1
|
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
|
2
|
Bao W, Wang H, Wen L, Wu J, Gu YC, Wang X, Zhao Y. NMR-Based Chiral Discrimination of Bulky Amines with a 19F-Tagged NNO Pincer Complex. Anal Chem 2024; 96:11448-11454. [PMID: 38960938 DOI: 10.1021/acs.analchem.4c01748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Within pharmaceutical research, ensuring the enantiomeric purity of chiral compounds is critical. Specifically, chiral amines are a crucial category of compounds, due to their extensive therapeutic uses. However, the enantiomeric analysis of these compounds, particularly those with significant steric hindrance, remains a challenge. To address this issue, our research introduces a novel chiral 19F-tagged NNO palladium pincer probe, strategically engineered with an open binding site to accommodate bulky amines. This probe facilitates the enantiodifferentiation of such amines, as evidenced by the distinct 19F NMR signals generated by the enantiomers. Moreover, our findings highlight the probe's applicability in the chiral discrimination of various psychoactive substances, underscoring its potential for the identification of illegal stimulant use and contributing to forensic investigations.
Collapse
Affiliation(s)
- Wenjing Bao
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Ling-Ling Road, Shanghai 200032, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Lixian Wen
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Ling-Ling Road, Shanghai 200032, China
| | - Jian Wu
- Instrumental Analysis Center, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yu-Cheng Gu
- Syngenta, Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, United Kingdom
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yanchuan Zhao
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Ling-Ling Road, Shanghai 200032, China
- Instrumental Analysis Center, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
3
|
Estave PM, Albertson SE, Karkhanis AN, Jones SR. Co-targeting the kappa opioid receptor and dopamine transporter reduces motivation to self-administer cocaine and partially reverses dopamine system dysregulation. Sci Rep 2024; 14:6509. [PMID: 38499566 PMCID: PMC10948819 DOI: 10.1038/s41598-024-53463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/31/2024] [Indexed: 03/20/2024] Open
Abstract
Cocaine disrupts dopamine (DA) and kappa opioid receptor (KOR) system activity, with long-term exposure reducing inhibiton of DA uptake by cocaine and increasing KOR system function. Single treatment therapies have not been successful for cocaine use disorder; therefore, this study focuses on a combination therapy targeting the dopamine transporter (DAT) and KOR. Sprague Dawley rats self-administered 5 days of cocaine (1.5 mg/kg/inf, max 40 inf/day, FR1), followed by 14 days on a progressive ratio (PR) schedule (0.19 mg/kg/infusion). Behavioral effects of individual and combined administration of phenmetrazine and nBNI were then examined using PR. Additionally, ex vivo fast scan cyclic voltammetry was then used to assess alterations in DA and KOR system activity in the nucleus accumbens before and after treatments. Chronic administration of phenmetrazine as well as the combination of phenmetrazine and nBNI-but not nBNI alone-significantly reduced PR breakpoints. In addition, the combination of phenmetrazine and nBNI partially reversed cocaine-induced neurodysregulations of the KOR and DA systems, indicating therapeutic benefits of targeting the DA and KOR systems in tandem. These data highlight the potential benefits of the DAT and KOR as dual-cellular targets to reduce motivation to administer cocaine and reverse cocaine-induced alterations of the DA system.
Collapse
Affiliation(s)
- Paige M Estave
- Department of Physiology and Pharmacology, Wake University Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA
| | - Steven E Albertson
- Department of Physiology and Pharmacology, Wake University Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA
| | - Anushree N Karkhanis
- Department of Psychology, Binghamton University - State University of New York, Binghamton, NY, 13902, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake University Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA.
| |
Collapse
|
4
|
Malik JA, Agrewala JN. Future perspectives of emerging novel drug targets and immunotherapies to control drug addiction. Int Immunopharmacol 2023; 119:110210. [PMID: 37099943 DOI: 10.1016/j.intimp.2023.110210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 04/28/2023]
Abstract
Substance Use Disorder (SUD) is one of the major mental illnesses that is terrifically intensifying worldwide. It is becoming overwhelming due to limited options for treatment. The complexity of addiction disorders is the main impediment to understanding the pathophysiology of the illness. Hence, unveiling the complexity of the brain through basic research, identification of novel signaling pathways, the discovery of new drug targets, and advancement in cutting-edge technologies will help control this disorder. Additionally, there is a great hope of controlling the SUDs through immunotherapeutic measures like therapeutic antibodies and vaccines. Vaccines have played a cardinal role in eliminating many diseases like polio, measles, and smallpox. Further, vaccines have controlled many diseases like cholera, dengue, diphtheria, Haemophilus influenza type b (Hib), human papillomavirus, influenza, Japanese encephalitis, etc. Recently, COVID-19 was controlled in many countries by vaccination. Currently, continuous effort is done to develop vaccines against nicotine, cocaine, morphine, methamphetamine, and heroin. Antibody therapy against SUDs is another important area where serious attention is required. Antibodies have contributed substantially against many serious diseases like diphtheria, rabies, Crohn's disease, asthma, rheumatoid arthritis, and bladder cancer. Antibody therapy is gaining immense momentum due to its success rate in cancer treatment. Furthermore, enormous advancement has been made in antibody therapy due to the generation of high-efficiency humanized antibodies with a long half-life. The advantage of antibody therapy is its instant outcome. This article's main highlight is discussing the drug targets of SUDs and their associated mechanisms. Importantly, we have also discussed the scope of prophylactic measures to eliminate drug dependence.
Collapse
Affiliation(s)
- Jonaid Ahmad Malik
- Immunology laboratory, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Javed N Agrewala
- Immunology laboratory, Indian Institute of Technology Ropar, Rupnagar, Punjab, India.
| |
Collapse
|
5
|
Hartikainen M, Taipale H, Tanskanen A, Mittendorfer-Rutz E, Lähteenvuo M, Tiihonen J. Association of Pharmacological Treatments and Hospitalization and Death in Individuals With Amphetamine Use Disorders in a Swedish Nationwide Cohort of 13 965 Patients. JAMA Psychiatry 2023; 80:31-39. [PMID: 36383348 PMCID: PMC9669925 DOI: 10.1001/jamapsychiatry.2022.3788] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022]
Abstract
Importance There are no medications approved by authorities for the treatment of amphetamine or methamphetamine dependence, and studies investigating the effectiveness of pharmacological treatments in hard outcomes, such as hospitalization and death, are lacking. Objective To investigate the association between pharmacotherapies and hospitalization and mortality outcomes in persons with amphetamine or methamphetamine use disorder. Design, Setting, and Participants This nationwide register-based cohort study was conducted from July 2006 to December 2018 with a median (IQR) follow-up time of 3.9 (1.0-6.1) years. Data were analyzed from December 1, 2021, to May 24, 2022. All residents aged 16 to 64 years living in Sweden with a registered first-time diagnosis of amphetamine or methamphetamine use disorder and without previous diagnoses of schizophrenia or bipolar disorder were identified from nationwide registers of inpatient care, specialized outpatient care, sickness absence, and disability pension. Exposures Medications for substance use disorders (SUDs) or for attention-deficit/hyperactive disorder, mood stabilizers, antidepressants, benzodiazepines and related drugs, and antipsychotics. Medication use vs nonuse was modeled with the PRE2DUP (from prescription drug purchases to drug use periods) method. Main Outcomes and Measures Primary outcomes were hospitalization due to SUD and any hospitalization or death, which were analyzed using within-individual models by comparing use and nonuse periods of 17 specific medications or medication classes in the same individual to minimize selection bias. The secondary outcome was all-cause mortality, studied using between-individual analysis as traditional Cox models. Results There were 13 965 individuals in the cohort (9671 [69.3%] male; mean [SD] age, 34.4 [13.0] years). During follow-up, 7543 individuals (54.0%) were taking antidepressants, 6101 (43.7%) benzodiazepines, 5067 (36.3%) antipsychotics, 3941 (28.2%) ADHD medications (1511 [10.8%] were taking lisdexamphetamine), 2856 (20.5%) SUD medications, and 1706 (12.2%) mood stabilizers. A total of 10 341 patients (74.0%) were hospitalized due to SUDs, 11 492 patients (82.3%) were hospitalized due to any cause or died, and 1321 patients (9.5%) died of any cause. Lisdexamphetamine was the only medication in this study that was significantly associated with a decrease in risk of 3 outcomes (adjusted hazard ratio [aHR], 0.82; 95% CI, 0.72-0.94 for SUD hospitalization; aHR, 0.86; 95% CI, 0.78-0.95 for any hospitalization or death; aHR, 0.43; 95% CI, 0.24-0.77 for all-cause mortality). Methylphenidate use also was associated with lower all-cause mortality (aHR, 0.56; 95% CI, 0.43-0.74). Use of benzodiazepines was associated with a significantly higher risk of SUD hospitalization (aHR, 1.17; 95% CI, 1.12-1.22), any hospitalization or death (aHR, 1.20; 95% CI, 1.17-1.24), and all-cause mortality (aHR, 1.39; 95% CI, 1.20-1.60). Use of antidepressants or antipsychotics was associated with a slight increase in risk of SUD hospitalization (aHR, 1.07; 95% CI, 1.03-1.11 and aHR, 1.05; 95% CI, 1.01-1.09) as well as any hospitalization or death (aHR, 1.10; 95% CI, 1.06-1.14 and aHR, 1.06; 95% CI, 1.03-1.10, respectively). Conclusions and Relevance In this study, use of lisdexamphetamine was associated with improved outcomes in persons with amphetamine or methamphetamine use disorders, encouraging the conduct of randomized clinical trials. Prescription benzodiazepine use was associated with poor outcomes.
Collapse
Affiliation(s)
- Milja Hartikainen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Heidi Taipale
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Antti Tanskanen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ellenor Mittendorfer-Rutz
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Markku Lähteenvuo
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Jari Tiihonen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
- Department of Clinical Neuroscience, Karolinska Institutet & Centre for Psychiatry Research, Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| |
Collapse
|
6
|
Jacobs DS, Blough BE, Kohut SJ. Reinforcing and Stimulant-Like Effects of Methamphetamine Isomers in Rhesus Macaques. J Pharmacol Exp Ther 2021; 378:124-132. [PMID: 33986037 PMCID: PMC8407528 DOI: 10.1124/jpet.121.000548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/10/2021] [Indexed: 11/22/2022] Open
Abstract
Monoamine releasers such as d-methamphetamine (d-MA) can reduce cocaine use in laboratory studies and have been forwarded for the management of cocaine use disorder (CUD). However, the proven abuse liability of d-MA has limited enthusiasm for clinical use. The levorotatory isomer of MA, l-MA, appears to have lesser stimulant effects, possibly due to its preferential norepinephrine-releasing properties compared with dopamine. The present study evaluated the abuse potential of l-MA by comparing its reinforcing effects with known stimulant drugs of abuse in nonhuman primates. Adult rhesus macaques (N = 4) responded for intravenous injections of cocaine, d-MA, methcathinone (MCAT), or l-MA under a fixed-ratio (FR) schedule of reinforcement; reinforcing effectiveness was evaluated using behavioral economic demand procedures. In a separate cohort (N = 9), daily activity and food-reinforced responding were assessed during 100 days of treatment with daily dosages of l-MA (2.3 mg/kg per day, i.v.) or d-MA (0.74 mg/kg per day, i.v.) previously shown to decrease cocaine self-administration. Results show that all drugs maintained self-administration, with peak injections reaching ∼100 inj per session for cocaine, MCAT, and d-MA and ∼50 inj per session for l-MA . In demand studies, self-administration of each drug gradually decreased as FR size increased. The exponential model of demand indicated that the reinforcing effectiveness of l-MA was significantly less than the other drugs studied. Chronic l-MA treatment did not appreciably alter daily activity and only transiently suppressed food-reinforced responding. These data, coupled with previous findings that l-MA effectively reduces stimulant self-administration, suggest that l-MA, or other norepinephrine-preferring releasers, may serve as agonist medication for CUD with lesser abuse liability than common psychostimulants. SIGNIFICANCE STATEMENT: Development of pharmacotherapies for cocaine use disorder remains a formidable challenge. Agonist-based therapies show promise, but enthusiasm is tempered by the abuse liability of previously proposed medications. This study evaluated the abuse liability and chronic treatment effects of methamphetamine's levorotatory isomer (l-MA). l-MA demonstrated lower abuse liability compared with commonly abused stimulants and produced few untoward effects. In the context of recent studies demonstrating that l-MA attenuates stimulant self-administration, these findings support l-MA's potential as a pharmacotherapy for stimulant addiction.
