1
|
Sved AF, Caggiula AR, Donny EC. Elucidating the reinforcing effects of nicotine: a tribute to Nadia Chaudhri. Psychopharmacology (Berl) 2023; 240:417-430. [PMID: 36329195 PMCID: PMC11188050 DOI: 10.1007/s00213-022-06266-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Nadia Chaudhri worked with us as a graduate student in the Center for Neuroscience at the University of Pittsburgh from 1999 until she earned her PhD in 2005, a time that coincided with the discovery in our lab of the dual reinforcing actions of nicotine, a concept that she played an important role in shaping. The research that was described in her doctoral thesis is among the foundational pillars of the now well-accepted notion that nicotine acts as both a primary reinforcer and an amplifier of other reinforcer stimuli. This reinforcement-enhancing action of nicotine is robust and likely to be a powerful driver of nicotine use. Below, we discuss the evidence that these two actions of nicotine - primary reinforcement and reinforcement enhancement - are distinct and dissociable, a finding that Nadia was closely associated with. We go on to address two other topics that greatly interested Nadia during that time, the generalizability of the reinforcement-enhancing action of nicotine to multiple classes of reinforcing stimuli and potential sex differences in the dual reinforcing actions of nicotine. The research has greatly expanded since Nadia's involvement, but the core ideas that she helped to develop remain central to the concept of the dual reinforcing actions of nicotine and its importance for understanding the drivers of nicotine use.
Collapse
Affiliation(s)
- Alan F Sved
- Departments of Neuroscience, Psychiatry and Psychology and the Center for Neuroscience, University of Pittsburgh, 210 Langley Hall, Pittsburgh, PA, 15260, USA.
| | - Anthony R Caggiula
- Departments of Psychology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Eric C Donny
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| |
Collapse
|
2
|
Green R, Roche DJO, Ray LA. The Effects of Menstrual Cycle Hormones on Responses to Varenicline and Naltrexone Among Female Heavy Drinking Smokers. Alcohol Alcohol 2022; 57:609-614. [PMID: 35470371 PMCID: PMC9465527 DOI: 10.1093/alcalc/agac017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/10/2022] [Accepted: 03/19/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Aims
Women often experience poorer smoking cessation outcomes in comparison to men. Menstrual cycle phase and sex hormones may influence smoking behavior and alter response to opioid antagonist medications. Less is known about the effects of sex hormones in response to pharmacotherapy for female heavy drinking smokers.
Methods
This study is a secondary analysis of premenopausal female heavy drinking smokers who completed a 12-week randomized clinical trial comparing varenicline plus placebo versus varenicline plus naltrexone for smoking cessation and drinking reduction. Participants (n = 26; total observations = 66) provided saliva samples for assays of progesterone (P4) and estradiol (E2) post-randomization at Weeks 4, 8 and 12. We examined the effects of P4/E2 ratio and medication on smoking and drinking outcomes.
Results
For drinking outcomes, there was a significant interaction for percent days abstinent (b = 0.017, P = 0.05), suggesting that greater P4/E2 ratio is associated with greater percent days abstinent for women assigned to the varenicline plus naltrexone condition. There were no interaction effects for the remaining drinking outcomes (P’s ≥ 0.12). Results found no significant interaction effect of P4/E2 ratio and medication on smoking abstinence (P = 0.19).
Conclusion
Our results imply that when women show a greater P4/E2 ratio, typically observed during the luteal phase of the menstrual cycle, they experience an added benefit of naltrexone, versus placebo, for drinking outcomes as shown by greater percent days abstinent. Additional studies in larger samples are warranted as sex hormones offer important information above and beyond comparing women versus men.
Collapse
Affiliation(s)
- ReJoyce Green
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Daniel J O Roche
- Department of Psychiatry, University of Maryland Baltimore, Baltimore, MD, USA
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| |
Collapse
|
3
|
Chen G, Ghazal M, Rahman S, Lutfy K. The impact of adolescent nicotine exposure on alcohol use during adulthood: The role of neuropeptides. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:53-93. [PMID: 34801174 DOI: 10.1016/bs.irn.2021.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Nicotine and alcohol abuse and co-dependence represent major public health crises. Indeed, previous research has shown that the prevalence of alcoholism is higher in smokers than in non-smokers. Adolescence is a susceptible period of life for the initiation of nicotine and alcohol use and the development of nicotine-alcohol codependence. However, there is a limited number of pharmacotherapeutic agents to treat addiction to nicotine or alcohol alone. Notably, there is no effective medication to treat this comorbid disorder. This chapter aims to review the early nicotine use and its impact on subsequent alcohol abuse during adolescence and adulthood as well as the role of neuropeptides in this comorbid disorder. The preclinical and clinical findings discussed in this chapter will advance our understanding of this comorbid disorder's neurobiology and lay a foundation for developing novel pharmacotherapies to treat nicotine and alcohol codependence.
Collapse
Affiliation(s)
- G Chen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States; Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - M Ghazal
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States
| | - K Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States.
| |
Collapse
|
4
|
Hartmann-Boyce J, Theodoulou A, Farley A, Hajek P, Lycett D, Jones LL, Kudlek L, Heath L, Hajizadeh A, Schenkels M, Aveyard P. Interventions for preventing weight gain after smoking cessation. Cochrane Database Syst Rev 2021; 10:CD006219. [PMID: 34611902 PMCID: PMC8493442 DOI: 10.1002/14651858.cd006219.pub4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Most people who stop smoking gain weight. This can discourage some people from making a quit attempt and risks offsetting some, but not all, of the health advantages of quitting. Interventions to prevent weight gain could improve health outcomes, but there is a concern that they may undermine quitting. OBJECTIVES To systematically review the effects of: (1) interventions targeting post-cessation weight gain on weight change and smoking cessation (referred to as 'Part 1') and (2) interventions designed to aid smoking cessation that plausibly affect post-cessation weight gain (referred to as 'Part 2'). SEARCH METHODS Part 1 - We searched the Cochrane Tobacco Addiction Group's Specialized Register and CENTRAL; latest search 16 October 2020. Part 2 - We searched included studies in the following 'parent' Cochrane reviews: nicotine replacement therapy (NRT), antidepressants, nicotine receptor partial agonists, e-cigarettes, and exercise interventions for smoking cessation published in Issue 10, 2020 of the Cochrane Library. We updated register searches for the review of nicotine receptor partial agonists. SELECTION CRITERIA Part 1 - trials of interventions that targeted post-cessation weight gain and had measured weight at any follow-up point or smoking cessation, or both, six or more months after quit day. Part 2 - trials included in the selected parent Cochrane reviews reporting weight change at any time point. DATA COLLECTION AND ANALYSIS Screening and data extraction followed standard Cochrane methods. Change in weight was expressed as difference in weight change from baseline to follow-up between trial arms and was reported only in people abstinent from smoking. Abstinence from smoking was expressed as a risk ratio (RR). Where appropriate, we performed meta-analysis using the inverse variance method for weight, and Mantel-Haenszel method for smoking. MAIN RESULTS Part 1: We include 37 completed studies; 21 are new to this update. We judged five studies to be at low risk of bias, 17 to be at unclear risk and the remainder at high risk. An intermittent very low calorie diet (VLCD) comprising full meal replacement provided free of charge and accompanied by intensive dietitian support significantly reduced weight gain at end of treatment compared with education on how to avoid weight gain (mean difference (MD) -3.70 kg, 95% confidence interval (CI) -4.82 to -2.58; 1 study, 121 participants), but there was no evidence of benefit at 12 months (MD -1.30 kg, 95% CI -3.49 to 0.89; 1 study, 62 participants). The VLCD increased the chances of abstinence at 12 months (RR 1.73, 95% CI 1.10 to 2.73; 1 study, 287 participants). However, a second study found that no-one completed the VLCD intervention or achieved abstinence. Interventions aimed at increasing acceptance of weight gain reported mixed effects at end of treatment, 6 months and 12 months with confidence intervals including both increases and decreases in weight gain compared with no advice or health education. Due to high heterogeneity, we did not combine the data. These interventions increased quit rates at 6 months (RR 1.42, 95% CI 1.03 to 1.96; 4 studies, 619 participants; I2 = 21%), but there was no evidence at 12 months (RR 1.25, 95% CI 0.76 to 2.06; 2 studies, 496 participants; I2 = 26%). Some pharmacological interventions tested for limiting post-cessation weight gain (PCWG) reduced weight gain at the end of treatment (dexfenfluramine, phenylpropanolamine, naltrexone). The effects of ephedrine and caffeine combined, lorcaserin, and chromium were too imprecise to give useful estimates of treatment effects. There was very low-certainty evidence that personalized weight management support reduced weight gain at end of treatment (MD -1.11 kg, 95% CI -1.93 to -0.29; 3 studies, 121 participants; I2 = 0%), but no evidence in the longer-term 12 months (MD -0.44 kg, 95% CI -2.34 to 1.46; 4 studies, 530 participants; I2 = 41%). There was low to very low-certainty evidence that detailed weight management education without personalized assessment, planning and feedback did not reduce weight gain and may have reduced smoking cessation rates (12 months: MD -0.21 kg, 95% CI -2.28 to 1.86; 2 studies, 61 participants; I2 = 0%; RR for smoking cessation 0.66, 95% CI 0.48 to 0.90; 2 studies, 522 participants; I2 = 0%). Part 2: We include 83 completed studies, 27 of which are new to this update. There was low certainty that exercise interventions led to minimal or no weight reduction compared with standard care at end of treatment (MD -0.25 kg, 95% CI -0.78 to 0.29; 4 studies, 404 participants; I2 = 0%). However, weight was reduced at 12 months (MD -2.07 kg, 95% CI -3.78 to -0.36; 3 studies, 182 participants; I2 = 0%). Both bupropion and fluoxetine limited weight gain at end of treatment (bupropion MD -1.01 kg, 95% CI -1.35 to -0.67; 10 studies, 1098 participants; I2 = 3%); (fluoxetine MD -1.01 kg, 95% CI -1.49 to -0.53; 2 studies, 144 participants; I2 = 38%; low- and very low-certainty evidence, respectively). There was no evidence of benefit at 12 months for bupropion, but estimates were imprecise (bupropion MD -0.26 kg, 95% CI -1.31 to 0.78; 7 studies, 471 participants; I2 = 0%). No studies of fluoxetine provided data at 12 months. There was moderate-certainty that NRT reduced weight at end of treatment (MD -0.52 kg, 95% CI -0.99 to -0.05; 21 studies, 2784 participants; I2 = 81%) and moderate-certainty that the effect may be similar at 12 months (MD -0.37 kg, 95% CI -0.86 to 0.11; 17 studies, 1463 participants; I2 = 0%), although the estimates are too imprecise to assess long-term benefit. There was mixed evidence of the effect of varenicline on weight, with high-certainty evidence that weight change was very modestly lower at the end of treatment (MD -0.23 kg, 95% CI -0.53 to 0.06; 14 studies, 2566 participants; I2 = 32%); a low-certainty estimate gave an imprecise estimate of higher weight at 12 months (MD 1.05 kg, 95% CI -0.58 to 2.69; 3 studies, 237 participants; I2 = 0%). AUTHORS' CONCLUSIONS Overall, there is no intervention for which there is moderate certainty of a clinically useful effect on long-term weight gain. There is also no moderate- or high-certainty evidence that interventions designed to limit weight gain reduce the chances of people achieving abstinence from smoking.
Collapse
Affiliation(s)
- Jamie Hartmann-Boyce
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Annika Theodoulou
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Amanda Farley
- Public Health, Epidemiology and Biostatistics, University of Birmingham, Birmingham, UK
| | - Peter Hajek
- Wolfson Institute of Preventive Medicine, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Deborah Lycett
- Faculty of Health and Life Sciences, Coventry University, Coventry, UK
| | - Laura L Jones
- Public Health, Epidemiology and Biostatistics, University of Birmingham, Birmingham, UK
| | - Laura Kudlek
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Laura Heath
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Anisa Hajizadeh
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | | | - Paul Aveyard
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
Mishra A, Maiti R, Mishra BR, Jena M. Comparative efficacy and safety of pharmacological interventions for smoking cessation in healthy adults: A network meta-analysis. Pharmacol Res 2021; 166:105478. [PMID: 33549729 DOI: 10.1016/j.phrs.2021.105478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/25/2021] [Accepted: 02/02/2021] [Indexed: 11/19/2022]
Abstract
Smoking is the leading cause of morbidity and mortality in different non-communicable diseases, and cessation leads to immense health benefits. The present network meta-analysis has been conducted to evaluate and compare the effects of available pharmacological interventions for smoking cessation in adults. A standard meta-analysis protocol was developed and after performing a comprehensive literature search on MEDLINE/PubMed, Cochrane databases, and International Clinical Trials Registry Platform, reviewers extracted data from 97 randomized controlled trials. PRISMA guidelines were followed in data extraction, analysis and reporting of findings. Random effects Bayesian network meta-analysis was done to pool the effects across the interventions. Network graph was built, and for closed triangles in the network graph, node splitting analysis was performed. The primary outcome measure was self-reported biochemically verified smoking abstinence at six months. The number of participants achieving continuous abstinence was reported. Data for the number of participants reporting at least one adverse event was also extracted, if available. Combination of nicotine receptor agonist and nicotine replacement therapy had a significant odd of 4.4 (95%CrI:2.2-8.7), bupropion and nicotine receptor agonist 4.0 (95%CrI:2.1-7.7), bupropion and nicotine replacement therapy 3.8 (95%CrI:2.3-6.2), combination nicotine replacement therapy has an odd of 2.6 (95%CrI:1.8-3.8), and nicotine receptor agonist had a significant odd of 2.7 (95%CrI:2.3-3.2) when compared to placebo (moderate quality of evidence) for continuous abstinence at 6 months. When compared with behavioural therapy, the odds ratio of interventions was not statistically significant. Combination of nicotine receptor agonist and nicotine replacement therapy has the highest probability of being the best treatment for abstinence from smoking.
