Abstract
Cigarette smoking is highly addictive and modern genetic research has identified robust genetic influences on nicotine dependence. An important step in translating these genetic findings is to identify the genetic factors affecting smoking cessation in order to enhance current smoking cessation treatments. We reviewed the significant genetic variants that predict nicotine dependence, smoking cessation, and response to cessation pharmacotherapy. These data suggest that genetic risks may predict smoking cessation outcomes and moderate the effect of pharmacological treatments. Some pharmacogenetic findings have been replicated in meta-analyses or multiple smoking cessation trials. The variation in efficacy between smokers with different genetic markers supports the notion that personalized smoking cessation intervention based upon genotype could maximize the efficiency of such treatment while minimizing side effects, thus influencing the number needed to treat (NNT) and the number needed to harm (NNH). In summary, as precision medicine is revolutionizing health care, smoking cessation may be one of the first areas where genetic variants identify individuals at increased risk. Genetic variants predict cessation failure, and this increased risk may be ameliorated by cessation pharmacotherapy.
Keywords: Smoking cessation, precision medicine, pharmacogenetics
1. INTRODUCTION
Cigarette smoking is a major global public health threat [1–4], and smoking cessation greatly diminishes the increased risk of mortality[3]. Nicotine dependence is a classic addictive disorder with symptoms of craving, withdrawal syndrome and heavy, uncontrollable use [5]. Nicotine dependence is also manifested by both quitting difficulty [6] and a high likelihood of lapses or relapses after a quit attempt [7–10]. Therefore, identification of the factors that contribute to smoking cessation difficulties is a critical step in understanding the biology of nicotine dependence, enhancing prediction of prognosis and treatment outcomes, and informing more effective cessation treatments.
Leveraging advances in genomics and data science, the goal of precision medicine is to tailor treatments based on an individual’s unique characteristics to maximize benefits and minimize side effects [11]. Growing research in understanding human genomes and identifying specific genetic markers for a disease will not only lead to improved understanding of the biology underlying the disease, but also improved clinical care. For example in clinical practice, it is common for patients to vary in efficacy or side effects to the same medication regimen such as tamoxifen for breast cancer risk reduction in patients with selected biomarkers [12]. Increasing evidence suggests that the risk benefit ratio of the medication may vary with a person’s genetic makeup.
Recent research has begun to determine biomarkers (such as genotype, nicotine and metabolite levels) that may have utility in predicting smoking cessation. Use of biomarkers in smoking cessation treatments has great promise as summarized by multiple reviews [13–16]. Different pharmacokinetic and pharmacodynamic markers have been implicated in personalized therapeutic approaches including variation in Cytochrome P450 2A6 (CYP2A6), nicotinic acetylcholine receptor genes (nAChRs) and other genes based on the Genome Wide Association Study (GWAS) discovery, although the precise mechanisms underlying many of these findings remain unknown [17, 18]. For example, variants in the genes encoding the α5-α3-β4 nicotinic receptor subunits show promise as a biomarker for predicting risk for developing nicotine dependence [19–22], lung cancer [23, 24], chronic obstructive pulmonary disease (COPD) [25], delayed smoking cessation [26], and possibly a differential response to pharmacologic treatment [27, 28].
Biomarkers are useful if they predict disease course or treatment outcomes. David and colleagues suggested key factors to consider when evaluating biomarkers: (1) analytical validation: determination of whether or not a biomarker is able to be measured accurately, precisely and reliably; (2) qualification: assessment of available evidence on associations between the biomarker and disease states; and (3) utilization: determination of adequate evidence to support applying the biomarker for a specific use [29]. For smoking cessation treatment, Bough and colleagues suggested 3 types of biomarkers: (1) diagnostic biomarkers, which can be used to identify the presence or absence of a specific disease state and aid in patient selection; (2) pharmacodynamics biomarkers, which can be used to report on the downstream effects of a pharmacotherapy and determining optimal dosing; (3) predictive biomarkers, which can be used to select patients who are likely to respond to therapy [30].