Collapse
Affiliation(s)
- David S Jacobs
- McLean Hospital - Harvard Medical School, Belmont, Massachusetts (D.S.J., S.J.K.), and Research Triangle Institute, Research Triangle Park, North Carolina (B.E.B.)
| | - Bruce E Blough
- McLean Hospital - Harvard Medical School, Belmont, Massachusetts (D.S.J., S.J.K.), and Research Triangle Institute, Research Triangle Park, North Carolina (B.E.B.)
| | - Stephen J Kohut
- McLean Hospital - Harvard Medical School, Belmont, Massachusetts (D.S.J., S.J.K.), and Research Triangle Institute, Research Triangle Park, North Carolina (B.E.B.)
| |
Collapse
|
7
|
Ezard N, Clifford B, Dunlop A, Bruno R, Carr A, Liu Z, Siefried KJ, Lintzeris N. Safety and tolerability of oral lisdexamfetamine in adults with methamphetamine dependence: a phase-2 dose-escalation study. BMJ Open 2021; 11:e044696. [PMID: 34006547 PMCID: PMC8137170 DOI: 10.1136/bmjopen-2020-044696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To examine the safety of an agonist-type treatment, lisdexamfetamine (LDX), at 250 mg/day among adults with methamphetamine (MA) dependence. DESIGN A dose-escalating, phase-2, open-label, single-group study of oral LDX at two Australian drug treatment services. SETTING The study was conducted at two Australian stimulant use disorder treatment clinics. PARTICIPANTS There were 16 participants: at least 18 years old, MA dependent for at least the preceding 2 years using ICD-10 criteria, reporting use of MA on at least 14 of the preceding 28 days. INTERVENTIONS Daily, supervised LDX of 100-250 mg, single-blinded to dose, ascending-descending regimen over 8 weeks (100-250 mg over 4 weeks; followed by 4-week dose reduction regimen, 250-100 mg). Participants were followed through to week 12. OUTCOMES Primary outcomes were safety, drug tolerability and regimen completion at the end of week 4. Participants were followed to week 12. Secondary outcomes included: change in MA use; craving; withdrawal; severity of dependence; risk behaviour; change in other substance use; medication acceptability; potential for non-prescription use; adherence and neurocognitive functioning. RESULTS Fourteen of 16 participants (87.5%) completed escalation to 250 mg/day. Two participants withdrew from the trial in the first week: one relocated away from the study site, the other self-withdrew due to a possible, known side effect of LDX (agitation). There was one serious adverse event of suicidal ideation which resolved. All other adverse events were mild or moderate in severity and known side effects of LDX. No participant was withdrawn due to adverse events. MA use decreased from a median of 21 days (IQR: 16-23) to 13 days (IQR: 11-17) over the 4-week escalation period (p=0.013). CONCLUSIONS LDX at a dose of up to 250 mg/day was safe and well tolerated by study participants, warranting larger trials as a pharmacotherapy for MA dependence. TRIAL REGISTRATION NUMBER ACTRN12615000391572.
Collapse
Affiliation(s)
- Nadine Ezard
- Alcohol and Drug Service, St Vincent's Hospital Sydney, Darlinghurst, New South Wales, Australia
- National Centre for Clinical Research on Emerging Drugs, C/O University of New South Wales Faculty of Medicine, Sydney, New South Wales, Australia
- National Drug and Alcohol Research Centre, University of New South Wales Faculty of Medicine, Sydney, New South Wales, Australia
- Drug and Alcohol Clinical Research and Improvement Network, C/O South East Sydney Local Health District, Sydney, New South Wales, Australia
| | - Brendan Clifford
- Alcohol and Drug Service, St Vincent's Hospital Sydney, Darlinghurst, New South Wales, Australia
- Susan Wakil School of Nursing & Midwifery, The University of Sydney, Sydney, New South Wales, Australia
| | - Adrian Dunlop
- Drug and Alcohol Clinical Research and Improvement Network, C/O South East Sydney Local Health District, Sydney, New South Wales, Australia
- Drug and Alcohol Clinical Services, Hunter New England Local Health District, Newcastle, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle Faculty of Health, Callaghan, New South Wales, Australia
| | - Raimondo Bruno
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Andrew Carr
- Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, New South Wales, Australia
| | - Zhixin Liu
- Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, New South Wales, Australia
- Mark Wainwright Analytical Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Krista J Siefried
- Alcohol and Drug Service, St Vincent's Hospital Sydney, Darlinghurst, New South Wales, Australia
- National Centre for Clinical Research on Emerging Drugs, C/O University of New South Wales Faculty of Medicine, Sydney, New South Wales, Australia
- National Drug and Alcohol Research Centre, University of New South Wales Faculty of Medicine, Sydney, New South Wales, Australia
| | - Nicholas Lintzeris
- Drug and Alcohol Clinical Research and Improvement Network, C/O South East Sydney Local Health District, Sydney, New South Wales, Australia
- Division of Addiction Medicine, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia
- The Langton Centre, South East Sydney Local Health District, Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Noroozi A, Motevalian SA, Zarrindast MR, Alaghband-Rad J, Akhondzadeh S. Adding extended-release methylphenidate to psychological intervention for treatment of methamphetamine dependence: A double-blind randomized controlled trial. Med J Islam Repub Iran 2020; 34:137. [PMID: 33437733 PMCID: PMC7787037 DOI: 10.34171/mjiri.34.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Indexed: 11/29/2022] Open
Abstract
Background: Iran has been faced with an emerging epidemic of methamphetamine (MA) use during recent years. No effective pharmacotherapy has been identified for MA treatment; and psychological interventions are the only available effective treatment. The aim of this study is to investigate the efficacy and safety of extended-release methylphenidate (ER-MTP) for the treatment of methamphetamine dependence. Methods: Sixty-two people with methamphetamine dependence, according to DSM-IV-TR, were randomly assigned to either fixed-dose extended-release methylphenidate (ER-MTP) (60 mg per day) or placebo for 12 weeks. All participants received twice-weekly cognitive behavioral treatment for stimulant dependence. Recent drug use and craving level were measured using weekly rapid urine test and craving visual analogue scale, respectively. The severity of addiction was measured using the Addiction Severity Index at baseline and study completion. Assessment of MA withdrawal was conducted using Amphetamine Withdrawal Questionnaire and Amphetamine Selective Severity Assessment at baseline, day 3, week 1, week 4 and week 12. Depression and high-risk behaviors assessed with the Beck Depression Inventory and the high-risk behavior questionnaire at baseline, weeks 4 and 12 of the study. SPSS software version 22 was used for data analysis and p<0.05 was considered significant. Results: Percent of weekly MA negative urine tests was not significantly different between groups during the course of the study (p=0.766). Two groups showed similar retention rates. Changes in MA craving, withdrawal, addiction severity, depression and high-risk behaviors were not significantly different between groups. No serious adverse event was observed. Conclusion: Our finding did not show the superiority of fixed-schedule ER-MTP over placebo when added to an intensive biweekly outpatient psychosocial treatment. Further studies using individually tailored flexible-dose regimes might provide new insights regarding the safety and efficacy of psychostimulant maintenance treatment for MA dependence.
Collapse
Affiliation(s)
- Alireza Noroozi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, Tehran, Iran
- Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Abbas Motevalian
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
- Medical Genomics Research Center, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Alaghband-Rad
- Department of Psychiatry, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahin Akhondzadeh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, Tehran, Iran
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
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
|
10
|
Minkiewicz M, Czoty PW, Blough BE, Nader MA. Evaluation of the Reinforcing Strength of Phendimetrazine Using a Progressive-Ratio Schedule of Reinforcement in Rhesus Monkeys. J Pharmacol Exp Ther 2020; 374:1-5. [PMID: 32269168 PMCID: PMC7288731 DOI: 10.1124/jpet.120.264952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/01/2020] [Indexed: 12/21/2022] Open
Abstract
Stimulant abuse is a persistent public health problem with no Food and Drug Administration-approved pharmacotherapy. Although monoamine-releasing drugs such as d-amphetamine can decrease cocaine self-administration in human and animal laboratory studies, their potential for abuse limits clinical utility. "Abuse-deterrent" formulations of monoamine releasers, such as prodrugs, hold greater clinical promise if their abuse potential is, as theorized, lower than that of cocaine. In these studies, we determined the reinforcing strength of phendimetrazine (PDM), a prodrug for the amphetamine-like monoamine releaser phenmetrazine; both drugs have been shown to decrease cocaine self-administration in laboratory animals. To date, no study has directly compared PDM (Schedule III) with cocaine (Schedule II) under progressive-ratio (PR) schedules of reinforcement, which are better suited than fixed-ratio schedules to directly compare reinforcing strength of drugs. Dose-response curves for cocaine (saline, 0.001-0.3 mg/kg per injection) and PDM (0.1-1.0 mg/kg per injection) were generated in six cocaine-experienced male rhesus monkeys during 4-hour sessions with a 20-minute limited hold (LH). Under these conditions, the maximum number of injections was not significantly different between cocaine and PDM. The reinforcing strength of doses situated on the peaks of the cocaine and PDM dose-effect curves were redetermined with a 60-minute LH. The mean number of injections increased for both drugs, but not for saline. Cocaine presentations resulted in significantly higher peak injections than PDM with a 60-minute LH, which is consistent with the lower scheduling of PDM. These results support PDM as Schedule III and highlight the importance of schedule parameters when comparing reinforcing strength of drugs using a PR schedule of reinforcement. SIGNIFICANCE STATEMENT: One strategy for reducing cocaine use is to identify a treatment that substitutes for cocaine but has lower abuse potential. In a rhesus monkey model of drug abuse, this study compared the reinforcing strength of cocaine and phendimetrazine, a drug that has been shown to decrease cocaine use in some studies.