Collapse
Affiliation(s)
- Archana Mishra
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rituparna Maiti
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| | - Biswa Ranjan Mishra
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Monalisa Jena
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| |
Collapse
|
6
|
Domi A, Barbier E, Adermark L, Domi E. Targeting the Opioid Receptors: A Promising Therapeutic Avenue for Treatment in “Heavy Drinking Smokers”. Alcohol Alcohol 2021; 56:127-138. [DOI: 10.1093/alcalc/agaa139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 01/12/2023] Open
Abstract
Abstract
Aims
Despite a general decline in tobacco use in the last decades, the prevalence of tobacco smoking in individuals with alcohol use disorder (AUD) remains substantial (45–50%). Importantly, the co-use of both substances potentiates the adverse effects, making it a significant public health problem. Substantial evidence suggests that AUD and Tobacco use disorder (TUD) may share common mechanisms. Targeting these mechanisms may therefore provide more effective therapy. Numerous studies describe a potential role of the endogenous opioid system in both AUD and TUD. Reviewing this literature, we aim to evaluate the efficacy of molecules that target the opioid system as promising therapeutic interventions for treating alcohol and tobacco co-use disorders.
Methods
We provide a synthesis of the current epidemiological knowledge of alcohol and tobacco co-use disorders. We evaluate clinical and preclinical research that focuses on the regulation of the endogenous opioid system in alcohol, nicotine, and their interactions.
Results
The epidemiological data confirm that smoking stimulates heavy drinking and facilitates alcohol craving. Pharmacological findings suggest that treatments that are efficacious in the dual addiction provide a beneficial treatment outcome in comorbid AUD and TUD. In this regard, MOP, DOP and NOP-receptor antagonists show promising results, while the findings prompt caution when considering KOP-receptor antagonists as a treatment option in alcohol and tobacco co-use disorders.
Conclusions
Existing literature suggests a role of the opioid system in sustaining the high comorbidity rates of AUD and TUD. Molecules targeting opioid receptors may therefore represent promising therapeutic interventions in ‘heavy drinking smokers.’
Collapse
Affiliation(s)
- Ana Domi
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy University of Gothenburg, Box 410, Gothenburg 405 30, Sweden
| | - Estelle Barbier
- Center for Social and Affective Neuroscience, Linköping University, Campus US, Entrance 65, Linköping 581 85, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy University of Gothenburg, Box 410, Gothenburg 405 30, Sweden
| | - Esi Domi
- Center for Social and Affective Neuroscience, Linköping University, Campus US, Entrance 65, Linköping 581 85, Sweden
| |
Collapse
|
7
|
Wang AL, Shi Z, Elman I, Langleben DD. Reduced cigarette smoking during injectable extended-release naltrexone treatment for opioid use disorder. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2020; 46:472-477. [PMID: 32379516 DOI: 10.1080/00952990.2020.1741001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The prevalence of tobacco cigarette smoking in the US has declined to approximately 15%, yet, it remains over 90% among individuals with opioid use disorder regardless of whether they are currently using opioids illicitly or as opioid substitution therapy. This disparity raises the question of whether opioids facilitate smoking among individuals with opioid use disorder and whether opioid antagonists may reduce it. OBJECTIVES Determine whether injectable extended-release naltrexone (XR-NTX) treatment of opioid use disorder patients is associated with a spontaneous smoking reduction. We hypothesized that treatment with XR-NTX for would lead to a reduction in smoking in tobacco cigarette smokers with opioid use disorder. METHODS We analyzed data from 64 tobacco cigarette smokers (38% female) with opioid use disorder who were induced on XR-NTX for prevention of relapse to opioids. The number of cigarettes smoked per day and opioid-related craving and withdrawal were assessed at baseline and during treatment. RESULTS Smoking was reduced from 14.4 ± 1.0 to 9.8 ± 1.0(p < 0.001) cigarettes per day after one month and 8.6 ± 1.1 cigarettes per day after two months of treatment. Daily cigarette consumption was positively correlated with the pre-treatment frequency of opioid use and opioid-related craving during the XR-NTX treatment. CONCLUSIONS XR-NTX treatment in smokers with opioid use disorder was associated with a 29% decline in daily cigarette consumption. Together with prior evidence of increased smoking during opioid agonist therapy, our finding suggests a pharmacodynamic interaction between nicotine and opioid systems that could influence treatment choices in this population. Our findings merit confirmation in a prospective controlled study. (NCT02324725 and NCT01587196).
Collapse
Affiliation(s)
- An-Li Wang
- Addiction Institute of Mount Sinai, Icahn School of Medicine at Mount Sinai , New York, NY, USA
| | - Zhenhao Shi
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA, USA
| | - Igor Elman
- Center for Pain and the Brain, Boston Children's Hospital , Boston, MA, USA
| | - Daniel D Langleben
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA, USA
| |
Collapse
|
8
|
Gendy MNS, Ibrahim C, Sloan ME, Le Foll B. Randomized Clinical Trials Investigating Innovative Interventions for Smoking Cessation in the Last Decade. Handb Exp Pharmacol 2020; 258:395-420. [PMID: 31267165 DOI: 10.1007/164_2019_253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Every year, billions of dollars are spent treating smoking and related conditions, yet smoking-related morbidity and mortality continue to rise. There are currently only three FDA-approved medications for smoking cessation: nicotine replacement therapy, bupropion, and varenicline. Although these medications increase abstinence rates, most individuals relapse following treatment. This chapter reviews clinical trials published within the past 10 years investigating novel smoking cessation pharmacotherapies. Among these pharmacotherapies, some showed promising results, such as cytisine and endocannabinoid modulators, whereas others failed to produce significant effects. More research is needed to develop drugs that produce higher rates of long-term abstinence and to determine which subgroups of patients benefit from a given treatment.
Collapse
Affiliation(s)
- Marie N S Gendy
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Christine Ibrahim
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Matthew E Sloan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada.
- Addictions Division, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Division of Brain and Therapeutics, Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
- Alcohol Research and Treatment Clinic, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Matthews AK, Steffen AD, Kuhns LM, Ruiz RA, Ross NA, Burke LA, Li CC, King AC. Evaluation of a Randomized Clinical Trial Comparing the Effectiveness of a Culturally Targeted and Nontargeted Smoking Cessation Intervention for Lesbian, Gay, Bisexual, and Transgender Smokers. Nicotine Tob Res 2019; 21:1506-1516. [PMID: 30169797 PMCID: PMC6821204 DOI: 10.1093/ntr/nty184] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/29/2018] [Indexed: 11/12/2022]
Abstract
PURPOSE To examine the benefits of a culturally targeted compared with a nontargeted smoking cessation intervention on smoking cessation outcomes among lesbian, gay, bisexual, and transgender (LGBT) smokers. METHODS A prospective randomized design was used to evaluate the added benefits of an LGBT culturally targeted Courage to Quit (CTQ-CT) smoking cessation treatment (N = 172) compared with the standard intervention (CTQ; N = 173). The smoking cessation program consisted of six treatment sessions combined with 8 weeks of nicotine replacement therapy. The primary smoking cessation outcome was 7-day point prevalence quit rates. Secondary outcomes examined included changes in nicotine dependence, nicotine withdrawal, cigarettes per day, smoking urges, self-efficacy, and readiness to quit. RESULTS Overall quit rates were 31.9% at 1 month, 21.1% at 3 months, 25.8% at 6 months, and 22.3% at 12 months. Quit rates did not differ between treatment groups [1 month OR = 0.81 (0.32, 2.09), 3 months OR = 0.65 (0.23, 1.78), 6 months OR = 0.45 (0.17, 1.21), 12 months OR = 0.70 (0.26, 1.91)]. Compared with baseline levels, all secondary smoking cessation outcomes measured were improved at 1 month and were maintained at 12-month follow-up. Compared with the CTQ, the CTQ-CT intervention was more highly rated on program effectiveness (d = 0.2, p = .011), intervention techniques (d = 0.2, p = .014), the treatment manual (d = 0.3, p < .001), and being targeted to the needs of LGBT smokers (d = 0.5, p < .0001). CONCLUSIONS LGBT smokers receiving the CTQ intervention achieved smoking cessation outcomes in the range reported for other demographic groups. Cultural targeting improved the acceptability of the intervention but did not confer any additional benefit for smoking cessation outcomes. IMPLICATIONS Study results have implications for understanding the benefits of culturally targeted compared with nontargeted smoking cessation interventions for improving smoking cessation outcomes among LGBT smokers. Shorter and longer term 7-day point prevalence quit rates associated with the targeted and nontargeted interventions were modest but comparable with other group-based interventions delivered in a community setting. Although cultural targeting improved the overall acceptability of the intervention, no added benefits were observed for the culturally targeted intervention on either the primary or secondary outcomes.
Collapse
Affiliation(s)
- Alicia K Matthews
- Department of Health Systems Sciences, College of Nursing, University of Illinois at Chicago, Chicago, IL
| | - Alana D Steffen
- Department of Health Systems Sciences, College of Nursing, University of Illinois at Chicago, Chicago, IL
| | - Lisa M Kuhns
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Raymond A Ruiz
- College of Medicine, Center for Clinical and Translational Science, University of Illinois at Chicago, Chicago, IL
| | - Nat A Ross
- Department of Research, Howard Brown Health Center, Chicago, IL
| | - Larisa A Burke
- Department of Health Systems Sciences, College of Nursing, University of Illinois at Chicago, Chicago, IL
| | - Chien Ching Li
- Department of Health Services Research, Rush University, Chicago, IL
| | - Andrea C King
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL
| |
Collapse
|
10
|
Green R, Bujarski S, Lim AC, Venegas A, Ray LA. Naltrexone and alcohol effects on craving for cigarettes in heavy drinking smokers. Exp Clin Psychopharmacol 2019; 27:257-264. [PMID: 30628813 PMCID: PMC7227763 DOI: 10.1037/pha0000252] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Naltrexone has been extensively studied for the treatment of alcohol use disorder. However, less is known about the effects of naltrexone on smoking outcomes in the context of alcohol use among East Asian individuals who have been suggested to differ in response to alcohol and to naltrexone. The present study is a secondary analysis that used a double-blind placebo-controlled design (n = 31) to examine the (a) effects of alcohol on basal craving for cigarettes, (b) effects of naltrexone on cigarette craving and alcohol craving during alcohol administration, and (c) relationship between craving for alcohol and cigarettes. Heavy drinking smokers of East Asian descent completed two counterbalanced intravenous alcohol administration sessions, one after taking naltrexone (50 mg) for five days and one after taking a placebo for five days. Self-reported subjective craving for cigarettes and for alcohol was recorded during each experimental session. Craving for cigarettes and alcohol increased significantly throughout the intravenous alcohol administration. A significant breath alcohol concentration (BrAC) × Medication interaction revealed that naltrexone blunted cigarette craving during alcohol administration, compared to placebo. Naltrexone significantly reduced craving for alcohol during alcohol administration in this group of heavy drinking smokers. Alcohol craving significantly predicted cigarette craving, however this effect did not vary across rising alcohol administration or by medication. These findings demonstrate that naltrexone reduces the urge to smoke and to drink during alcohol administration. Clinical studies are needed to further ascertain whether naltrexone may be of benefit to this distinct subgroup of heavy drinking smokers. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
Affiliation(s)
- ReJoyce Green
- Department of Psychology, University of California, Los Angeles
| | | | - Aaron C Lim
- Department of Psychology, University of California, Los Angeles
| | | | - Lara A Ray
- Department of Psychology, University of California, Los Angeles
| |
Collapse
|
11
|
Roche DJO, Trela CJ, Argos M, Jasmine F, Kibriya MG, Ahsan H, King AC. Lack of Association between Opioid-Receptor Genotypes and Smoking Cessation Outcomes in a Randomized, Controlled Naltrexone Trial. Alcohol Alcohol 2019; 54:559-565. [PMID: 31206155 PMCID: PMC7963143 DOI: 10.1093/alcalc/agz046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 05/08/2019] [Accepted: 05/18/2019] [Indexed: 02/01/2023] Open
Abstract
AIMS The present study examined how variation in mu- (OPRM1), kappa- (OPRK), and delta- (OPRD) opioid receptor genes may influence the efficacy of naltrexone in the context of a smoking cessation trial. METHODS The study's primary objective was to examine the association of the Asn40Asp OPRM1 single nucleotide polymorphism (SNP) with naltrexone's effects on smoking quit rate, weight gain, and heavy drinking behavior during a double-blind, randomized clinical trial in 280 adult DSM-IV nicotine-dependent participants. The secondary goal of the study was to examine the relationship of 20 additional SNPs of OPRM1, OPRK, and OPRD with the aforementioned outcomes. RESULTS Results indicated a null association between any opioid-receptor gene SNP and naltrexone's effects on smoking quit rate, weight gain, and heavy drinking behavior in this sample of nicotine dependent participants. CONCLUSIONS In sum, these results do not suggest that genetic variation in opioid-receptors is related to treatment responses to naltrexone in a smoking cessation trial.
