department of Psychiatry, University of Pennsylvania, Philadelphia
| | - Casey H. Halpern
- Department of Neurosurgery, Stanford University, Stanford, California
| |
Collapse
|
9
|
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
|
10
|
Improving translation of animal models of addiction and relapse by reverse translation. Nat Rev Neurosci 2020; 21:625-643. [PMID: 33024318 DOI: 10.1038/s41583-020-0378-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2020] [Indexed: 12/13/2022]
Abstract
Critical features of human addiction are increasingly being incorporated into complementary animal models, including escalation of drug intake, punished drug seeking and taking, intermittent drug access, choice between drug and non-drug rewards, and assessment of individual differences based on criteria in the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV). Combined with new technologies, these models advanced our understanding of brain mechanisms of drug self-administration and relapse, but these mechanistic gains have not led to improvements in addiction treatment. This problem is not unique to addiction neuroscience, but it is an increasing source of disappointment and calls to regroup. Here we first summarize behavioural and neurobiological results from the animal models mentioned above. We then propose a reverse translational approach, whose goal is to develop models that mimic successful treatments: opioid agonist maintenance, contingency management and the community-reinforcement approach. These reverse-translated 'treatments' may provide an ecologically relevant platform from which to discover new circuits, test new medications and improve translation.
Collapse
|
11
|
Kearns DN, Silberberg A. Opening the cocaine economy by providing within-session access to a cheaper source of cocaine makes demand for it more elastic. Behav Pharmacol 2020; 31:448-457. [PMID: 31625978 PMCID: PMC7182474 DOI: 10.1097/fbp.0000000000000510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Previous studies found that opening the cocaine economy by providing postsession access to cocaine had no effect on animals' demand for cocaine, whereas postsession access to saccharin or food made demand for these nondrug reinforcers more elastic. It is possible that there was no effect of economy type on cocaine taking in these earlier studies because of the delay to the postsession cocaine in the open economy. The present experiment tested whether forming an open economy by providing additional within-session cocaine, rather than postsession cocaine, would make rats' demand for cocaine more elastic. Saccharin was used as a nondrug comparison reinforcer. Three groups of rats pressed one lever for cocaine and one for saccharin on an ascending series of fixed ratio (FR) schedules where the number of responses required per reinforcer increased from 1 to 48 over sessions. In the open cocaine and open saccharin economy groups, rats had occasional access during the session to a third lever where cocaine or saccharin reinforcers, respectively, were always available on an FR-1 schedule. The main finding was that demand for cocaine was more elastic in the open cocaine economy group than in either of the other groups. Demand for saccharin was more elastic in the open saccharin economy group than in the open cocaine economy group. This study shows that cocaine taking is sensitive to economy type when the additional source of cocaine in an open economy is available close in time to when rats work for cocaine.
Collapse
Affiliation(s)
- David N Kearns
- Psychology Department, American University, Washington DC, USA
| | | |
Collapse
|
12
|
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
|
13
|
Bardo MT, Denehy ED, Hammerslag LR, Dwoskin LP, Blough BE, Landavazo A, Bergman J, Kohut SJ. Effects of methamphetamine isomers on d-methamphetamine self-administration and food-maintained responding in male rats. Psychopharmacology (Berl) 2019; 236:3557-3565. [PMID: 31346628 PMCID: PMC6895396 DOI: 10.1007/s00213-019-05327-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 07/08/2019] [Indexed: 11/25/2022]
Abstract
RATIONALE Methamphetamine (METH) abuse is generally attributed to the d-isomer. Self-administration of l-METH has been examined only in rhesus monkeys with a history of cocaine self-administration or drug-naïve rats using high toxic doses. OBJECTIVES In this study, the ability of l-METH and, for comparison, d-METH to engender self-administration in experimentally naïve rats, as well as to decrease d-METH self-administration and food-maintained responding, was examined. METHODS Male Sprague-Dawley rats were used in 3 separate experiments. In experiment 1, the acquisition of l- or d-METH self-administration followed by dose-response determinations was studied. In experiment 2, rats were trained to self-administer d-METH (0.05 mg/kg/infusion) and, then, various doses of l- or d-METH were given acutely prior to the session; the effect of repeated l-METH (30 mg/kg) also was examined. In experiment 3, rats were trained to respond for food reinforcement and, then, various doses of l- or d-METH were given acutely prior to the session; the effect of repeated l-METH (3 mg/kg) also was examined. RESULTS Reliable acquisition of l- and d-METH self-administration was obtained at unit doses of 0.5 and 0.05 mg/kg/infusion respectively. The dose-response function for l-METH self-administration was flattened and shifted rightward compared with d-METH self-administration, with peak responding for l- and d-METH occurring at unit doses of 0.17 and 0.025 respectively. l-METH also was approximately 10-fold less potent than d-METH in decreasing d-METH self-administration and 2-fold lower in decreasing food-maintained responding. Tolerance did not occur to repeated l-METH pretreatments on either measure. CONCLUSIONS As a potential pharmacotherapeutic, l-METH has less abuse liability than d-METH and its efficacy in decreasing d-METH self-administration and food-maintained responding is sustained with repeated treatment.