Collapse
Affiliation(s)
- Molly Minkiewicz
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina (M.M., P.W.C., M.A.N.) and Discovery Sciences, Research Triangle Institute, Research Triangle Park, North Carolina (B.E.B.)
| | - Paul W Czoty
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina (M.M., P.W.C., M.A.N.) and Discovery Sciences, Research Triangle Institute, Research Triangle Park, North Carolina (B.E.B.)
| | - Bruce E Blough
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina (M.M., P.W.C., M.A.N.) and Discovery Sciences, Research Triangle Institute, Research Triangle Park, North Carolina (B.E.B.)
| | - Michael A Nader
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina (M.M., P.W.C., M.A.N.) and Discovery Sciences, Research Triangle Institute, Research Triangle Park, North Carolina (B.E.B.)
| |
Collapse
|
11
|
Levin FR, Mariani JJ, Pavlicova M, Choi CJ, Mahony AL, Brooks DJ, Bisaga A, Dakwar E, Carpenter KM, Naqvi N, Nunes EV, Kampman K. Extended release mixed amphetamine salts and topiramate for cocaine dependence: A randomized clinical replication trial with frequent users. Drug Alcohol Depend 2020; 206:107700. [PMID: 31753736 PMCID: PMC6980777 DOI: 10.1016/j.drugalcdep.2019.107700] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Cocaine use disorder (CUD) remains a substantial public health problem with no clearly effective pharmacotherapy available. In a prior trial, combined amphetamine and topiramate treatment significantly reduced cocaine use among individuals demonstrating the most frequent use at baseline. This trial targeted such frequent users. METHODS A double-blind, randomized placebo-controlled trial, testing the combination of mixed amphetamine salts extended-release (MAS-ER) and topiramate or placebo over a 12-week medication phase was conducted. The two-site outpatient trial included 127 adults (96 males) with CUD using at least 9 days in the prior month. MAS-ER was titrated to a maximum dose of 60 mg/day and topiramate to a maximum dose of 100 mg twice/day. The primary outcome was the proportion of individuals who achieved three consecutive abstinent weeks at the end of the study (EOS) as measured by urine toxicology and self-report. RESULTS The proportion of participants achieving three abstinent weeks at the EOS was significantly (P = .03) larger in the treatment (14.1%) compared to the placebo group (0.0%), while controlling for baseline cocaine use, sex, current alcohol use disorder, and site. Of note, due to conservative cardiac safety-parameters a considerable number of individuals in the treatment group were discontinued from study medication (20.3%). CONCLUSIONS While these findings provide further evidence that the combination of MAS-ER and topiramate is efficacious in promoting abstinence in CUD adults with frequent use it remains possible that the combination treatment is no more effective than either treatment alone. Despite this, the study provides a valuable "proof of concept."
Collapse
Affiliation(s)
- Frances R Levin
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA; Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 630 West 168(th) Street, New York, NY 10032 USA.
| | - John J Mariani
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA; Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 630 West 168(th) Street, New York, NY 10032 USA
| | - Martina Pavlicova
- Department of Biostatistics, Columbia University, 722 West 168(th) Street, New York, NY 10032 USA
| | - C Jean Choi
- New York State Psychiatric Institute, Division of Biostatistics, 1051 Riverside Drive, New York, NY 10032 USA
| | - Amy L Mahony
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA
| | - Daniel J Brooks
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA
| | - Adam Bisaga
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA; Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 630 West 168(th) Street, New York, NY 10032 USA
| | - Elias Dakwar
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA; Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 630 West 168(th) Street, New York, NY 10032 USA
| | - Kenneth M Carpenter
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA; Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 630 West 168(th) Street, New York, NY 10032 USA
| | - Nasir Naqvi
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA; Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 630 West 168(th) Street, New York, NY 10032 USA
| | - Edward V Nunes
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA; Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 630 West 168(th) Street, New York, NY 10032 USA
| | - Kyle Kampman
- Department of Psychiatry, University of Pennsylvania, 3535 Market Street, Philadelphia, PA, 19104 USA
| |
Collapse
|
12
|
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
|
13
|
Solinas M, Belujon P, Fernagut PO, Jaber M, Thiriet N. Dopamine and addiction: what have we learned from 40 years of research. J Neural Transm (Vienna) 2018; 126:481-516. [PMID: 30569209 DOI: 10.1007/s00702-018-1957-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/17/2018] [Indexed: 12/22/2022]
Abstract
Among the neurotransmitters involved in addiction, dopamine (DA) is clearly the best known. The critical role of DA in addiction is supported by converging evidence that has been accumulated in the last 40 years. In the present review, first we describe the dopaminergic system in terms of connectivity, functioning and involvement in reward processes. Second, we describe the functional, structural, and molecular changes induced by drugs within the DA system in terms of neuronal activity, synaptic plasticity and transcriptional and molecular adaptations. Third, we describe how genetic mouse models have helped characterizing the role of DA in addiction. Fourth, we describe the involvement of the DA system in the vulnerability to addiction and the interesting case of addiction DA replacement therapy in Parkinson's disease. Finally, we describe how the DA system has been targeted to treat patients suffering from addiction and the result obtained in clinical settings and we discuss how these different lines of evidence have been instrumental in shaping our understanding of the physiopathology of drug addiction.
Collapse
Affiliation(s)
- Marcello Solinas
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.
| | - Pauline Belujon
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Pierre Olivier Fernagut
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Mohamed Jaber
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
- CHU de Poitiers, Poitiers, France
| | - Nathalie Thiriet
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| |
Collapse
|
14
|
Lee NK, Jenner L, Harney A, Cameron J. Pharmacotherapy for amphetamine dependence: A systematic review. Drug Alcohol Depend 2018; 191:309-337. [PMID: 30173086 DOI: 10.1016/j.drugalcdep.2018.06.038] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 06/20/2018] [Accepted: 06/21/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Demand for treatment for amphetamine use is increasing internationally. Establishing effective pharmacotherapy provides broader treatment options for people who are dependent on amphetamine and may encourage engagement in evidence-based behavioral treatment. This study aimed to identify medicines that have potential in improving treatment outcomes for people who are dependent on amphetamines. METHODS Medline, PsycINFO, Embase and the Cochrane Database of Systematic Reviews were searched from 1997 to 2012 and again from 2013 to 2016. Studies on medications for amphetamine/methamphetamine dependence treatment were selected and assessed by two independent researchers. A meta-narrative review approach was used to synthesize results. RESULTS A total of 49 studies investigating 20 potential pharmacotherapies were eligible for inclusion. Of these, 35 studies related to 33 level II quality randomized controlled trials (RCTs). Five medications were subject to multiple RCTs. Four of these medicines demonstrated some limited evidence of benefit for reducing amphetamine use: methylphenidate (as reported in three studies), bupropion (in three studies), modafinil (two studies), and naltrexone (one study). Four RCTs of dexamphetamine suggest its benefit on secondary outcomes such as treatment retention, but not for reducing amphetamine use. Six other medicines indicate the potential for efficacy, but the number of studies is too small to draw conclusions. CONCLUSIONS No medicine has as yet demonstrated sufficient, consistent evidence of effectiveness to support its use in routine treatment. High study drop-out and poor medication adherence limits the strength of evidence and raises important clinical questions about how to improve treatment engagement and outcomes.
Collapse
Affiliation(s)
- Nicole K Lee
- National Drug Research Institute, Curtin University, 7 Parker Place, Bentley, WA, 6102, Australia; 360Edge Consulting, P.O. Box 359, Elwood, 3184, Victoria, Australia.
| | - Linda Jenner
- 360Edge Consulting, P.O. Box 359, Elwood, 3184, Victoria, Australia
| | - Angela Harney
- 360Edge Consulting, P.O. Box 359, Elwood, 3184, Victoria, Australia
| | - Jacqui Cameron
- 360Edge Consulting, P.O. Box 359, Elwood, 3184, Victoria, Australia; Department of General Practice, University of Melbourne, 200 Berkeley Street, Carlton, Victoria, 3053, Australia
| |
Collapse
|
15
|
Ganti SS, Nguyen HX, Murnane KS, Blough BE, Banga AK. Transdermal formulation of 4-benzylpiperidine for cocaine-use disorder. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Ganti SS, Bhattaccharjee SA, Murnane KS, Blough BE, Banga AK. Formulation and evaluation of 4-benzylpiperidine drug-in-adhesive matrix type transdermal patch. Int J Pharm 2018; 550:71-78. [PMID: 30125654 DOI: 10.1016/j.ijpharm.2018.08.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/02/2018] [Accepted: 08/16/2018] [Indexed: 12/15/2022]
Abstract
The objective of our study was to develop a transdermal patch of 4-benzylpiperidine and to evaluate its in vitro transdermal permeation profile. Appropriate pressure sensitive adhesives and additives were selected based on solubility and slide crystallization studies. Release liners and backing membranes were selected based on their ability to peel without leaving a residue and their affinity to formulation respectively. Drug-in-adhesive patches developed were investigate for their in vitro drug permeation over 48 h across dermatomed human skin using Franz diffusion cells. Silicone based pressure sensitive adhesive along with colloidal silicon dioxide as viscosity builder, fluoropolymer coated membranes as the release liner and polyester based membranes as backing were chosen to develop a drug in silicone adhesive patch. Polyisobutylene adhesive based patch was developed with drug in polyisobutylene adhesive, along with oleic acid and oleyl alcohol as permeation enhancers, polyester for the release liner and polyethylene as backing. Among the patches developed, polyisobutylene adhesive based patch with higher drug concentration exhibited superior transdermal permeation (1608.5 ± 53.4 µg/cm2 over 48 h). The final patch was further tested for uniformity in coat weight, shear strength, tack and peel adhesion.
Collapse
Affiliation(s)
- Sindhu S Ganti
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Sonalika A Bhattaccharjee
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Kevin S Murnane
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Bruce E Blough
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - Ajay K Banga
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA.
| |
Collapse
|
17
|
|
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
|
Antinori S, Fattore L, Saba P, Fratta W, Gessa GL, Devoto P. Levodopa prevents the reinstatement of cocaine self-administration in rats via potentiation of dopamine release in the medial prefrontal cortex. Addict Biol 2018; 23:556-568. [PMID: 28429835 DOI: 10.1111/adb.12509] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 01/04/2023]
Abstract
Dopamine agonists have been proposed as therapeutic tools for cocaine addiction. We have recently demonstrated that indirect dopamine agonists, including levodopa (L-DOPA), markedly increase cocaine-induced dopamine release in the medial prefrontal cortex (mPFC) of rats leading to the suppression of cocaine-seeking behavior. This study was aimed to understand the behavioral and neurochemical effects of L-DOPA on cocaine-taking and cocaine-seeking in rats. After reaching a stable pattern of intravenous cocaine self-administration under a continuous fixed ratio (FR-1) schedule of reinforcement, male rats were treated with L-DOPA at different steps of the self-administration protocol. We found that L-DOPA reduced cocaine self-administration under FR-1 schedule of reinforcement and decreased the breaking points and the amount of cocaine self-administered under the progressive ratio schedule of reinforcement. Levodopa also decreased cocaine-seeking behavior both in a saline substitution test and in the cue priming-induced reinstatement test, without affecting general motor activity. Importantly, L-DOPA greatly potentiated cocaine-induced dopamine release in the mPFC of self-administering rats while reducing their cocaine intake. In the same brain area, L-DOPA also increased dopamine levels during cue priming-induced reinstatement of cocaine-seeking behavior. The potentiating effect was also evident in the mPFC but not nucleus accumbens core of drug-naïve rats passively administered with cocaine. Altogether, these findings demonstrate that L-DOPA efficaciously reduces the reinforcing and motivational effects of cocaine likely potentiating dopamine transmission in the mPFC. Its ability to prevent cue priming-induced reinstatement of cocaine-seeking suggests that it might be effective in reducing the risk to relapse to cocaine in abstinent patients.