Collapse
Affiliation(s)
- Daniel J O Roche
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Constantine J Trela
- Department of Psychological Sciences, University of Missouri, 210 McAlester Hall, Columbia, MO 65211, USA
| | - Maria Argos
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago School of Public Health, Chicago, IL 60612, USA
| | - Farzana Jasmine
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago School of Public Health, Chicago, IL 60612, USA
- Department of Public Health Studies, University of Chicago, Chicago, IL 60637, USA
| | - Muhammad G Kibriya
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago School of Public Health, Chicago, IL 60612, USA
| | - Habibul Ahsan
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago School of Public Health, Chicago, IL 60612, USA
| | - Andrea C King
- Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
12
|
Gómez-Coronado N, Walker AJ, Berk M, Dodd S. Current and Emerging Pharmacotherapies for Cessation of Tobacco Smoking. Pharmacotherapy 2018; 38:235-258. [PMID: 29250815 DOI: 10.1002/phar.2073] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tobacco use disorder is a chronic illness. With its high comorbidity rate, it is a major cause of years of life lost or years lived with disability; however, it is also considered the most preventable cause of death in developed countries. Since the development of nicotine replacement therapy (NRT) in 1978, treatment options have continued to evolve and expand. Despite this, currently available treatments remain insufficient, with less than 25% of smokers remaining abstinent 1 year after treatment. In this article, we review existing and emerging smoking cessation pharmacotherapies, with a special emphasis on the most promising agents that are currently being investigated. A search of the Cochrane Database of Systematic Reviews and the PubMed, Ovid, and ClinicalTrials.gov databases (August 2 to September 1, 2017) was undertaken for articles on smoking cessation pharmacotherapies, applying no language restrictions. More than 40 pharmacotherapies were reviewed including conventional pharmacotherapies-NRT, bupropion, and varenicline (all approved by the U.S. Food and Drug Administration as first-line treatment of smoking cessation)-and novel therapies: cytisine, N-acetylcysteine, cycloserine, memantine, baclofen, topiramate, galantamine, and bromocriptine. Studies of combination NRT and varenicline showed the greatest smoking cessation rates. Clonidine and nortriptyline are second-line treatments used when first-line treatments fail or are contraindicated, or by patient preference. Some novel therapies, especially acetylcholinesterase inhibitors, cytisine, and N-acetylcysteine, display promising results. Because the results of randomized clinical trials were reported using varied end points and outcome measures, direct comparisons between different pharmacotherapies cannot easily be evaluated. Additional high-quality randomized double-blind placebo-controlled trials with long-term follow-up, using validated sustained abstinence measures, are needed to find more effective smoking cessation aids.
Collapse
Affiliation(s)
- Nieves Gómez-Coronado
- Unidad de Gestión Clinica Salud Mental, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Adam J Walker
- IMPACT Strategic Research Centre, Deakin University, Geelong, Victoria, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, Deakin University, Geelong, Victoria, Australia.,Barwon Health, University Hospital Geelong, Geelong, Victoria, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia.,Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Seetal Dodd
- IMPACT Strategic Research Centre, Deakin University, Geelong, Victoria, Australia.,Barwon Health, University Hospital Geelong, Geelong, Victoria, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia.,Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
13
|
Böttcher B, Seeber B, Leyendecker G, Wildt L. Impact of the opioid system on the reproductive axis. Fertil Steril 2017; 108:207-213. [DOI: 10.1016/j.fertnstert.2017.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022]
|
14
|
Norman H, D'Souza MS. Endogenous opioid system: a promising target for future smoking cessation medications. Psychopharmacology (Berl) 2017; 234:1371-1394. [PMID: 28285326 DOI: 10.1007/s00213-017-4582-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 02/24/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Nicotine addiction continues to be a health challenge across the world. Despite several approved medications, smokers continue to relapse. Several human and animal studies have evaluated the role of the endogenous opioid system as a potential target for smoking cessation medications. METHODS In this review, studies that have elucidated the role of the mu (MORs), delta (DORs), and kappa (KORs) opioid receptors in nicotine reward, nicotine withdrawal, and reinstatement of nicotine seeking will be discussed. Additionally, the review will discuss discrepancies in the literature and therapeutic potential of the endogenous opioid system, and suggest studies to address gaps in knowledge with respect to the role of the opioid receptors in nicotine dependence. RESULTS Data available till date suggest that blockade of the MORs and DORs decreased the rewarding effects of nicotine, while activation of the MORs and DORs decreased nicotine withdrawal-induced aversive effects. In contrast, activation of the KORs decreased the rewarding effects of nicotine, while blockade of the KORs decreased nicotine withdrawal-induced aversive effects. Interestingly, blockade of the MORs and KORs attenuated reinstatement of nicotine seeking. In humans, MOR antagonists have shown benefits in select subpopulations of smokers and further investigation is required to realize their full therapeutic potential. CONCLUSION Future work must assess the influence of polymorphisms in opioid receptor-linked genes in nicotine dependence, which will help in both identifying individuals vulnerable to nicotine addiction and the development of opioid-based smoking cessation medications. Overall, the endogenous opioid system continues to be a promising target for future smoking cessation medications.
Collapse
Affiliation(s)
- Haval Norman
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH, 45810, USA
| | - Manoranjan S D'Souza
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH, 45810, USA.
| |
Collapse
|
15
|
Dermody SS, Hendershot CS. A Critical Review of the Effects of Nicotine and Alcohol Coadministration in Human Laboratory Studies. Alcohol Clin Exp Res 2017; 41:473-486. [PMID: 28247555 DOI: 10.1111/acer.13321] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/21/2016] [Indexed: 11/29/2022]
Abstract
Simultaneous use of cigarettes and alcohol is common and may be driven by nicotine increasing alcohol self-administration or vice versa. To better evaluate the causal nature of this relationship, we systematically reviewed human experimental laboratory studies that coadministered nicotine and alcohol with control conditions. Searches of PubMed/MEDLINE and PsycINFO databases and study bibliographies identified 30 studies that met our inclusion criteria. Research methodologies were critically reviewed. Effects of coadministration on drug self-administration and related factors such as craving, subjective response, motivation, and heart rate are reported. Results most strongly supported that alcohol increases nicotine and cigarette self-administration, whereas, depending on the context, nicotine increased, decreased, or had no effect on alcohol self-administration. Craving and subjective drug effects were also impacted by coadministration. Interaction effects of nicotine and alcohol on self-administration and subjective responses were reported infrequently. The effects may be moderated by a number of factors, including dose of administered drug and sex. Recommendations are made for future research, and clinical and policy implications of findings are discussed.
Collapse
Affiliation(s)
- Sarah S Dermody
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Christian S Hendershot
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Department of Psychology, University of Toronto, Toronto, Ontario, Canada.,Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Nicotine enhancement and reinforcer devaluation: Interaction with opioid receptors. Pharmacol Biochem Behav 2016; 150-151:1-7. [DOI: 10.1016/j.pbb.2016.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/10/2016] [Accepted: 08/17/2016] [Indexed: 01/11/2023]
|
17
|
Huskamp HA, Greenfield SF, Stuart EA, Donohue JM, Duckworth K, Kouri EM, Song Z, Chernew ME, Barry CL. Effects of Global Payment and Accountable Care on Tobacco Cessation Service Use: An Observational Study. J Gen Intern Med 2016; 31:1134-40. [PMID: 27177915 PMCID: PMC5023596 DOI: 10.1007/s11606-016-3718-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/04/2016] [Accepted: 04/15/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Tobacco use is the leading cause of preventable death and disability. New payment and delivery system models including global payment and accountable care have the potential to increase use of cost-effective tobacco cessation services. OBJECTIVE To examine how the Alternative Quality Contract (AQC) established in 2009 by Blue Cross Blue Shield of Massachusetts (BCBSMA) has affected tobacco cessation service use. DESIGN We used 2006-2011 BCBSMA claims and enrollment data to compare adults 18-64 years in AQC provider organizations to adults in non-AQC provider organizations. We examined the AQC's effects on all enrollees; a subset at high risk of tobacco-related complications due to certain medical conditions; and behavioral health service users. MAIN MEASURES We examined use of: (1) any cessation treatment (pharmacotherapy or counseling); (2) varenicline or bupropion; (3) nicotine replacement therapies (NRTs); (4) cessation counseling; and (4) combination therapy (pharmacotherapy plus counseling). We also examined duration of pharmacotherapy use and number of counseling visits among users. KEY RESULTS Rates of tobacco cessation treatment use were higher following implementation of the AQC relative to the comparison group overall (2.02 vs. 1.87 %, p < 0.0001), among enrollees at risk for tobacco-related complications (4.97 vs. 4.66 %, p < 0.0001), and among behavioral health service users (3.67 vs. 3.25 %, p < 0.0001). Statistically significant increases were found for use of varenicline or bupropion alone, counseling alone, and combination therapy, but not for NRT use, pharmacotherapy duration, or number of counseling visits among users. CONCLUSIONS In its initial three years, the AQC was associated with increases in use of tobacco cessation services.
Collapse
Affiliation(s)
- Haiden A Huskamp
- Department of Health Care Policy, Harvard Medical School, 180 Longwood Avenue, Boston, MA, 02115, USA.
| | - Shelly F Greenfield
- McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | | | - Julie M Donohue
- University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | | | - Elena M Kouri
- Department of Health Care Policy, Harvard Medical School, 180 Longwood Avenue, Boston, MA, 02115, USA
| | - Zirui Song
- Massachusetts General Hospital, Boston, MA, USA
| | - Michael E Chernew
- Department of Health Care Policy, Harvard Medical School, 180 Longwood Avenue, Boston, MA, 02115, USA
| | - Colleen L Barry
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
18
|
Abstract
Addiction is a major public health problem with few efficacious and safe treatments. The goal of this review is to provide an evidence-based assessment of the therapeutic role of the opioid antagonist naltrexone across the addiction spectrum-substance-based and behavioral. The PubMed database was searched for randomized, placebo-controlled clinical trials that investigated the oral or intramuscular long-acting formulation of naltrexone in substance use disorders or behavioral addictions such as pathological gambling, kleptomania, and trichotillomania. Thirty-nine efficacy studies were retrieved, covering alcohol use disorder (n = 22), opioid use disorder (n = 6), nicotine use disorder (n = 5), stimulant use disorder (n = 2), gambling disorder (n = 2), trichotillomania (n = 1), and kleptomania (n = 1). Despite the very different presentations within and between both addiction categories, the data, as a whole, show consistency in favor of naltrexone's relative efficacy and safety. Given the potential benefit and good tolerability revealed in the studies, the high morbidity associated with addiction, and the dearth of alternate treatments, naltrexone would seem like an underutilized treatment option. Further, naltrexone's seemingly broad anti-addiction efficacy supports a shared role for brain opioid pathways in the pathophysiology of addiction, broadly defined. More studies investigating the efficacy and tolerability of naltrexone and other opioid modulators are warranted. Studies should also further examine the effect of combining psychotherapy with naltrexone, as well as the potential role of naltrexone in treating comorbid addictions.
Collapse
|
19
|
Mooney ME, Schmitz JM, Allen S, Grabowski J, Pentel P, Oliver A, Hatsukami DK. Bupropion and naltrexone for smoking cessation: A double-blind randomized placebo-controlled clinical trial. Clin Pharmacol Ther 2016; 100:344-52. [PMID: 27213949 DOI: 10.1002/cpt.402] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 05/12/2016] [Accepted: 05/16/2016] [Indexed: 11/06/2022]
Abstract
Combination of non-nicotine pharmacotherapies has been underexamined for cigarette smoking cessation. A randomized, double-blind, parallel-group double-dummy study evaluated two medications, bupropion (BUP) and naltrexone (NTX), in treatment-seeking cigarette smokers (N = 121) over a 7-week treatment intervention with 6-month follow-up. Smokers were randomized to either BUP (300 mg/day) + placebo (PBO) or BUP (300 mg/day) + NTX (50 mg/day). The primary outcome was biochemically verified (saliva cotinine, carbon monoxide) 7-day, point-prevalence abstinence. BUP + NTX was associated with significantly higher point-prevalence abstinence rates after 7-weeks of treatment (BUP + NTX, 54.1%; BUP + PBO, 33.3%), P = 0.0210, but not at 6-month follow-up (BUP + NTX, 27.9%; BUP + PBO, 15.0%), P = 0.09. Continuous abstinence rates did not differ, P = 0.0740 (BUP + NTX, 26.2%; BUP + PBO, 13.3%). Those receiving BUP + NTX reported reduced nicotine withdrawal, P = 0.0364. The BUP + NTX combination was associated with elevated rates of some side effects, but with no significant difference in retention between the groups.