Collapse
Affiliation(s)
- M T Bardo
- Department of Psychology, University of Kentucky, Lexington, KY, 40536, USA.
| | - E D Denehy
- Department of Psychology, University of Kentucky, Lexington, KY, 40536, USA
| | - L R Hammerslag
- Department of Psychology, University of Kentucky, Lexington, KY, 40536, USA
| | - L P Dwoskin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA
| | - B E Blough
- Research Triangle Institute, Center for Drug Discovery, Research Triangle Park, NC, USA
| | - A Landavazo
- Research Triangle Institute, Center for Drug Discovery, Research Triangle Park, NC, USA
| | - J Bergman
- McLean Hospital - Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, USA
| | - S J Kohut
- McLean Hospital - Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, USA
| |
Collapse
|
14
|
Siciliano CA, Mauterer MI, Fordahl SC, Jones SR. Modulation of striatal dopamine dynamics by cocaine self-administration and amphetamine treatment in female rats. Eur J Neurosci 2019; 50:2740-2749. [PMID: 31111573 DOI: 10.1111/ejn.14437] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 05/05/2019] [Accepted: 05/10/2019] [Indexed: 11/29/2022]
Abstract
Despite decades of research into the neurobiological basis of cocaine abuse, pharmacotherapeutic treatments for cocaine addiction have been largely ineffective. Converging evidence from preclinical research and from outpatient clinical trials suggest that treatment with amphetamine is efficacious in reducing cocaine intake. Although it has been suggested that amphetamine treatment reduces cocaine intake as an agonist replacement therapy, we have shown recently that multiple aspects of dopamine signaling are altered by cocaine self-administration and returned to pre-cocaine function by amphetamine treatment in the nucleus accumbens of male rats. Here, we sought to determine if these effects were also evident in female subjects, and across regions of the striatum. Female rats performed 5 days of cocaine self-administration (1.5 mg kg-1 inj-1 , 40 inj/day) and were treated with a single amphetamine (0.56 mg/kg) or saline infusion 1 hr prior to killing. We then used ex vivo fast-scan cyclic voltammetry in the nucleus accumbens core or dorsolateral caudate-putamen to examine dopamine signaling and cocaine potency. We found that in the nucleus accumbens core, cocaine self-administration decreased dopamine uptake rate and cocaine potency, and both alterations were restored by amphetamine treatment. In the dorsolateral caudate-putamen, neither cocaine self-administration nor amphetamine treatment altered dopamine uptake; however, cocaine potency was decreased by self-administration and returned to control levels by amphetamine. Together, these findings support a role for amphetamine treatment for cocaine addiction outside of agonist replacement therapy, and suggest that the development of cocaine tolerance is similar across sexes.
Collapse
Affiliation(s)
- Cody A Siciliano
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina.,Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts
| | - Madelyn I Mauterer
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Steve C Fordahl
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina.,Department of Nutrition, University of North Carolina at Greensboro, Greensboro, North Carolina
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
15
|
|
16
|
Ezard N, Dunlop A, Hall M, Ali R, McKetin R, Bruno R, Phung N, Carr A, White J, Clifford B, Liu Z, Shanahan M, Dolan K, Baker AL, Lintzeris N. LiMA: a study protocol for a randomised, double-blind, placebo controlled trial of lisdexamfetamine for the treatment of methamphetamine dependence. BMJ Open 2018; 8:e020723. [PMID: 30030312 PMCID: PMC6059315 DOI: 10.1136/bmjopen-2017-020723] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Methamphetamine dependence is a growing public health concern. There is currently no pharmacotherapy approved for methamphetamine dependence. Lisdexamfetamine (LDX) dimesylate, used in the treatment of attention-deficit hyperactivity disorder and binge eating disorder, has potential as an agonist therapy for methamphetamine dependence, and possible benefits of reduced risk of aberrant use due to its novel formulation. METHODS AND ANALYSIS A double-blind randomised controlled trial will be used to evaluate the efficacy of LDX in reducing methamphetamine use. The target sample is 180 participants with methamphetamine dependence of ≥2 years, using ≥14 days out of the previous 28, who have previously attempted but not responded to treatment for methamphetamine use. Participants will be randomly assigned to receive either a 15-week intervention consisting of induction (1 week of 150 mg LDX or placebo), maintenance (12 weeks of 250 mg LDX or placebo) and reduction (1 week of 150 mg LDX or placebo and 1 week of 50 mg LDX or placebo). All participants will be given access to four sessions of cognitive-behavioural therapy as treatment as usual and receive a 4-week follow-up appointment. The primary outcomes are efficacy (change from baseline in days of methamphetamine use by self-report for the last 28 days at week 13 and urinalyses confirmation of methamphetamine use) and safety (treatment-related adverse events). Secondary outcomes are total number of days of self-report methamphetamine use over the 12-week active treatment, longest period of abstinence during treatment period, percentage of achieving ≥21 days abstinence, craving, withdrawal, dependence, retention, bloodborne virus transmission risk behaviour, criminal behaviour, as well measures of abuse liability, physical and mental health, other substance use, cognitive performance, psychosocial functioning, treatment retention and satisfaction. Additionally, the study will assess the cost-effectiveness of LDX relative to the placebo control. ETHICS AND DISSEMINATION The study has been approved by the Human Research Ethics Committee of St. Vincent's Hospital, Sydney, Australia (HREC/16/SVH/222). Contact the corresponding author for the full trial protocol. TRIAL REGISTRATION NUMBER ACTRN12617000657325; Pre-results.