Opinions differ as to whether pharmacogenetic testing should be implemented in the clinic at this time because clinical utility and cost-effectiveness require further investigation. However, allowing physicians to select medications for individuals based, at least in part, on genetic factors that predispose to treatment success or failure rather than on a trial and error basis may ultimately improve successful smoking cessation rates [31]. In the post-GWAS era, translational research from several disciplines, including behavioral science, ethics and economics, should be performed in parallel with ongoing genome-wide association studies for smoking behavior and pharmacogenetic trials. This critical step will enable translation and implementation of genetic insights into clinical practice to reduce the global burden of smoking [32].
2. Genetics of Nicotine Dependence
Multiple recent large scale genetic meta-analyses based on tens of thousands of subjects of European descent confirmed the association of 15q25.1 with smoking heaviness, defined by cigarettes per day [19–22], with the most robust associations being reported for rs16969968 and rs1051730, two highly correlated variants (p<5.57*10−72) [21]. In the CHRNA5-A3-B4 region, at least two independent signals have been identified [20, 33]. The first signal, tagged by rs16969968, a variant that results in an amino acid change in the α5 nicotinic cholinergic receptor (CHRNA5), alters nicotinic receptor conductance in vitro [34, 35]. A second, distinct signal tagged by rs680244 is associated with variability in CHRNA5 mRNA levels [36]. Animal studies of the association between CHRNA5 and nicotine dependence has shed light on potential biological mechanisms underlying addiction [37–39]. Individuals of European descent have one of the three common haplotypes in the region spanning CHRNA5 and the 3’ end of CHRNA3 [33], which can be defined by these two variants: rs16969968 and rs680244 [36]. These three common haplotypes represent different risk levels of nicotine dependence: low-risk (H1, frequency 21%), intermediate-risk (H2, frequency 44%), and high-risk (H3, frequency 36%) haplotypes. The association of CHRNA5 and heavy smoking extends from individuals of European ancestry to diverse ancestry backgrounds [40]. In a recent large meta-analysis of cigarettes per day (CPD) in 32,389 individuals of African ancestry, rs2036527 in CHRNA5 is found as associated with heavy smoking [41].
More genome wide association studies (GWAS) examined different smoking phenotypes. Growing GWAS evidence supports CHRNA5 as a robust top predictor for different smoking phenotypes defined as cigarettes smoked per day [19–22], nicotine dependence as defined by Fagerstrom Test for Nicotine Dependence (FTND) [42], carbon monoxide levels [43], or cotinine levels [44],
Dissection of phenotypic features associated with CHRNA5 reveals that CHRNA5 risk gene is associated with a smoker profile of compulsive heavy smoking with craving which is best captured by high cigarettes per day instead of a DSM diagnosis of tobacco dependence [45]. Research suggests nicotinic receptor variants are associated with greater odds of nicotine dependence according to cigarettes per day (CPD) and to a lesser extent Time to First Cigarette (TTF) [46]. It is possible different phenotypic characteristics of nicotine dependence are associated with different risk genes discovered in recent GWAS of nicotine dependence [47]. Identification of such a phenotypic cluster can be a pivotal step for further pharmacogenetic studies with specific genetic targets [48].
Additional genes (CYP2A6 and other nicotinic receptor genes) have also been shown as associated with heavy smoking. Multiple signals are independently associated with heavy smoking and nicotine dependence. For example, variants in CYP2A6 has been shown to predict risk for developing nicotine dependence [21, 22, 49], possibly lung cancer [50, 51] and chronic obstructive pulmonary disease (COPD) [52], delayed smoking cessation [53–55], and possibly a differential response to pharmacologic treatment [56]. Moreover, evidence suggests that a metabolic biomarker, nicotine metabolism ratio (NMR), and CHRNA5 may have independent and additive effects on nicotine dependence [57]. A recent GWAS of a nicotine metabolism biomarker, NMR, identified variants in CYP2A6 which explained 30% of variance in the nicotine metabolism biomarker, NMR [58].