Collapse
Affiliation(s)
- Silvia Antinori
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences; University of Cagliari; Italy
| | - Liana Fattore
- Institute of Neuroscience-Cagliari; National Research Council (CNR); Italy
- Center of Excellence ‘Neurobiology of Addiction’; University of Cagliari; Italy
| | - Pierluigi Saba
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences; University of Cagliari; Italy
| | - Walter Fratta
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences; University of Cagliari; Italy
- Center of Excellence ‘Neurobiology of Addiction’; University of Cagliari; Italy
| | - Gian Luigi Gessa
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences; University of Cagliari; Italy
- Institute of Neuroscience-Cagliari; National Research Council (CNR); Italy
- ‘Guy Everett Laboratory’; University of Cagliari; Italy
| | - Paola Devoto
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences; University of Cagliari; Italy
- Center of Excellence ‘Neurobiology of Addiction’; University of Cagliari; Italy
- ‘Guy Everett Laboratory’; University of Cagliari; Italy
| |
Collapse
|
20
|
Herbrink M, Thijssen B, Hillebrand MJX, Rosing H, Schellens JHM, Nuijen B, Beijnen JH. Development and validation of a high-performance liquid chromatography-tandem mass spectrometry assay for the quantification of Dexamphetamine in human plasma. J Pharm Biomed Anal 2017; 148:259-264. [PMID: 29059615 DOI: 10.1016/j.jpba.2017.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/12/2017] [Indexed: 11/18/2022]
Abstract
Dexamphetamine is registered for the treatment of attention deficit hyperactivity disorder and narcolepsy. Current research has highlighted the possible application of dexamphetamine in the treatment of cocaine addiction. To support clinical pharmacologic trials a new simple, fast, and sensitive assay for the quantification of dexamphetamine in human plasma using liquid chromatography tandem mass spectrometry (LC-MS/MS) was developed. Additionally, it is the first reported LC-MS assay with these advantages to be fully validated according to current US FDA and EMA guidelines. Human plasma samples were collected on an outpatient basis and stored at nominally -20°C. The analyte and the internal standard (stable isotopically labeled dexamphetamine) were extracted using double liquid-liquid extraction (plasma-organic and organic-water) combined with snap-freezing. The aqueous extract was filtered and 2μL was injected on a C18-column with isocratic elution and analyzed with triple quadrupole mass spectrometry in positive ion mode. The validated concentration range was from 2.5-250ng/mL and the calibration model was linear. A weighting factor of 1 over the squared concentration was applied and correlation coefficients of 0.997 or better were obtained. At all concentrations the bias was within ±15% of the nominal concentrations and imprecision was ≤15%. All results were within the acceptance criteria of the latest US FDA guidance and EMA guidelines on method validation. In conclusion, the developed method to quantify dexamphetamine in human plasma was fit to support a clinical study with slow-release dexamphetamine.
Collapse
Affiliation(s)
- M Herbrink
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| | - B Thijssen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| | - M J X Hillebrand
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| | - H Rosing
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| | - J H M Schellens
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands; Department of Pharmacoepidemiology and Clinical Pharmacology, Science Faculty, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - B Nuijen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| | - J H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands; Department of Pharmacoepidemiology and Clinical Pharmacology, Science Faculty, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| |
Collapse
|
21
|
Cook J, Lloyd-Jones M, Arunogiri S, Ogden E, Bonomo Y. Managing attention deficit hyperactivity disorder in adults using illicit psychostimulants: A systematic review. Aust N Z J Psychiatry 2017. [PMID: 28639480 DOI: 10.1177/0004867417714878] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
CONTEXT Attention deficit hyperactivity disorder and stimulant use disorder commonly co-exist, and appropriate treatments have not been well established. OBJECTIVE To provide guidance for treatment of co-existing attention deficit hyperactivity disorder and stimulant use disorder. DATA SOURCES A systematic review of published English articles using MEDLINE, EMBASE, CINAHL, PsycINFO and Cochrane, utilising consistent search terms. STUDY SELECTION Randomised controlled trials, comparing any treatment arm with a control group, for participants meeting Diagnostic and Statistical Manual of Mental Disorders or equivalent criteria for both attention deficit hyperactivity disorder and stimulant use disorder. RESULTS Eight trials were identified for inclusion in this review. Four of eight studies showed improvement in attention deficit hyperactivity disorder outcome measures compared with placebo. Two of six studies that reported substance use outcomes showed improvement in treatment arms compared with placebo. Studies to show effect tended to be those with the highest treatment dosage. CONCLUSION Evidence for the efficacy of treatment of patients with comorbid stimulant use disorder and attention deficit hyperactivity disorder is limited. Promising outcomes need replication in further studies utilising higher treatment dosage.
Collapse
Affiliation(s)
- Jon Cook
- 1 Drug Health Services, Western Health, Footscray, VIC, Australia.,2 Youth Support + Advocacy Service, Fitzroy, VIC, Australia
| | - Martyn Lloyd-Jones
- 3 Department of Addiction Medicine, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia.,4 Delmont Private Hospital, Glen Iris, VIC, Australia
| | - Shalini Arunogiri
- 5 Turning Point, Eastern Health, Fitzroy, VIC, Australia.,6 Monash University, Melbourne, VIC, Australia
| | - Edward Ogden
- 3 Department of Addiction Medicine, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia.,7 Swinburne University of Technology, Melbourne, VIC, Australia.,8 Youth Health and Rehabilitation Service (YHaRS), Fitzroy, VIC, Australia
| | - Yvonne Bonomo
- 3 Department of Addiction Medicine, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia.,9 Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
22
|
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
|
23
|
Abuse Potential of Oral Phendimetrazine in Cocaine-dependent Individuals: Implications for Agonist-like Replacement Therapy. J Addict Med 2017; 10:156-65. [PMID: 26933876 DOI: 10.1097/adm.0000000000000206] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Phendimetrazine is a prodrug for the monoamine releaser phenmetrazine-a drug with known abuse potential. Preclinical studies suggest that phendimetrazine has limited abuse potential and may have promise as an agonist-like replacement therapy for cocaine dependence. This study evaluated the abuse potential of phendimetrazine in humans. METHODS Nine cocaine-dependent individuals (N = 9) were enrolled to investigate the abuse potential of phendimetrazine and d-amphetamine, using a double-blind, placebo-controlled, within-subject design. Subjective and cardiovascular effects of oral phendimetrazine (35, 70, and 105 mg), d-amphetamine (10, 20, and 30 mg), and placebo were assessed in quasi-random order across 8 sessions lasting for approximately 8 hours each. RESULTS d-Amphetamine (20 and 30 mg) significantly increased cardiovascular measures in a time and dose-related manner, but phendimetrazine did not systematically alter cardiovascular measures. Although d-amphetamine and phendimetrazine significantly increased ratings indicative of abuse potential (eg, drug liking) and stimulant-like effects relative to placebo, these increases were generally small in magnitude, with phendimetrazine producing significant effects on fewer abuse-related measures and at fewer time points than d-amphetamine. CONCLUSIONS These preliminary findings suggest that oral phendimetrazine and d-amphetamine may have limited abuse potential in cocaine-dependent individuals. These findings collectively emphasize that the clinical utility of medications to treat cocaine-use disorders should be weighed carefully against their potential for abuse and diversion, with careful attention paid to evaluating abuse potential in a clinically relevant population of interest. Future studies are needed to further elucidate the potential utility of phendimetrazine as an agonist-like replacement therapy for cocaine dependence.
Collapse
|
24
|
Ezard N, Dunlop A, Clifford B, Bruno R, Carr A, Bissaker A, Lintzeris N. Study protocol: a dose-escalating, phase-2 study of oral lisdexamfetamine in adults with methamphetamine dependence. BMC Psychiatry 2016; 16:428. [PMID: 27905916 PMCID: PMC5134059 DOI: 10.1186/s12888-016-1141-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 11/25/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The treatment of methamphetamine dependence is a continuing global health problem. Agonist type pharmacotherapies have been used successfully to treat opioid and nicotine dependence and are being studied for the treatment of methamphetamine dependence. One potential candidate is lisdexamfetamine, a pro-drug for dexamphetamine, which has a longer lasting therapeutic action with a lowered abuse potential. The purpose of this study is to determine the safety of lisdexamfetamine in this population at doses higher than those currently approved for attention deficit hyperactivity disorder or binge eating disorder. METHODS/DESIGN This is a phase 2 dose escalation study of lisdexamfetamine for the treatment of methamphetamine dependence. Twenty individuals seeking treatment for methamphetamine dependence will be recruited at two Australian drug and alcohol services. All participants will undergo a single-blinded ascending-descending dose regime of 100 to 250 mg lisdexamfetamine, dispensed daily on site, over an 8-week period. Participants will be offered counselling as standard care. For the primary objectives the outcome variables will be adverse events monitoring, drug tolerability and regimen completion. Secondary outcomes will be changes in methamphetamine use, craving, withdrawal, severity of dependence, risk behaviour and other substance use. Medication acceptability, potential for non-prescription use, adherence and changes in neurocognition will also be measured. DISCUSSION Determining the safety of lisdexamfetamine will enable further research to develop pharmacotherapies for the treatment of methamphetamine dependence. TRIAL REGISTRATION Australian and New Zealand Clinical Trials Registry ACTRN12615000391572 Registered 28th April 2015.
Collapse
Affiliation(s)
- Nadine Ezard
- Alcohol and Drug Service, O’Brien Centre, St Vincent’s Hospital, Sydney, Darlinghurst, 2010 NSW Australia ,St Vincent’s Hospital Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
| | - Adrian Dunlop
- Drug and Alcohol Clinical Services, Hunter New England Local Health District, Newcastle Community Health Centre, Newcastle, 2302 NSW Australia
| | - Brendan Clifford
- Alcohol and Drug Service, O'Brien Centre, St Vincent's Hospital, Sydney, Darlinghurst, 2010, NSW, Australia.
| | - Raimondo Bruno
- School of Medicine, University of Tasmania, Private Bag 30, Hobart, 7001 TAS Australia
| | - Andrew Carr
- Centre for Applied Medical Research, St Vincent’s Hospital, 390 Victoria Street, Darlinghurst, 2010 NSW Australia
| | - Alexandra Bissaker
- Alcohol and Drug Service, O’Brien Centre, St Vincent’s Hospital, Sydney, Darlinghurst, 2010 NSW Australia
| | - Nicholas Lintzeris
- South East Sydney Local Health District, The Langton Centre, 591 South Dowling St, Surry Hills, 2010 NSW Australia ,Discipline of Addiction Medicine and Lambert initiative in Cannabinoid Therapeutics, University of Sydney, Sydney, 2006 NSW Australia
| |
Collapse
|
25
|
Wardle MC, Vincent JN, Suchting R, Green CE, Lane SD, Schmitz JM. Anhedonia Is Associated with Poorer Outcomes in Contingency Management for Cocaine Use Disorder. J Subst Abuse Treat 2016; 72:32-39. [PMID: 27646197 DOI: 10.1016/j.jsat.2016.08.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 08/22/2016] [Accepted: 08/31/2016] [Indexed: 12/17/2022]
Abstract
This study explored anhedonia (lack of interest or pleasure in non-drug rewards) as a potentially modifiable individual difference associated with the effectiveness of Contingency Management (CM). It also tested the hypothesis that a dopaminergic drug, levodopa (L-DOPA), would improve the effectiveness of CM, particularly in individuals high in anhedonia. The study was a single-site, randomized, double-blind, parallel group, 12-week trial comparing L-DOPA with placebo, with both medication groups receiving voucher-based CM targeting cocaine-negative urines. Participants were N=85 treatment-seeking adults with CUD. Anhedonia was measured at baseline using a validated self-report measure and a progressive ratio behavioral measure. Treatment Effectiveness Score (TES) was defined as the total number of cocaine-negative urines submitted. Analyses based on Frequentist general linear models were not significant, but Bayesian analyses indicated a high probability (92.6%) that self-reported anhedonia was associated with poor treatment outcomes (lower TES). L-DOPA did not significantly improve outcomes, nor was the effect of L-DOPA moderated by anhedonia. While the study failed to replicate positive findings from previous studies of L-DOPA in combination with CM, it does provide preliminary evidence that anhedonia may be a modifiable individual difference associated with poorer CM outcomes.
