1
|
Betts JM, Cook JW, Kobinsky KH, Baker TB, Jorenby DE, Piper ME. Understanding the motivational mechanisms for smoking and vaping among dual users and exclusive smokers. Drug Alcohol Depend 2024; 264:112436. [PMID: 39341015 PMCID: PMC11527565 DOI: 10.1016/j.drugalcdep.2024.112436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/08/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Understanding the motivational processes that influence e-cigarette use in a laboratory setting may help elucidate mechanisms that support long-term ecigarette use, which could have significant clinical and public health consequences. METHODS Secondary analyses were conducted on data from exclusive smokers (N=47) and dual users (N=88) who underwent a laboratory ad lib use session. Participants were given 10minutes to smoke (exclusive smokers) or vape (dual users) as much as they wanted. Withdrawal was assessed pre- and post-use. Smoking and vaping behavior was coded from session videos. Person-level predictors included cigarette/ecigarette craving-relief expectancies, demographics, and cigarette/e-cigarette use and dependence. Smoking and vaping status was assessed at Year 1 using self-reported 30-day point prevalence. Data were analyzed using general linear models and logistic regressions. RESULTS Both groups reported reductions in withdrawal after product use, including cigarette craving. Baseline e-cigarette craving-relief expectancies, pre-session ecigarette craving, heaviness of e-cigarette use, and relative e-cigarette dependence were significant univariate predictors of continued vaping in dual users at Year 1 (ORs>1.04, ps<.05). Dual users and exclusive smokers did not differ on use behavior (i.e., average number of puffs, ps>.16). CONCLUSIONS E-cigarette use alleviated withdrawal, including cigarette and e-cigarette craving, in dual users. Laboratory use behavior did not differ between dual users using e-cigarettes and exclusive smokers using cigarettes. Greater e-cigarette craving-relief expectancies, e-cigarette craving, heaviness of e-cigarette use, and morning product use pattern ('relative dependence') may reflect mechanisms that sustain e-cigarette use.
Collapse
Affiliation(s)
- Jennifer M Betts
- Center for Tobacco Research and Intervention, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans' Hospital, Madison, WI, USA.
| | - Jessica W Cook
- Center for Tobacco Research and Intervention, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans' Hospital, Madison, WI, USA
| | - Kate H Kobinsky
- Center for Tobacco Research and Intervention, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Timothy B Baker
- Center for Tobacco Research and Intervention, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Douglas E Jorenby
- Center for Tobacco Research and Intervention, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Megan E Piper
- Center for Tobacco Research and Intervention, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
2
|
Yang K, McLaughlin I, Shaw JK, Quijano-Cardé N, Dani JA, De Biasi M. CHRNA5 gene variation affects the response of VTA dopaminergic neurons during chronic nicotine exposure and withdrawal. Neuropharmacology 2023; 235:109547. [PMID: 37116611 PMCID: PMC10249248 DOI: 10.1016/j.neuropharm.2023.109547] [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: 03/03/2023] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 04/30/2023]
Abstract
Nicotine is the principal psychoactive component in tobacco that drives addiction through its action on neuronal nicotinic acetylcholine receptors (nAChR). The nicotinic receptor gene CHRNA5, which encodes the α5 subunit, is associated with nicotine use and dependence. In humans, the CHRNA5 missense variant rs16969968 (G > A) is associated with increased risk for nicotine dependence and other smoking-related phenotypes. In rodents, α5-containing nAChRs in dopamine (DA) neurons within the ventral tegmental area (VTA) powerfully modulate nicotine reward and reinforcement. Although the neuroadaptations caused by long-term nicotine exposure are being actively delineated at both the synaptic and behavioral levels, the contribution of α5-containing nAChRs to the cellular adaptations associated with long-term nicotine exposure remain largely unknown. To gain insight into the mechanisms behind the influence of α5-containing nAChRs and the rs16969968 polymorphism on nicotine use and dependence, we used electrophysiological approaches to examine changes in nAChR function arising in VTA neurons during chronic nicotine exposure and multiple stages of nicotine withdrawal. Our results demonstrate that CHRNA5 mutation leads to profound changes in VTA nAChR function at baseline, during chronic nicotine exposure, and during short-term and prolonged withdrawal. Whereas nAChR function was suppressed in DA neurons from WT mice undergoing withdrawal relative to drug-naïve or nicotine-drinking mice, α5-null mice exhibited an increase in nAChR function during nicotine exposure that persisted throughout 5-10 weeks of withdrawal. Re-expressing the hypofunctional rs16969968 CHRNA5 variant in α5-null VTA DA neurons did not rescue the phenotype, with α5-SNP neurons displaying a similar increased response to ACh during nicotine exposure and early stages of withdrawal. These results demonstrate the importance of VTA α5-nAChRs in the response to nicotine and implicate them in the time course of withdrawal.
Collapse
Affiliation(s)
- Kechun Yang
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ian McLaughlin
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jessica K Shaw
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Natalia Quijano-Cardé
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John A Dani
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Mariella De Biasi
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
3
|
Serretti A. Anhedonia and Depressive Disorders. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2023; 21:401-409. [PMID: 37424409 PMCID: PMC10335915 DOI: 10.9758/cpn.23.1086] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/01/2023] [Indexed: 07/11/2023]
Abstract
Anhedonia is a core symptom of depression and of several psychiatric disorders. Anhedonia has however expanded from its original definition to encompass a spectrum of reward processing deficits that received much interest in the last decades. It is a relevant risk factor for possible suicidal behaviors, and that it may operate as an independent risk factor for suicidality apart from the episode severity. Anhedonia has also been linked to inflammation with a possible reciprocal deleterious effect on depression. Its neurophysiological bases mainly include alterations in striatal and prefrontal areas, with dopamine being the most involved neurotransmitter. Anhedonia is thought to have a significant genetic component and polygenic risk scores are a possible tool for predicting an individual's risk for developing anhedonia. Traditional antidepressants, such as selective serotonin reuptake inhibitors, showed a limited benefit on anhedonia, also considering their potential pro-anhedonic effect in some subjects. Other treatments may be more effective in treating anhedonia, such as agomelatine, vortioxetine, ketamine and transcranial magnetic stimulation. Psychotherapy is also widely supported, with cognitive-behavioral therapy and behavioral activation both showing benefit. In conclusion, a large body of evidence suggests that anhedonia is, at least partially, independent from depression, therefore it needs careful assessment and targeted treatment.
Collapse
Affiliation(s)
- Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
4
|
McNealy KR, Weyrich L, Bevins RA. The co-use of nicotine and prescription psychostimulants: A review of their behavioral and neuropharmacological interactions. Drug Alcohol Depend 2023; 248:109906. [PMID: 37216808 PMCID: PMC10361216 DOI: 10.1016/j.drugalcdep.2023.109906] [Citation(s) in RCA: 1] [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: 02/13/2023] [Revised: 04/05/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Nicotine is commonly co-used with other psychostimulants. These high co-use rates have prompted much research on interactions between nicotine and psychostimulant drugs. These studies range from examination of illicitly used psychostimulants such as cocaine and methamphetamine to prescription psychostimulants used to treat attention deficit hyperactivity disorder (ADHD) such as methylphenidate (Ritalin™) and d-amphetamine (active ingredient of Adderall™). However, previous reviews largely focus on nicotine interactions with illicitly used psychostimulants with sparse mention of prescription psychostimulants. The currently available epidemiological and laboratory research, however, suggests high co-use between nicotine and prescription psychostimulants, and that these drugs interact to modulate use liability of either drug. The present review synthesizes epidemiological and experimental human and pre-clinical research assessing the behavioral and neuropharmacological interactions between nicotine and prescription psychostimulants that may contribute to high nicotine-prescription psychostimulant co-use. METHODS We searched databases for literature investigating acute and chronic nicotine and prescription psychostimulant interactions. Inclusion criteria were that participants/subjects had to experience nicotine and a prescription psychostimulant compound at least once in the study, in addition to assessment of their interaction. RESULTS AND CONCLUSIONS Nicotine clearly interacts with d-amphetamine and methylphenidate in a variety of behavioral tasks and neurochemical assays assessing co-use liability across preclinical, clinical, and epidemiological research. The currently available research suggests research gaps examining these interactions in women/female rodents, in consideration of ADHD symptoms, and how prescription psychostimulant exposure influences later nicotine-related outcomes. Nicotine has been less widely studied with alternative ADHD pharmacotherapy bupropion, but we also discuss this research.
Collapse
Affiliation(s)
- Kathleen R McNealy
- Department of Psychology, University of Nebraska-Lincoln, 238 Burnett Hall, Lincoln, NE68588-0308, USA.
| | - Lucas Weyrich
- Institute for Human Neuroscience, Boys Town National Research Hospital, 14090 Mother Teresa Ln, Boys Town, NE68010, USA; Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE687178, USA
| | - Rick A Bevins
- Department of Psychology, University of Nebraska-Lincoln, 238 Burnett Hall, Lincoln, NE68588-0308, USA
| |
Collapse
|
5
|
Allain AE, Aribo O, Medrano MC, Fournier ML, Bertrand SS, Caille S. Impact of acute and chronic nicotine administration on midbrain dopaminergic neuron activity and related behaviors in TRPV1 knock-out juvenile mice. Eur J Neurosci 2021; 55:697-713. [PMID: 34939238 DOI: 10.1111/ejn.15577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/03/2022]
Abstract
The addictive properties of nicotine, the main alkaloid in tobacco and tobacco-derived products, largely depend on its action on the activity of midbrain dopamine (DA) neurons. The Transient Receptor Potential Vanilloid 1 (TRPV1) channel has also been examined as an emerging contributor to addiction-related symptoms due to its ability to modulate midbrain neurons. Thus, the objective of our study was to explore the role of TRPV1 receptors (TRPV1Rs) on nicotine-induced behaviors and associated response of DA neuron activity. Both wild type juvenile mice and juvenile mice with invalidation of the TRPV1R gene were exposed to acute or chronic nicotine 0.3 mg/kg administration. We analyzed locomotor activity in response to the drug. In addition, we performed cell-attached and whole-cell recordings from ventral tegmental area (VTA) neurons after nicotine exposure. Our results showed that the genetic deletion of TRPV1Rs reduced nicotine-induced locomotor sensitization. In addition, it provided evidence in support of TRPV1Rs being regulators of inhibitory synaptic transmission in the VTA. However, TRPV1Rs did not seem to modulate either nicotine-induced conditioning place preference or nicotine-evoked electrical activity of DA neurons. In conclusion, TRPV1Rs modulate nicotine-induced psychomotor sensitization in mice independently of a control on VTA DA neuron activity. Thus, TRPV1R control may depend on another key player of the mesolimbic circuit.