Collapse
Affiliation(s)
- Nadine Ezard
- Alcohol and Drug Service, St Vincents Hospital, Sydney, New South Wales, Australia
- University of New South Wales, Randwick, New South Wales, Australia
| | - Adrian Dunlop
- Drug and Alcohol Clinical Services, Hunter New England, Newcastle, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Michelle Hall
- Drug and Alcohol Clinical Services, Hunter New England, Newcastle, New South Wales, Australia
| | - Robert Ali
- University of New South Wales, Randwick, New South Wales, Australia
- University of Adelaide, Adelaide, South Australia, Australia
| | | | - Raimondo Bruno
- University of South Australia, Adelaide, South Australia, Australia
- University of Tasmania, Hobart, Tasmania, Australia
| | - Nghi Phung
- Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Andrew Carr
- Alcohol and Drug Service, St Vincents Hospital, Sydney, New South Wales, Australia
- University of New South Wales, Randwick, New South Wales, Australia
| | - Jason White
- University of South Australia, Adelaide, South Australia, Australia
| | - Brendan Clifford
- Alcohol and Drug Service, St Vincents Hospital, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Zhixin Liu
- University of New South Wales, Randwick, New South Wales, Australia
| | - Marian Shanahan
- National Drug and Alcohol Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kate Dolan
- National Drug and Alcohol Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Amanda L Baker
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Nicholas Lintzeris
- University of Sydney, Sydney, New South Wales, Australia
- South East Sydney Local Health District, Sydney, New South Wales, Australia
| |
Collapse
|
17
|
Abstract
Treatment-resistant mood disorders and anxiety disorders require intensive treatment, but treatment options should balance benefits and adverse effects or other potential detrimental effects on patients, including the risk of developing prescription medication addiction. Some of the newer treatment modalities for mood and anxiety disorders may have similar properties to benzodiazepines. The goal of this review was to identify the potential for developing dependence on the novel treatment approaches to treatment-resistant depression and refractory anxiety disorders. PubMed, MEDLINE, PsycINFO, Ovid, Cochrane Library, and Google Scholar were searched. Ketamine is effective in improving symptoms of major depressive disorder, but with no sustained benefits. Long-term use of oral or intranasal ketamine formulations may be associated with the risk of developing dependence. Augmentation of stimulant medication is usually effective for residual symptoms of depression, but the effects are usually short lasting and there is a potential for abuse. Synthetic cannabinoids and medicinal cannabis are increasingly being prescribed for a number of medical conditions, including anxiety disorders, without enough evidence about their efficacy and with the risk of patients developing dependence. In summary, benzodiazepines, ketamine, stimulant medications, and cannabinoids have some common characteristics, including short-lasting benefits and the risk of developing prescription medication addiction with longer use. All of these treatments may raise ethical dilemmas about the appropriateness of prescribing these medications in the long run for patients with depression and anxiety disorders.
Collapse
Affiliation(s)
- Dusan Kolar
- Mood Disorders Research and Treatment Service, Department of Psychiatry, Queen’s University, Kingston, Ontario, Canada
| |
Collapse
|
18
|
Pharmacokinetics of Sustained-Release Oral Dexamphetamine Sulfate in Cocaine and Heroin-Dependent Patients. J Clin Psychopharmacol 2018; 38:212-217. [PMID: 29620701 DOI: 10.1097/jcp.0000000000000862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Research has shown that sustained-release (SR) dexamphetamine is a promising agonist treatment for cocaine dependence. However, little is known about the pharmacokinetics (PKs) of SR oral dexamphetamine. This study examined the PKs of a new SR dexamphetamine formulation in cocaine plus heroin-dependent patients currently in heroin-assisted treatment. METHODS The study was designed as an open-label PK study in 2 cohorts: n = 5 with once daily 60 mg and n = 7 with once daily 30 mg SR oral dexamphetamine. Five days of blood plasma dexamphetamine concentrations measured with liquid chromatography-mass spectrometry with PK parameter estimates using noncompartmental analysis. RESULTS Twelve cocaine-dependent plus heroin-dependent patients in heroin-assisted treatment were included. The initial cohort 1 dose of 60 mg once daily was adjusted to 30 mg after mild to moderate adverse events. After oral administration, tmax values (coefficient of variation %) were 6.0 (17.0%) and 6.3 (16.3%) hours and t1/2 were 11 (24.6%) and 12 (25.4%) hours for 60 mg and 30 mg SR dexamphetamine, respectively. At steady state, CSSmax values were reached at 100 (27.5%) ng/mL and 58.4 (14.4%) ng/mL, whereas CSSmin values were 39.5 (38.9%) ng/mL and 21.8 (19.8%) ng/mL for 60 mg and 30 mg, respectively. CONCLUSIONS The investigated SR formulation of dexamphetamine showed favorable slow-release characteristics in cocaine and heroin-dependent patients. A dose-proportional steady-state concentration was achieved within 3 days. These findings support the suitability of the SR formulation in the treatment of cocaine dependence.
Collapse
|
19
|
Hussain Y, Krishnamurthy S. Piracetam attenuates binge eating disorder related symptoms in rats. Pharmacol Biochem Behav 2018; 169:35-47. [DOI: 10.1016/j.pbb.2018.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 04/10/2018] [Accepted: 04/11/2018] [Indexed: 01/19/2023]
|
20
|
Negus SS, Banks ML. Pharmacokinetic-Pharmacodynamic (PKPD) Analysis with Drug Discrimination. Curr Top Behav Neurosci 2018; 39:245-259. [PMID: 27571746 PMCID: PMC5446801 DOI: 10.1007/7854_2016_36] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Discriminative stimulus and other drug effects are determined by the concentration of drug at its target receptor and by the pharmacodynamic consequences of drug-receptor interaction. For in vivo procedures such as drug discrimination, drug concentration at receptors in a given anatomical location (e.g., the brain) is determined both by the dose of drug administered and by pharmacokinetic processes of absorption, distribution, metabolism, and excretion that deliver drug to and from that anatomical location. Drug discrimination data are often analyzed by strategies of dose-effect analysis to determine parameters such as potency and efficacy. Pharmacokinetic-Pharmacodynamic (PKPD) analysis is an alternative to conventional dose-effect analysis, and it relates drug effects to a measure of drug concentration in a body compartment (e.g., venous blood) rather than to drug dose. PKPD analysis can yield insights on pharmacokinetic and pharmacodynamic determinants of drug action. PKPD analysis can also facilitate translational research by identifying species differences in pharmacokinetics and providing a basis for integrating these differences into interpretation of drug effects. Examples are discussed here to illustrate the application of PKPD analysis to the evaluation of drug effects in rhesus monkeys trained to discriminate cocaine from saline.