3. Genetics of Smoking Cessation
Compared to smoking quantity as defined by cigarettes per day, smoking cessation is a much more complex behavioral phenotype, which may be defined variably as a simple contrast of former versus current smokers, age of quitting smoking, or days to relapse in a well-controlled smoking cessation trial. Using a crude phenotypic contrast (current vs. former smokers), genome wide association study of smoking cessation identified DBH as associated with cessation [21]. Using more refined definitions of smoking cessation such as days to relapse in smoking cessation treatment trials or age of quitting smoking, the CHRNA5-CHRNA3-CHRNB4 variants have been shown as associated with cessation outcomes, although the level of association is not as strong as the genetic association with smoking heaviness measures. Multiple studies show an association between the CHRNA5-CHRNA3-CHRNB4 region and successful smoking cessation [26–28, 59–64]. These studies found that the same genetic risk variants that contributed to smoking heaviness and nicotine dependence also predicted smoking cessation. Yet, other studies failed to confirm this association [65–68]. One large genome-wide association meta-analysis, that strongly supported the association between 15q25.1 and smoking heaviness, reported a modest association with current versus former smoking as a measure of smoking cessation below genome wide level of significance [21]. Uhl and colleagues [67], in a genome-wide association of three treatment cohorts, did not identify any nicotinic receptor genes as predictors of smoking cessation in smokers receiving nicotine replacement therapy. Variations in study design (with or without a placebo group), ascertainment, definitions of smoking cessation (time to relapse, abstinence, or the contrast of former vs. current smoker), and study power may explain these inconsistent findings.
3.1. CHRNA5-CHRNA3-CHRNB4 and smoking cessation
3.1.1. CHRNA5-CHRNA3-CHRNB4 Genetic Variants Predict lung cancer, chronic obstructive pulmonary disease (COPD), coronary artery disease (CAD), and mortality
GWAS of lung cancer reveals CHRNA5 as one of the top signals for increased lung cancer risks [23]. However, whether the risk gene exerts a direct or indirect effect via smoking on increased lung cancer risk is still unknown [24, 69, 70]. CHRNA5 has been shown to not only predict increase in lung cancer risk, but also predict earlier age of lung cancer diagnosis [71]. In a large meta-analysis, Chen and colleagues [26] showed the clinical significance of the CHRNA5 variant rs16969968 and that it predicts delayed smoking cessation and an earlier age of lung cancer diagnosis. Among smokers with lung cancer diagnoses, the rs16969968 genotype (AA) was associated with a four-year earlier median age of diagnosis compared with the low-risk genotype (GG). Smokers with high-risk AA genotypes were diagnosed with lung cancer at age 61, four years earlier than age 65 for those with low-risk GG genotypes. These findings underscore the potential clinical and public health importance of rs16969968 in CHRNA5 in relation to smoking cessation success and lung cancer risk [26]. Importantly, CHRNA5 is also associated with chronic obstructive pulmonary disease (COPD) [25, 72], coronary artery disease (CAD) [73, 74], and increased mortality in large scale population studies [75, 76].
3.1.2. CHRNA5-CHRNA3-CHRNB4 Genetic Variants Predict Age of Smoking Cessation
CHRNA5-CHRNA3-CHRNB4 haplotypes have been found to be associated with age of self-reported smoking cessation in a community-based sample [27]. In a large meta-analysis, Chen and colleagues [26] showed the clinical significance of the CHRNA5 variant rs16969968 in predicting smoking cessation. The rs16969968 genotype (AA) was associated with a four-year delay (age 52 delayed to 56) in median age of smoking cessation compared with the low-risk genotype (GG). CHRNA5 predicts delayed smoking cessation even in special high-risk population such as patients with myocardial infarction (MI). Among patients who were ever smokers and hospitalized with acute MI, the high-risk CHRNA5 allele was associated with lower likelihood of quitting before hospitalization and significantly less abstinence one year after hospitalization with MI. The CHRNA5 rs16969968 genotype may therefore identify patients who would benefit from aggressive, tailored smoking cessation intervention [77].