Collapse
Affiliation(s)
- Anne-Emilie Allain
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France.,Univ. Bordeaux, CNRS, PHYCELL Platform INCIA, UMR 5287, Bordeaux, France
| | - Oceane Aribo
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
| | | | | | - Sandrine S Bertrand
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France.,Univ. Bordeaux, CNRS, PHYCELL Platform INCIA, UMR 5287, Bordeaux, France
| | | |
Collapse
|
6
|
Liautaud MM, Kechter A, Bello MS, Guillot CR, Oliver JA, Banks DE, D’Orazio LM, Leventhal AM. Anhedonia in tobacco withdrawal among African-American smokers. Exp Clin Psychopharmacol 2021; 29:511-523. [PMID: 34110886 PMCID: PMC8511043 DOI: 10.1037/pha0000474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Anhedonia-diminished interest and pleasure in response to rewards-may be a symptom of tobacco withdrawal that is understudied in priority populations. This experiment investigated the magnitude and correlates of various dimensions of anhedonia during tobacco withdrawal among African-American (AA) smokers-a population subject to health disparities. AA smokers (N = 607; ≥ 10 cigarettes/day, 37.8% female, M[SD] age = 50.0[10.6] years) completed self-report measures assessing expected pleasure from (i.e., consummatory anhedonia) and desire to engage in (i.e., anticipatory anhedonia) various types of hypothetically experienced rewards at counterbalanced 16-hr tobacco deprived and nondeprived lab visits. Other tobacco withdrawal symptom measures (e.g., craving, negative affect, hunger) were also assessed. Tobacco deprivation most robustly increased scores on a composite measure of consummatory anhedonia directed toward various reward domains (i.e., hobbies, sensory experiences, social activities; d = .32, p < .001). Deprivation modestly increased consummatory and anticipatory anhedonia directed toward sexual rewards (ds = .09-.12, ps < .02), did not significantly change anhedonia toward food rewards, and decreased anhedonia directed toward psychoactive drug rewards (i.e., increased desire for and pleasure from drugs; ds = -.21 to -.19, ps < .001). Deprivation-induced changes in anhedonia were modestly correlated with other withdrawal symptoms (average |r|s = .04-.23) and were amplified among participants with higher nicotine dependence and lower positive affect-related traits (|β|s = .10-.12, ps < .01). Some dimensions of anhedonia may be genuine expressions of acute tobacco withdrawal in AA smokers. Applying multi-dimensional anhedonia conceptualizations might advance basic knowledge and treatment of tobacco use disorder, improve smoking cessation outcomes, and address tobacco-related health disparities facing AA smokers. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
| | - Afton Kechter
- University of Southern California, Department of Preventive Medicine
| | | | | | - Jason A. Oliver
- Duke University School of Medicine, Department of Psychiatry and Behavioral Sciences
| | - Devin E. Banks
- University of Missouri Saint Louis, Department of Psychological Sciences
| | | | - Adam M. Leventhal
- University of Southern California, Department of Preventive Medicine
- University of Southern California, Department of Psychology
| |
Collapse
|
7
|
Harris AC. Further pharmacological characterization of a preclinical model of the early development of nicotine withdrawal. Drug Alcohol Depend 2021; 226:108870. [PMID: 34216863 DOI: 10.1016/j.drugalcdep.2021.108870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/24/2021] [Accepted: 06/10/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Establishing preclinical models of the development of nicotine withdrawal following acute nicotine exposure could inform tobacco addiction-related research, treatment, and policy. To this end, this lab has previously reported that rats exhibit withdrawal-like elevations in intracranial self-stimulation (ICSS) thresholds (anhedonia-like behavior) following acute nicotine exposure. The goal of this study was to provide further pharmacological characterization of ICSS as a measure of spontaneous and antagonist-precipitated withdrawal from acute nicotine. METHODS AND RESULTS Rats exhibited a small increase in ICSS thresholds over time following a single nicotine injection (1.0 mg/kg, s.c.), suggesting a modest spontaneous withdrawal effect (Experiment 1). In Experiment 2, the antidepressant bupropion (5.0 mg/kg, i.p.), which is used to treat tobacco addiction and attenuates nicotine withdrawal in both humans and rodents, blocked elevations in ICSS thresholds induced by a single injection of nicotine (0.5 mg/kg, s.c.) followed ≈ 2 h later by the non-selective, non-competitive nicotinic acetylcholine receptor (nAChR) antagonist mecamylamine (3.0 mg/kg, s.c.). In Experiment 3a, s.c. administration of the competitive, relatively selective α4ß2 nAChR antagonist dihydro-beta-erythroidine (DHßE) (5.6 mg/kg, but not 3.0 mg/kg) following each of 5 daily injections of nicotine (0.5 mg/kg, s.c.) elevated ICSS thresholds. Mecamylamine (3.0 mg/kg, s.c.) also elevated ICSS thresholds when administered following all 5 daily nicotine injections (0.5 mg/kg, s.c., Experiment 3b). CONCLUSIONS These findings provide further characterization of elevations in ICSS thresholds as a measure of withdrawal from acute nicotine exposure. Further use of these models may be useful for understanding the early development of nicotine withdrawal.
Collapse
Affiliation(s)
- Andrew C Harris
- Hennepin Healthcare Research Institute, 701 Park Avenue, Minneapolis, MN 55415, United States; Departments of Medicine and Psychology, University of Minnesota Minneapolis, MN, United States.
| |
Collapse
|
8
|
Langmia IM, Just KS, Yamoune S, Brockmöller J, Masimirembwa C, Stingl JC. CYP2B6 Functional Variability in Drug Metabolism and Exposure Across Populations-Implication for Drug Safety, Dosing, and Individualized Therapy. Front Genet 2021; 12:692234. [PMID: 34322158 PMCID: PMC8313315 DOI: 10.3389/fgene.2021.692234] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022] Open
Abstract
Adverse drug reactions (ADRs) are one of the major causes of morbidity and mortality worldwide. It is well-known that individual genetic make-up is one of the causative factors of ADRs. Approximately 14 million single nucleotide polymorphisms (SNPs) are distributed throughout the entire human genome and every patient has a distinct genetic make-up which influences their response to drug therapy. Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of antiretroviral, antimalarial, anticancer, and antidepressant drugs. These drug classes are commonly in use worldwide and face specific population variability in side effects and dosing. Parts of this variability may be caused by single nucleotide polymorphisms (SNPs) in the CYP2B6 gene that are associated with altered protein expression and catalytic function. Population variability in the CYP2B6 gene leads to changes in drug metabolism which may result in adverse drug reactions or therapeutic failure. So far more than 30 non-synonymous variants in CYP2B6 gene have been reported. The occurrence of these variants show intra and interpopulation variability, thus affecting drug efficacy at individual and population level. Differences in disease conditions and affordability of drug therapy further explain why some individuals or populations are more exposed to CYP2B6 pharmacogenomics associated ADRs than others. Variabilities in drug efficacy associated with the pharmacogenomics of CYP2B6 have been reported in various populations. The aim of this review is to highlight reports from various ethnicities that emphasize on the relationship between CYP2B6 pharmacogenomics variability and the occurrence of adverse drug reactions. In vitro and in vivo studies evaluating the catalytic activity of CYP2B6 variants using various substrates will also be discussed. While implementation of pharmacogenomic testing for personalized drug therapy has made big progress, less data on pharmacogenetics of drug safety has been gained in terms of CYP2B6 substrates. Therefore, reviewing the existing evidence on population variability in CYP2B6 and ADR risk profiles suggests that, in addition to other factors, the knowledge on pharmacogenomics of CYP2B6 in patient treatment may be useful for the development of personalized medicine with regards to genotype-based prescription.
Collapse
Affiliation(s)
- Immaculate M. Langmia
- Institute of Clinical Pharmacology, University Hospital of Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Katja S. Just
- Institute of Clinical Pharmacology, University Hospital of Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Sabrina Yamoune
- Institute of Clinical Pharmacology, University Hospital of Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Jürgen Brockmöller
- Department of Clinical Pharmacology, University Medical Center Göttingen, Georg-August University, Göttingen, Germany
| | - Collen Masimirembwa
- African Institute of Biomedical Science and Technology (AiBST), Harare, Zimbabwe
| | - Julia C. Stingl
- Institute of Clinical Pharmacology, University Hospital of Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| |
Collapse
|
9
|
Underner M, Perriot J, Peiffer G, Ruppert AM, de Chazeron I, Jaafari N. [Combinations of pharmacological treatments in smoking cessation. A systematic review]. Rev Mal Respir 2021; 38:706-720. [PMID: 34215484 DOI: 10.1016/j.rmr.2021.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/30/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The effectiveness of the three validated smoking cessation medications, nicotine replacement therapy, varenicline and bupropion, may be insufficient, in hard-core smokers. OBJECTIVES This systematic review investigates the efficacy of combinations of different medications in smoking abstinence and their tolerability. RESULTS Three randomized controlled trials (RCTs) compared the combined medications with varenicline and nicotine patches vs. varenicline; two found an increase in abstinence rates with the combined medications. In one study, the beneficial effect was only observed in heavy smokers. The four RCTs comparing the combined medications with varenicline and bupropion (vs. varenicline) demonstrated an increase in abstinence rates with the combined medications, most often in heavy smokers who are very dependent on tobacco. The results of the three RCTs comparing the combined medications with bupropion and nicotine replacement therapy vs. varenicline were discordant. Three studies included other molecules (mecamylamine, selegiline, sertraline, buspirone). Combined medications were well tolerated. CONCLUSION Combination treatments can achieve higher smoking abstinence rates than monotherapies, especially in smokers who have failed to quit (Hard-core smokers). Treatment with a combination of varenicline and nicotine replacement therapy is a therapeutic option in smoking cessation.