Collapse
Affiliation(s)
- Margaret C Wardle
- Center for Neurobehavioral Research on Addiction, University of Texas Medical Center at Houston, 1941 East Rd., Houston, TX, 77054, USA.
| | - Jessica N Vincent
- Center for Neurobehavioral Research on Addiction, University of Texas Medical Center at Houston, 1941 East Rd., Houston, TX, 77054, USA
| | - Robert Suchting
- Center for Neurobehavioral Research on Addiction, University of Texas Medical Center at Houston, 1941 East Rd., Houston, TX, 77054, USA
| | - Charles E Green
- Center for Neurobehavioral Research on Addiction, University of Texas Medical Center at Houston, 1941 East Rd., Houston, TX, 77054, USA
| | - Scott D Lane
- Center for Neurobehavioral Research on Addiction, University of Texas Medical Center at Houston, 1941 East Rd., Houston, TX, 77054, USA; Graduate School of Biomedical Sciences, University of Texas Medical Center at Houston, 1941 East Rd., Houston, TX, 77054, USA
| | - Joy M Schmitz
- Center for Neurobehavioral Research on Addiction, University of Texas Medical Center at Houston, 1941 East Rd., Houston, TX, 77054, USA; Graduate School of Biomedical Sciences, University of Texas Medical Center at Houston, 1941 East Rd., Houston, TX, 77054, USA
| |
Collapse
|
26
|
Bolin BL, Alcorn JL, Reynolds AR, Lile JA, Rush CR. Human drug discrimination: A primer and methodological review. Exp Clin Psychopharmacol 2016; 24:214-28. [PMID: 27454673 PMCID: PMC4965187 DOI: 10.1037/pha0000077] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Drug-discrimination procedures empirically evaluate the control that internal drug states exert over behavior. They provide a highly selective method to investigate the neuropharmacological underpinnings of the interoceptive effects of drugs. Historically, drug discrimination has been one of the most widely used assays in the field of behavioral pharmacology. Drug-discrimination procedures have also been adapted for use with humans and are conceptually similar to preclinical drug-discrimination techniques in that a behavior is differentially reinforced contingent on the presence or absence of a specific interoceptive drug stimulus. This review gives some general history and background concerning the major theoretical concepts and principles of drug-discrimination research as well as its relevance to substance-use disorders. This article also provides a procedural overview and discusses key methodological issues that must be considered when designing and conducting a human drug-discrimination study. Although drug discrimination is unequivocally one of the most sophisticated and useful behavioral assays to investigate the underlying neuropharmacology of drugs in vivo, enthusiasm for its use has steadily declined in the last decade and a half. We conclude by commenting on the current state of drug-discrimination research and suggest potential avenues for future drug-discrimination research. (PsycINFO Database Record
Collapse
Affiliation(s)
- B. Levi Bolin
- Department of Behavioral Science, University of Kentucky College of Medicine, 140 Medical Behavioral Science Building, Lexington, KY, 40536-0086, U.S.A
| | - Joseph L. Alcorn
- Department of Behavioral Science, University of Kentucky College of Medicine, 140 Medical Behavioral Science Building, Lexington, KY, 40536-0086, U.S.A
| | - Anna R. Reynolds
- Department of Behavioral Science, University of Kentucky College of Medicine, 140 Medical Behavioral Science Building, Lexington, KY, 40536-0086, U.S.A
| | - Joshua A. Lile
- Department of Behavioral Science, University of Kentucky College of Medicine, 140 Medical Behavioral Science Building, Lexington, KY, 40536-0086, U.S.A,Department of Psychology, University of Kentucky College of Arts and Sciences, 110 Kastle Hall, Lexington, KY 40506-0044, U.S.A,Department of Psychiatry, University of Kentucky College of Medicine, 3470 Blazer Parkway, Lexington, KY 40509, U.S.A
| | - Craig R. Rush
- Department of Behavioral Science, University of Kentucky College of Medicine, 140 Medical Behavioral Science Building, Lexington, KY, 40536-0086, U.S.A,Department of Psychology, University of Kentucky College of Arts and Sciences, 110 Kastle Hall, Lexington, KY 40506-0044, U.S.A,Department of Psychiatry, University of Kentucky College of Medicine, 3470 Blazer Parkway, Lexington, KY 40509, U.S.A
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Stoops WW, Strickland JC, Hays LR, Rayapati AO, Lile JA, Rush CR. Safety and tolerability of intranasal cocaine during phendimetrazine maintenance. Psychopharmacology (Berl) 2016; 233:2055-2063. [PMID: 26932737 PMCID: PMC4864128 DOI: 10.1007/s00213-016-4260-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/19/2016] [Indexed: 01/09/2023]
Abstract
RATIONALE Phendimetrazine appears to have limited abuse potential and reduces cocaine self-administration in preclinical studies. No human studies have evaluated the safety and tolerability of cocaine in combination with phendimetrazine, which is a necessary next step in evaluating the efficacy of phendimetrazine for treating cocaine use disorder. OBJECTIVES This study determined the safety and tolerability of acute cocaine doses during chronic phendimetrazine treatment. METHODS Ten subjects completed this within-subject, placebo-controlled, inpatient study. Subjects were maintained on ascending oral phendimetrazine doses (0, 70, 140, and 210 mg/day). After at least seven maintenance days at each dose, subjects received ascending doses of intranasal cocaine (0, 10, 20, 40, and 80 mg), separated by 90 min, within one session. RESULTS Cocaine produced prototypical cardiovascular and subject-rated effects (e.g., increased blood pressure and ratings of like drug). The cardiovascular effects of cocaine alone were not clinically significant for an acute drug response (e.g., average heart rate did not approach tachycardia, 100 beats/min). Phendimetrazine enhanced peak heart rate following placebo and low cocaine doses, but these effects were also not clinically significant. Phendimetrazine was otherwise devoid of effects alone and did not alter the subject-rated effects of cocaine or hypothetical demand for cocaine on a purchase task. CONCLUSIONS Cocaine was safe and well tolerated during maintenance on a threefold range of phendimetrazine doses. Given this safety profile, the reduced abuse potential of phendimetrazine and promising preclinical research, future human laboratory studies, and possibly clinical trials should evaluate the efficacy of phendimetrazine for reducing cocaine use.
Collapse
Affiliation(s)
- William W Stoops
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Room 140, Lexington, KY, 40536-0086, USA.
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY, 40509-1810, USA.
- Department of Psychology, University of Kentucky College of Arts and Sciences, Kastle Hall, Lexington, KY, 40506-0044, USA.
| | - Justin C Strickland
- Department of Psychology, University of Kentucky College of Arts and Sciences, Kastle Hall, Lexington, KY, 40506-0044, USA
| | - Lon R Hays
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY, 40509-1810, USA
| | - Abner O Rayapati
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY, 40509-1810, USA
| | - Joshua A Lile
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Room 140, Lexington, KY, 40536-0086, USA
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY, 40509-1810, USA
- Department of Psychology, University of Kentucky College of Arts and Sciences, Kastle Hall, Lexington, KY, 40506-0044, USA
| | - Craig R Rush
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Room 140, Lexington, KY, 40536-0086, USA
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY, 40509-1810, USA
- Department of Psychology, University of Kentucky College of Arts and Sciences, Kastle Hall, Lexington, KY, 40506-0044, USA
| |
Collapse
|
30
|
Reed SC, Evans SM. The effects of oral d-amphetamine on impulsivity in smoked and intranasal cocaine users. Drug Alcohol Depend 2016; 163:141-52. [PMID: 27114203 PMCID: PMC4880502 DOI: 10.1016/j.drugalcdep.2016.04.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 04/07/2016] [Accepted: 04/09/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND Effective treatments for cocaine use disorders remain elusive. Two factors that may be related to treatment failures are route of cocaine used and impulsivity. Smoked cocaine users are more likely to have poorer treatment outcomes compared to intranasal cocaine users. Further, cocaine users are impulsive and impulsivity is associated with poor treatment outcomes. While stimulants are used to treat Attention Deficit Hyperactivity Disorder (ADHD) and attenuate certain cocaine-related behaviors, few studies have comprehensively examined whether stimulants can reduce behavioral impulsivity in cocaine users, and none examined route of cocaine use as a factor. METHODS The effects of immediate release oral d-amphetamine (AMPH) were examined in 34 cocaine users (13 intranasal, 21 smoked). Participants had three separate sessions where they were administered AMPH (0, 10, or 20mg) and completed behavioral measures of impulsivity and risk-taking and subjective measures of abuse liability. RESULTS Smoked cocaine users were more impulsive on the Delayed Memory Task, the GoStop task and the Delay Discounting Task than intranasal cocaine users. Smoked cocaine users also reported more cocaine craving and negative mood than intranasal cocaine users. AMPH produced minimal increases on measures of abuse liability (e.g., Drug Liking). CONCLUSIONS Smoked cocaine users were more impulsive than intranasal cocaine users on measures of impulsivity that had a delay component. Additionally, although AMPH failed to attenuate impulsive responding, there was minimal evidence of abuse liability in cocaine users. These preliminary findings need to be confirmed in larger samples that control for route and duration of cocaine use.
Collapse
Affiliation(s)
- Stephanie Collins Reed
- Division on Substance Abuse, New York State Psychiatric Institute and Department of Psychiatry, Columbia University Medical Center, 1051 Riverside Drive, Unit 66, New York, NY 10032 USA.
| | - Suzette M Evans
- Division on Substance Abuse, New York State Psychiatric Institute and Department of Psychiatry, Columbia University Medical Center, 1051 Riverside Drive, Unit 66, New York, NY 10032 USA
| |
Collapse
|
31
|
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
|
32
|
Phillips TJ, Mootz JRK, Reed C. Identification of Treatment Targets in a Genetic Mouse Model of Voluntary Methamphetamine Drinking. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:39-85. [PMID: 27055611 DOI: 10.1016/bs.irn.2016.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Methamphetamine has powerful stimulant and euphoric effects that are experienced as rewarding and encourage use. Methamphetamine addiction is associated with debilitating illnesses, destroyed relationships, child neglect, violence, and crime; but after many years of research, broadly effective medications have not been identified. Individual differences that may impact not only risk for developing a methamphetamine use disorder but also affect treatment response have not been fully considered. Human studies have identified candidate genes that may be relevant, but lack of control over drug history, the common use or coabuse of multiple addictive drugs, and restrictions on the types of data that can be collected in humans are barriers to progress. To overcome some of these issues, a genetic animal model comprised of lines of mice selectively bred for high and low voluntary methamphetamine intake was developed to identify risk and protective alleles for methamphetamine consumption, and identify therapeutic targets. The mu opioid receptor gene was supported as a target for genes within a top-ranked transcription factor network associated with level of methamphetamine intake. In addition, mice that consume high levels of methamphetamine were found to possess a nonfunctional form of the trace amine-associated receptor 1 (TAAR1). The Taar1 gene is within a mouse chromosome 10 quantitative trait locus for methamphetamine consumption, and TAAR1 function determines sensitivity to aversive effects of methamphetamine that may curb intake. The genes, gene interaction partners, and protein products identified in this genetic mouse model represent treatment target candidates for methamphetamine addiction.