Collapse
Affiliation(s)
- S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| |
Collapse
|
21
|
Abstract
Pain is essential for avoidance of tissue damage and for promotion of healing. Notwithstanding the survival value, pain brings about emotional suffering reflected in fear and anxiety, which in turn augment pain thus giving rise to a self-sustaining feedforward loop. Given such reciprocal relationships, the present article uses neuroscientific conceptualizations of fear and anxiety as a theoretical framework for hitherto insufficiently understood pathophysiological mechanisms underlying chronic pain. To that end, searches of PubMed-indexed journals were performed using the following Medical Subject Headings' terms: pain and nociception plus amygdala, anxiety, cognitive, fear, sensory, and unconscious. Recursive sets of scientific and clinical evidence extracted from this literature review were summarized within the following key areas: (1) parallelism between acute pain and fear and between chronic pain and anxiety; (2) all are related to the evasion of sensory-perceived threats and are subserved by subcortical circuits mediating automatic threat-induced physiologic responses and defensive actions in conjunction with higher order corticolimbic networks (e.g., thalamocortical, thalamo-striato-cortical and amygdalo-cortical) generating conscious representations and valuation-based adaptive behaviors; (3) some instances of chronic pain and anxiety conditions are driven by the failure to diminish or block respective nociceptive information or unconscious treats from reaching conscious awareness; and (4) the neural correlates of pain-related conscious states and cognitions may become autonomous (i.e., dissociated) from the subcortical activity/function leading to the eventual chronicity. Identifying relative contributions of the diverse neuroanatomical sources, thus, offers prospects for the development of novel preventive, diagnostic, and therapeutic strategies in chronic pain patients.
Collapse
Affiliation(s)
- Igor Elman
- Boonshoft School of Medicine, Wright State University, Dayton VA Medical Center, Dayton, OH, United States
| | - David Borsook
- Harvard Medical School, Center for Pain and the Brain, Boston Children's Hospital, Massachusetts General Hospital, McLean Hospital, Boston, MA, United States
| |
Collapse
|
22
|
Zernig G, Hiemke C. Making the Case for 'Power Abuse Disorder' as a Nosologic Entity. Pharmacology 2017; 100:50-63. [PMID: 28467994 DOI: 10.1159/000475600] [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] [Received: 02/03/2017] [Accepted: 04/10/2017] [Indexed: 12/18/2022]
Abstract
The development of societies and cultures arguably is based on the ability of human primates to form hierarchies in which some individuals acquire and wield power, that is, control resources and influence and control the behavior of their conspecifics. In the following, we focus on the type of human primate power wielding that (a) harms and (b) produces excessive negative emotions in (1) the victim(s) of the power wielder and (2) the power wielder her/himself. If such a harmful behavior of the power wielder is not accompanied by an ethically justifiable benefit for the involved human primate groups, it can be considered "power abuse." We propose to term the associated behaviors, cognitions, and emotions of the power wielder as "power abuse disorder" (PAD). This behavior results from what we consider addictive behavior of the power abuse disordered (PADed) power wielder. PAD can be diagnosed on the basis of the World Health Organization's criteria for "dependence syndrome" as listed in the International Classification of Diseases version 10. We will demonstrate that many PADed individuals may very likely carry the Zeitgeist diagnosis "burnout." This article reviews the current understanding of the neural correlates of PAD and suggests future research. Based on the available evidence, PAD seems to be associated with a dysfunction of the mesocorticolimbic dopamine system, rendering PADed individuals vulnerable for psychostimulant abuse/dependence, and suggesting specific pharmacotherapeutic approaches to treat PAD.
Collapse
Affiliation(s)
- Gerald Zernig
- Experimental Psychiatry Unit, Department of Psychiatry 1, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
23
|
Banks ML, Snyder RW, Fennell TR, Negus SS. Role of d-amphetamine and d-methamphetamine as active metabolites of benzphetamine: Evidence from drug discrimination and pharmacokinetic studies in male rhesus monkeys. Pharmacol Biochem Behav 2017; 156:30-38. [PMID: 28373066 PMCID: PMC5485908 DOI: 10.1016/j.pbb.2017.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 11/19/2022]
Abstract
Benzphetamine is a Schedule III anorectic agent that is a prodrug for d-amphetamine and d-methamphetamine and may have utility as an "agonist" medication for cocaine use disorder treatment. This study evaluated the pharmacokinetic-pharmacodynamic profile of benzphetamine using a drug discrimination procedure in rhesus monkeys. The potency and time course of cocaine-like discriminative stimulus effects were compared for benzphetamine (10-18mg/kg, intramuscular (IM)) and d-amphetamine (0.032-0.32mg/kg, IM) in monkeys (n=3-4) trained to discriminate IM cocaine (0.32mg/kg) from saline in a two-key food-reinforced discrimination procedure. Parallel pharmacokinetic studies in the same monkeys determined plasma benzphetamine, d-methamphetamine and/or d-amphetamine levels for correlation with behavioral effects. d-Amphetamine produced dose-dependent, time-dependent, and full cocaine-like effects, i.e. ≥90% cocaine-appropriate responding, in all monkeys without altering response rates. The time course of d-amphetamine's cocaine-like discriminative stimulus effects correlated with plasma d-amphetamine levels. Benzphetamine was 180-fold less potent than d-amphetamine and produced full cocaine-like effects in only 2 of 4 monkeys while significantly decreasing response rates. Benzphetamine administration increased plasma d-methamphetamine (peak at 100min) and d-amphetamine (peak at 24h) levels, but the time course of behavioral effects did not correlate with increased levels of benzphetamine, d-methamphetamine or d-amphetamine. These results suggest that benzphetamine yields d-amphetamine and d-methamphetamine as active metabolites in rhesus monkeys, but generation of these metabolites is not sufficient to account for benzphetamine behavioral effects. The incomplete cocaine substitution profile and protracted d-amphetamine plasma levels suggest that benzphetamine may still warrant further evaluation as a candidate pharmacotherapy for cocaine use disorder treatment.