Collapse
Affiliation(s)
- M Underner
- Unité de recherche clinique, université de Poitiers, centre hospitalier Henri-Laborit, 370, avenue Jacques-Cœur, CS 10587, 86021 Poitiers, France.
| | - J Perriot
- CLAT 63, dispensaire Emile-Roux, centre de tabacologie, 63100 Clermont-Ferrand, France
| | - G Peiffer
- Service de pneumologie, CHR Metz-Thionville, 57038 Metz, France
| | - A-M Ruppert
- Service de pneumologie et oncologie thoracique, GRC n(o) 04 Theranoscan, unité de tabacologie, Sorbonne Université, hôpital Tenon, Assistance publique-Hôpitaux de Paris, 4, rue de la Chine, 75970 Paris cedex 20, France
| | - I de Chazeron
- Service de psychiatrie-addictologie, CMP-B, CHU de Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - N Jaafari
- Unité de recherche clinique, université de Poitiers, centre hospitalier Henri-Laborit, 370, avenue Jacques-Cœur, CS 10587, 86021 Poitiers, France
| |
Collapse
|
10
|
Chellian R, Behnood-Rod A, Wilson R, Kamble SH, Sharma A, McCurdy CR, Bruijnzeel AW. Adolescent nicotine and tobacco smoke exposure enhances nicotine self-administration in female rats. Neuropharmacology 2020; 176:108243. [PMID: 32702403 DOI: 10.1016/j.neuropharm.2020.108243] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/24/2020] [Accepted: 07/13/2020] [Indexed: 12/30/2022]
Abstract
Most people start experimenting with tobacco products or e-cigarettes in early adolescence and become habitual smokers in late adolescence or adulthood. These studies investigated if exposure to tobacco smoke or nicotine during early and mid-adolescence affects nicotine intake in late adolescence and early adulthood. Male and female rats were exposed to tobacco smoke from low- and high-nicotine SPECTRUM cigarettes or nicotine (0.3 mg/kg, twice a day) from postnatal day (P) 24-42. The self-administration sessions started at P55. The rats self-administered nicotine for 14-15 days under a fixed-ratio 1 schedule, and on the first day, the maximum number of infusions was twenty. Exposure to smoke from high, but not low, nicotine cigarettes during adolescence increased nicotine self-administration in female but not male rats. Adolescent treatment with nicotine facilitated nicotine self-administration. On the first day of nicotine self-administration, nicotine-treated rats reached the maximum number of infusions before the saline-treated control rats. Nicotine intake was also higher in the nicotine-treated female rats than in the saline-treated females. There was no sex difference in nicotine intake in controls when the data from the studies were combined. Smoke exposure led to a dose-dependent increase in plasma nicotine and cotinine levels in adolescent rats. Exposure to smoke from high-nicotine cigarettes and 0.3 mg/kg of nicotine led to plasma nicotine and cotinine levels that are similar to those in tobacco users. These findings indicate that in females, but not males, exposure to nicotine during adolescence may facilitate smoking and e-cigarette use later in life.
Collapse
Affiliation(s)
| | - Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Ryann Wilson
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Shyam H Kamble
- Department of Pharmaceutics, University of Florida, Gainesville, FL, USA; Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA
| | - Abhisheak Sharma
- Department of Pharmaceutics, University of Florida, Gainesville, FL, USA; Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA
| | - Christopher R McCurdy
- Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA; Department of Medicinal Chemistry, University of Florida, Gainesville, FL, USA
| | | |
Collapse
|
11
|
Bupropion increases activation in nucleus accumbens during anticipation of monetary reward. Psychopharmacology (Berl) 2019; 236:3655-3665. [PMID: 31342097 DOI: 10.1007/s00213-019-05337-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/16/2019] [Indexed: 12/21/2022]
Abstract
RATIONALE Bupropion is used for major depressive disorder, smoking cessation aid, and obesity. It blocks reuptake of dopamine and noradrenaline and antagonizes nicotinic acetylcholine receptor. Animal studies showed that bupropion enhanced rewarding effects. In addition, bupropion has the potential to treat patients with reward processing dysfunction. However, neural substrates underlying the bupropion effects on reward function in human subjects are not fully understood. OBJECTIVES We investigated single-dose administration of bupropion on neural response of reward anticipation in healthy subjects using a monetary incentive delay (MID) task by functional magnetic resonance imaging (fMRI), especially focusing on nucleus accumbens (NAc) activity to non-drug reward stimuli under bupropion treatment. METHODS We used a randomized placebo-controlled within-subject crossover design. Fifteen healthy adults participated in two series of an fMRI study, taking either placebo or bupropion. The participants performed the MID task during the fMRI scanning. The effects of bupropion on behavioral performance and blood oxygenation level-dependent (BOLD) signal in NAc during anticipation of monetary gain were analyzed. RESULTS We found that bupropion significantly increased BOLD responses in NAc during monetary reward anticipation. The increased BOLD responses in NAc were observed with both low and high reward incentive cues. There was no significant difference between placebo and bupropion in behavioral performance. CONCLUSIONS Our findings provide support for the notion that bupropion enhances non-drug rewarding effects, suggesting a possible mechanism underlying therapeutic effects for patients with motivational deficit.
Collapse
|
12
|
Combination therapy of varenicline and bupropion in smoking cessation: A meta-analysis of the randomized controlled trials. Compr Psychiatry 2019; 95:152125. [PMID: 31669972 DOI: 10.1016/j.comppsych.2019.152125] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/21/2019] [Accepted: 09/03/2019] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The effects of the combination therapy of varenicline and bupropion in smoking cessation are still controversial. METHODS Databases including PubMed, EMBASE, Cochrane Library and Web of Science were scanned without time and language limitation. Subgroup analysis was performed to assess the effect of combination therapy in smokers with different level of nicotine dependence and cigarette consumption. RESULTS Four randomized controlled trials involving a total of 1230 smokers were included. Compared with varenicline monotherapy, combination treatment with varenicline and bupropion could significantly improve the abstinence rate at the end of treatment (RR 1.153, 95% CI 1.019 to 1.305, P=0.024). The benefit existed at 6months follow-up (RR 1.231, 95% CI 1.017 to 1.490, P=0.033), disappeared at 12months follow-up (RR 1.130, 95% CI 0.894 to 1.428, P=0.305), and mainly concentrated in highly dependent smokers (RR 1.631, 95% CI 1.290 to 2.061, P<0.001) and heavy smokers (RR 1.515, 95% CI 1.226 to 1.873, P<0.001) rather than individuals with low nicotine dependence (RR 0.989, 95% CI 0.815 to 1.199, P=0.907) or low cigarette consumption (RR 0.985, 95% CI 0.800 to 1.212, P=0.252). For safety outcomes, the combination treatment was associated with more anxiety (RR 1.717, 95% CI 1.176 to 2.505, P=0.005) and insomnia (RR 1.268, 95% CI 1.076 to 1.494, P=0.005) symptoms compared with varenicline monotherapy. CONCLUSION Compared with varenicline monotherapy, combination treatment with varenicline and bupropion can significantly improve the abstinence rate at the end of treatment and 6months follow-up, mainly in highly dependent smokers and heavy smokers.
Collapse
|
13
|
Skånland SS, Cieślar-Pobuda A. Off-label uses of drugs for depression. Eur J Pharmacol 2019; 865:172732. [PMID: 31622593 DOI: 10.1016/j.ejphar.2019.172732] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 02/08/2023]
Abstract
The prescription of drugs for depression is rising rapidly. One of the reasons for this trend is their many off-label uses. Up to one third of all prescriptions are for non-indicated use, which in addition to drug repurposing includes different dosing or duration than those recommended. In this review, we elaborate on what antidepressants can treat besides depression. The five classes of drugs for depression are introduced, and their mechanisms of action and serious side effects are described. The most common off-label uses of antidepressants are discussed, with a special focus on treating eating disorders, sleep problems, smoking cessation and managing chronic pain. Depression is often a comorbidity when antidepressants are chosen as therapy, but good therapeutic effects have been observed for other conditions also when depression is not involved. Finally, a new type of antidepressant developed from the hallucinogenic "party drug" ketamine is briefly introduced. This recent development suggests that antidepressants will keep playing a central role in medicine for years to come.
Collapse
Affiliation(s)
- Sigrid S Skånland
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; The K. G. Jebsen Centre for B Cell Malignancies, Institute for Clinical Medicine, University of Oslo, Oslo, Norway; K. G. Jebsen Centre for Cancer Immunotherapy, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Artur Cieślar-Pobuda
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; K. G. Jebsen Centre for Cancer Immunotherapy, Institute for Clinical Medicine, University of Oslo, Oslo, Norway; Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway.
| |
Collapse
|
14
|
Tran AX, Ho TT, Varghese Gupta S. Role of CYP2B6 pharmacogenomics in bupropion-mediated smoking cessation. J Clin Pharm Ther 2018; 44:174-179. [PMID: 30578565 DOI: 10.1111/jcpt.12783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/25/2018] [Accepted: 11/18/2018] [Indexed: 01/11/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Pharmacogenomics holds promise in smoking cessation because of its potential to shed light on the complexity of drug metabolism and improve treatments using therapeutic agents. The cytochrome P450 2B6 gene (CYP2B6) encodes CYP2B6 enzyme that has been found to mediate the hydroxylation of bupropion, a smoking cessation aid. CYP2B6 exhibits a range of polymorphic variants that alter the pharmacokinetics and pharmacodynamics of bupropion. Genetic variations in CYP2B6 may influence the risk of adverse effects or efficacy of treatment with bupropion. The objective of this review was to investigate the influence of pharmacogenomics on smoking cessation therapy. METHODS A thorough literature search was conducted on PubMed, SCOPUS and EMBASE using keywords related to bupropion, smoking cessation, pharmacogenomics and CYP2B6. Research and review articles, case reports and clinical and preclinical studies pertinent to the research topic were identified, evaluated and summarized. Cited articles within the above-mentioned sources also provided pertinent information. RESULTS There is strong literature evidence to prove that CYP2B6 polymorphisms affect pharmacokinetic and pharmacodynamic properties of bupropion, thus affecting the therapeutic outcome of smoking cessation therapy. WHAT IS NEW AND CONCLUSIONS Complete understanding of pharmacogenetic variation of bupropion pharmacokinetics and pharmacodynamics will be beneficial for designing safer and more personalized smoking cessation therapy with improved outcomes.