3.1.3 CYP2A6 and smoking cessation
Variation in nicotine metabolism, and variation in the gene that encodes the primary nicotine metabolism enzyme, cytochrome P450 2A6 (CYP2A6), are robustly associated with cigarette consumption [22, 78–80]. Several studies have reported an influence of nicotine metabolic rate upon cessation [53–55], although the predictive relation between nicotine metabolism and different treatment regimens remains unclear. In general, smokers who are slow nicotine metabolizers (both genetic and phenotypic) had significantly higher plasma nicotine levels and higher quit rates with both placebo and nicotine gum treatments [54, 81, 82].
4. Gene-environment Interplay of Smoking Cessation
The genetic risk association varies with potential moderators for the genetic risk such as the impact of disease/symptom, use of cessation medication, and influence of environmental risk factors. Some studies show an association between CHRNA5 and smoking cessation [59–63], whereas other studies do not [65–67]. It is likely that the expression of this genetic risk on cessation varies with several factors, such as developing a smoking-related disease such as COPD or lung cancer [26], use of cessation pharmacotherapy [27], and environmental influences on smoking cessation such as living with a partner who smokes [83]. The effect of this genetic locus is seen most clearly in subjects who have strong environmental influences to quit smoking, no smoking-related disorder, and no use of cessation medicine. If CHRNA5 risk alleles causes early development of smoking-related disease such as lung cancer, the disease development encourages earlier quitting than would otherwise occur. Therefore, the genetic effect of delaying cessation is offset by its effect of accelerating disease development which may promote quitting [26]. Similarly, use of cessation pharmacotherapy may reduce the effect of CHRNA5 on cessation difficulty since pharmacotherapy appears to alter this genetic risk [27].
5. Genetics of Smoking Cessation Treatment Response
Recent evidence suggests that the efficacy of smoking cessation pharmacotherapy can vary across patients based on their genotypes or metabolic markers.
5.1. CHRNA5-CHRNA3-CHRNB4 predicts response to medication
Growing evidence suggests that CHRNA5 predicts response to cessation pharmacotherapy. Sarginson and colleagues [62] conducted a pharmacogenetic analysis that showed a significant association between rs8192475 (R37H) in CHRNA3, higher craving after quitting, and increased withdrawal symptoms over time. They identified two markers for point prevalence abstinence, CHRNA5 SNP rs680244 and CHRNB4 SNP rs12914008, and provided support for the role of the CHRNA5/A3/B4 subunits in determining number of cigarettes smoked and response to smoking cessation therapy. Munafo and colleagues [61] reported a weak association between CHRNA5 variant and short-term smoking cessation in treatment seeking smokers, which does not seem to operate only among those receiving nicotine replacement therapy. Further, in a retrospective candidate pharmacogenetics analysis, several genes were examined with respect to different pharmacotherapies: In the treatment arm with varenicline, continuous abstinence (weeks 9–12) was associated with multiple nAChR subunit genes (including CHRNB2, CHRNA5, and CHRNA4). In the treatment arm with bupropion, abstinence was associated with CYP2B6. Incidence of nausea was associated with several nAChR subunit genes [63]. More recently, both CHRNB4 promoter SNP rs3813567 and CHRNA5 rs680244 genotypes were associated with both point prevalence abstinence and post-quit craving in a selegiline trial. These findings highlight the potential of CHRNA5-CHRNA3-CHRNB4 genetic markers in identifying those likely to respond to pharmacotherapy for smoking cessation [84]. In a recent examination of a large smoking cessation trial by Chen and colleagues [27], smokers receiving cessation counseling with placebo medication, the high-risk CHRNA5 rs16969968-rs680244 haplotype that is associated with heavy smoking predicts failed abstinence in comparison to the low-risk haplotype . Pharmacological treatment significantly increased the likelihood of abstinence in individuals with the high-risk haplotype, but exerted little effects in individuals with the low-risk haplotype. This is reflected by a significant interaction between treatment (placebo versus active treatment) and CHRNA5 haplotypes. Across the active pharmacologic treatment conditions, these genetic variants do not predict abstinence, and this reduced genetic effect with pharmacological treatments suggests that cessation treatments differ in effectiveness across the haplotypes and most strongly mitigate the genetic risks for cessation difficulty. A subsequent meta-analysis of the Pharmacogenetics of Nicotine Addiction Treatment (PNAT) Consortium confirms a similar pharmacogenetic interaction that patient responses to nicotine replacement therapy (NRT) are moderated by CHRNA5 genetic variants [28].