Collapse
Affiliation(s)
- T J Phillips
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States; Veterans Affairs Portland Health Care System, Portland, OR, United States.
| | - J R K Mootz
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States
| | - C Reed
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
33
|
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
|
34
|
Kohut SJ, Bergman J, Blough BE. Effects of L-methamphetamine treatment on cocaine- and food-maintained behavior in rhesus monkeys. Psychopharmacology (Berl) 2016; 233:1067-75. [PMID: 26713332 PMCID: PMC4761269 DOI: 10.1007/s00213-015-4186-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
RATIONALE Monoamine releasers with prominent dopaminergic actions, e.g., D-methamphetamine (D-MA), significantly reduce cocaine use and craving in clinical and preclinical laboratory studies. However, D-MA and related drugs also display high abuse potential, which limits their acceptability as agonist replacement medications for the management of Cocaine Use Disorder. OBJECTIVES The L-isomer of methamphetamine (L-MA), unlike D-MA, has preferential noradrenergic actions and is used medicinally with low, if any, abuse liability. The present study was conducted to determine whether L-MA could serve as an agonist replacement medication by both mimicking interoceptive effects of cocaine and decreasing intravenous (IV) cocaine self-administration. METHODS Separate groups (N = 4-5) of rhesus monkeys were studied to determine whether L-MA could (1) substitute for cocaine in subjects that discriminated intramuscular (IM) cocaine (0.4 mg/kg) from saline and (2) decrease IV cocaine self-administration under a second-order FR2(VR16:S) schedule of reinforcement. RESULTS L-MA, like D-MA but with approximately 5-fold lesser potency, substituted for cocaine in drug discrimination experiments in a dose-dependent manner. In IV self-administration studies, 5-10-day treatments with continuously infused L-MA (0.032-0.32 mg/kg/h, IV) dose-dependently decreased cocaine-maintained responding; the highest dosage reduced cocaine intake to levels of saline self-administration without appreciable effects on food-maintained responding. CONCLUSIONS These results indicate that L-MA both shares discriminative stimulus effects with cocaine and reduces cocaine self-administration in a behaviorally selective manner. L-MA and other compounds with a similar pharmacological profile deserve further evaluation for the management of Cocaine Use Disorder.
Collapse
Affiliation(s)
- Stephen J. Kohut
- Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - Jack Bergman
- Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - Bruce E. Blough
- Center for Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, NC, USA
| |
Collapse
|
35
|
Decker AM, Partilla JS, Baumann MH, Rothman RB, Blough BE. The biogenic amine transporter activity of vinylogous amphetamine analogs. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00245e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Vinylogous amphetamine analog S-6 is a potent dual dopamine/serotonin (DA/5-HT) releaser with no activity at 5-HT2 receptors.
Collapse
Affiliation(s)
| | - John S. Partilla
- Medicinal Chemistry Section
- Intramural Research Program
- National Institute on Drug Abuse
- National Institutes of Health
- Baltimore
| | - Michael H. Baumann
- Medicinal Chemistry Section
- Intramural Research Program
- National Institute on Drug Abuse
- National Institutes of Health
- Baltimore
| | - Richard B. Rothman
- Medicinal Chemistry Section
- Intramural Research Program
- National Institute on Drug Abuse
- National Institutes of Health
- Baltimore
| | | |
Collapse
|
36
|
Dopaminergic mediation of the discriminative stimulus functions of modafinil in rats. Psychopharmacology (Berl) 2015; 232:4411-9. [PMID: 26374456 DOI: 10.1007/s00213-015-4065-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 08/30/2015] [Indexed: 10/23/2022]
Abstract
RATIONALE Modafinil is a wake-promoting drug with FDA approval for the treatment of excessive daytime sleepiness that has been prescribed for ADHD and recently assessed as a potential treatment for psychostimulant dependence. Previous research indicates that modafinil modestly increases locomotor activity and produces similar discriminative stimulus effects to psychostimulants in rodents, although the subjective effects of modafinil are reportedly distinct from those of cocaine or amphetamine in humans with a history of psychostimulant abuse. OBJECTIVES The current study employed drug discrimination procedures in rats to examine the pharmacological actions contributing to modafinil's discriminative stimulus functions. METHODS Eight male Sprague-Dawley rats were trained to discriminate intragastric administration of 256 mg/kg modafinil from vehicle (5% arabic gum) under a FR 20 schedule of food reinforcement. Substitution tests were conducted with various dopaminergic agents (d-amphetamine, cocaine, PNU-91356A, GBR 12909, methylphenidate) and nondopaminergic agents (nicotine, ethanol). Antagonist tests were conducted with the selective D1 antagonist, SCH 39166, and the nonselective D2 antagonist, haloperidol. RESULTS Rats trained to discriminate modafinil displayed complete stimulus generalization to cocaine, methylphenidate, and GBR 12909 and the discrimination was completely blocked by both SCH 39166 and haloperidol. Evidence for significant partial substitution was obtained with d-amphetamine, PNU-91356A, and nicotine. CONCLUSIONS Results strongly support the role of dopaminergic mechanisms in the discriminative stimulus functions of modafinil, although further evaluation regarding the contribution of other neurotransmitter systems to these effects should be continued. The findings are discussed in light of clinical research efforts with modafinil as a treatment for psychostimulant dependence.
Collapse
|
37
|
Pilot study of the effects of lisdexamfetamine on cocaine use: A randomized, double-blind, placebo-controlled trial. Drug Alcohol Depend 2015; 153:94-103. [PMID: 26116930 PMCID: PMC4509923 DOI: 10.1016/j.drugalcdep.2015.05.042] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 05/28/2015] [Accepted: 05/31/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Amphetamine analogs have been demonstrated to have some efficacy in reducing use in cocaine dependent individuals. However, these agents also have potential for abuse. Lisdexamfetamine (LDX), a lysine+dextroamphetamine formulation, has been approved for the treatment of Attention-Deficit/Hyperactivity Disorder (ADHD) and as a prodrug, has less abuse potential. OBJECTIVE This pilot study sought to evaluate the safety, tolerability, and efficacy of LDX as a candidate treatment for cocaine dependence. METHODS A randomized, double-blind, placebo-controlled parallel group study served to evaluate LDX in 43 cocaine-dependent individuals: (1) placebo (PBO; 0mg, n=21), (2) LDX (70mg, n=22). Participants received medication for 14 weeks. Cocaine use was determined based on urine analysis for benzoylecgonine (BE; a cocaine metabolite). RESULTS Retention rates were higher though not significantly different in the PBO (71.4%) than the LDX condition (57.1%). Compared to those in the PBO condition, those receiving LDX were more likely to report experiencing (ps<0.05) diarrhea (45.5% vs. 14.3%), headaches (45.5% vs. 9.5%), and anxiety (31.8% vs. 4.8%). No differences in medication conditions were observed for blood pressure, heart rate, or body weight. In the randomized sample, no differences in cocaine use were seen. Those receiving LDX reported significantly less craving for cocaine than participants receiving PBO. CONCLUSIONS LDX did not significantly reduce cocaine use compared to PBO in the randomized sample.
Collapse
|
38
|
Negus SS, Henningfield J. Agonist Medications for the Treatment of Cocaine Use Disorder. Neuropsychopharmacology 2015; 40:1815-25. [PMID: 25563633 PMCID: PMC4839506 DOI: 10.1038/npp.2014.322] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/04/2014] [Accepted: 12/04/2014] [Indexed: 11/09/2022]
Affiliation(s)
- S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Jack Henningfield
- Pinney Associates, Bethesda, MD, USA
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Levin FR, Mariani JJ, Specker S, Mooney M, Mahony A, Brooks DJ, Babb D, Bai Y, Eberly LE, Nunes EV, Grabowski J. Extended-Release Mixed Amphetamine Salts vs Placebo for Comorbid Adult Attention-Deficit/Hyperactivity Disorder and Cocaine Use Disorder: A Randomized Clinical Trial. JAMA Psychiatry 2015; 72:593-602. [PMID: 25887096 PMCID: PMC4456227 DOI: 10.1001/jamapsychiatry.2015.41] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IMPORTANCE Adult attention-deficit/hyperactivity disorder (ADHD) is prevalent but often unrecognized, in part because it tends to co-occur with other disorders such as substance use disorders. Cocaine use disorder is one such disorder with high co-occurrence of ADHD. OBJECTIVE To examine whether treatment of co-occurring ADHD and cocaine use disorder with extended-release mixed amphetamine salts is effective at both improving ADHD symptoms and reducing cocaine use. DESIGN, SETTING, AND PARTICIPANTS Thirteen-week, randomized, double-blind, 3-arm, placebo-controlled trial of participants meeting DSM-IV-TR criteria for both ADHD and cocaine use disorder conducted between December 1, 2007, and April 15, 2013, at 2 academic health center substance abuse treatment research sites. One hundred twenty-six adults diagnosed as having comorbid ADHD and cocaine use disorder were randomized to extended-release mixed amphetamine salts or placebo. Analysis was by intent-to-treat population. INTERVENTIONS Participants received extended-release mixed amphetamine salts (60 or 80 mg) or placebo daily for 13 weeks and participated in weekly individual cognitive behavioral therapy. MAIN OUTCOMES AND MEASURES For ADHD, percentage of participants achieving at least a 30% reduction in ADHD symptom severity, measured by the Adult ADHD Investigator Symptom Rating Scale; for cocaine use, cocaine-negative weeks (by self-report of no cocaine use and weekly benzoylecgonine urine screens) during maintenance medication (weeks 2-13) and percentage of participants achieving abstinence for the last 3 weeks. RESULTS More patients achieved at least a 30% reduction in ADHD symptom severity in the medication groups (60 mg: 30 of 40 participants [75.0%]; odds ratio [OR] = 5.23; 95% CI, 1.98-13.85; P < .001; and 80 mg: 25 of 43 participants [58.1%]; OR = 2.27; 95% CI, 0.94-5.49; P = .07) compared with placebo (17 of 43 participants [39.5%]). The odds of a cocaine-negative week were higher in the 80-mg group (OR = 5.46; 95% CI, 2.25-13.27; P < .001) and 60-mg group (OR = 2.92; 95% CI, 1.15-7.42; P = .02) compared with placebo. Rates of continuous abstinence in the last 3 weeks were greater for the medication groups than the placebo group: 30.2% for the 80-mg group (OR = 11.87; 95% CI, 2.25-62.62; P = .004) and 17.5% for the 60-mg group (OR = 5.85; 95% CI, 1.04-33.04; P = .04) vs 7.0% for placebo. CONCLUSIONS AND RELEVANCE Extended-release mixed amphetamine salts in robust doses along with cognitive behavioral therapy are effective for treatment of co-occurring ADHD and cocaine use disorder, both improving ADHD symptoms and reducing cocaine use. The data suggest the importance of screening and treatment of ADHD in adults presenting with cocaine use disorder. TRIAL REGISTRATION clinicaltrials.gov Identifier:NCT00553319.