Collapse
Affiliation(s)
- Andrew X Tran
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Teresa T Ho
- Department of Pharmacotherapeutics and Clinical Research, University of South Florida College of Pharmacy, Tampa, Florida
| | - Sheeba Varghese Gupta
- Department of Pharmaceutical Sciences, University of South Florida College of Pharmacy, Tampa, Florida
| |
Collapse
|
15
|
Semenova S, Jin X, McClure-Begley TD, Tadman MP, Marks MJ, Markou A. Differential effects of withdrawal from intermittent and continuous nicotine exposure on reward deficit and somatic aspects of nicotine withdrawal and expression of α4β2* nAChRs in Wistar male rats. Pharmacol Biochem Behav 2018; 171:54-65. [PMID: 29908200 DOI: 10.1016/j.pbb.2018.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/03/2018] [Accepted: 06/12/2018] [Indexed: 10/14/2022]
Abstract
BACKGROUND Chronic nicotine exposure produces neuroadaptations in brain reward systems and α4β2 nicotinic acetylcholine receptors (nAChRs) in the corticolimbic brain areas. We previously demonstrated opposite effects of nicotine exposure delivered by self-administration or pumps on brain reward thresholds that can be attributed to the different temporal pattern and contingency of nicotine exposure. We investigated the effects of these two factors on reward thresholds and somatic signs during nicotine withdrawal, and on nAChRs binding in corticolimbic brain areas. METHODS The intracranial self-stimulation procedure was used to assess reward thresholds in rats prepared with pumps delivering various doses of nicotine continuously or intermittently. Separate group of rats were randomly exposed to nicotine via pumps (non-contingent) or nicotine self-administration (contingent) to determine [125I]-epibatidine binding at α4β2* nAChRs. RESULTS Withdrawal from continuous non-contingent nicotine exposure led to significant elevations in thresholds and increases in somatic signs in rats, while there was no significant effect of withdrawal from intermittent non-contingent nicotine exposure at the same doses. nAChRs were upregulated during withdrawal from continuous non-contingent nicotine exposure. α4β2* nAChRs were upregulated in the ventral tegmental area and prelimbic cortex during withdrawal from non-contingent intermittent exposure and in the nucleus accumbens during withdrawal from contingent intermittent nicotine exposure to the same dose. CONCLUSIONS During non-contingent nicotine exposure, the temporal pattern of nicotine delivery differentially affected thresholds and somatic signs of withdrawal. Upregulation of α4β2* nAChRs was brain site-specific and depended on both temporal pattern and contingency of nicotine exposure.
Collapse
Affiliation(s)
- Svetlana Semenova
- Department of Psychiatry, School of Medicine, University California San Diego, 9500 Gilman Drive, M/C 0603, La Jolla, CA 92093, USA.
| | - Xinchun Jin
- Department of Psychiatry, School of Medicine, University California San Diego, 9500 Gilman Drive, M/C 0603, La Jolla, CA 92093, USA
| | | | | | - Michael J Marks
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309, USA; Department of Psychology and Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Athina Markou
- Department of Psychiatry, School of Medicine, University California San Diego, 9500 Gilman Drive, M/C 0603, La Jolla, CA 92093, USA
| |
Collapse
|
16
|
Phillips AG, Geyer MA, Robbins TW. Effective Use of Animal Models for Therapeutic Development in Psychiatric and Substance Use Disorders. Biol Psychiatry 2018; 83:915-923. [PMID: 29478700 DOI: 10.1016/j.biopsych.2018.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/13/2017] [Accepted: 01/12/2018] [Indexed: 12/28/2022]
Abstract
Athina Markou and others argue forcefully for the adoption of a "translational-back translational strategy" for central nervous system drug discovery involving novel application of drugs with established safety profiles in proof-of-principle studies in humans, which in turn encourage parallel studies using experimental animals to provide vital data on the neural systems and neuropharmacological mechanisms related to the actions of the candidate drugs. Encouraged by the increasing adoption of drug-development strategies involving reciprocal information exchange between preclinical animal studies and related clinical research programs, this review presents additional compelling examples related to the following: 1) the treatment of cognitive deficits that define attention-deficit/hyperactivity disorder; 2) the development of fast-acting antidepressants based on promising clinical effects with low doses of the anesthetic ketamine; and 3) new and effective medications for the treatment of substance misuse. In the context of addressing the unmet medical need for new and effective drugs for treatment of mental ill health, now may be the time to launch major new academic-industry consortia committed to open access of all preclinical and clinical data generated by this research.
Collapse
Affiliation(s)
- Anthony G Phillips
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Mark A Geyer
- Department of Psychiatry, University of California-San Diego, La Jolla, California
| | - Trevor W Robbins
- Department of Psychology and Behavioral and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Rose JE, Behm FM. Combination Varenicline/Bupropion Treatment Benefits Highly Dependent Smokers in an Adaptive Smoking Cessation Paradigm. Nicotine Tob Res 2018; 19:999-1002. [PMID: 29054128 PMCID: PMC5896474 DOI: 10.1093/ntr/ntw283] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/22/2016] [Indexed: 11/14/2022]
Abstract
Introduction This study replicated and extended results of a previous trial, which found that combination varenicline/bupropion treatment increased smoking abstinence in smokers who were male, highly dependent, and who did not respond to prequit nicotine patch treatment with a >50% reduction in expired-air carbon monoxide in the first week. Methods One hundred and twenty-two male nicotine patch nonresponders and 52 responders were identified. Smokers in each group were randomized to receive 12 weeks of varenicline plus bupropion treatment versus varenicline plus placebo. The primary outcome was continuous smoking abstinence at weeks 8–11 after the target quit date. Results For smokers with a high level of dependence, judged by having a baseline Fagerstrom Test for Nicotine Dependence (FTND) score ≥ 6 and cigarette consumption ≥ 20/d, combination varenicline/bupropion treatment increased the abstinence rate relative to varenicline alone: 71.0% versus 43.8% (odds ratio = 3.14; 95% confidence interval = 1.11–8.92, p [one tailed] = .016). In contrast, less dependent smokers did not show a benefit of combination treatment relative to varenicline (abstinence rates of 32.1% vs. 45.6%, respectively); there was a significant interaction of treatment and dependence level. Patch nonresponders tended to benefit the most from combination treatment, which was well tolerated overall. Conclusions Combination varenicline/bupropion treatment proved significantly more efficacious than varenicline alone among highly dependent male smokers. These results, together with prior studies, support an adaptive treatment paradigm that assigns smoking cessation treatment according to baseline smoker characteristics and initial response to nicotine patch treatment. Implications This study replicated, in a prospective manner, an important and surprising retrospective finding from a previous clinical trial, which showed that a specific subpopulation of smokers benefited substantially from receiving a combination treatment of varenicline plus bupropion, relative to varenicline plus placebo. Specifically, male smokers having high baseline nicotine dependence (FTND score ≥ 6 and cigarette consumption ≥ 20/d), showed a marked increase in smoking abstinence rate on combination pharmacotherapy. The present study likewise found an enhancement in end-of-treatment abstinence rate in this subgroup, from 43.8% to 71.0%. The adaptive treatment paradigm, which classifies smokers based on initial dependence level and response to prequit nicotine patch treatment, may be used to identify target populations of smokers whose success can be enhanced by intervening with combination pharmacotherapy before the quit-smoking date. Trial Registration ClinicalTrials.gov identifier: NCT01806779.
Collapse
Affiliation(s)
- Jed E Rose
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC
| | - Frédérique M Behm
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC
| |
Collapse
|
18
|
|
19
|
Spierling SR, Zorrilla EP. Don't stress about CRF: assessing the translational failures of CRF 1antagonists. Psychopharmacology (Berl) 2017; 234:1467-1481. [PMID: 28265716 PMCID: PMC5420464 DOI: 10.1007/s00213-017-4556-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/27/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Dr. Athina Markou sought treatments for a common neural substrate shared by depression and drug dependence. Antagonists of corticotropin-releasing factor (CRF) receptors, a target of interest to her, have not reached the clinic despite strong preclinical rationale and sustained translational efforts. METHODS We explore potential causes for the failure of CRF1 antagonists and review recent findings concerning CRF-CRF1 systems in psychopathology. RESULTS Potential causes for negative outcomes include (1) poor safety and efficacy of initial drug candidates due to bad pharmacokinetic and physicochemical properties, (2) specificity problems with preclinical screens, (3) the acute nature of screens vs. late-presenting patients, (4) positive preclinical results limited to certain models and conditions with dynamic CRF-CRF1 activation not homologous to tested patients, (5) repeated CRF1 activation-induced plasticity that reduces the importance of ongoing CRF1 agonist stimulation, and (6) therapeutic silencing which may need to address CRF2 receptor or CRF-binding protein molecules, constitutive CRF1 activity, or molecules that influence agonist-independent activity or to target structural regions other than the allosteric site bound by all drug candidates. We describe potential markers of activation towards individualized treatment, human genetic, and functional data that still implicate CRF1 systems in emotional disturbance, sex differences, and suggestive clinical findings for CRF1 antagonists in food craving and CRF-driven HPA-axis overactivation. CONCLUSION The therapeutic scope of selective CRF1 antagonists now appears narrower than had been hoped. Yet, much remains to be learned about CRF's role in the neurobiology of dysphoria and addiction and the potential for novel anti-CRF therapies therein.
Collapse
Affiliation(s)
- Samantha R Spierling
- Committee on the Neurobiology of Addictive Disorders, SP30-2400, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Eric P Zorrilla
- Committee on the Neurobiology of Addictive Disorders, SP30-2400, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
20
|
Vogeler T, McClain C, Evoy KE. Combination bupropion SR and varenicline for smoking cessation: a systematic review. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 42:129-39. [PMID: 26809272 DOI: 10.3109/00952990.2015.1117480] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Tobacco is the leading cause of preventable death in the world. Current cessation medications include nicotine replacement therapy (NRT), varenicline, and bupropion, while combination therapy primarily entails NRT with either varenicline or bupropion. However, recent studies have examined varenicline and bupropion in combination. OBJECTIVES A systematic review assessing the efficacy and safety of combination varenicline and bupropion was conducted. METHODS PubMed and Clinicaltrials.gov were searched using terms: "varenicline combination", "bupropion combination", "bupropion AND varenicline", and "bupropion AND varenicline combination smoking cessation", yielding four studies including 1193 total patients. RESULTS Combination therapy yielded greater efficacy than varenicline monotherapy in two randomized controlled trials and one retrospective outcomes study. One single-arm Phase II trial provided additional efficacy and safety data. Of the prospective trials, one displayed a greater 4-week smoking abstinence for weeks 8-11 with combination (39.8%) versus monotherapy (25.9%) (OR = 1.89; 95% CI = 1.07-3.35). The other demonstrated greater prolonged abstinence (continuous abstinence from week 2) at 12 weeks (OR = 1.49; 95% CI = 1.05-2.12) and 26 weeks (OR = 1.52; 95% CI = 1.04-2.22), though results were not significant at 52 weeks in this study. The retrospective study displayed higher success rates (continuous abstinence rates at 52 weeks) with combination varenicline and bupropion (55.0%; compared to varenicline monotherapy (32.1%), p < 0.001). Subgroup analyses suggest that this combination may be more beneficial in males and patients with higher baseline nicotine dependence. CONCLUSION To the authors' knowledge, this is the first review conducted to compile current literature on this novel pharmacotherapy combination for smoking cessation. Combination bupropion SR and varenicline displayed greater efficacy in smoking cessation than varenicline monotherapy, though further safety analysis is warranted to rule out additive psychiatric adverse effects.