Collapse
Affiliation(s)
- M E Mooney
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, USA.
| | - J M Schmitz
- Department of Psychiatry and Behavioral Sciences, University of Texas, Houston, Houston, Texas, USA
| | - S Allen
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, USA
| | - J Grabowski
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, USA
| | - P Pentel
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, USA
| | - A Oliver
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, USA
| | - D K Hatsukami
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
20
|
Roche DJO, Ray LA, Yardley MM, King AC. Current insights into the mechanisms and development of treatments for heavy drinking cigarette smokers. CURRENT ADDICTION REPORTS 2016; 3:125-137. [PMID: 27162709 PMCID: PMC4859339 DOI: 10.1007/s40429-016-0081-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is a strong association between cigarette smoking and alcohol use at the epidemiological, behavioral, and molecular levels, and this co-use creates substantial impediments to smoking cessation among smokers who are also heavy drinkers. Compared with individuals who only smoke, those who both drink and smoke heavily experience more severe health consequences and have greater difficulty in quitting smoking. During smoking abstinence, greater alcohol use is associated with decreased odds of smoking cessation, and smokers are substantially more likely to experience a smoking lapse during drinking episodes. As heavy drinking smokers are less responsive to the currently available pharmacological treatments, this subgroup of high-risk substance users possesses a unique clinical profile and treatment needs. Thus, treatment development for heavy drinking smokers represents a significant and understudied research area within the field of smoking cessation. This review will briefly describe findings from epidemiological, behavioral, and molecular studies illustrating alcohol and tobacco co-use and identify how the behavioral and neurobiological mechanisms underlying the interaction of alcohol and nicotine may inform the development of targeted treatments for this unique population of smokers.
Collapse
Affiliation(s)
- Daniel J O Roche
- University of California, Los Angeles, Department of Psychology, Los Angeles, CA, 90095, USA
| | - Lara A Ray
- University of California, Los Angeles, Department of Psychology, Los Angeles, CA, 90095, USA; University of California, Los Angeles, Department of Psychiatry and Biobehavioral Sciences, Los Angeles, CA, 90095, USA
| | - Megan M Yardley
- University of California, Los Angeles, Department of Psychology, Los Angeles, CA, 90095, USA
| | - Andrea C King
- University of Chicago, Department of Psychiatry and Behavioral Neuroscience, Chicago, IL 60637, USA
| |
Collapse
|
21
|
McLaughlin PJ, Zagon IS. Duration of opioid receptor blockade determines biotherapeutic response. Biochem Pharmacol 2015; 97:236-46. [DOI: 10.1016/j.bcp.2015.06.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/17/2015] [Indexed: 12/20/2022]
|
22
|
Roche DJO, Bujarski S, Hartwell E, Green R, Ray LA. Combined varenicline and naltrexone treatment reduces smoking topography intensity in heavy-drinking smokers. Pharmacol Biochem Behav 2015; 134:92-8. [PMID: 25933795 DOI: 10.1016/j.pbb.2015.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/24/2015] [Accepted: 04/22/2015] [Indexed: 11/15/2022]
Abstract
Heavy drinking smokers constitute a distinct sub-population of smokers for whom traditional smoking cessation therapies may not be effective. Recent evidence suggested that combined varenicline (VAR) and naltrexone (NTX) therapy may be more efficacious than either monotherapy alone in reducing smoking and drinking-related behavior in this population. The manner in which individuals smoke a cigarette (i.e., smoking topography) may be predictive of smoking cessation outcomes, yet the effects of smoking pharmacotherapies on puffing behavior have not been thoroughly examined. Therefore, the current double-blind medication study examined the effects of VAR alone (1mg BID), low dose NTX alone (25mg QD), the combination of VAR+NTX, and placebo on smoking topography measures in heavy drinking, non-treatment seeking daily smokers (n=120). After a 9-day titration period, participants completed a laboratory session in which they smoked their first cigarette of the day using a smoking topography device following 12h of nicotine abstinence and consumption of an alcoholic beverage (BrAC=0.06g/dl). The primary measures were puff count, volume, duration, and velocity and inter-puff interval (IPI). Independent of medication group, puff velocity and IPI increased, while puff volume and duration decreased, over the course of the cigarette. The active medication groups, vs. the placebo group, had significantly blunted puff duration and velocity slopes over the course of the cigarette, and this effect was particularly evident in the VAR+NTX group. Additionally, the VAR+NTX group demonstrated lower average IPI than the monotherapy groups and lower average puff volume than all other groups. These results suggest that smoking pharmacotherapies, particularly the combination of VAR+NTX, alter smoking topography in heavy drinking smokers, producing a pattern of less intense puffing behavior. As smoking topography has been predictive of the ability to quit smoking, future studies should examine how smoking pharmacotherapies' effects on puffing behavior relate to smoking cessation outcomes.
Collapse
Affiliation(s)
- Daniel J O Roche
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Spencer Bujarski
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Emily Hartwell
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - ReJoyce Green
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA 90095, United States; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
23
|
Abstract
There is a high prevalence of comorbid tobacco use and alcohol use disorder (AUD), affecting more than 6 million people in the US. Globally, tobacco and alcohol use rank fourth and fifth, respectively, for disability-adjusted life-years lost. Levels of alcohol use are higher in smokers than nonsmokers, and the prevalence of smoking is higher in heavy drinkers compared with nondrinkers. This relationship is driven by many different factors, including genetics, neurobiological mechanisms, conditioning processes, and psychosocial influences. Although this unique population tends to experience more negative health consequences, more severe AUD, and poorer response to treatment than those with either AUD or tobacco use disorder alone, there are currently no available treatment protocols tailored to this comorbid condition. In this review, we provide a comprehensive review of ongoing clinical research into smoking cessation options for heavy-drinking smokers (HDS) through an evaluation of the effect of promising novel pharmacotherapies as well as combination therapies, including varenicline, naltrexone, the combination of varenicline and naltrexone, and the combination of naltrexone and nicotine replacement therapy (NRT). These treatments are considered in light of the standard of care for smoking cessation, and seek to improve upon the available guidelines for this sizeable subgroup of smokers, namely those smokers who drink heavily.
Collapse
Affiliation(s)
- Megan M Yardley
- Department of Psychology, University of California, Los Angeles, 1285 Franz Hall, Box 951563, Los Angeles, CA, 90095-1563, USA
| | - Michael M Mirbaba
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, 1285 Franz Hall, Box 951563, Los Angeles, CA, 90095-1563, USA.
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Asvat Y, Cao D, Africk JJ, Matthews A, King A. Feasibility and effectiveness of a community-based smoking cessation intervention in a racially diverse, urban smoker cohort. Am J Public Health 2014; 104 Suppl 4:S620-7. [PMID: 25100429 DOI: 10.2105/ajph.2014.302097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES We evaluated the feasibility, acceptability, and effectiveness, in addition to the effects of a psychoeducation-based orientation on smoking cessation knowledge for Courage to Quit (CTQ), an evidence-based smoking cessation intervention disseminated to racially diverse, urban community sites in Chicago, Illinois. METHODS Smokers (n = 1494; 55% African American) enrolled in 6-session full (n = 945) or 3-session short (n = 549) versions of CTQ in 2008 to 2012. RESULTS Orientation improved knowledge of efficacious and nonefficacious treatments. Acceptability was outstanding: more than 90% of participants would recommend CTQ. Feasibility was good: completion rates were 53% in the full and 75% in the short programs. Intent-to-treat quit rates were 19% in the full and 17% in the short programs (completer quit rates were 36% and 22%, respectively). Among completers, smoking cessation medication use was associated with higher quit rates. There were no racial disparities: African Americans and Whites showed similar completion and quit rates. Predictors of successful quitting were higher readiness to quit and smoking cessation medication use. CONCLUSIONS CTQ is moderately successful in the short term as delivered in community-based settings for urban-dwelling, largely minority smokers. Further evaluation of longer-term outcomes and cost effectiveness is warranted.
Collapse
Affiliation(s)
- Yasmin Asvat
- Yasmin Asvat is with the Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL. Dingcai Cao is with the Department of Ophthalmology, University of Illinois, Chicago. Joel J. Africk is with the Respiratory Health Association of Metropolitan Chicago. Alicia Matthews is with the College of Nursing, University of Illinois, Chicago. Andrea King is with the Department of Psychiatry & Behavioral Neuroscience, University of Chicago, Chicago, IL
| | | | | | | | | |
Collapse
|
25
|
Ray LA, Courtney KE, Ghahremani DG, Miotto K, Brody A, London ED. Varenicline, low dose naltrexone, and their combination for heavy-drinking smokers: human laboratory findings. Psychopharmacology (Berl) 2014; 231:3843-53. [PMID: 24733235 PMCID: PMC4161630 DOI: 10.1007/s00213-014-3519-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 02/26/2014] [Indexed: 01/20/2023]
Abstract
RATIONALE Heavy-drinking smokers constitute a sizeable and hard-to-treat subgroup of smokers, for whom tailored smoking cessation therapies are not yet available. OBJECTIVES The present study used a double-blind, randomized, 2 × 2 medication design, testing varenicline alone (VAR; 1 mg twice daily), low dose naltrexone alone (L-NTX; 25 mg once daily), varenicline plus naltrexone, and placebo for effects on cigarette craving and subjective response to alcohol and cigarettes in a sample (n = 130) of heavy-drinking daily smokers (≥10 cigarettes/day). METHODS All participants were tested after a 9-day titration period designed to reach a steady state on the target medication. Testing was completed at 12 h of nicotine abstinence, after consuming a standard dose of alcohol (target breath alcohol concentration = 0.06 g/dl) and after smoking the first cigarette of the day. RESULTS The combination of VAR + L-NTX was superior to placebo, and at times superior to monotherapy, in attenuating cigarette craving, cigarette and alcohol "high," and in reducing ad-lib consumption of both cigarettes and alcohol during the 9-day medication titration period. CONCLUSIONS These preliminary findings indicate that clinical studies of the combination of VAR + L-NTX for heavy drinkers trying to quit smoking are warranted and may ultimately improve clinical care for this sizeable and treatment-resistant subgroup of smokers.
Collapse
Affiliation(s)
- Lara A Ray
- Department of Psychology, University of California, 1285 Franz Hall, Box 951563, Los Angeles, CA, 90095-1563, USA,
| | | | | | | | | | | |
Collapse
|
26
|
Lin SK. Pharmacological means of reducing human drug dependence: a selective and narrative review of the clinical literature. Br J Clin Pharmacol 2014; 77:242-52. [PMID: 23701272 DOI: 10.1111/bcp.12163] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 04/28/2013] [Indexed: 12/20/2022] Open
Abstract
Substance abuse or addictive disorder is a global problem. A greater understanding of the associated changes in brain pathophysiology supports the notion that pharmacological treatments are part of the necessary treatment options. Craving is a core symptom of addictive disorder. It refers to a strong desire to use drugs again either to re-experience positive effects or to diminish negative experiences. Currently there are a number of medicines that are effective in the treatment of addictive disorders. These medications can either be for substitution (same pharmacological effect as the abused substance) or anticraving (decrease the craving of the abused substance). In this MEDLNE based review, specific compounds (naltrexone, acamprosate, topiramate, disulfiram, baclofen, N-acetylcysteine and bupropion) were selected that are known to diminish desire to use (anticraving effect) and that have been trialled for a number of different substance addictive disorders. Their therapeutic potential in clinical practice is discussed in light of their efficacy.
Collapse
Affiliation(s)
- Shih-Ku Lin
- Department of Psychiatry, Taipei City Hospital and Psychiatric Center, Taipei, Taiwan; Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
27
|
Abstract
INTRODUCTION Smokers' treatment expectancies may influence their choice of a particular medication as well as their medication experience. AIMS This study examined the role of smokers' treatment expectancies to their smoking cessation outcomes in a completed, randomized, placebo-controlled trial of naltrexone for smoking cessation, controlling for perceptions of treatment assignment. METHODS Treatment seeking cigarette smokers (N = 315) were randomized to receive either naltrexone (50 mg) or placebo in combination with nicotine patch and behavioral counseling. Expectancies for naltrexone as a smoking cessation aid were assessed at baseline and 4 weeks after the quit date. RESULTS More positive baseline medication expectancies predicted higher quit rates at one month in the naltrexone (OR =1.45, p =.04) group but were associated with lower quit rates in the placebo group (OR =.66, p =.03). Maintaining and/or increasing positive medication expectancies in the first month of treatment was associated with better pill adherence during this interval in the naltrexone group (ps <.05). Positive baseline medication expectancies were also associated with the perception of having received naltrexone over placebo among all participants. CONCLUSIONS Positive medication expectancies in smokers may contribute to better treatment response. Assessing treatment expectancies and attempting to maintain or improve them may be important for the delivery, evaluation, and targeting of smoking cessation treatments.