Medication efficacy is often represented by the number-needed-to-treat (NNT). The NNT is 7 when computed across all individuals regardless of their haplotype status, supporting the established effect of pharmacotherapy. However, the NNT varies widely depending on the individual’s haplotype. Based on their absolute risks, the NNT is 4 for smokers with the high-risk haplotype, 7 for smokers with the intermediate-risk haplotype, and >1000 for smokers with the low-risk haplotype [27, 85]. An NNT of 4 is an impressive finding, compared to the NNT’s of many existing pharmacotherapies [86–88]. The wide variation in NNT between smokers with different haplotypes supports the notion that personalized smoking cessation intervention based upon genotype, could meaningfully increase the efficiency of such treatment.
The pharmacogenetic associations could be ameliorated by different factors such as effective treatments or extremely high or low cessation rates. This is reflected in a recent meta-analysis to examine only smokers receiving nicotine replacement therapy without a control group where rs16969968 or rs1051730 showed no association with smoking abstinence [89]. In another recent trial of smokers of European ancestry, CHRNA5-A3-B4 gene variants exhibited a trending, but not significant association with smoking cessation in smokers receiving placebo; however, there was no association with another robust predictor, nicotine metabolism ratio (NMR), in this same group [68]. In an effort to synthesize existing pharmacogenetics findings, Chen and colleagues [90] examined results from two large smoking cessation trials conducted in different institutes and demonstrated inconsistent genotypic associations in the placebo arms across these different trials. This inconsistency highlights the need to compare the most effective pharmacotherapies in the same trial with the same placebo control to establish pharmacogenetic evidence to aid decisions on medication choice for patients trying to quit smoking. In fact, interpretation of any results of pharmacogenetics analyses highlights the importance of a placebo control group in testing whether medication efficacy differ with genetic markers [91].
Pharmacogenetic evidence beyond European ancestry is important but quite limited at this time. In recent clinical trials of African Americans, Zhu and colleagues reported that CHRNA5-A3-B4 variants influence smoking abstinence during active pharmacotherapy [64]. Medication adherence, another important predictor for cessation outcome, also has been examined for genetic associations in smokers of European ancestry. Secondary analysis of data from a pharmacogenetic smoking cessation trial showed a weak potential association between CHRNA5 rs1051730 genotype and adherence to prescribed NRT dose [92].
5.2. Nicotine metabolism biomarkers (NMR, CYP2A6, CYP2B6) predicts response to treatment
Previous studies of nicotine metabolism and cessation treatment have examined a proxy for CYP2A6 activity, Nicotine Metabolite Ratio (NMR), the ratio of two stable nicotine metabolites, cotinine: 3-hydroxycotine, measured in the blood, urine, or saliva of current smokers [53–55, 93, 94]. CYP2A6 is highly polymorphic, with reduced function alleles producing significantly slower rates of nicotine metabolism. Relatively common variants define multiple CYP2A6 haplotypes in European populations [95], and the large majority of inter-individual variation in metabolism of nicotine to cotinine can be explained by seven polymorphisms among European Americans [96]. Chen, Bloom, and colleagues showed that CYP2A6-defined nicotine metabolic function moderated the effect of smoking cessation pharmacotherapy on smoking relapse with pharmacotherapy significantly slowing relapse in fast, but not slow, metabolizers. Nicotine replacement therapy was effective amongst individuals with fast, but not slow, CYP2A6-defined nicotine metabolism. In contrast, the effect of bupropion on relapse likelihood was unlikely affected by nicotine metabolism as estimated from CYP2A6 genotype [56].