Collapse
Affiliation(s)
- Tiffany Vogeler
- a Palm Beach Atlantic University , West Palm Beach , FL , USA
| | | | - Kirk E Evoy
- c College of Pharmacy, University of Texas at Austin , San Antonio , TX , USA
| |
Collapse
|
21
|
Neuroscience of nicotine for addiction medicine: novel targets for smoking cessation medications. PROGRESS IN BRAIN RESEARCH 2015; 223:191-214. [PMID: 26806777 DOI: 10.1016/bs.pbr.2015.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Morbidity and mortality associated with tobacco smoking constitutes a significant burden on healthcare budgets all over the world. Therefore, promoting smoking cessation is an important goal of health professionals and policy makers throughout the world. Nicotine is a major psychoactive component in tobacco that is largely responsible for the widespread addiction to tobacco. A majority of the currently available FDA-approved smoking cessation medications act via neuronal nicotinic receptors. These medications are effective in approximately half of all the smokers, who want to quit and relapse among abstinent smokers continues to be high. In addition to relapse among abstinent smokers, unpleasant effects associated with nicotine withdrawal are a major motivational factor in continued tobacco smoking. Over the last two decades, animal studies have helped in identifying several neural substrates that are involved in nicotine-dependent behaviors including those associated with nicotine withdrawal and relapse to tobacco smoking. In this review, first the role of specific brain regions/circuits that are involved in nicotine dependence will be discussed. Next, the review will describe the role of specific nicotinic receptor subunits in nicotine dependence. Finally, the review will discuss the role of classical neurotransmitters (dopamine, serotonin, noradrenaline, glutamate, and γ-aminobutyric acid) as well as endogenous opioid and endocannabinoid signaling in nicotine dependence. The nicotinic and nonnicotinic neural substrates involved in nicotine-dependent behaviors can serve as possible targets for future smoking cessation medications.
Collapse
|
22
|
Hall FS, Der-Avakian A, Gould TJ, Markou A, Shoaib M, Young JW. Negative affective states and cognitive impairments in nicotine dependence. Neurosci Biobehav Rev 2015; 58:168-85. [PMID: 26054790 DOI: 10.1016/j.neubiorev.2015.06.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 02/13/2015] [Accepted: 06/03/2015] [Indexed: 10/23/2022]
Abstract
Smokers have substantial individual differences in quit success in response to current treatments for nicotine dependence. This observation may suggest that different underlying motivations for continued tobacco use across individuals and nicotine cessation may require different treatments in different individuals. Although most animal models of nicotine dependence emphasize the positive reinforcing effects of nicotine as the major motivational force behind nicotine use, smokers generally report that other consequences of nicotine use, including the ability of nicotine to alleviate negative affective states or cognitive impairments, as reasons for continued smoking. These states could result from nicotine withdrawal, but also may be associated with premorbid differences in affective and/or cognitive function. Effects of nicotine on cognition and affect may alleviate these impairments regardless of their premorbid or postmorbid origin (e.g., before or after the development of nicotine dependence). The ability of nicotine to alleviate these symptoms would thus negatively reinforce behavior, and thus maintain subsequent nicotine use, contributing to the initiation of smoking, the progression to dependence and relapse during quit attempts. The human and animal studies reviewed here support the idea that self-medication for pre-morbid and withdrawal-induced impairments may be more important factors in nicotine addiction and relapse than has been previously appreciated in preclinical research into nicotine dependence. Given the diverse beneficial effects of nicotine under these conditions, individuals might smoke for quite different reasons. This review suggests that inter-individual differences in the diverse effects of nicotine associated with self-medication and negative reinforcement are an important consideration in studies attempting to understand the causes of nicotine addiction, as well as in the development of effective, individualized nicotine cessation treatments.
Collapse
Affiliation(s)
- F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA.
| | - Andre Der-Avakian
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Thomas J Gould
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Athina Markou
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Mohammed Shoaib
- Institute of Neuroscience, Newcastle University, Newcastle, UK
| | - Jared W Young
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
23
|
Abstract
Many patients with major depressive disorder (MDD) only partially respond, and some have no clinically meaningful response, to current widely used antidepressant drugs. Due to the purported role of dopamine in the pathophysiology of depression, triple-reuptake inhibitors (TRIs) that simultaneously inhibit serotonin (5-HT), norepinephrine (NE) and dopamine reuptake could be a useful addition to the armamentarium of treatments for MDD. A TRI should more effectively activate mesolimbic dopamine-related reward-networks, restore positive mood and reduce potent 5-HT reuptake blockade associated "hypodopaminergic" adverse effects of decreased libido, weight gain and "blunting" of emotions. On the other hand, dopaminergic effects raise concern over abuse liability and TRIs may have many of the cardiovascular effects associated with NET inhibition. Several clinical development programs for potential TRI antidepressants have failed to demonstrate significantly greater efficacy than placebo or standard of care. Successful late-stage clinical development of a TRI is more likely if experimental research studies in the target population of depressed patients have demonstrated target engagement that differentially and dose-dependently improves assessments of reward-network dysfunction relative to existing antidepressants. TRI treatment could be individualized on the basis of predictive markers such as the burden of decreased positive mood symptoms and/or neuroimaging evidence of reward network dysfunction. This review focuses on how the next generation of monoamine-based treatments could be efficiently developed to address unmet medical need in MDD.
Collapse
|
24
|
Hall BJ, Slade S, Wells C, Rose JE, Levin ED. Bupropion-varenicline interactions and nicotine self-administration behavior in rats. Pharmacol Biochem Behav 2015; 130:84-9. [PMID: 25616031 DOI: 10.1016/j.pbb.2015.01.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 01/06/2015] [Accepted: 01/14/2015] [Indexed: 11/30/2022]
Abstract
Varenicline and bupropion each have been shown to significantly improve cessation of tobacco addiction in humans. They act through different mechanisms and the question about the potential added efficacy with their combined used has arisen. Preclinical animal models of nicotine addiction can help with the evaluation of this combined approach and what dose combinations of varenicline and bupropion may be useful for enhancing tobacco cessation. In this study, we investigated the interacting dose-effect functions of varenicline and bupropion in a rat model of nicotine self-administration. Young adult female Sprague-Dawley rats were allowed to self-administer nicotine in 1-h sessions under an FR1 reinforcement schedule. Varenicline (0.3, 1. 3 mg/kg) and bupropion (8.33, 25, 75 mg/kg) were administered alone or together 15 min before each session. The vehicle saline was the control. Higher doses of each drug alone reduced nicotine self-administration compared to control with reductions of 62% and 75% with 3 mg/kg varenicline and 75 mg/kg bupropion respectively. Lower dose varenicline which does not by itself reduce nicotine self-administration, significantly augmented bupropion effects. The 0.3 mg/kg varenicline dose combined with the 25 and 75 mg/kg bupropion doses caused greater reductions of nicotine self-administration than either dose of bupropion given alone. However, higher dose varenicline did not have this effect. Lower dose bupropion did not augment varenicline effects. Only the high bupropion dose significantly enhanced the varenicline effect. Likewise, combining 1 mg/kg varenicline with 75 mg/kg bupropion reduced self-administration to a greater extent than either dose alone. These results demonstrate that combination therapy with varenicline and bupropion may be more beneficial than monotherapy with either drug alone.
Collapse
Affiliation(s)
- Brandon J Hall
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, USA
| | - Susan Slade
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, USA
| | - Corinne Wells
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, USA
| | - Jed E Rose
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, USA
| | - Edward D Levin
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, USA.
| |
Collapse
|
25
|
Abstract
There is abundant evidence that the dopamine (DA) neurons that project to the nucleus accumbens play a central role in neurobiological mechanisms underpinning drug dependence. This chapter considers the ways in which these projections facilitate the addiction to nicotine and tobacco. It focuses on the complimentary roles of the two principal subdivisions of the nucleus accumbens, the accumbal core and shell, in the acquisition and maintenance of nicotine-seeking behavior. The ways in which tonic and phasic firing of the neurons contributes to the ways in which the accumbens mediate the behavioral responses to nicotine are also considered. Experimental studies suggest that nicotine has relatively weak addictive properties which are insufficient to explain the powerful addictive properties of tobacco smoke. This chapter discusses hypotheses that seek to explain this conundrum. They implicate both discrete sensory stimuli closely paired with the delivery of tobacco smoke and contextual stimuli habitually associated with the delivery of the drug. The mechanisms by which each type of stimulus influence tobacco dependence are hypothesized to depend upon the increased DA release and overflow, respectively, in the two subdivisions of the accumbens. It is suggested that a majority of pharmacotherapies for tobacco dependence are not more successful because they fail to address this important aspect of the dependence.
Collapse
Affiliation(s)
- David J K Balfour
- Medical Research Institute, Division of Neuroscience, Ninewells Hospital and Medical School, Dundee, DD1 9SY, Scotland,
| |
Collapse
|
26
|
Neurobiological Bases of Cue- and Nicotine-induced Reinstatement of Nicotine Seeking: Implications for the Development of Smoking Cessation Medications. Curr Top Behav Neurosci 2015; 24:125-54. [PMID: 25638336 DOI: 10.1007/978-3-319-13482-6_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A better understanding of the neurobiological factors that contribute to relapse to smoking is needed for the development of efficacious smoking cessation medications. Reinstatement procedures allow the preclinical assessment of several factors that contribute to relapse in humans, including re-exposure to nicotine via tobacco smoking and the presentation of stimuli that were previously associated with nicotine administration (i.e., conditioned stimuli). This review provides an integrated discussion of the results of animal studies that used reinstatement procedures to assess the efficacy of pharmacologically targeting various neurotransmitter systems in attenuating the cue- and nicotine-induced reinstatement of nicotine seeking. The results of these animal studies have increased our understanding of the neurobiological processes that mediate the conditioned effects of stimuli that trigger reinstatement to nicotine seeking. Thus, these findings provide important insights into the neurobiological substrates that modulate relapse to tobacco smoking in humans and the ongoing search for novel efficacious smoking cessation medications.