Collapse
|
28
|
Ray LA, Courtney KE, Ghahremani DG, Miotto K, Brody A, London ED. Varenicline, naltrexone, and their combination for heavy-drinking smokers: preliminary neuroimaging findings. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2014; 41:35-44. [PMID: 24949564 DOI: 10.3109/00952990.2014.927881] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
RATIONALE Heavy drinking smokers constitute a sizeable and hard-to-treat subgroup of smokers, for whom tailored smoking cessation therapies are not yet available. OBJECTIVE The present study used a double-blind, randomized, 2 × 2 medication design, testing varenicline alone (VAR; 1 mg twice daily), naltrexone alone (NTX; 25 mg once daily), varenicline plus naltrexone, and placebo for effects on neural activation to cigarette cues in a sample (n = 40) of heavy drinking daily smokers (≥10 cigarettes/day). METHODS All participants were tested after a 10-12-day titration period designed to reach steady state on the target medication. Participants underwent functional neuroimaging (fMRI) for examination of brain responses to visual smoking-related (vs. neutral) cues. RESULTS Region of interest (ROI) analyses of brain responses to Cigarette vs. Neutral Cues indicated that the combination of VAR + NTX was associated with reduced activation of the bilateral anterior cingulate cortex as compared to placebo and to NTX alone. Exploratory whole-brain analyses also indicated significant differences in brain activation during cigarette cues in the active medications versus placebo condition. All medications suppressed left nucleus accumbens activation relative to placebo, suggesting the possibility that both medications, either alone or in combination, reduce neural signals associated with appetitive behavior. CONCLUSIONS Although preliminary, these neuroimaging findings indicate that clinical studies of the combination of VAR + NTX for heavy drinkers trying to quit smoking may be warranted.
Collapse
|
29
|
Matthews AK, McConnell EA, Li CC, Vargas MC, King A. Design of a comparative effectiveness evaluation of a culturally tailored versus standard community-based smoking cessation treatment program for LGBT smokers. BMC Psychol 2014; 2:12. [PMID: 25566383 PMCID: PMC4269994 DOI: 10.1186/2050-7283-2-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 05/15/2014] [Indexed: 11/30/2022] Open
Abstract
Background Smoking prevalence rates among the lesbian, gay, bisexual, and transgender (LGBT) population are significantly higher than the general population. However, there is limited research on smoking cessation treatments in this group, particularly on culturally targeted interventions. Moreover, there are few interventions that address culturally specific psychosocial variables (e.g., minority stress) that may influence outcomes. This paper describes the protocol for a comparative effectiveness trial testing an evidence-based smoking cessation program, Courage to Quit, against a culturally tailored version for LGBT smokers, and examines the role of culturally specific psychosocial variables on cessation outcomes. Methods/Design To examine the effectiveness of a culturally targeted versus standard smoking cessation intervention, the study utilizes a 2-arm block, randomized, control trial (RCT) design. Adult LGBT participants (n = 400) are randomized to one of the two programs each consisting of a six-session group program delivered in a community center and an eight week supply of the transdermal nicotine patch. Four individualized telephone counseling sessions occur at weeks 2, 5, 7, and 9, at times of greatest risk for relapse. Study outcome measures are collected at baseline, and 1, 3, 6, and 12 months post quit date. Primary outcomes are expired air carbon monoxide verified 7-day point-prevalence quit rates at each measurement period. Secondary outcomes assess changes in cravings, withdrawal symptoms, smoking cessation self-efficacy, and treatment adherence. Additionally, study staff examines the role of culturally specific psychosocial variables on cessation outcomes using path analysis. Discussion Determining the efficacy of culturally specific versus standard evidence based approaches to smoking cessation is a critical issue facing the field today. This study provides a model for the development and implementation of a culturally tailored smoking cessation intervention for LGBT participants and addresses a gap in the field by examining the role of culturally psychosocial variables associated with cessation outcomes. Trial registration U.S. National Institutes of Health Clinical Trials NCT01633567 Registered 30 May 2012.
Collapse
Affiliation(s)
- Alicia K Matthews
- University of Illinois at Chicago (UIC), Chicago, IL USA ; College of Nursing, University of Illinois at Chicago, 845 S. Damen Avenue, Chicago, IL 60612 USA
| | | | | | | | | |
Collapse
|
30
|
Allen SS, Allen AM, Tosun N, Lunos S, al'Absi M, Hatsukami D. Smoking- and menstrual-related symptomatology during short-term smoking abstinence by menstrual phase and depressive symptoms. Addict Behav 2014; 39:901-6. [PMID: 24594903 DOI: 10.1016/j.addbeh.2014.01.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/16/2014] [Accepted: 01/29/2014] [Indexed: 12/01/2022]
Abstract
Menstrual phase and depressive symptoms are known to minimize quit attempts in women. Therefore, the influence of these factors on smoking- and menstrual-related symptomatology during acute smoking cessation was investigated in a controlled cross-over lab-study. Participants (n=147) completed two six-day testing weeks during their menstrual cycle with testing order randomly assigned (follicular vs. luteal). The testing week consisted of two days of ad libitum smoking followed by four days of biochemically verified smoking abstinence. Daily symptomatology measures were collected. Out of the 11 total symptoms investigated, six were significantly associated with menstrual phase and nine were significantly associated with level of depressive symptoms. Two significant interactions were noted indicating that there may be a stronger association between depressive symptoms with negative affect and premenstrual pain during the follicular phase compared to the luteal phase. Overall, these observations suggest that during acute smoking abstinence in premenopausal smokers, there is an association between depressive symptoms and symptomatology whereas menstrual phase appears to have less of an effect. Further study is needed to determine the effect of these observations on smoking cessation outcomes, as well as to define the mechanism of menstrual phase and depressive symptoms on smoking-related symptomatology.
Collapse
Affiliation(s)
- Sharon S Allen
- Department of Family Medicine & Community Health Medical School, University of Minnesota, 420 Delaware Street SE, Room A682, Minneapolis, MN 55455, United States
| | - Alicia M Allen
- Department of Family Medicine & Community Health, Medical School, University of Minnesota, 717 Delaware Street SE, Room 422, Minneapolis, MN 55414, United States
| | - Nicole Tosun
- Department of Family Medicine & Community Health, Medical School, University of Minnesota, 717 Delaware Street SE, Room 261, Minneapolis, MN 55414, United States
| | - Scott Lunos
- Biostatistical Design and Analysis Center, Clinical and Translational Science Institute, University of Minnesota, 717 Delaware Street SE, Suite 132, Minneapolis, MN 55414, United States
| | - Mustafa al'Absi
- Department of Behavioral Sciences, Medical School, University of Minnesota, Duluth 1035 University Drive, 236 SMed, D601A, Duluth, MN 55812, United States
| | - Dorothy Hatsukami
- Department of Psychiatry, Medical School, University of Minnesota, 717 Delaware Street SE, Room 256, Minneapolis, MN 55414, United States
| |
Collapse
|
31
|
Elrashidi MY, Ebbert JO. Emerging drugs for the treatment of tobacco dependence: 2014 update. Expert Opin Emerg Drugs 2014; 19:243-60. [PMID: 24654737 DOI: 10.1517/14728214.2014.899580] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Tobacco dependence remains a global epidemic and the largest preventable cause of morbidity and mortality around the world. Smoking cessation has benefits at all ages but remains challenging for several reasons, among which are the complexities of nicotine addiction and limitations of available pharmacotherapies. AREAS COVERED This review summarizes current and emerging pharmacotherapies for the treatment of tobacco dependence, including first- and second-line recommended agents. Medications with alternative primary indications that have been investigated as potential treatments for tobacco dependence are also discussed. Articles reviewed were obtained through searches of PubMed, Ovid MEDLINE, ClinicalTrials.gov and the Pharmaprojects database. EXPERT OPINION Current evidence suggests that the two most effective pharmacotherapies to treat tobacco dependence are varenicline and combination nicotine replacement therapy. Alternative agents investigated demonstrate mixed rates of success in achieving long-term abstinence from smoking. No single pharmacotherapy will serve as a universally successful treatment given the complex underpinnings of tobacco dependence and individuality of smokers. The ultimate goal of tobacco research with respect to pharmacotherapeutic development continues to be providing clinicians with an armamentarium of drugs to choose from allowing for tailoring of treatment for smokers.
Collapse
|
32
|
David SP, Chu IM, Lancaster T, Stead LF, Evins AE, Prochaska JJ. Systematic review and meta-analysis of opioid antagonists for smoking cessation. BMJ Open 2014; 4:e004393. [PMID: 24633528 PMCID: PMC3963070 DOI: 10.1136/bmjopen-2013-004393] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/29/2014] [Accepted: 02/20/2014] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES This meta-analysis sought to evaluate the efficacy of opioid antagonists in promoting long-term smoking cessation. Post-treatment abstinence was examined as a secondary outcome and effects on withdrawal symptoms, craving and reduced consumption were also explored. DESIGN The search strategy for this meta-analysis included clinical trials (published and unpublished data) in the Cochrane Tobacco Addiction Group Specialized Register and MEDLINE. PARTICIPANTS Adult smokers. INTERVENTIONS We included randomised trials comparing opioid antagonists to placebo or an alternative therapy for smoking cessation and reported data on abstinence for a minimum of 6 months. PRIMARY AND SECONDARY OUTCOME MEASURES Outcomes included smoking abstinence at long-term follow-up (primary); abstinence at end of treatment (secondary); and effects on withdrawal, craving and smoking consumption (exploratory). RESULTS 8 trials with a total of 1213 participants were included. Half the trials examined the benefit of adding naltrexone versus placebo to nicotine replacement therapy (NRT). There was no significant difference between naltrexone and placebo alone (relative risk (RR) 1.00; 95% CI 0.66 to 1.51) or as an adjunct to NRT (RR 0.95; 95% CI 0.70 to 1.30), with an overall pooled estimate of RR 0.97; 95% CI 0.76 to 1.24. Findings for naltrexone effects on withdrawal, craving and reduced smoking were equivocal. CONCLUSIONS The findings indicate no beneficial effect of naltrexone alone or as an adjunct to NRT on short-term or long-term smoking abstinence. While further trials may narrow the confidence limits, they are unlikely to appreciably alter the conclusion.
Collapse
Affiliation(s)
- Sean P David
- Center for Education in Family & Community Medicine, Stanford University, Stanford, California, USA
- Stanford General Medical Disciplines, Stanford University, Stanford, California, USA
| | - Isabella M Chu
- Stanford General Medical Disciplines, Stanford University, Stanford, California, USA
| | - Tim Lancaster
- Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Lindsay F Stead
- Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - A Eden Evins
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Judith J Prochaska
- Stanford Prevention Research Center, Stanford University, Stanford, California, USA
| |
Collapse
|
33
|
King A, Cao D, Zhang L, Rueger SY. Effects of the opioid receptor antagonist naltrexone on smoking and related behaviors in smokers preparing to quit: a randomized controlled trial. Addiction 2013; 108:1836-44. [PMID: 23714324 PMCID: PMC3775903 DOI: 10.1111/add.12261] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 08/10/2012] [Accepted: 05/20/2013] [Indexed: 01/16/2023]
Abstract
AIMS To determine if naltrexone affects smoking behaviors in smokers preparing to quit, and whether or not such pre-quit responses predict post-quit date outcomes. DESIGN Double-blind, placebo-controlled, randomized study. The current study focused on smoking-related outcomes in the pre-quit phase, which was 1 week prior to the quit date, and these findings were linked with reductions in the same outcomes demonstrated in the post-quit phase published previously for this randomized controlled trial (RCT) in mediation analyses. SETTING Community sample of adult smokers desiring to quit in Chicago, Illinois, USA. PARTICIPANTS Participants were 315 smokers randomized to naltrexone (n = 161; mean age = 42.58 years; 60% Caucasian) or placebo (n = 154; mean age = 41.32 years; 55% Caucasian). MEASUREMENTS The difference from baseline in the number of cigarettes smoked during the pre-quit phase interval was the primary outcome. Secondary pre-quit outcomes were assessed using Likert scales of subjective responses and consumption of cigarettes, alcohol and food. Number of cigarettes smoked, alcoholic drinks consumed and the Brief Questionnaire of Smoking Urges were assessed in the post-quit phase. FINDINGS Relative to placebo, naltrexone decreased the number of cigarettes smoked (-4.21 versus -2.93, P < 0.05), smoking urge (P = 0.02) and number of alcoholic drinks consumed (P = 0.04). Exploratory mediation analyses linking outcomes of the pre-quit and post-quit phases found that naltrexone's effects on reducing smoking urge, cigarettes smoked and alcoholic drinks consumed in the pre-quit phase demonstrated full mediation of their respective effects during the post-quit phase. CONCLUSIONS Naltrexone taken in the week before a quit attempt reduces cigarette consumption, urges to smoke and alcohol consumption relative to placebo. The size of the effect mediates statistically the size of similar effects after the quit date.