Collapse
Affiliation(s)
- Matthew L Banks
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Rodney W Snyder
- Department of Pharmacology & Toxicology, Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - Timothy R Fennell
- Department of Pharmacology & Toxicology, Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - S Stevens Negus
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
24
|
Weyandt LL, Oster DR, Marraccini ME, Gudmundsdottir BG, Munro BA, Rathkey ES, McCallum A. Prescription stimulant medication misuse: Where are we and where do we go from here? Exp Clin Psychopharmacol 2016; 24:400-414. [PMID: 27690507 PMCID: PMC5113141 DOI: 10.1037/pha0000093] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Prescription stimulants, including methylphenidate (e.g., Ritalin) and amphetamine compounds (e.g., dextroamphetamine; Adderall), have been approved by the U.S. Food and Drug Administration for the treatment of attention-deficit/hyperactivity disorder (ADHD) and are classified by the United States Drug Enforcement Administration as Schedule II medications because of their high potential for abuse and dependence (Drug Enforcement Administration, U.S. Department of Justice, 2015). Despite the potential health and judicial consequences, misuse of prescription stimulants, typically defined as taking stimulants without a valid prescription, or use of stimulants other than as prescribed, has become a serious problem in the United States and abroad, especially on college campuses. The purpose of the present article is to review historical information concerning prescription stimulants and to summarize the literature with respect to misuse among adults, particularly college students, including risk factors, mediators and moderators, and motivations for prescription stimulant misuse. In addition, evidence is presented concerning the question of whether prescription stimulants truly enhance cognitive functioning in individuals with and without ADHD, and the ethical and professional implications of these findings are explored. Lastly, recommendations for addressing prescription stimulant misuse and suggestions for future research are advanced. (PsycINFO Database Record
Collapse
|
25
|
Castells X, Cunill R, Pérez‐Mañá C, Vidal X, Capellà D. Psychostimulant drugs for cocaine dependence. Cochrane Database Syst Rev 2016; 9:CD007380. [PMID: 27670244 PMCID: PMC6457633 DOI: 10.1002/14651858.cd007380.pub4] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cocaine dependence is a severe disorder for which no medication has been approved. Like opioids for heroin dependence, replacement therapy with psychostimulants could be an effective therapy for treatment. OBJECTIVES To assess the effects of psychostimulants for cocaine abuse and dependence. Specific outcomes include sustained cocaine abstinence and retention in treatment. We also studied the influence of type of drug and comorbid disorders on psychostimulant efficacy. SEARCH METHODS This is an update of the review previously published in 2010. For this updated review, we searched the Cochrane Drugs and Alcohol Group Trials Register, CENTRAL, MEDLINE, Embase and PsycINFO up to 15 February 2016. We handsearched references of obtained articles and consulted experts in the field. SELECTION CRITERIA We included randomised parallel group controlled clinical trials comparing the efficacy of a psychostimulant drug versus placebo. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 26 studies involving 2366 participants. The included studies assessed nine drugs: bupropion, dexamphetamine, lisdexamfetamine, methylphenidate, modafinil, mazindol, methamphetamine, mixed amphetamine salts and selegiline. We did not consider any study to be at low risk of bias for all domains included in the Cochrane 'Risk of bias' tool. Attrition bias was the most frequently suspected potential source of bias of the included studies. We found very low quality evidence that psychostimulants improved sustained cocaine abstinence (risk ratio (RR) 1.36, 95% confidence interval (CI) 1.05 to 1.77, P = 0.02), but they did not reduce cocaine use (standardised mean difference (SMD) 0.16, 95% CI -0.02 to 0.33) among participants who continued to use it. Furthermore, we found moderate quality evidence that psychostimulants did not improve retention in treatment (RR 1.00, 95% CI 0.93 to 1.06). The proportion of adverse event-induced dropouts and cardiovascular adverse event-induced dropouts was similar for psychostimulants and placebo (RD 0.00, 95% CI -0.01 to 0.01; RD 0.00, 95% CI -0.02 to 0.01, respectively). When we included the type of drug as a moderating variable, the proportion of patients achieving sustained cocaine abstinence was higher with bupropion and dexamphetamine than with placebo. Psychostimulants also appeared to increase the proportion of patients achieving sustained cocaine and heroin abstinence amongst methadone-maintained, dual heroin-cocaine addicts. Retention to treatment was low, though, so our results may be compromised by attrition bias. We found no evidence of publication bias. AUTHORS' CONCLUSIONS This review found mixed results. Psychostimulants improved cocaine abstinence compared to placebo in some analyses but did not improve treatment retention. Since treatment dropout was high, we cannot rule out the possibility that these results were influenced by attrition bias. Existing evidence does not clearly demonstrate the efficacy of any pharmacological treatment for cocaine dependence, but substitution treatment with psychostimulants appears promising and deserves further investigation.