Collapse
|
27
|
Rose JE, Behm FM. Combination treatment with varenicline and bupropion in an adaptive smoking cessation paradigm. Am J Psychiatry 2014; 171:1199-205. [PMID: 24934962 PMCID: PMC4557205 DOI: 10.1176/appi.ajp.2014.13050595] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The authors assessed the efficacy and safety of combination treatment with varenicline and sustained-release bupropion for smokers who, based on an assessment of initial smoking reduction prior to the quit date, were deemed unlikely to achieve abstinence using nicotine patch treatment. METHOD In a randomized, double-blind, parallel-group adaptive treatment trial, the authors identified 222 cigarette smokers who failed to show a reduction of more than 50% in smoking after 1 week of nicotine patch treatment. Smokers were randomly assigned to receive 12 weeks of varenicline plus bupropion or varenicline plus placebo. The primary outcome measure was continuous smoking abstinence at weeks 8-11 after the target quit date. RESULTS Both treatments were well tolerated. Participants who received the combination treatment had a significantly higher abstinence rate than those who received varenicline plus placebo (39.8% compared with 25.9%; odds ratio=1.89; 95% CI=1.07, 3.35). Combination treatment had a significantly greater effect on abstinence rate in male smokers (odds ratio=4.26; 95% CI=1.73, 10.49) than in female smokers (odds ratio=0.94; 95% CI=0.43, 2.05). It also had a significantly greater effect in highly nicotine-dependent smokers (odds ratio=3.51, 95% CI=1.64, 7.51) than in smokers with lower levels of dependence (odds ratio=0.71, 95% CI=0.28, 1.80). CONCLUSIONS Among smokers who did not show a sufficient initial response to prequit nicotine patch treatment, combination treatment with varenicline and bupropion proved more efficacious than varenicline alone for male smokers and for smokers with a high degree of nicotine dependence.
Collapse
Affiliation(s)
- Jed E. Rose
- To whom correspondence should be addressed at: Center for Smoking Cessation, Duke University Medical Center, Durham NC 27705, phone: (919) 668-5055, fax: (919) 668-5088,
| | | |
Collapse
|
28
|
Simmons SJ, Gould TJ. Involvement of neuronal β2 subunit-containing nicotinic acetylcholine receptors in nicotine reward and withdrawal: implications for pharmacotherapies. J Clin Pharm Ther 2014; 39:457-67. [PMID: 24828779 DOI: 10.1111/jcpt.12171] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 04/14/2014] [Indexed: 11/28/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Tobacco smoking remains a major health problem. Nicotine binds to nicotinic acetylcholine receptors (nAChRs), which can cause addiction and withdrawal symptoms upon cessation of nicotine administration. Pharmacotherapies for nicotine addiction target brain alterations that underlie withdrawal symptoms. This review will delineate the involvement of the β2 subunit of neuronal nAChRs in nicotine reward and in generating withdrawal symptoms to better understand the efficacy of smoking cessation pharmacotherapies. COMMENT Chronic nicotine desensitizes and upregulates β2 subunit-containing nAChRs, and the prolonged upregulation of receptors may underlie symptoms of withdrawal. Experimental research has demonstrated that the β2 subunit of neuronal nAChRs is necessary for generating nicotine reward and withdrawal symptoms. WHAT IS NEW AND CONCLUSION Smoking cessation pharmacotherapies act on β2 subunit-containing nAChRs to reduce nicotine reward and withdrawal symptom severity.
Collapse
Affiliation(s)
- Steven J Simmons
- Department of Psychology, Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | | |
Collapse
|
29
|
Iijima M, Koike H, Chaki S. Effect of an mGlu2/3 receptor antagonist on depressive behavior induced by withdrawal from chronic treatment with methamphetamine. Behav Brain Res 2013; 246:24-8. [PMID: 23473878 DOI: 10.1016/j.bbr.2013.02.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 02/21/2013] [Accepted: 02/26/2013] [Indexed: 11/30/2022]
Abstract
Withdrawal from chronic treatment with a psychostimulant precipitates behavioral and physiological conditions similar to the symptoms of major depressive disorder (MDD). Accumulated studies have indicated that withdrawal from a psychostimulant in rodents elicits depressive phenotypes including despair and anhedonia. Recently, the modulation of the group II metabotropic glutamate (mGlu2/3) receptor has been proposed as a novel therapeutic approach to MDD. In the present study, we investigated the effect of an mGlu2/3 receptor antagonist, LY341495, on the depressive behavior induced by withdrawal from chronic treatment with a psychostimulant, methamphetamine (MAP) (5.0mg/kg/day×5 days). The rats were then tested for depressive behavior using the forced swimming test. Withdrawal from chronic treatment with MAP increased the immobility time during the forced swimming test, indicating increased depressive behavior. Systemically administered LY341495 counteracted the depressive behavior induced by withdrawal from chronic treatment with MAP. Moreover, we found that the microinjection of LY341495 into the nucleus accumbens (NAc) also counteracted the increase in the immobility time caused by withdrawal from chronic treatment with MAP. Taken together, the present results suggested that the blockade of the mGlu2/3 receptor may prevent the depressive symptoms induced by withdrawal from a psychostimulant and that the blockade of the mGlu2/3 receptor in the NAc may contribute to the antidepressant-like effects of the mGlu2/3 receptor antagonist in this test.
Collapse
Affiliation(s)
- Michihiko Iijima
- Discovery Pharmacology І, Molecular Function and Pharmacology Laboratories, Taisho Pharmaceutical Co. Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama, Japan
| | | | | |
Collapse
|
30
|
Abstract
Tobacco dependence is an addiction with high rates of relapse, resulting in multiple quit attempts in individuals who are trying to stop smoking. How these multiple cycles of smoking and withdrawal contribute to nicotine dependence, long-term alterations in brain reward systems, and nicotine receptor regulation is unknown. Therefore, to evaluate how multiple exposures of nicotine and withdrawal periods modulate rewarding properties of nicotine, we used intracranial self-stimulation to measure alterations in the threshold of brain stimulation reward. In addition, we employed the conditioned place preference (CPP) paradigm to evaluate positive context conditioning following each withdrawal period and measured levels of neuronal nicotinic receptors in cortex, striatum, and hippocampus. We found that repeated nicotine exposure and withdrawal enhanced brain stimulation reward and reward sensitivity to acute injections of nicotine. This increased reward was reflected by enhanced CPP to nicotine. Chronic nicotine is known to up-regulate nAChRs (nicotinic acetylcholine receptors) and we found that this up-regulation was maintained for up to 8 days of withdrawal in the striatum and in the hippocampus, but not in the cortex, of animals exposed to multiple nicotine exposure and withdrawal periods. These results demonstrate that repeated exposures to nicotine, followed by withdrawal, induce a persistent increase in both brain reward function and sensitivity to the hedonic value of nicotine and long-lasting up-regulation of neuronal nicotinic receptors. Together, these data suggest that a continuing increase in brain reward function and enhanced sensitivity to nicotine reward following repeated withdrawal periods may be one reason why smokers relapse frequently.
Collapse
|
31
|
Breivogel C, Jamerson B. Passion flower extract antagonizes the expression of nicotine locomotor sensitization in rats. PHARMACEUTICAL BIOLOGY 2012; 50:1310-1316. [PMID: 22873194 DOI: 10.3109/13880209.2012.674535] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
CONTEXT Nicotine, a bioactive component of tobacco, is highly addictive. Numerous therapies have been developed or are currently under investigation for smoking cessation, and all have met with limited success and/or side effects, indicating the need for additional therapies. OBJECTIVE This study examines the ability of a commerically-available aqueous extract of Passiflora incarnata Linn. (Passifloraceae) to ameliorate the signs of nicotine sensitization using a rat model. MATERIALS AND METHODS Rats were administered 0.4 mg/kg nicotine or vehicle once a day for four consecutive days. Nicotine adminstration produces sensitization of locomotor activity, a phenomenon implicated in the development of nicotine dependence. On the fifth day, locomotor activity of the subjects was monitored as rats from each treatment group were administered 800 mg/kg of Passiflora incarnata extract (or its vehicle) followed by a challenge dose of 0.4 mg/kg nicotine. RESULTS When given to rats sensitized to nicotine for 4 days, the challenge dose of nicotine increased locomotor activity by more than 2-fold over activity following nicotine challenge in rats treated with vehicle during the sensitization phase. The difference was significant from 15-40 min after nicotine administration. Rats sensitized to nicotine then treated with Passiflora incarnata extract prior to the nicotine challenge exhibited a level of locomotor activity the same as the vehicle-treated controls. DISCUSSION Passiflora incarnata extract did antagonize the expression of nicotine locomotor sensitization. CONCLUSION Passiflora incarnata extract should be examined in future studies to evaluate its potential for treating nicotine addiction in humans.
Collapse
Affiliation(s)
- Chris Breivogel
- Department of Pharmaceutical Sciences, Campbell University College of Pharmacy & Health Sciences, Buies Creek, NC 27506, USA.
| | | |
Collapse
|
32
|
Radke AK, Gewirtz JC. Increased dopamine receptor activity in the nucleus accumbens shell ameliorates anxiety during drug withdrawal. Neuropsychopharmacology 2012; 37:2405-15. [PMID: 22692565 PMCID: PMC3442355 DOI: 10.1038/npp.2012.97] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A number of lines of evidence suggest that negative emotional symptoms of withdrawal involve reduced activity in the mesolimbic dopamine system. This study examined the contribution of dopaminergic signaling in structures downstream of the ventral tegmental area to withdrawal from acute morphine exposure, measured as potentiation of the acoustic startle reflex. Systemic administration of the general dopamine receptor agonist apomorphine or a cocktail of the D1-like receptor agonist SKF82958 and the D2-like receptor agonist quinpirole attenuated potentiated startle during morphine withdrawal. This effect was replicated by apomorphine infusion into the nucleus accumbens shell. Finally, apomorphine injection was shown to relieve startle potentiation during nicotine withdrawal and conditioned place aversion to morphine withdrawal. These results suggest that transient activation of the ventral tegmental area mesolimbic dopamine system triggers the expression of anxiety and aversion during withdrawal from multiple classes of abused drugs.