Collapse
Affiliation(s)
- Andrea King
- University of Chicago, Department of Psychiatry & Behavioral Neuroscience. Chicago, IL
| | - Dingcai Cao
- University of Illinois at Chicago, Department of Ophthalmology & Visual Sciences, Chicago, IL
| | - Lingjiao Zhang
- University of Chicago, Department of Psychiatry & Behavioral Neuroscience. Chicago, IL
| | - Sandra Yu Rueger
- University of Chicago, Department of Psychiatry & Behavioral Neuroscience. Chicago, IL,Wheaton College, Department of Psychology, Wheaton, IL
| |
Collapse
|
34
|
Raffa RB, Baron S, Bhandal JS, Brown T, Song K, Tallarida CS, Rawls SM. Opioid receptor types involved in the development of nicotine physical dependence in an invertebrate (Planaria) model. Pharmacol Biochem Behav 2013; 112:9-14. [PMID: 24084318 DOI: 10.1016/j.pbb.2013.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 09/18/2013] [Accepted: 09/21/2013] [Indexed: 11/18/2022]
Abstract
Recent data suggest that opioid receptors are involved in the development of nicotine physical dependence in mammals. Evidence in support of a similar involvement in an invertebrate (Planaria) is presented using the selective opioid receptor antagonist naloxone, and the more receptor subtype-selective antagonists CTAP (D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2) (μ, MOR), naltrindole (δ, DOR), and nor-BNI (norbinaltorphimine) (κ, KOR). Induction of physical dependence was achieved by 60-min pre-exposure of planarians to nicotine and was quantified by abstinence-induced withdrawal (reduction in spontaneous locomotor activity). Known MOR and DOR subtype-selective opioid receptor antagonists attenuated the withdrawal, as did the non-selective antagonist naloxone, but a KOR subtype-selective antagonist did not. An involvement of MOR and DOR, but not KOR, in the development of nicotine physical dependence or in abstinence-induced withdrawal was thus demonstrated in a sensitive and facile invertebrate model.
Collapse
Affiliation(s)
- Robert B Raffa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
van der Meer RM, Willemsen MC, Smit F, Cuijpers P. Smoking cessation interventions for smokers with current or past depression. Cochrane Database Syst Rev 2013:CD006102. [PMID: 23963776 DOI: 10.1002/14651858.cd006102.pub2] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Individuals with current or past depression are often smokers who are more nicotine dependent, more likely to suffer from negative mood changes after nicotine withdrawal, and more likely to relapse to smoking after quitting than the general population, which contributes to their higher morbidity and mortality from smoking-related illnesses. It remains unclear what interventions can help them to quit smoking. OBJECTIVES To evaluate the effectiveness of smoking cessation interventions, with and without specific mood management components, in smokers with current or past depression. SEARCH METHODS In April 2013, we searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE, PsycINFO, other reviews, and asked experts for information on trials. SELECTION CRITERIA Criteria for including studies in this review were that they had to be randomised controlled trials (RCTs) comparing smoking cessation interventions in adult smokers with current or past depression. Depression was defined as major depression or depressive symptoms. We included studies where subgroups of participants with depression were identified, either pre-stated or post hoc. The outcome was abstinence from smoking after six months or longer follow-up. We preferred prolonged or continuous abstinence and biochemically validated abstinence where available. DATA COLLECTION AND ANALYSIS When possible, we estimated pooled risk ratios (RRs) with the Mantel-Haenszel method (fixed-effect model). We also performed subgroup analyses, by length of follow-up, depression measurement, depression group in study, antidepressant use, published or unpublished data, format of intervention, level of behavioural support, additional pharmacotherapy, type of antidepressant medication, and additional nicotine replacement therapy (NRT). MAIN RESULTS Forty-nine RCTs were included of which 33 trials investigated smoking cessation interventions with specific mood management components for depression. In smokers with current depression, meta-analysis showed a significant positive effect for adding psychosocial mood management to a standard smoking cessation intervention when compared with standard smoking cessation intervention alone (11 trials, N = 1844, RR 1.47, 95% CI 1.13 to 1.92). In smokers with past depression we found a similar effect (13 trials, N = 1496, RR 1.41, 95% CI 1.13 to 1.77). Meta-analysis resulted in a positive effect, although not significant, for adding bupropion compared with placebo in smokers with current depression (5 trials, N = 410, RR 1.37, 95% CI 0.83 to 2.27). There were not enough trial data to evaluate the effectiveness of fluoxetine and paroxetine for smokers with current depression. Bupropion (4 trials, N = 404, RR 2.04, 95% CI 1.31 to 3.18) might significantly increase long-term cessation among smokers with past depression when compared with placebo, but the evidence for bupropion is relatively weak due to the small number of studies and the post hoc subgroups for all the studies. There were not enough trial data to evaluate the effectiveness of fluoxetine, nortriptyline, paroxetine, selegiline, and sertraline in smokers with past depression.Twenty-three of the 49 trials investigated smoking cessation interventions without specific components for depression. There was heterogeneity between the trials which compared psychosocial interventions with standard smoking cessation counselling for both smokers with current and past depression. Therefore, we did not estimate a pooled effect. One trial compared nicotine replacement therapy (NRT) versus placebo in smokers with current depression and found a positive, although not significant, effect (N = 196, RR 2.64, 95% CI 0.93 to 7.45). Meta-analysis also found a positive, although not significant, effect for NRT versus placebo in smokers with past depression (3 trials, N = 432, RR 1.17, 95% CI 0.85 to 1.60). Three trials compared other pharmacotherapy versus placebo and six trials compared other interventions in smokers with current or past depression. Due to heterogeneity between the interventions of the included trials we did not estimate pooled effects. AUTHORS' CONCLUSIONS Evidence suggests that adding a psychosocial mood management component to a standard smoking cessation intervention increases long-term cessation rates in smokers with both current and past depression when compared with the standard intervention alone. Pooled results from four trials suggest that use of bupropion may increase long-term cessation in smokers with past depression. There was no evidence found for the use of bupropion in smokers with current depression. There was not enough evidence to evaluate the effectiveness of the other antidepressants in smokers with current or past depression. There was also not enough evidence to evaluate the group of trials that investigated interventions without specific mood management components for depression, including NRT and psychosocial interventions.
Collapse
|
36
|
Abstract
BACKGROUND The reinforcing properties of nicotine may be mediated through release of various neurotransmitters both centrally and systemically. People who smoke report positive effects such as pleasure, arousal, and relaxation as well as relief of negative affect, tension, and anxiety. Opioid (narcotic) antagonists are of particular interest to investigators as potential agents to attenuate the rewarding effects of cigarette smoking. OBJECTIVES To evaluate the efficacy of opioid antagonists in promoting long-term smoking cessation. The drugs include naloxone and the longer-acting opioid antagonist naltrexone. SEARCH METHODS We searched the Cochrane Tobacco Addiction Group Specialised Register for trials of naloxone, naltrexone and other opioid antagonists and conducted an additional search of MEDLINE using 'Narcotic antagonists' and smoking terms in April 2013. We also contacted investigators, when possible, for information on unpublished studies. SELECTION CRITERIA We considered randomised controlled trials comparing opioid antagonists to placebo or an alternative therapeutic control for smoking cessation. We included in the meta-analysis only those trials which reported data on abstinence for a minimum of six months. We also reviewed, for descriptive purposes, results from short-term laboratory-based studies of opioid antagonists designed to evaluate psycho-biological mediating variables associated with nicotine dependence. DATA COLLECTION AND ANALYSIS We extracted data in duplicate on the study population, the nature of the drug therapy, the outcome measures, method of randomisation, and completeness of follow-up. The main outcome measure was abstinence from smoking after at least six months follow-up in patients smoking at baseline. Abstinence at end of treatment was a secondary outcome. We extracted cotinine- or carbon monoxide-verified abstinence where available. Where appropriate, we performed meta-analysis, pooling risk ratios using a Mantel-Haenszel fixed-effect model. MAIN RESULTS Eight trials of naltrexone met inclusion criteria for meta-analysis of long-term cessation. One trial used a factorial design so five trials compared naltrexone versus placebo and four trials compared naltrexone plus nicotine replacement therapy (NRT) versus placebo plus NRT. Results from 250 participants in one long-term trial remain unpublished. No significant difference was detected between naltrexone and placebo (risk ratio (RR) 1.00; 95% confidence interval (CI) 0.66 to 1.51, 445 participants), or between naltrexone and placebo as an adjunct to NRT (RR 0.95; 95% CI 0.70 to 1.30, 768 participants). The estimate was similar when all eight trials were pooled (RR 0.97; 95% CI 0.76 to 1.24, 1213 participants). In a secondary analysis of abstinence at end of treatment, there was also no evidence of any early treatment effect, (RR 1.03; 95% CI 0.88 to 1.22, 1213 participants). No trials of naloxone or buprenorphine reported abstinence outcomes. AUTHORS' CONCLUSIONS Based on data from eight trials and over 1200 individuals, there was no evidence of an effect of naltrexone alone or as an adjunct to NRT on long-term smoking abstinence, with a point estimate strongly suggesting no effect and confidence intervals that make a clinically important effect of treatment unlikely. Although further trials might narrow the confidence intervals they are unlikely to be a good use of resources.
Collapse
Affiliation(s)
- Sean P David
- Center for Education in Family & Community Medicine, Stanford University, Stanford, California, USA.
| | | | | | | | | |
Collapse
|
37
|
Abstract
BACKGROUND The opioid antagonist naltrexone has shown promise to reduce weight gain during active treatment, but longer-term studies have not been conducted. The goal was to examine effects of naltrexone on weight gain over long-term follow-up in men and women who quit smoking. METHODS Weight was examined at baseline and 6- and 12-month follow-up in the two largest randomized, double-blind, placebo-controlled trials of naltrexone in nicotine dependence. For 6-12 weeks after the quit date, participants were randomly assigned to receive naltrexone or placebo. Behavioral counseling and open-label nicotine patch were also included for the first 4-6 weeks. Of the 700 participants in the combined intent-to-treat sample, there were 159 (77 women) biochemically verified abstinent smokers at 6 months, and 115 (57 women) of them remained abstinent at 12 months. Changes in weight (in kilograms or in percentage) and body mass index from baseline to the follow-ups were assessed for these participants. RESULTS Weight gain was significantly lower for women treated with naltrexone compared with placebo (6 months, 3.3 vs. 5.5 kg; 12 months, 5.9 vs. 7.4 kg, respectively). Increases in body mass index and percentage body weight gain were also significantly lower in women treated with naltrexone versus placebo. These effects were not observed in men. CONCLUSION The results provide evidence for naltrexone as the first pharmacotherapy to reduce postsmoking cessation weight gain among women.
Collapse
|
38
|
Hitsman B, Papandonatos GD, McChargue DE, DeMott A, Herrera MJ, Spring B, Borrelli B, Niaura R. Past major depression and smoking cessation outcome: a systematic review and meta-analysis update. Addiction 2013; 108:294-306. [PMID: 23072580 PMCID: PMC3593055 DOI: 10.1111/add.12009] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 07/03/2012] [Accepted: 10/04/2012] [Indexed: 11/29/2022]
Abstract
AIMS To update our prior meta-analysis that showed past major depression (MD+) to be unrelated to smoking cessation outcome. METHODS Eligible trials included 14 from our original review and 28 identified through an updated systematic review (2000-2009). We coded for assessment of past MD, exclusion for recent MD episode (MDE; ≤6 months versus no exclusion), duration/modality of cognitive behavioral treatment (CBT; face-to-face versus self-help) and other factors. To minimize influence of experimental treatments that may selectively benefit MD+ smokers we analyzed placebo/lowest intensity control arms only. Study-specific ORs for the effect of past MD on short-term (≤3 months) and long-term (≥6 months) abstinence were estimated and combined using random effects. Two-way interaction models of past MD with study methodology and treatment factors were used to evaluate hypothesized moderators of the past MD-abstinence association. RESULTS MD+ smokers had 17% lower odds of short-term abstinence (n = 35, OR = 0.83, 95% CI = 0.72-0.95, P = 0.009) and 19% lower odds of long-term abstinence (n = 38, OR = 0.81, 95% CI = 0.67-0.97, P = 0.023) than MD- smokers after excluding the sole study of varenicline because of its antidepressant properties. The association between past MD and abstinence was affected by methodological (recent MDE exclusion, type of MD assessment) and treatment (CBT modality) factors. CONCLUSIONS Past major depression has a modest adverse effect on abstinence during and after smoking cessation treatment. An increased focus on the identification of effective treatments or treatment adaptations that eliminate this disparity in smoking cessation for MD+ smokers is needed.