Collapse
Affiliation(s)
- Xavier Castells
- Universitat de GironaUnit of Clinical Pharmacology, TransLab Research Group, Department of Medical SciencesEmili Grahit, 77GironaCataloniaSpain17071
| | - Ruth Cunill
- Parc Sanitari Sant Joan de DéuParc Sanitari Sant Joan de Déu‐NumanciaBarcelonaCatalunyaSpain08735
| | - Clara Pérez‐Mañá
- Universitat Autònoma de BarcelonaIntegrative Pharmacology and Systems Neuroscience Research Group, Hospital del Mar Research Institute‐IMIM, Parc de Salut Mar, and Department of Pharmacology, Therapeutics and ToxicologyDoctor Aiguader 88BarcelonaCataloniaSpain08003
| | - Xavier Vidal
- Hospital Universitari Vall d'Hebron, Universitat Autònoma de BarcelonaDepartment of Clinical PharmacologyPasseig Vall d'Hebron 119‐129BarcelonaCataloniaSpain08035
| | - Dolors Capellà
- Faculty of Medicine, Universitat de GironaUnit of Clinical Pharmacology, TransLab Research Group, Department of Medical SciencesGironaSpain
| | | |
Collapse
|
26
|
Johnson AR, Banks ML, Blough BE, Lile JA, Nicholson KL, Negus SS. Development of a translational model to screen medications for cocaine use disorder I: Choice between cocaine and food in rhesus monkeys. Drug Alcohol Depend 2016; 165:103-10. [PMID: 27264165 PMCID: PMC4939093 DOI: 10.1016/j.drugalcdep.2016.05.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/09/2016] [Accepted: 05/24/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Homologous cocaine self-administration procedures in laboratory animals and humans may facilitate translational research for medications development to treat cocaine dependence. This study, therefore, sought to establish choice between cocaine and an alternative reinforcer in rhesus monkeys responding under a procedure back-translated from previous human studies and homologous to a human laboratory procedure described in a companion paper. METHODS Four rhesus monkeys with chronic indwelling intravenous catheters had access to cocaine injections (0, 0.043, 0.14, or 0.43mg/kg/injection) and food (0, 1, 3, or 10 1g banana-flavored food pellets). During daily 5h sessions, a single cocaine dose and a single food-reinforcer magnitude were available in 10 30-min trials. During the initial "sample" trial, the available cocaine and food reinforcer were delivered non-contingently. During each of the subsequent nine "choice" trials, responding could produce either the cocaine or food reinforcer under an independent concurrent progressive-ratio schedule. RESULTS Preference was governed by the cocaine dose and food-reinforcer magnitude, and increasing cocaine doses produced dose-dependent increases in cocaine choice at all food-reinforcer magnitudes. Effects of the candidate medication lisdexamfetamine (0.32-3.2mg/kg/day) were then examined on choice between 0.14mg/kg/injection cocaine and 10 pellets. Under baseline conditions, this reinforcer pair maintained an average of approximately 6 cocaine and 3 food choices. Lisdexamfetamine dose-dependently decreased cocaine choice in all monkeys, but food choice was not significantly altered. CONCLUSIONS These results support utility of this procedure in rhesus monkeys as one component of a platform for translational research on medications development to treat cocaine use disorder.
Collapse
Affiliation(s)
- Amy R. Johnson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA
| | - Matthew L. Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA
| | - Bruce E. Blough
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC
| | - Joshua A. Lile
- Departments of Behavioral Science, Psychiatry, and Psychology, University of Kentucky, Lexington, KY
| | - Katherine L. Nicholson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA
| | - S. Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
27
|
Development of a translational model to screen medications for cocaine use disorder II: Choice between intravenous cocaine and money in humans. Drug Alcohol Depend 2016; 165:111-9. [PMID: 27269368 PMCID: PMC4939714 DOI: 10.1016/j.drugalcdep.2016.05.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/22/2016] [Accepted: 05/02/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND A medication for treating cocaine use disorder has yet to be approved. Laboratory-based evaluation of candidate medications in animals and humans is a valuable means to demonstrate safety, tolerability and initial efficacy of potential medications. However, animal-to-human translation has been hampered by a lack of coordination. Therefore, we designed homologous cocaine self-administration studies in rhesus monkeys (see companion article) and human subjects in an attempt to develop linked, functionally equivalent procedures for research on candidate medications for cocaine use disorder. METHODS Eight (N=8) subjects with cocaine use disorder completed 12 experimental sessions in which they responded to receive money ($0.01, $1.00 and $3.00) or intravenous cocaine (0, 3, 10 and 30mg/70kg) under independent, concurrent progressive-ratio schedules. Prior to the completion of 9 choice trials, subjects sampled the cocaine dose available during that session and were informed of the monetary alternative value. RESULTS The allocation of behavior varied systematically as a function of cocaine dose and money value. Moreover, a similar pattern of cocaine choice was demonstrated in rhesus monkeys and humans across different cocaine doses and magnitudes of the species-specific alternative reinforcers. The subjective and cardiovascular responses to IV cocaine were an orderly function of dose, although heart rate and blood pressure remained within safe limits. CONCLUSIONS These coordinated studies successfully established drug versus non-drug choice procedures in humans and rhesus monkeys that yielded similar cocaine choice behavior across species. This translational research platform will be used in future research to enhance the efficiency of developing interventions to reduce cocaine use.