Collapse
Affiliation(s)
- Anna K Radke
- Graduate Program in Neuroscience, Minneapolis, MN, USA
| | - Jonathan C Gewirtz
- Graduate Program in Neuroscience, Minneapolis, MN, USA,Department of Neuroscience, Minneapolis, MN, USA,Department of Psychology, University of Minnesota, Minneapolis, MN, USA,Department of Psychology, University of Minnesota, N-218 Elliott Hall, 75 East River Road, Minneapolis, MN 55455, USA, Tel: +1 612 625 6653, Fax: +1 612 626 2079 E-mail:
| |
Collapse
|
33
|
Grabus SD, Carroll FI, Damaj MI. Bupropion and its main metabolite reverse nicotine chronic tolerance in the mouse. Nicotine Tob Res 2012; 14:1356-61. [PMID: 22589419 DOI: 10.1093/ntr/nts088] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
INTRODUCTION Although the antidepressant bupropion is prescribed to aid in smoking cessation, little is known concerning its mechanisms of action in this regard. One factor that might influence quit success is nicotine tolerance, which could promote high levels of nicotine intake in order to maintain nicotine's subjective effects (thereby making attempts to reduce smoking more difficult). METHODS To explore whether bupropion and its active hydroxymetabolite modulate nicotine tolerance, mice were injected for 14 days with saline or nicotine. On Day 14, animals received saline, (2S,3S)-hydroxybupropion, or bupropion at different doses. On Day 15, mice were assayed on test day for nicotine-induced analgesia and hypothermia. RESULTS Animals chronically injected with saline + nicotine developed tolerance to nicotine's effects in both assays. Administration of bupropion and (2S,3S)-hydroxybupropion dose-dependently reversed chronic nicotine tolerance. CONCLUSIONS These results indicate that bupropion's ability to promote smoking cessation may be partly due to its attenuation of nicotine tolerance since both measured responses of nicotine (antinociception and hypothermia) are mediated to a large extent by neuronal α4β2* nicotine receptors.
Collapse
Affiliation(s)
- Sheri D Grabus
- Department of Pharmacology & Toxicology, Virginia Commonwealth University Medical Campus, Richmond, VA 23298-0524, USA
| | | | | |
Collapse
|
34
|
|
35
|
Der-Avakian A, Markou A. The neurobiology of anhedonia and other reward-related deficits. Trends Neurosci 2011; 35:68-77. [PMID: 22177980 DOI: 10.1016/j.tins.2011.11.005] [Citation(s) in RCA: 676] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 11/06/2011] [Accepted: 11/17/2011] [Indexed: 01/07/2023]
Abstract
Anhedonia, or markedly diminished interest or pleasure, is a hallmark symptom of major depression, schizophrenia and other neuropsychiatric disorders. Over the past three decades, the clinical definition of anhedonia has remained relatively unchanged, although cognitive psychology and behavioral neuroscience have expanded our understanding of other reward-related processes. Here, we review the neural bases of the construct of anhedonia that reflects deficits in hedonic capacity and also closely linked to the constructs of reward valuation, decision-making, anticipation and motivation. The neural circuits subserving these reward-related processes include the ventral striatum, prefrontal cortical regions, and afferent and efferent projections. An understanding of anhedonia and other reward-related constructs will facilitate the diagnosis and treatment of disorders that include reward deficits as key symptoms.
Collapse
Affiliation(s)
- Andre Der-Avakian
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA 92093-0603, USA
| | | |
Collapse
|
36
|
Hadjiconstantinou M, Neff NH. Nicotine and endogenous opioids: Neurochemical and pharmacological evidence. Neuropharmacology 2011; 60:1209-20. [DOI: 10.1016/j.neuropharm.2010.11.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 11/03/2010] [Accepted: 11/11/2010] [Indexed: 10/18/2022]
|
37
|
Danielson K, Truman P, Kivell BM. The effects of nicotine and cigarette smoke on the monoamine transporters. Synapse 2011; 65:866-79. [DOI: 10.1002/syn.20914] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 12/23/2010] [Indexed: 11/11/2022]
|
38
|
Vlachou S, Paterson NE, Guery S, Kaupmann K, Froestl W, Banerjee D, Finn MG, Markou A. Both GABA(B) receptor activation and blockade exacerbated anhedonic aspects of nicotine withdrawal in rats. Eur J Pharmacol 2011; 655:52-8. [PMID: 21262222 DOI: 10.1016/j.ejphar.2011.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 12/07/2010] [Accepted: 01/07/2011] [Indexed: 01/19/2023]
Abstract
Nicotine dependence is maintained by the aversive, depression-like effects of nicotine withdrawal and the rewarding effects of acute nicotine. GABA(B) receptor antagonists exhibit antidepressant-like effects in rodents, whereas GABA(B) receptor agonists attenuate the rewarding effects of nicotine. Recent studies with GABA(B) receptor positive modulators showed that these compounds represent potentially improved medications for the treatment of nicotine dependence because of fewer side-effects than GABA(B) receptor agonists. Thus, GABA(B) receptor agonists and antagonists, and GABA(B) receptor positive modulators may have efficacy as smoking cessation aids by targeting different aspects of nicotine dependence and withdrawal. The present study assessed the effects of the GABA(B) receptor agonist CGP44532, the GABA(B) receptor antagonist CGP56433A, and the GABA(B) receptor positive modulator BHF177 on the anhedonic aspects of nicotine withdrawal. Rats were prepared with stimulating electrodes in the posterior lateral hypothalamus. After establishing stable intracranial self-stimulation (ICSS) thresholds, rats were prepared with subcutaneous osmotic minipumps delivering either nicotine or saline for 7 or 14days. ICSS thresholds were assessed 6h post-pump removal. Thirty hours after pump removal, CGP44532, CGP56433A, and BHF177 were administered 30min prior to ICSS testing. Both GABA(B) receptor activation (CGP44532 and BHF177) and blockade (CGP56433A) elevated ICSS thresholds in all groups, resulting in exacerbated effects of nicotine withdrawal in the nicotine-treated groups. These similar effects of GABA(B) receptor activation and blockade on the anhedonic depression-like aspects of nicotine withdrawal were surprising and perhaps reflect differential efficacy of these compounds at presynaptic hetero- and autoreceptors, as well as postsynaptic, GABA(B) receptors.
Collapse
Affiliation(s)
- Styliani Vlachou
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA 92093-0603, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Paterson NE. Translational research in addiction: toward a framework for the development of novel therapeutics. Biochem Pharmacol 2011; 81:1388-407. [PMID: 21216239 DOI: 10.1016/j.bcp.2010.12.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 12/13/2010] [Accepted: 12/15/2010] [Indexed: 12/16/2022]
Abstract
The development of novel substance use disorder (SUD) therapeutics is insufficient to meet the medical needs of a growing SUD patient population. The identification of translatable SUD models and tests is a crucial step in establishing a framework for SUD therapeutic development programs. The present review begins by identifying the clinical features of SUDs and highlights the narrow regulatory end-point required for approval of a novel SUD therapeutic. A conceptual overview of dependence is provided, followed by identification of potential intervention targets in the addiction cycle. The main components of the addiction cycle provide the framework for a discussion of preclinical models and their clinical analogs, all of which are focused on isolated behavioral end-points thought to be relevant to the persistence of compulsive drug use. Thus, the greatest obstacle to successful development is the gap between the multiplicity of preclinical and early clinical end-points and the regulatory end-point of sustained abstinence. This review proposes two pathways to bridging this gap: further development and validation of the preclinical extended access self-administration model; inclusion of secondary end-points comprising all of the measures highlighted in the present discussion in Phase 3 trials. Further, completion of the postdictive validation of analogous preclinical and clinical assays is of high priority. Ultimately, demonstration of the relevance and validity of a variety of end-points to the ultimate goal of abstinence will allow researchers to identify truly relevant therapeutic mechanisms and intervention targets, and establish a framework for SUD therapeutic development that allows optimal decision-making and resource allocation.
Collapse
Affiliation(s)
- Neil E Paterson
- Behavioral Pharmacology, PsychoGenics, Inc., 765 Old Saw Mill River Rd., Tarrytown, NY 10591, USA.
| |
Collapse
|
40
|
Hayase T. Depression-related anhedonic behaviors caused by immobilization stress: a comparison with nicotine-induced depression-like behavioral alterations and effects of nicotine and/or "antidepressant" drugs. J Toxicol Sci 2011; 36:31-41. [DOI: 10.2131/jts.36.31] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
41
|
|
42
|
Hadjiconstantinou M, Duchemin AM, Zhang H, Neff NH. Enhanced dopamine transporter function in striatum during nicotine withdrawal. Synapse 2010; 65:91-8. [DOI: 10.1002/syn.20820] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
43
|
Brody AL, London ED, Olmstead RE, Allen-Martinez Z, Shulenberger S, Costello MR, Abrams AL, Scheibal D, Farahi J, Shoptaw S, Mandelkern MA. Smoking-induced change in intrasynaptic dopamine concentration: effect of treatment for Tobacco Dependence. Psychiatry Res 2010; 183:218-24. [PMID: 20682457 PMCID: PMC2947623 DOI: 10.1016/j.pscychresns.2009.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 06/12/2009] [Accepted: 06/17/2009] [Indexed: 11/29/2022]
Abstract
The aim of this study was to determine whether standard treatments for Tobacco Dependence affect smoking-induced changes in intrasynaptic dopamine (DA) concentration. Forty-three otherwise healthy adult cigarette smokers (10 to 40 cigarettes per day) were treated with either practical group counseling (PGC) psychotherapy (n=14), bupropion HCl (n=14), or matching pill placebo (n=15) (random assignment) for 8 weeks. Before and after treatment, each subject underwent a bolus-plus-continuous-infusion (11)C-raclopride positron emission tomography (PET) scanning session, during which he or she smoked a regular cigarette. The PET scanning outcome measure of interest was percent change in smoking-induced (11)C-raclopride binding potential (BP(ND)) in the ventral caudate/nucleus accumbens (VCD/NAc), as an indirect measure of DA release. Although the entire study sample had a smaller mean smoking-induced reduction in VCD/NAc BP(ND) after treatment (compared to before treatment), this change was highly correlated with smaller total cigarette puff volumes (and not other treatment variables). These data indicate that smoking-induced DA release is dose-dependent, and is not significantly affected by reductions in daily smoking levels or treatment type.
Collapse
Affiliation(s)
- Arthur L Brody
- Department of Psychiatry, UCLA, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Varenicline, an alpha4beta2 nicotinic acetylcholine receptor partial agonist, is the newest drug in the armamentarium of tobacco-addiction treatment. Improved smoking quit rates with varenicline are seen in comparison with other pharmacotherapies or behavioral treatments alone. Efficacy, tolerability and safety have been demonstrated in healthy smokers and in smokers with cardiovascular or pulmonary comorbidity, as well as in smokeless tobacco users. Varenicline is started 1 week before a target quit date, uptitrated to 1 mg twice daily, and continued for 12-24 weeks. Post-marketing reports have led to labeling changes to monitor patients for change in behavior, hostility, agitation, depressed mood and suicide-related events. Varenicline's pharmacological profile does not clearly explain an association with these adverse events. A review of placebo-controlled studies found that varenicline was not associated with self-reported neuropsychiatric symptoms, with the exception of sleep disorders. Data in smokers with psychiatric problems are limited.