Collapse
Affiliation(s)
- Frances R. Levin
- Division of Substance Abuse, New York State Psychiatric Institute, New York2Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York, New York
| | - John J. Mariani
- Division of Substance Abuse, New York State Psychiatric Institute, New York2Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Sheila Specker
- Department of Psychiatry, Medical School, University of Minnesota, Minneapolis
| | - Marc Mooney
- Department of Psychiatry, Medical School, University of Minnesota, Minneapolis
| | - Amy Mahony
- Division of Substance Abuse, New York State Psychiatric Institute, New York
| | - Daniel J. Brooks
- Division of Substance Abuse, New York State Psychiatric Institute, New York
| | - David Babb
- Department of Psychiatry, Medical School, University of Minnesota, Minneapolis
| | - Yun Bai
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis
| | - Lynn E. Eberly
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis
| | - Edward V. Nunes
- Division of Substance Abuse, New York State Psychiatric Institute, New York2Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York, New York
| | - John Grabowski
- Department of Psychiatry, Medical School, University of Minnesota, Minneapolis
| |
Collapse
|
40
|
Nuijten M, Blanken P, van den Brink W, Hendriks V. Modafinil in the treatment of crack-cocaine dependence in the Netherlands: Results of an open-label randomised controlled feasibility trial. J Psychopharmacol 2015; 29:678-87. [PMID: 25922424 DOI: 10.1177/0269881115582151] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Crack-cocaine dependence is a serious disorder with no approved pharmacological treatment. Modafinil is a promising medication with increased cocaine abstinence and reduced craving in some previous studies. In the present study, we examined the acceptance, safety and potential benefits of modafinil as an add-on treatment to cognitive behavioural therapy (CBT) in crack-cocaine dependent patients. METHODS Sixty-five crack-cocaine dependent outpatients participated in an open-label, randomised feasibility trial. Patients were randomised to receive either 12-week individual CBT plus 400 mg/day modafinil or 12-week individual CBT only. The primary outcome measure was CBT treatment retention. Secondary outcomes included modafinil adherence, tolerability and safety, use of cocaine and other substances, cocaine craving, health, social functioning and patient satisfaction. RESULTS Modafinil adherence was low, with only 10% treatment completers. Intent-to-treat analyses showed that modafinil did not improve CBT treatment retention or any of the secondary cocaine-related outcomes. Both groups showed similar, large reductions in cocaine use during the study treatment. Post hoc exploratory analyses within the CBT plus modafinil group showed significantly larger baseline to week 12 reductions in cocaine use days in high (⩾ 8 weeks) modafinil adherent patients. CONCLUSIONS Acceptance and benefits of modafinil were not demonstrated in the present study. Since reduction in cocaine use was observed in high modafinil adherent patients, further research in the treatment of cocaine dependence, in which modafinil adherence is optimised, is warranted.
Collapse
Affiliation(s)
- Mascha Nuijten
- Parnassia Addiction Research Centre (PARC, Brijder Addiction Treatment), The Hague, the Netherlands
| | - Peter Blanken
- Parnassia Addiction Research Centre (PARC, Brijder Addiction Treatment), The Hague, the Netherlands
| | - Wim van den Brink
- Amsterdam Institute for Addiction Research, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Vincent Hendriks
- Parnassia Addiction Research Centre (PARC, Brijder Addiction Treatment), The Hague, the Netherlands
| |
Collapse
|
41
|
Banks ML, Hutsell BA, Blough BE, Poklis JL, Negus SS. Preclinical Assessment of Lisdexamfetamine as an Agonist Medication Candidate for Cocaine Addiction: Effects in Rhesus Monkeys Trained to Discriminate Cocaine or to Self-Administer Cocaine in a Cocaine Versus Food Choice Procedure. Int J Neuropsychopharmacol 2015; 18:pyv009. [PMID: 25618405 PMCID: PMC4458439 DOI: 10.1093/ijnp/pyv009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/20/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Chronic amphetamine treatment decreases cocaine consumption in preclinical and human laboratory studies and in clinical trials. Lisdexamfetamine is an amphetamine prodrug in which L-lysine is conjugated to the terminal nitrogen of d-amphetamine. Prodrugs may be advantageous relative to their active metabolites due to slower onsets and longer durations of action; however, lisdexamfetamine treatment's efficacy in decreasing cocaine consumption is unknown. METHODS This study compared lisdexamfetamine and d-amphetamine effects in rhesus monkeys using two behavioral procedures: (1) a cocaine discrimination procedure (training dose = 0.32mg/kg cocaine, i.m.); and (2) a cocaine-versus-food choice self-administration procedure. RESULTS In the cocaine-discrimination procedure, lisdexamfetamine (0.32-3.2mg/kg, i.m.) substituted for cocaine with lower potency, slower onset, and longer duration of action than d-amphetamine (0.032-0.32mg/kg, i.m.). Consistent with the function of lisdexamfetamine as an inactive prodrug for amphetamine, the time course of lisdexamfetamine effects was related to d-amphetamine plasma levels by a counter-clockwise hysteresis loop. In the choice procedure, cocaine (0-0.1mg/kg/injection, i.v.) and food (1g banana-flavored pellets) were concurrently available, and cocaine maintained a dose-dependent increase in cocaine choice under baseline conditions. Treatment for 7 consecutive days with lisdexamfetamine (0.32-3.2mg/kg/day, i.m.) or d-amphetamine (0.032-0.1mg/kg/h, i.v.) produced similar dose-dependent rightward shifts in cocaine dose-effect curves and decreases in preference for 0.032mg/kg/injection cocaine. CONCLUSIONS Lisdexamfetamine has a slower onset and longer duration of action than amphetamine but retains amphetamine's efficacy to reduce the choice of cocaine in rhesus monkeys. These results support further consideration of lisdexamfetamine as an agonist-based medication candidate for cocaine addiction.
Collapse
Affiliation(s)
- Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA (Drs Banks, Hutsell, Negus, and Mr Poklis); Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA (Drs Banks and Negus); Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC (Dr Blough)
| | - Blake A Hutsell
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA (Drs Banks, Hutsell, Negus, and Mr Poklis); Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA (Drs Banks and Negus); Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC (Dr Blough)
| | - Bruce E Blough
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA (Drs Banks, Hutsell, Negus, and Mr Poklis); Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA (Drs Banks and Negus); Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC (Dr Blough)
| | - Justin L Poklis
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA (Drs Banks, Hutsell, Negus, and Mr Poklis); Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA (Drs Banks and Negus); Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC (Dr Blough)
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA (Drs Banks, Hutsell, Negus, and Mr Poklis); Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA (Drs Banks and Negus); Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC (Dr Blough)
| |
Collapse
|
42
|
Rezaei F, Emami M, Zahed S, Morabbi MJ, Farahzadi M, Akhondzadeh S. Sustained-release methylphenidate in methamphetamine dependence treatment: a double-blind and placebo-controlled trial. Daru 2015; 23:2. [PMID: 25588930 PMCID: PMC4298048 DOI: 10.1186/s40199-015-0092-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/04/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The objective of this randomized, double-blind, placebo-controlled study was to evaluate the efficacy of sustained-release methylphenidate (MPH-SR) in treatment of methamphetamine dependence. METHODS Fifty-six individuals who met DSM-IV-TR criteria for methamphetamine dependence participated in this 10-week trial. The participants were randomly allocated into two groups and received 18 to 54 mg/day sustained-released methylphenidate or placebo for 10 weeks. Craving was evaluated by a visual analogue craving scale every week. Urinary screening test for methamphetamine was carried out each week. The Beck Depression Inventory-II (BDI-II) was used to monitor participant depressive symptoms at baseline and bi-weekly during the treatment period. RESULTS At the end of the trial, the MPH-SR group was less methamphetamine positive compared to the placebo group and the difference was significant (p = 0.03). By the end of the study, MPH-SR group showed significantly less craving scores compared to the placebo group [MD (95% CI) = -10.28(0.88-19.18), t(54) = 2.19, p = 0.03]. There was greater improvement in the depressive symptoms scores in the intervention group compared to the placebo group [MD (95% CI) =2.03(0.31-3.75), t (54) =2.37, p = 0.02]. CONCLUSION Sustained-released methylphenidate was safe and well tolerated among active methamphetamine users and significantly reduced methamphetamine use, craving and depressive symptoms. TRIAL REGISTRATION IRCT201202281556N38.
Collapse
Affiliation(s)
- Farzin Rezaei
- Department of psychiatry, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Maryam Emami
- Department of psychiatry, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Shakiba Zahed
- Department of Health Education and Health Promotion, Faculty of Health, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammad-Javad Morabbi
- Department of Neuroscience, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammadhadi Farahzadi
- Department of Neuroscience, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran.
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, South Kargar Street, Tehran, 13337, Iran.
| |
Collapse
|
43
|
Effectiveness of secondary prevention and treatment interventions for crack-cocaine abuse: a comprehensive narrative overview of English-language studies. THE INTERNATIONAL JOURNAL OF DRUG POLICY 2015; 26:352-63. [PMID: 25662894 DOI: 10.1016/j.drugpo.2015.01.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 12/30/2014] [Accepted: 01/06/2015] [Indexed: 11/18/2022]
Abstract
There are an estimated several million crack-cocaine users globally; use is highest in the Americas. Most crack users are socio-economically marginalized (e.g., homeless), and feature elevated risks for morbidity (e.g., blood-borne viruses), mortality and crime/violence involvement, resulting in extensive burdens. No comprehensive reviews of evidence-based prevention and/or treatment interventions specifically for crack use exist. We conducted a comprehensive narrative overview of English-language studies on the efficacy of secondary prevention and treatment interventions for crack (cocaine) abuse/dependence. Literature searches (1990-2014) using pertinent keywords were conducted in main scientific databases. Titles/abstracts were reviewed for relevance, and full studies were included in the review if involving a primary prevention/treatment intervention study comprising a substantive crack user sample. Intervention outcomes considered included drug use, health risks/status (e.g., HIV or sexual risks) and select social outcome indicators. Targeted (e.g., behavioral/community-based) prevention measures show mixed and short-term effects on crack use/HIV risk outcomes. Material (e.g., safer crack use kit distribution) interventions also document modest efficacy in risk reduction; empirical assessments of environmental (e.g., drug consumption facilities) for crack smokers are not available. Diverse psycho-social treatment (including contingency management) interventions for crack abuse/dependence show some positive but also limited/short-term efficacy, yet likely constitute best currently available treatment options. Ancillary treatments show little effects but are understudied. Despite ample studies, pharmaco-therapeutic/immunotherapy treatment agents have not produced convincing evidence; select agents may hold potential combined with personalized approaches and/or psycho-social strategies. No comprehensively effective 'gold-standard' prevention/treatment interventions for crack abuse exist; concerted research towards improved interventions is urgently needed.