Collapse
|
28
|
Czoty PW, Stoops WW, Rush CR. Evaluation of the "Pipeline" for Development of Medications for Cocaine Use Disorder: A Review of Translational Preclinical, Human Laboratory, and Clinical Trial Research. Pharmacol Rev 2016; 68:533-62. [PMID: 27255266 PMCID: PMC4931869 DOI: 10.1124/pr.115.011668] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cocaine use disorder is a persistent public health problem for which no widely effective medications exist. Self-administration procedures, which have shown good predictive validity in estimating the abuse potential of drugs, have been used in rodent, nonhuman primate, and human laboratory studies to screen putative medications. This review assessed the effectiveness of the medications development process regarding pharmacotherapies for cocaine use disorder. The primary objective was to determine whether data from animal and human laboratory self-administration studies predicted the results of clinical trials. In addition, the concordance between laboratory studies in animals and humans was assessed. More than 100 blinded, randomized, fully placebo-controlled studies of putative medications for cocaine use disorder were identified. Of the 64 drugs tested in these trials, only 10 had been examined in both human and well-controlled animal laboratory studies. Within all three stages, few studies had been conducted for each drug and when multiple studies had been conducted conclusions were sometimes contradictory. Overall, however, there was good concordance between animal and human laboratory results when the former assessed chronic drug treatment. Although only seven of the ten reviewed drugs showed fully concordant results across all three types of studies reviewed, the analysis revealed several subject-related, procedural, and environmental factors that differ between the laboratory and clinical trial settings that help explain the disagreement for other drugs. The review closes with several recommendations to enhance translation and communication across stages of the medications development process that will ultimately speed the progress toward effective pharmacotherapeutic strategies for cocaine use disorder.
Collapse
Affiliation(s)
- Paul W Czoty
- Wake Forest School of Medicine, Winston-Salem, North Carolina (P.W.C.); and University of Kentucky, Lexington, Kentucky (W.W.S., C.R.R.)
| | - William W Stoops
- Wake Forest School of Medicine, Winston-Salem, North Carolina (P.W.C.); and University of Kentucky, Lexington, Kentucky (W.W.S., C.R.R.)
| | - Craig R Rush
- Wake Forest School of Medicine, Winston-Salem, North Carolina (P.W.C.); and University of Kentucky, Lexington, Kentucky (W.W.S., C.R.R.)
| |
Collapse
|
29
|
Nuijten M, Blanken P, van de Wetering B, Nuijen B, van den Brink W, Hendriks VM. Sustained-release dexamfetamine in the treatment of chronic cocaine-dependent patients on heroin-assisted treatment: a randomised, double-blind, placebo-controlled trial. Lancet 2016; 387:2226-34. [PMID: 27015909 DOI: 10.1016/s0140-6736(16)00205-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Heroin-assisted treatment is effective for methadone treatment-refractory heroin-dependent patients, but continued comorbid cocaine dependence remains problematic. Sustained-release dexamfetamine is a promising agonist pharmacotherapy for cocaine dependence and we aimed to assess its acceptance, efficacy, and safety. METHODS In this multicentre, randomised, double-blind, placebo-controlled trial, patients who were treatment-refractory, as indicated by at least two earlier failed treatments aimed at reducing or abstaining from cocaine use, and who regularly (≥8 days/month) used crack-cocaine were enrolled from four heroin-assisted treatment centres in the Netherlands. Eligible patients were randomly assigned (1:1) to receive either 12 weeks of daily, supervised prescription of 60 mg/day oral sustained-release dexamfetamine or placebo in addition to co-prescribed methadone and diacetylmorphine. Randomisation was done by the collaborating pharmacist, using a computer-generated random number sequence with stratification by treatment centre in blocks of four per stratum. Randomisation was masked to patients, staff, and researchers throughout the study. The primary outcome was the number of self-reported days of cocaine use during study treatment, assessed every 4 weeks. Primary and safety analyses were done in the intention-to-treat population. The study was registered with the European Union Drug Regulating Authorities Clinical Trials (EUdraCT 2013-004024-11) and with The Netherlands Trial Register (NTR2576). FINDINGS Between Aug 8, 2014, and Feb 27, 2015, 111 patients were assessed for eligibility, of whom 73 were enrolled and randomised; 38 patients were assigned to the sustained-release dexamfetamine group and 35 to the placebo group. Sustained-release dexamfetamine treatment resulted in significantly fewer days of cocaine use than placebo treatment (mean 44·9 days [SD 29·4] vs 60·6 days [24·3], respectively [95% CI of difference 3·1-28·4]; p=0·031; Cohen's standardised effect size d=0·58). One or more adverse events were reported by 28 (74%) patients in the dexamfetamine group and by 16 (46%) patients in the placebo group. Most adverse events were transient and well-tolerated. INTERPRETATION Sustained-release dexamfetamine is a well accepted, effective, and safe agonist pharmacotherapy for comorbid treatment-refractory cocaine dependence in heroin-dependent patients in heroin-assisted treatment. Future research should aim to replicate these findings in chronic cocaine-dependent and other stimulant-dependent patients in more routine treatment settings, including strategies to optimise treatment adherence like medication management interventions and contingency management. FUNDING Netherlands Organisation for Health Research and Development.