Collapse
|
45
|
Treadway MT, Zald DH. Reconsidering anhedonia in depression: lessons from translational neuroscience. Neurosci Biobehav Rev 2010; 35:537-55. [PMID: 20603146 DOI: 10.1016/j.neubiorev.2010.06.006] [Citation(s) in RCA: 968] [Impact Index Per Article: 69.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 06/25/2010] [Accepted: 06/28/2010] [Indexed: 01/14/2023]
Abstract
Anhedonia is a core symptom of major depressive disorder (MDD), the neurobiological mechanisms of which remain poorly understood. Despite decades of speculation regarding the role of dopamine (DA) in anhedonic symptoms, empirical evidence has remained elusive, with frequent reports of contradictory findings. In the present review, we argue that this has resulted from an underspecified definition of anhedonia, which has failed to dissociate between consummatory and motivational aspects of reward behavior. Given substantial preclinical evidence that DA is involved primarily in motivational aspects of reward, we suggest that a refined definition of anhedonia that distinguishes between deficits in pleasure and motivation is essential for the purposes of identifying its neurobiological substrates. Moreover, bridging the gap between preclinical and clinical models of anhedonia may require moving away from the conceptualization of anhedonia as a steady-state, mood-like phenomena. Consequently, we introduce the term "decisional anhedonia" to address the influence of anhedonia on reward decision-making. These proposed modifications to the theoretical definition of anhedonia have implications for research, assessment and treatment of MDD.
Collapse
Affiliation(s)
- Michael T Treadway
- Department of Psychology, Vanderbilt University, Nashville, TN 37240, USA.
| | | |
Collapse
|
46
|
Association of the histidine-triad nucleotide-binding protein-1 (HINT1) gene variants with nicotine dependence. THE PHARMACOGENOMICS JOURNAL 2010; 11:251-7. [PMID: 20514075 DOI: 10.1038/tpj.2010.41] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The histidine triad nucleotide-binding protein-1 gene (HINT1) is implicated in schizophrenia and in the behavioral effects of morphine and amphetamine. Because nicotine dependence (ND) is highly comorbid with schizophrenia and other substance abuse, we examined the association of HINT1 with ND. Association analyses from two independent samples show that HINT1 gene variants are associated with ND phenotypes. Furthermore, human postmortem mRNA expression shows that smoking status and genotype influence HINT1 expression in the brain. In animal studies, western blot analyses show an increase of HINT1 protein level in the mouse nucleus accumbens (NAc) after chronic nicotine exposure. This increase was reduced after treatment with the nicotinic-receptor antagonist mecamylamine, and 24 and 72 h after cessation of nicotine treatment. These results indicate a genetic association between HINT1 variants and ND, and indicate that nicotine-induced modulation of HINT1 level may be involved in mechanisms of excess smoking.
Collapse
|
47
|
Carroll FI, Blough BE, Mascarella SW, Navarro HA, Eaton JB, Lukas RJ, Damaj MI. Synthesis and biological evaluation of bupropion analogues as potential pharmacotherapies for smoking cessation. J Med Chem 2010; 53:2204-14. [PMID: 20158204 DOI: 10.1021/jm9017465] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bupropion (2a) analogues were synthesized and tested for their ability to inhibit monoamine uptake and to antagonize the effects of human alpha3beta4*, alpha4beta2, alpha4beta4, and alpha1* nAChRs. The analogues were evaluated for their ability to block nicotine-induced effects in four tests in mice. Nine analogues showed increased monoamine uptake inhibition. Similar to 2a, all but one analogue show inhibition of nAChR function selective for human alpha3beta4*-nAChR. Nine analogues have higher affinity at alpha3beta4*-nAChRs than 2a. Four analogues also had higher affinity for alpha4beta2 nAChR. Analogues 2r, 2m, and 2n with AD(50) values of 0.014, 0.015, and 0.028 mg/kg were 87, 81, and 43 times more potent than 2a in blocking nicotine-induced antinociception in the tail-flick test. Analogue 2x with IC(50) values of 31 and 180 nM for DA and NE, respectively, and with IC(50) of 0.62 and 9.8 microm for antagonism of alpha3beta4 and alpha4beta2 nAChRs had the best overall in vitro profile relative to 2a.
Collapse
Affiliation(s)
- F Ivy Carroll
- Center for Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, North Carolina 27709-2194, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Madden GJ, Kalman D. Effects of bupropion on simulated demand for cigarettes and the subjective effects of smoking. Nicotine Tob Res 2010; 12:416-22. [PMID: 20194522 DOI: 10.1093/ntr/ntq018] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
INTRODUCTION The biobehavioral mechanism(s) mediating bupropion's efficacy are not well understood. Behavioral economic measures such as demand curves have proven useful in investigations of the reinforcing effects of drugs of abuse. Behavioral economic measures may also be used to measure the effect of pharmacotherapies on the reinforcing effects of drugs of abuse. METHODS The effects of bupropion on simulated demand for cigarettes were investigated in a placebo-controlled double-blind clinical trial. Participants reported the number of cigarettes they would purchase and consume in a single day at a range of prices. The effects of medication on the subjective effects of smoking were also explored. RESULTS Demand for cigarettes was well described by an exponential demand equation. Bupropion did not significantly decrease the maximum number of cigarettes that participants said they would smoke in a single day nor did it significantly alter the relation between price per cigarette and demand. Baseline demand elasticity did not predict smoking cessation, but changes in elasticity following 1 week of treatment did. Medication group had no effect on any subjective effects of smoking. DISCUSSION Bupropion had no significant effects on demand for cigarettes. The exponential demand equation, recently introduced in behavioral economics, proved amenable to human simulated demand and might be usefully employed in other pharmacotherapy studies as it provides a potentially useful measure of changes in the essential value of the drug as a reinforcer. Such changes may be useful in predicting the efficacy of medications designed to reduce drug consumption.
Collapse
Affiliation(s)
- Gregory J Madden
- Department of Applied Behavioral Science, University of Kansas, 1000 Sunnyside Ave, Lawrence, KS 66045, USA.
| | | |
Collapse
|
49
|
Egerton A, Shotbolt JP, Stokes PRA, Hirani E, Ahmad R, Lappin JM, Reeves SJ, Mehta MA, Howes OD, Grasby PM. Acute effect of the anti-addiction drug bupropion on extracellular dopamine concentrations in the human striatum: an [11C]raclopride PET study. Neuroimage 2010; 50:260-6. [PMID: 19969097 PMCID: PMC4135078 DOI: 10.1016/j.neuroimage.2009.11.077] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Revised: 11/25/2009] [Accepted: 11/26/2009] [Indexed: 11/19/2022] Open
Abstract
Bupropion is an effective medication in treating addiction and is widely used as an aid to smoking cessation. Bupropion inhibits striatal dopamine reuptake via dopamine transporter blockade, but it is unknown whether this leads to increased extracellular dopamine levels at clinical doses in man. The effects of bupropion on extracellular dopamine levels in the striatum were investigated using [(11)C]raclopride positron emission tomography (PET) imaging in rats administered saline, 11 or 25 mg/kg bupropion i.p. and in healthy human volunteers administered either placebo or 150 mg bupropion (Zyban Sustained-Release). A cognitive task was used to stimulate dopamine release in the human study. In rats, bupropion significantly decreased [(11)C]raclopride specific binding in the striatum, consistent with increases in extracellular dopamine concentrations. In man, no significant decreases in striatal [(11)C]raclopride specific binding were observed. Levels of dopamine transporter occupancy in the rat at 11 and 25 mg/kg bupropion i.p. were higher than predicted to occur in man at the dose used. Thus, these data indicate that, at the low levels of dopamine transporter occupancy achieved in man at clinical doses, bupropion does not increase extracellular dopamine levels. These findings have important implications for understanding the mechanism of action underlying bupropions' therapeutic efficacy and for the development of novel treatments for addiction and depression.
Collapse
Affiliation(s)
- Alice Egerton
- Psychiatric Imaging, Medical Research Council Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Costello MR, Mandelkern MA, Shoptaw S, Shulenberger S, Baker SK, Abrams AL, Xia C, London ED, Brody AL. Effects of treatment for tobacco dependence on resting cerebral glucose metabolism. Neuropsychopharmacology 2010; 35:605-12. [PMID: 19865076 PMCID: PMC2813904 DOI: 10.1038/npp.2009.165] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
While bupropion HCl and practical group counseling (PGC) are commonly used treatments for tobacco dependence, the effects of these treatments on brain function are not well established. For this study, 54 tobacco-dependent cigarette smokers underwent resting (18)F-fluorodeoxyglucose-positron emission tomography (FDG-PET) scanning before and after 8 weeks of treatment with bupropion HCl, PGC, or pill placebo. Using Statistical Parametric Mapping (SPM 2), changes in cerebral glucose metabolism from before to after treatment were compared between treatment groups and correlations were determined between amount of daily cigarette usage and cerebral glucose metabolism. Compared with placebo, the two active treatments (bupropion HCl and PGC) had reductions in glucose metabolism in the posterior cingulate gyrus. Further analysis suggested that PGC had a greater effect than bupropion HCl on glucose metabolism in this region. We also found positive correlations between daily cigarette use and glucose metabolism in the left occipital gyrus and parietal-temporal junction. There were no significant negative correlations between daily cigarette use and glucose metabolism. Our findings suggest that bupropion HCl and PGC reduce neural activity much as the performance of a goal-oriented task does in the default mode network of the brain, including the posterior cingulate gyrus. Thus, this study supports the theory that active treatments for tobacco dependence move the brain into a more goal-oriented state.
Collapse
Affiliation(s)
- Matthew R Costello
- Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA 90095, USA.
| | - Mark A Mandelkern
- Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, USA,UCI Department of Physics, Irvine, CA, USA
| | - Stephen Shoptaw
- UCLA Department of Psychiatry and Biobehavioral Sciences, Los Angeles, CA, USA
| | | | - Stephanie K Baker
- Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, USA
| | - Anna L Abrams
- Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, USA
| | - Catherine Xia
- Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, USA
| | - Edythe D London
- UCLA Department of Psychiatry and Biobehavioral Sciences, Los Angeles, CA, USA,UCLA Brain Research Institute, Los Angeles, CA, USA,UCLA Department of Molecular and Medical Pharmacology, Los Angeles, CA, USA
| | - Arthur L Brody
- Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, USA,UCLA Department of Psychiatry and Biobehavioral Sciences, Los Angeles, CA, USA,UCLA Brain Research Institute, Los Angeles, CA, USA,Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA, Tel: +1 310 268 4778, Fax: +1 310 206 2802, E-mail:
| |
Collapse
|