Collapse
|
44
|
Oteo Pérez A, Benschop A, Blanken P, Korf DJ. Criminal involvement and crime specialization among crack users in the Netherlands. Eur Addict Res 2015; 21:53-62. [PMID: 25402472 DOI: 10.1159/000363737] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 05/18/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Crack users in the Netherlands are an ageing and diverse population with longstanding criminal careers. Our aim was to assess factors associated with current criminal involvement and specialization in selling drugs, property crime and violence. METHOD A sample of 1,039 frequent crack users was recruited in three major Dutch cities, combining respondent-driven sampling with random institutional sampling. Bivariate and logistic regression analyses were performed to find factors associated with current criminality. RESULTS A total of 431 participants (41.5%) had engaged in crime in the past 30 days, mostly selling drugs (68.9%), followed by property crimes (34.4%) and a few cases of violent crime (9.7%). Younger age, homelessness, heavier patterns of use and a more prolific criminal justice history were associated with current criminality. Those receiving welfare benefits tended to be more likely to specialize only in selling drugs as opposed to (also) property crimes. CONCLUSION Reducing drug use among criminally involved crack users and addressing their housing conditions could have a significant impact on reducing drug-related crime. Welfare benefits might act as protective factor against committing property crimes but not against the selling of drugs.
Collapse
Affiliation(s)
- Alberto Oteo Pérez
- Bonger Institute of Criminology, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
45
|
Abstract
Agonist replacement may be a viable treatment approach for managing stimulant use disorders. This study sought to determine the effects of D-amphetamine maintenance on methamphetamine self-administration in stimulant using human participants. We predicted that D-amphetamine maintenance would reduce methamphetamine self-administration. Eight participants completed the protocol, which tested 2 D-amphetamine maintenance conditions in counterbalanced order (0 and 40 mg/d). Participants completed 4 experimental sessions under each maintenance condition in which they first sampled 1 of 4 doses of intranasal methamphetamine (0, 10, 20, or 30 mg). Participants then had the opportunity to respond on a computerized progressive-ratio task to earn portions of the sampled methamphetamine dose. Subject-rated drug effect and physiological measures were completed at regular intervals prior to and after sampling methamphetamine. Methamphetamine was self-administered as an orderly function of dose regardless of the maintenance condition. Methamphetamine produced prototypical subject-rated effects on 12 items of the drug-effects questionnaires, 8 of which were attenuated by D-amphetamine maintenance (eg, increased ratings were attenuated on items such as Any Effect, Like Drug, and Willing to Take Again on the Drug Effect Questionnaire). Methamphetamine produced significant increases in systolic blood pressure, which were attenuated by D-amphetamine maintenance compared to placebo maintenance. Methamphetamine was well tolerated during D-amphetamine maintenance and no adverse events occurred. Although D-amphetamine attenuated some subject-rated effects of methamphetamine, the self-administration results are concordant with those of clinical trials showing that D-amphetamine did not reduce methamphetamine use. Unique pharmacological approaches may be needed for treating amphetamine use disorders.
Collapse
|
46
|
Forray A, Sofuoglu M. Future pharmacological treatments for substance use disorders. Br J Clin Pharmacol 2014; 77:382-400. [PMID: 23039267 DOI: 10.1111/j.1365-2125.2012.04474.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 09/23/2012] [Indexed: 12/20/2022] Open
Abstract
Substance use disorders represent a serious public health and social issue worldwide. Recent advances in our understanding of the neurobiological basis of the addictive processes have led to the development of a growing number of pharmacological agents to treat addictions. Despite this progress, there are no approved pharmacological treatments for cocaine, methamphetamine and cannabis addiction. Moving treatment development to the next stage will require novel ways of approaching substance use disorders. One such novel approach is to target individual vulnerabilities, such as cognitive function, sex differences and psychiatric comorbidities. This review provides a summary of promising pharmacotherapies for alcohol, opiate, stimulant and nicotine addictions. Many medications that target positive and negative reinforcement of drugs, as well as individual vulnerabilities to addiction, are in different phases of development. Clinical trials testing the efficacy of these medications for substance use disorder are warranted.
Collapse
Affiliation(s)
- Ariadna Forray
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
47
|
Ling W, Chang L, Hillhouse M, Ang A, Striebel J, Jenkins J, Hernandez J, Olaer M, Mooney L, Reed S, Fukaya E, Kogachi S, Alicata D, Holmes N, Esagoff A. Sustained-release methylphenidate in a randomized trial of treatment of methamphetamine use disorder. Addiction 2014; 109:1489-500. [PMID: 24825486 PMCID: PMC4127124 DOI: 10.1111/add.12608] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/14/2014] [Accepted: 05/02/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND AIMS No effective pharmacotherapy for methamphetamine (MA) use disorder has yet been found. This study evaluated sustained-release methylphenidate (MPH-SR) compared with placebo (PLA) for treatment of MA use disorder in people also undergoing behavioral support and motivational incentives. DESIGN This was a randomized, double-blind, placebo-controlled design with MPH-SR or PLA provided for 10 weeks (active phase) followed by 4 weeks of single-blind PLA. Twice-weekly clinic visits, weekly group counseling (CBT) and motivational incentives (MI) for MA-negative urine drug screens (UDS) were included. SETTING Treatment sites were in Los Angeles, California (LA) and Honolulu, Hawaii (HH), USA. PARTICIPANTS A total of 110 MA-dependent (via DSM-IV) participants (LA = 90; HH = 20). MEASUREMENTS The primary outcome measure is self-reported days of MA use during the last 30 days of the active phase. Included in the current analyses are drug use (UDS and self-report), retention, craving, compliance (dosing, CBT, MI), adverse events and treatment satisfaction. FINDINGS No difference was found between treatment groups in self-reported days of MA use during the last 30 days of the active phase (P = 0.22). In planned secondary outcomes analyses, however, the MPH group had fewer self-reported MA use days from baseline through the active phase compared with the PLA group (P = 0.05). The MPH group also had lower craving scores and fewer marijuana-positive UDS than the PLA group in the last 30 days of the active phase. The two groups had similar retention, other drug use, adverse events and treatment satisfaction. CONCLUSIONS Methylphenidate may lead to a reduction in concurrent methamphetamine use when provided as treatment for patients undergoing behavioral support for moderate to severe methamphetamine use disorder, but this requires confirmation.
Collapse
Affiliation(s)
- Walter Ling
- University of California, Los Angeles, Integrated Substance Abuse
Programs
| | - Linda Chang
- University of Hawaii, Neuroscience and MRI Research Group
| | - Maureen Hillhouse
- University of California, Los Angeles, Integrated Substance Abuse
Programs
| | - Alfonso Ang
- University of California, Los Angeles, Integrated Substance Abuse
Programs
| | - Joan Striebel
- University of California, Los Angeles, Integrated Substance Abuse
Programs
| | - Jessica Jenkins
- University of California, Los Angeles, Integrated Substance Abuse
Programs
| | - Jasmin Hernandez
- University of California, Los Angeles, Integrated Substance Abuse
Programs
| | - Mary Olaer
- University of California, Los Angeles, Integrated Substance Abuse
Programs
| | - Larissa Mooney
- University of California, Los Angeles, Integrated Substance Abuse
Programs
| | - Susan Reed
- University of California, Los Angeles, Integrated Substance Abuse
Programs
| | - Erin Fukaya
- University of Hawaii, Neuroscience and MRI Research Group
| | | | - Daniel Alicata
- University of Hawaii, Neuroscience and MRI Research Group
| | | | | |
Collapse
|
48
|
Stoops WW, Rush CR. Agonist replacement for stimulant dependence: a review of clinical research. Curr Pharm Des 2014; 19:7026-35. [PMID: 23574440 DOI: 10.2174/138161281940131209142843] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 04/04/2013] [Indexed: 11/22/2022]
Abstract
Stimulant use disorders are an unrelenting public health concern worldwide. Agonist replacement therapy is among the most effective strategies for managing substance use disorders including nicotine and opioid dependence. The present paper reviewed clinical data from human laboratory self-administration studies and clinical trials to determine whether agonist replacement therapy is a viable strategy for managing cocaine and/or amphetamine use disorders. The extant literature suggests that agonist replacement therapy may be effective for managing stimulant use disorders, however, the clinical selection of an agonist replacement medication likely needs to be based on the pharmacological mechanism of the medication and the stimulant abused by patients. Specifically, dopamine releasers appear most effective for reducing cocaine use whereas dopamine reuptake inhibitors appear most effective for reducing amphetamine use.
Collapse
Affiliation(s)
- William W Stoops
- Department of Behavioral Science, University of Kentucky Medical Center, Lexington, KY 40536- 0086.
| | | |
Collapse
|
49
|
Nuijten M, Blanken P, van den Brink W, Hendriks V. Treatment of crack-cocaine dependence with topiramate: a randomized controlled feasibility trial in The Netherlands. Drug Alcohol Depend 2014; 138:177-84. [PMID: 24629631 DOI: 10.1016/j.drugalcdep.2014.02.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/17/2014] [Accepted: 02/17/2014] [Indexed: 01/14/2023]
Abstract
BACKGROUND Crack-cocaine dependence is a complex disorder with limited treatment options. Topiramate is one of the promising medications with reported reductions in cocaine use and craving in former studies. The present study evaluated the acceptance and effectiveness of topiramate as an add-on to cognitive behavioral therapy (CBT) in crack-cocaine dependent patients. METHODS Seventy-four crack-cocaine dependent outpatients participated in an open-label, randomized feasibility trial. They were randomized to receive either 12-week CBT plus topiramate (200mg/day) or 12-week CBT only. The primary outcome measure was treatment retention. Secondary outcomes included medication adherence, safety, cocaine and other substance use, health, social functioning, and patient satisfaction. RESULTS Adherence to topiramate treatment was low. In the intent-to-treat analyses, topiramate neither improved treatment retention nor reduced cocaine and other substance use. Post hoc, exploratory analyses suggested a moderation effect of comorbid opioid dependence, with a significant effect of topiramate on cocaine use reduction only in crack-cocaine dependent patients with comorbid opioid dependence. CONCLUSIONS Topiramate was safe and well-tolerated in this sample of crack-cocaine dependent patients, but efficacy was not supported probably due to low acceptance of the treatment. Given the equivocal results of previous studies and the negative findings in our study, the potential of topiramate in the treatment of cocaine dependence seems limited.
Collapse
Affiliation(s)
- Mascha Nuijten
- Parnassia Addiction Research Centre (PARC, Brijder Addiction Treatment), PO Box 53002, 2505 AA The Hague, The Netherlands.
| | - Peter Blanken
- Parnassia Addiction Research Centre (PARC, Brijder Addiction Treatment), PO Box 53002, 2505 AA The Hague, The Netherlands
| | - Wim van den Brink
- Amsterdam Institute for Addiction Research, Department of Psychiatry, Academic Medical Centre, University of Amsterdam, PO Box 22660, 1100 DD Amsterdam, The Netherlands
| | - Vincent Hendriks
- Parnassia Addiction Research Centre (PARC, Brijder Addiction Treatment), PO Box 53002, 2505 AA The Hague, The Netherlands
| |
Collapse
|
50
|
Psychostimulant addiction treatment. Neuropharmacology 2014; 87:150-60. [PMID: 24727297 DOI: 10.1016/j.neuropharm.2014.04.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/14/2014] [Accepted: 04/01/2014] [Indexed: 01/29/2023]
Abstract
Treatment of psychostimulant addiction has been a major, and not fully met, challenge. For opioid addiction, there is strong evidence for the effectiveness of several medications. For psychostimulants, there is no corresponding form of agonist maintenance that has met criteria for regulatory approval or generally accepted use. Stimulant-use disorders remain prevalent and can result in both short-term and long-term adverse consequences. The mainstay of treatment remains behavioral interventions. In this paper, we discuss those interventions and some promising candidates in the search for pharmacological interventions. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
|