Collapse
Affiliation(s)
- Mascha Nuijten
- Parnassia Addiction Research Centre (PARC, Brijder Addiction Treatment), The Hague, Netherlands.
| | - Peter Blanken
- Parnassia Addiction Research Centre (PARC, Brijder Addiction Treatment), The Hague, Netherlands
| | | | - Bastiaan Nuijen
- Antoni van Leeuwenhoek/The Netherlands Cancer Institute, Medical Centre Slotervaart, Amsterdam, Netherlands
| | - Wim van den Brink
- Amsterdam Institute for Addiction Research, Department of Psychiatry, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Vincent M Hendriks
- Parnassia Addiction Research Centre (PARC, Brijder Addiction Treatment), The Hague, Netherlands; Curium, Leiden University Medical Centre, Department of Child and Adolescent Psychiatry, Leiden University, Leiden, Netherlands
| |
Collapse
|
30
|
Roncero C, Abad AC, Padilla-Mata A, Ros-Cucurull E, Barral C, Casas M, Grau-López L. Psychotic Symptoms Associated with the use of Dopaminergic Drugs, in Patients with Cocaine Dependence or Abuse. Curr Neuropharmacol 2016; 15:315-323. [PMID: 27009114 PMCID: PMC5412693 DOI: 10.2174/1570159x14666160324144912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/29/2015] [Accepted: 03/10/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND In the field of dual diagnosis, physicians are frequently presented with pharmacological questions. Questions about the risk of developing psychotic symptoms in cocaine users who need treatment with dopaminergic drugs could lead to an undertreatment. OBJECTIVE Review the presence of psychotic symptoms in patients with cocaine abuse/dependence, in treatment with dopaminergic drugs. METHODS Systematic PubMed searches were conducted including December 2014, using the keywords: "cocaine", dopaminergic drug ("disulfuram-methylphenidate-bupropion-bromocriptine-sibutramineapomorphine- caffeine") and ("psychosis-psychotic symptoms-delusional-paranoia"). Articles in English, Spanish, Portuguese, French, and Italian were included. Articles in which there was no history of cocaine abuse/dependence, absence of psychotic symptoms, systematic reviews, and animal studies, were excluded. RESULTS 313 papers were reviewed. 7 articles fulfilled the inclusion-exclusion criteria. There is a clinical trial including 8 cocaine-dependent patients using disulfiram in which 3 of them presented psychotic symptoms and 6 case-reports: disulfuram (1), methylphenidate (1), disulfiram with methylphenidate (2), and bupropion (2), reporting psychotic symptoms, especially delusions of reference and persecutory ideation. CONCLUSION Few cases have been described, which suggests that the appearance of these symptoms is infrequent. The synergy of dopaminergic effects or the dopaminergic sensitization in chronic consumption are the explanatory theories proposed by the authors. In these cases, a relationship was found between taking these drugs and the appearance of psychotic symptoms. Given the low number of studies found, further research is required. The risk of psychotic symptoms seems to be acceptable if we compare it with the benefits for the patients but a closer monitoring seems to be advisable.
Collapse
Affiliation(s)
- Carlos Roncero
- Department of Psychiatry, Vall d'Hebron University Hospital Passeig Vall d´Hebron, 119-129, 08035 Barcelona, EU, Spain
| | | | | | | | | | | | | |
Collapse
|
31
|
Czoty PW, Blough BE, Fennell TR, Snyder RW, Nader MA. Attenuation of cocaine self-administration by chronic oral phendimetrazine in rhesus monkeys. Neuroscience 2016; 324:367-76. [PMID: 26964683 DOI: 10.1016/j.neuroscience.2016.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/09/2016] [Accepted: 03/02/2016] [Indexed: 11/18/2022]
Abstract
Chronic treatment with the monoamine releaser d-amphetamine has been consistently shown to decrease cocaine self-administration in laboratory studies and clinical trials. However, the abuse potential of d-amphetamine is an obstacle to widespread clinical use. Approaches are needed that exploit the efficacy of the agonist approach but avoid the abuse potential associated with dopamine releasers. The present study assessed the effectiveness of chronic oral administration of phendimetrazine (PDM), a pro-drug for the monoamine releaser phenmetrazine (PM), to decrease cocaine self-administration in four rhesus monkeys. Each day, monkeys pressed a lever to receive food pellets under a 50-response fixed-ratio (FR) schedule of reinforcement and self-administered cocaine (0.003-0.56 mg/kg per injection, i.v.) under a progressive-ratio (PR) schedule in the evening. After completing a cocaine self-administration dose-response curve, sessions were suspended and PDM was administered (1.0-9.0 mg/kg, p.o., b.i.d.). Cocaine self-administration was assessed using the PR schedule once every 7 days while food-maintained responding was studied daily. When a persistent decrease in self-administration was observed, the cocaine dose-effect curve was re-determined. Daily PDM treatment decreased cocaine self-administration by 30-90% across monkeys for at least 4 weeks. In two monkeys, effects were completely selective for cocaine. Tolerance developed to initial decreases in food-maintained responding in the third monkey and in the fourth subject, fluctuations were observed that were lower in magnitude than effects on cocaine self-administration. Cocaine dose-effect curves were shifted down and/or rightward in three monkeys. These data provide further support for the use of agonist medications for cocaine abuse, and indicate that the promising effects of d-amphetamine extend to a more clinically viable pharmacotherapy.
Collapse
Affiliation(s)
- P W Czoty
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States.
| | - B E Blough
- Discovery Sciences, Research Triangle Institute, Research Triangle Park, NC 27709, United States
| | - T R Fennell
- Discovery Sciences, Research Triangle Institute, Research Triangle Park, NC 27709, United States
| | - R W Snyder
- Discovery Sciences, Research Triangle Institute, Research Triangle Park, NC 27709, United States
| | - M A Nader
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States; Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|