Collapse
Affiliation(s)
- Brian Hitsman
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Matthews AK, Conrad M, Kuhns L, Vargas M, King AC. Project Exhale: preliminary evaluation of a tailored smoking cessation treatment for HIV-positive African American smokers. AIDS Patient Care STDS 2013; 27:22-32. [PMID: 23305259 DOI: 10.1089/apc.2012.0253] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study examined the feasibility, acceptability, and outcomes of a culturally tailored smoking cessation intervention for HIV-positive African American male smokers. Eligible smokers were enrolled in a seven-session group-based treatment combined with nicotine patch. The mean age of participants was M=46 years. The majority were daily smokers (71%), smoked a mentholated brand (80%), and averaged 8.6 (standard deviation [SD]=8.1) cigarettes per day. Baseline nicotine dependency scores (M=5.8) indicated a moderate to high degree of physical dependence. Of the 31 participants enrolled, the majority completed treatment (≥3 sessions; 68%), 1-month follow-up (74%), and 3-month follow-up (87%) interviews. Program acceptability scores were strong. However, adherence to the patch was low, with 39% reporting daily patch use. The majority of participants (80%, n=24) made a quit attempt. Furthermore, over the course of the intervention, smoking urge, cigarettes smoked, nicotine dependence, withdrawal symptoms, and depression scores all significantly decreased. Follow-up quit rates at 1 and 3 months ranged from 6% to 24%, with treatment completers having better outcomes. This first of its kind intervention for HIV-positive African American male smokers was feasible, acceptable, and showed benefit for reducing smoking behaviors and depression scores. Smoking cessation outcomes were on par with other similar programs. A larger trial is needed to address limitations and to confirm benefits.
Collapse
Affiliation(s)
- Alicia K. Matthews
- College of Nursing, University of Illinois, Chicago, Illinois
- Department of Research, Howard Brown Health Center, Chicago, Illinois
| | - Megan Conrad
- Department of Psychology, University of Illinois, Chicago, Illinois
| | - Lisa Kuhns
- Children's Memorial Hospital, Chicago, Illinois
| | - Maria Vargas
- Department of Research, Howard Brown Health Center, Chicago, Illinois
| | - Andrea C. King
- Department of Psychiatry & Behavioral Neuroscience, The University of Chicago, Chicago, Illinois
| |
Collapse
|
40
|
Naltrexone effects on cortisol secretion in women and men in relation to a family history of alcoholism: studies from the Oklahoma Family Health Patterns Project. Psychoneuroendocrinology 2012; 37:1922-8. [PMID: 22575355 PMCID: PMC3449011 DOI: 10.1016/j.psyneuen.2012.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 04/09/2012] [Accepted: 04/11/2012] [Indexed: 02/02/2023]
Abstract
Naltrexone evokes a cortisol response through its blockade of central opioid receptors on the hypothalamic-pituitary-adrenocortical axis (HPA). The magnitude of this cortisol response may be useful as a probe for central opioid activity in different groups of subjects. Accordingly, the present study examined the effect of opioid blockade on the HPA in 70 women and 58 men with (N=41) and without (N=87) a family history of alcoholism, using a randomized, placebo-controlled, double blind administration of oral naltrexone (50mg). Saliva cortisol was sampled at baseline prior to placebo or naltrexone and again every 30 min over the next 180 min. Women had significantly larger cortisol responses to naltrexone than did the men, F=6.88, p<0.0001. There were no significant differences in cortisol response between groups differing in family history of alcoholism, F=0.65, p>0.69. The present results confirm that women have much greater central opioid restraint on the HPA than men do and that this endogenous restraint is unmasked by opioid blockade. However the results provide no evidence of a differential central opioid tonus in persons with a family history of alcoholism at this dose of naltrexone. The cortisol response to naltrexone may be a useful probe for central opioid activity in women and to a lesser degree in men.
Collapse
|
41
|
King AC, Cao D, O'Malley SS, Kranzler HR, Cai X, deWit H, Matthews AK, Stachoviak RJ. Effects of naltrexone on smoking cessation outcomes and weight gain in nicotine-dependent men and women. J Clin Psychopharmacol 2012; 32:630-6. [PMID: 22926596 PMCID: PMC4640209 DOI: 10.1097/jcp.0b013e3182676956] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study examined whether the opioid receptor antagonist naltrexone is efficacious in smoking cessation and whether sex moderates the response. We assessed smoking quit rates and weight gain in a double-blind randomized trial comparing oral naltrexone (n = 162) with placebo (n = 154) in nicotine-dependent participants who wanted to quit smoking. The medication was gradually titrated up to 50 mg during the week before the quit date and then maintained at this dose for 12 weeks. For the first 4 weeks after the quit date, all participants received a nicotine patch to mitigate tobacco withdrawal and attended weekly individual cognitive-behavioral smoking cessation counseling sessions. After this time, participants continued with naltrexone or placebo through 12 weeks. Follow-up assessments were conducted at 26 and 52 weeks. During treatment, naltrexone (vs placebo) increased quit rates, attenuated smoking urge, and reduced weight gain. At follow-up, after medication discontinuation, the effect of naltrexone on improving quit rates was no longer evident. Men and women experienced different benefits from naltrexone; men showed greater reductions in smoking, whereas women showed greater reductions in weight gain. In sum, naltrexone showed acute efficacy in treating nicotine dependence, but after the medication was discontinued, the effect on quit rate was not maintained. Further study of naltrexone in smoking cessation treatment and reduction of cessation-related weight gain, as well as preclinical investigation of mechanisms underlying sex differences, is warranted.
Collapse
Affiliation(s)
- Andrea C King
- Department of Psychiatry & Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Multiple studies in animal models and humans suggest that the endogenous opioid system is an important neurobiological substrate for nicotine addictive properties. In this study, we evaluated the participation of δ-opioid receptors in different behavioral responses of nicotine by using δ-opioid receptor knockout mice. Acute nicotine administration induced hypolocomotion and antinociception in wild-type mice, which were similar in knockout animals. The development of tolerance to nicotine-induced antinociception was also similar in both genotypes. In agreement, the expression and functional activity of δ-opioid receptors were not modified in the different layers of the spinal cord and brain areas evaluated after chronic nicotine treatment. The somatic manifestation of the nicotine withdrawal syndrome precipitated by mecamylamine was also similar in wild-type and δ-opioid receptor knockout mice. In contrast, nicotine induced a conditioned place preference in wild-type animals that was abolished in knockout mice. Moreover, a lower percentage of acquisition of intravenous nicotine self-administration was observed in mice lacking δ-opioid receptors as well as in wild-type mice treated with the selective δ-opioid receptor antagonist naltrindole. Accordingly, in-vivo microdialysis studies revealed that the enhancement in dopamine extracellular levels induced by nicotine in the nucleus accumbens was reduced in mutant mice. In summary, the present results show that δ-opioid receptors are involved in the modulation of nicotine rewarding effects. However, this opioid receptor does not participate either in several acute effects of nicotine or in the development of tolerance and physical dependence induced by chronic nicotine administration.
Collapse
|
43
|
Schnoll RA, Wileyto EP, Lerman C. Extended duration therapy with transdermal nicotine may attenuate weight gain following smoking cessation. Addict Behav 2012; 37:565-8. [PMID: 22244706 DOI: 10.1016/j.addbeh.2011.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 11/07/2011] [Accepted: 12/19/2011] [Indexed: 11/28/2022]
Abstract
AIM People who quit smoking often gain 11-12 lb, on average, which can frequently lead to a relapse to smoking. This study evaluated whether extended vs. standard duration treatment with nicotine patch helps those able to quit smoking to reduce cessation-induced weight gain and explored nicotine patch adherence as a mediator of treatment effects. DESIGN AND SETTING We examined data from a completed randomized placebo-controlled clinical trial of extended (24 weeks) vs. standard (8 weeks plus 16 weeks of placebo) transdermal nicotine patch therapy. Changes in measured weight over 24 weeks were compared across the two treatment arms, controlling for gender, baseline smoking rate, and previous weight. Adherence to patch use was assessed using self-report of daily use over 24 weeks. PARTICIPANTS 139 clinical trial participants who were confirmed to be abstinent at weeks 8 and 24. FINDINGS Compared to participants who received 8 weeks of nicotine patch therapy, participants who received 24 weeks of treatment showed significantly less weight gain from pre-treatment to week 24 (β=-4.76, 95% CI: -7.68 to -1.84, p=.002) and significantly less weight gain from week 8 to week 24 (β=-2.31, 95% CI: -4.39 to -0.23, p=.03). Extended treatment increased patch adherence which, in turn, reduced weight gain; patch adherence accounted for 20% of the effect of treatment arm on weight gain. CONCLUSION Compared to 8 weeks of transdermal nicotine therapy, 24 weeks of patch treatment may help to reduce the weight gain that is typical among smokers who are able to achieve abstinence from tobacco use. Extended treatment increased nicotine patch adherence which, in turn, reduced weight gain.
Collapse
Affiliation(s)
- Robert A Schnoll
- Department of Psychiatry, Abramson Cancer Center, and Annenberg Public Policy Center, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | | | | |
Collapse
|
44
|
Salameh P, Khayat G, Waked M. Lower Prevalence of Cigarette and Waterpipe Smoking, But a Higher Risk of Waterpipe Dependence in Lebanese Adult Women Than in Men. Women Health 2012; 52:135-50. [DOI: 10.1080/03630242.2012.656885] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
45
|
Farley AC, Hajek P, Lycett D, Aveyard P. Interventions for preventing weight gain after smoking cessation. Cochrane Database Syst Rev 2012; 1:CD006219. [PMID: 22258966 DOI: 10.1002/14651858.cd006219.pub3] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Most people who stop smoking gain weight. There are some interventions that have been designed to reduce weight gain when stopping smoking. Some smoking cessation interventions may also limit weight gain although their effect on weight has not been reviewed. OBJECTIVES To systematically review the effect of: (1) Interventions targeting post-cessation weight gain on weight change and smoking cessation.(2) Interventions designed to aid smoking cessation that may also plausibly affect weight on post-cessation weight change. SEARCH METHODS Part 1 - We searched the Cochrane Tobacco Addiction Group's Specialized Register and CENTRAL in September 2011.Part 2 - In addition we searched the included studies in the following "parent" Cochrane reviews: nicotine replacement therapy (NRT), antidepressants, nicotine receptor partial agonists, cannabinoid type 1 receptor antagonists and exercise interventions for smoking cessation published in Issue 9, 2011 of the Cochrane Library. SELECTION CRITERIA Part 1 - We included trials of interventions that were targeted at post-cessation weight gain and had measured weight at any follow up point and/or smoking cessation six or more months after quit day.Part 2 - We included trials that had been included in the selected parent Cochrane reviews if they had reported weight gain at any time point. DATA COLLECTION AND ANALYSIS We extracted data on baseline characteristics of the study population, intervention, outcome and study quality. Change in weight was expressed as difference in weight change from baseline to follow up between trial arms and was reported in abstinent smokers only. Abstinence from smoking was expressed as a risk ratio (RR). We used the most rigorous definition of abstinence available in each trial. Where appropriate, we performed meta-analysis using the inverse variance method for weight and Mantel-Haenszel method for smoking using a fixed-effect model. MAIN RESULTS Part 1: Some pharmacological interventions tested for limiting post cessation weight gain (PCWG) resulted in a significant reduction in WG at the end of treatment (dexfenfluramine (Mean difference (MD) -2.50 kg, 95% confidence interval (CI) -2.98 to -2.02, 1 study), phenylpropanolamine (MD -0.50 kg, 95% CI -0.80 to -0.20, N=3), naltrexone (MD -0.78 kg, 95% CI -1.52 to -0.05, N=2). There was no evidence that treatment reduced weight at 6 or 12 months (m). No pharmacological intervention significantly affected smoking cessation rates.Weight management education only was associated with no reduction in PCWG at end of treatment (6 or 12m). However these interventions significantly reduced abstinence at 12m (Risk ratio (RR) 0.66, 95% CI 0.48 to 0.90, N=2). Personalised weight management support reduced PCWG at 12m (MD -2.58 kg, 95% CI -5.11 to -0.05, N=2) and was not associated with a significant reduction of abstinence at 12m (RR 0.74, 95% CI 0.39 to 1.43, N=2). A very low calorie diet (VLCD) significantly reduced PCWG at end of treatment (MD -3.70 kg, 95% CI -4.82 to -2.58, N=1), but not significantly so at 12m (MD -1.30 kg, 95% CI -3.49 to 0.89, N=1). The VLCD increased chances of abstinence at 12m (RR 1.73, 95% CI 1.10 to 2.73, N=1). There was no evidence that cognitive behavioural therapy to allay concern about weight gain (CBT) reduced PCWG, but there was some evidence of increased PCWG at 6m (MD 0.74, 95% CI 0.24 to 1.24). It was associated with improved abstinence at 6m (RR 1.83, 95% CI 1.07 to 3.13, N=2) but not at 12m (RR 1.25, 95% CI 0.83 to 1.86, N=2). However, there was significant statistical heterogeneity.Part 2: We found no evidence that exercise interventions significantly reduced PCWG at end of treatment (MD -0.25 kg, 95% CI -0.78 to 0.29, N=4) however a significant reduction was found at 12m (MD -2.07 kg, 95% CI -3.78 to -0.36, N=3).Both bupropion and fluoxetine limited PCWG at the end of treatment (bupropion MD -1.12 kg, 95% CI -1.47 to -0.77, N=7) (fluoxetine MD -0.99 kg, 95% CI -1.36 to -0.61, N=2). There was no evidence that the effect persisted at 6m (bupropion MD -0.58 kg, 95% CI -2.16 to 1.00, N=4), (fluoxetine MD -0.01 kg, 95% CI -1.11 to 1.10, N=2) or 12m (bupropion MD -0.38 kg, 95% CI -2.00 to 1.24, N=4). There were no data on WG at 12m for fluoxetine.Overall, treatment with NRT attenuated PCWG at the end of treatment (MD -0.69 kg, 95% CI -0.88 to -0.51, N=19), with no strong evidence that the effect differed for the different forms of NRT. There was evidence of significant statistical heterogeneity caused by one study which reported a 4.3 kg reduction in PCWG due to NRT. With this study removed, the difference in weight change at end of treatment was -0.45 kg (95% CI -0.66 to -0.27, N=18). There was no evidence of an effect on PCWG at 12m (MD -0.42 kg, 95% CI -0.92 to 0.08, N=15).We found evidence that varenicline significantly reduced PCWG at end of treatment (MD -0.41 kg, 95% CI -0.63 to -0.19, N=11), but this effect was not maintained at 6 or 12m. Three studies compared the effect of bupropion to varenicline. Participants taking bupropion gained significantly less weight at the end of treatment (-0.51 kg (95% CI -0.93 to -0.09 kg), N=3). Direct comparison showed no significant difference in PCWG between varenicline and NRT. AUTHORS' CONCLUSIONS Although some pharmacotherapies tested to limit PCWG show evidence of short-term success, other problems with them and the lack of data on long-term efficacy limits their use. Weight management education only, is not effective and may reduce abstinence. Personalised weight management support may be effective and not reduce abstinence, but there are too few data to be sure. One study showed a VLCD increased abstinence but did not prevent WG in the longer term. CBT to accept WG did not limit PCWG and may not promote abstinence in the long term. Exercise interventions significantly reduced weight in the long term, but not the short term. More studies are needed to clarify whether this is an effect of treatment or a chance finding. Bupropion, fluoxetine, NRT and varenicline reduce PCWG while using the medication. Although this effect was not maintained one year after stopping smoking, the evidence is insufficient to exclude a modest long-term effect. The data are not sufficient to make strong clinical recommendations for effective programmes to prevent weight gain after cessation.
Collapse
Affiliation(s)
- Amanda C Farley
- Primary Care Clinical Sciences, University of Birmingham, Birmingham, UK
| | | | | | | |
Collapse
|
46
|
King AC, Cao D, Southard CC, Matthews A. Racial differences in eligibility and enrollment in a smoking cessation clinical trial. Health Psychol 2011; 30:40-8. [PMID: 21299293 DOI: 10.1037/a0021649] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The purpose of this study was to compare the recruitment, eligibility screening, and enrollment of African American and White smokers into an intensive smoking cessation intervention trial [The Chicago STOP Smoking Trial (C-STOP)]. METHODS We compared demographic, smoking, substance use, and medical/psychiatric screening data from the recruitment records of 1,189 non-Hispanic, African American and White smokers screened for eligibility in the last year of a randomized pharmacological and behavioral smoking cessation trial. The study took place at a large urban medical center and two satellite locations within the Chicago metropolitan area. RESULTS Interest levels in the study were high among African American smokers, with twice as many African Americans as Whites contacting study staff for information and an initial screening. However, African Americans were nearly three times as likely not to be enrolled in the trial as Whites, because of higher ineligibility rates, failure to attend a screening session, and lower participation rates even among those meeting eligibility requirements. CONCLUSIONS Racial differences were observed nearly at all levels of enrollment determination. These critical barriers to inclusion of African Americans in smoking cessation research limit our understanding of treatment efficacy and ultimately the ability to reduce the health disparities in tobacco-related disease experienced by African Americans.
Collapse
Affiliation(s)
- Andrea C King
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
47
|
Liu X, Jernigan C. Activation of the opioid μ1, but not δ or κ, receptors is required for nicotine reinforcement in a rat model of drug self-administration. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:146-53. [PMID: 20965223 PMCID: PMC3019243 DOI: 10.1016/j.pnpbp.2010.10.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 10/11/2010] [Accepted: 10/11/2010] [Indexed: 12/17/2022]
Abstract
There has long been an interest in examining the involvement of opioid neurotransmission in nicotine rewarding process and addiction to nicotine. Over the past 3 decades, however, clinical effort to test the effectiveness of nonselective opioid antagonists (mainly naloxone and naltrexone) for smoking cessation has yielded equivocal results. In light of the fact that there are three distinctive types of receptors mediating actions of the endogenous opioid peptides, this study, using a rat model of nicotine self-administration, examined involvement of different opioid receptors in the reinforcement of nicotine by selective blockade of the μ1, the δ, and the κ opioid receptors. Male Sprague-Dawley rats were trained in daily 1h sessions to intravenously self-administer nicotine (0.03 mg/kg/infusion) on a fixed-ratio 5 schedule. After establishment of stable nicotine self-administration behavior, the effects of the opioid antagonists were tested. Separate groups of rats were used to test the effects of naloxanazine (selective for μ1 receptors, 0, 5 and 15 mg/kg), naltrindole (selective for δ receptors, 0, 0.5 and 5mg/kg), and 5'-guanidinonaltrindole (GNTI, selective for κ receptors, 0, 0.25 and 1mg/kg). In each individual drug group, the 3 drug doses were tested by using a within-subject and Latin-Square design. The effects of these antagonists on food self-administering behavior were also examined in the same rats in each respective drug group after retrained for food self-administration. Pretreatment with naloxonazine, but not naltrindole or GNTI, significantly reduced responses on the active lever and correspondingly the number of nicotine infusions. None of these antagonists changed lever-pressing behavior for food reinforcement. These results indicate that activation of the opioid μ1, but not the δ or the κ, receptors is required for the reinforcement of nicotine and suggest that opioid neurotransmission via the μ1 receptors would be a promising target for the development of opioid ligands for smoking cessation.
Collapse
Affiliation(s)
- Xiu Liu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | | |
Collapse
|
48
|
Sánchez-Johnsen LA, Carpentier MR, King AC. Race and sex associations to weight concerns among urban African American and Caucasian smokers. Addict Behav 2011; 36:14-7. [PMID: 20832176 PMCID: PMC3402031 DOI: 10.1016/j.addbeh.2010.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 07/06/2010] [Accepted: 08/02/2010] [Indexed: 11/22/2022]
Abstract
This study compared general weight concerns (Drive for Thinness, Body Dissatisfaction, and Cognitive Restraint) and smoking-specific weight concerns among adult African American and Caucasian women and men smokers enrolled in a smoking cessation clinical trial. Participants were 119 African Americans (73 female) and 182 Caucasians (90 female). Results revealed that general weight concerns were higher in Caucasians versus African Americans, and in women compared with men but there were no race by sex interactions. Drive for Thinness and Body Dissatisfaction was higher in women compared with men, and Cognitive Restraint was highest in Caucasian women. Finally, smoking-specific weight concerns were higher in Caucasian women than both Caucasian and African American men, with African American women intermediate. Results indicate that while Caucasian women preparing to quit smoking exhibited the highest levels of concern about weight, smoking-specific weight concerns, and certain sub-components of general weight concerns were also prevalent among African Americans and Caucasians. Future research is needed to elucidate how race and sex differences in weight concerns may impact smoking cessation.
Collapse
Affiliation(s)
- Lisa A.P. Sánchez-Johnsen
- Department of Psychiatry & Behavioral Neuroscience, The University of Chicago, 5841 South Maryland Avenue, MC 3077, Chicago, Illinois 60637, U.S.A,
| | - Michelle R. Carpentier
- Department of Psychiatry & Behavioral Neuroscience, The University of Chicago, 5841 South Maryland Avenue, MC 3077, Chicago, Illinois 60637, U.S.A,
| | - Andrea C. King
- Department of Psychiatry & Behavioral Neuroscience, The University of Chicago, 5841 South Maryland Avenue, MC 3077, Chicago, Illinois 60637, U.S.A,
| |
Collapse
|
49
|
Berrendero F, Robledo P, Trigo JM, Martín-García E, Maldonado R. Neurobiological mechanisms involved in nicotine dependence and reward: participation of the endogenous opioid system. Neurosci Biobehav Rev 2010; 35:220-31. [PMID: 20170672 PMCID: PMC2908214 DOI: 10.1016/j.neubiorev.2010.02.006] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 02/08/2010] [Accepted: 02/09/2010] [Indexed: 01/07/2023]
Abstract
Nicotine is the primary component of tobacco that maintains the smoking habit and develops addiction. The adaptive changes of nicotinic acetylcholine receptors produced by repeated exposure to nicotine play a crucial role in the establishment of dependence. However, other neurochemical systems also participate in the addictive effects of nicotine including glutamate, cannabinoids, GABA and opioids. This review will cover the involvement of these neurotransmitters in nicotine addictive properties, with a special emphasis on the endogenous opioid system. Thus, endogenous enkephalins and beta-endorphins acting on mu-opioid receptors are involved in nicotine-rewarding effects, whereas opioid peptides derived from prodynorphin participate in nicotine aversive responses. An up-regulation of mu-opioid receptors has been reported after chronic nicotine treatment that could counteract the development of nicotine tolerance, whereas the down-regulation induced on kappa-opioid receptors seems to facilitate nicotine tolerance. Endogenous enkephalins acting on mu-opioid receptors also play a role in the development of physical dependence to nicotine. In agreement with these actions of the endogenous opioid system, the opioid antagonist naltrexone has shown to be effective for smoking cessation in certain sub-populations of smokers.
Collapse
Affiliation(s)
- Fernando Berrendero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, University Pompeu Fabra, PRBB, C/Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Patricia Robledo
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, University Pompeu Fabra, PRBB, C/Doctor Aiguader 88, 08003 Barcelona, Spain
- Municipal Institute of Medical Research (IMIM), Barcelona, Spain
| | - José Manuel Trigo
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, University Pompeu Fabra, PRBB, C/Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Elena Martín-García
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, University Pompeu Fabra, PRBB, C/Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, University Pompeu Fabra, PRBB, C/Doctor Aiguader 88, 08003 Barcelona, Spain
| |
Collapse
|
50
|
Epperson CN, Toll B, Wu R, Amin Z, Czarkowski KA, Jatlow P, Mazure CM, O’Malley SS. Exploring the impact of gender and reproductive status on outcomes in a randomized clinical trial of naltrexone augmentation of nicotine patch. Drug Alcohol Depend 2010; 112:1-8. [PMID: 20561758 PMCID: PMC2946976 DOI: 10.1016/j.drugalcdep.2010.04.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 04/14/2010] [Accepted: 04/15/2010] [Indexed: 10/19/2022]
Abstract
In a series of exploratory analyses, we examined the roles of gender, reproductive status and negative affect on smoking abstinence in subjects participating in a large (n=385) 6-week randomized clinical trial (RCT) of nicotine patch therapy, with varying doses of oral naltrexone (0mg, 25mg, 50mg, 100mg) treatment. Negative affect was assessed daily during the first post-quit week via telephone interactive voice response (IVR). Weight and adverse events were recorded weekly. In the intent to treat sample, the effects of dose on continuous abstinence were non-significant in the overall model for men and women. In the 295 study completers, there was a significant effect of dose on continuous abstinence in women only (F=8.53, p=0.04). In the 100mg group, 71% of women were continuously abstinent compared to 41% in the placebo group (p<0.05). Women in the active naltrexone groups gained less weight (F=2.91, df=3, p=0.04). Women in the 100mg vs. placebo group were less adherent with medication (F=3.19, p<0.05). These effects were not significant in men. Naltrexone treatment condition (100mg vs. placebo, p=0.02, odds ratio (OR)=0.28), gender (OR=0.55 p=0.09), and IVR ratings of negative affect (OR 1.02, p=0.04) predicted abstinence at Week 1 in study completers. Menstrual cycle status on quit day had a modest affect on abstinence. These data suggest that naltrexone dose, gender, and negative affect play a role in smoking abstinence, particularly in the early stages of treatment. When used in conjunction with nicotine replacement therapy, naltrexone dose may be important in women.
Collapse
Affiliation(s)
- C. Neill Epperson
- University of Pennsylvania School of Medicine Department of Psychiatry, Philadelphia, PA 19104 USA,University of Pennsylvania School of Medicine Department of Obstetrics and Gynecology; Philadelphia, PA 19104 USA
| | - Benjamin Toll
- Yale University School of Medicine Department of Psychiatry, New Haven, CT 06520 USA
| | - Ran Wu
- Yale University School of Medicine Department of Psychiatry, New Haven, CT 06520 USA
| | - Zenab Amin
- Yale University School of Medicine Department of Psychiatry, New Haven, CT 06520 USA
| | - Kathryn A. Czarkowski
- University of Pennsylvania School of Medicine Department of Psychiatry, Philadelphia, PA 19104 USA
| | - Peter Jatlow
- Yale University School of Medicine Department of Laboratory Medicine; New Haven, CT 06520 USA
| | - Carolyn M. Mazure
- Yale University School of Medicine Department of Psychiatry, New Haven, CT 06520 USA,Yale University, Department of Psychology, New Haven, CT 06520 USA,Yale University, Women’s Health Research at Yale; New Haven, CT 06520 USA
| | - Stephanie S. O’Malley
- Yale University School of Medicine Department of Psychiatry, New Haven, CT 06520 USA,Yale University, Department of Psychology, New Haven, CT 06520 USA
| |
Collapse
|