Metabolic markers, rather than genetic variants, may be used to optimize treatment choice for individual smokers and to improve treatment outcomes. Lerman and colleagues examined nicotine metabolism ratio (NMR) and reported findings from a recent trial that at end of treatment, varenicline was more efficacious than nicotine patch in normal metabolizers, but not in slow metabolizers [97]. This study suggested that treating normal metabolizers with varenicline and slow metabolizers with nicotine patch could optimize quit rates while minimizing side-effects [97]. However, in this trial normal vs. slow metabolizers did not differ in abstinence, which raised concern of inconsistency with existing evidence suggesting a higher quit rate for slow metabolizers [81, 82] . Another study of a community-based sample of treatment seeking smokers who received transdermal nicotine showed that faster nicotine metabolizers were significantly less likely to quit smoking although there is no control comparison in this trial [82].
Another study of smokers who received behavioral counseling and drug therapy (bupropion, nicotine replacement therapy, and/or varenicline) showed that patients with CYP2B6*4 (rs2279343) variant had lower success rate with bupropion. Likely, the CYP2B6*4 variant influences the pharmacological activity of bupropion, and CYP2B6*4 may be a genetic marker for individualized bupropion pharmacotherapy [98].
5.3. Other markers and smoking cessation treatment response
Additional genetic and imaging biomarkers reported in respect to smoking cessation treatment response requires further evidence of replication and biological investigation [99–106]. However, few of these candidate genes are present within regions of suggestive or significant linkage or overlap with genome-wide linkage or association studies of tobacco dependence or smoking cessation [107].
There have been attempts to create polygenic risk scores to predict smoking cessation or treatment response, although their variant selection is based on candidate gene studies, or subthreshold associations, and not based on genetic signals surpassing GWAS association thresholds. For example, an ‘additive genetic efficacy score’ (AGES) based on dopamine functional polymorphisms appears to predict lapse to smoking following a quit attempt [108]. Rose, Uhl, and colleagues developed quit-success genotype score based on many variants arising from pooled GWAS results in smokers receiving nicotine replacement therapy [109, 110]. Different neuroimaging markers have also been examined [111]. For example, lower pre-treatment brain nAChR density has been shown as associated with a greater chance of quitting smoking with NRT or placebo [112]. Cigarette smoking leads to upregulation of nicotinic acetylcholine receptors (nAChRs) in the human brain, including the common α4β2* nAChR subtype. Smokers with less upregulation of available α4β2* nAChRs have a greater likelihood of quitting with treatment than smokers with more upregulation. While the costly, time-consuming PET procedure used here is not likely to be used clinically, simpler methods for examining α4β2* nAChR upregulation may be developed in the future to help determine which smokers need more intensive treatment [113].
6. Translation and Feasibility
Despite growing evidence of genetic markers as predictor of prognosis and treatment response, translation into clinical practice requires much work due to many factors such as inconsistent evidence, little data in diverse ancestry populations, lack of available CLIA-certified fast genotyping for clinical use, and need for research on cost-effectiveness [2, 14, 17, 44, 54, 114, 115] . To implement precision medicine for smoking cessation treatment, there are potential barriers. Some people remain skeptical for the value of genetic testing while proven interventions for treating tobacco dependence, including simple cost-effective measures, such as quit lines which are underutilized [116]. Furthermore, whether the use of personalized genomic information can be used to promote behavior change remains a topic of debate [117].
7. CONCLUSIONS
Multiple studies underscore the relation between the genetic markers, nicotine dependence, smoking-related diseases, and smoking cessation. First, current evidence demonstrates that smokers with the high-risk α5 nicotinic cholinergic receptor CHRNA5 genotypes appear more biologically predisposed to have difficulty quitting without pharmacologic treatment and acceleration of multiple health risks including earlier mortality, and this risk may be ameliorated by effective pharmacological treatment. Second, current evidence strongly suggests a significant interaction between CHRNA5 genotypes or nicotine metabolism ratio (NMR) and pharmacotherapy on cessation success. This evidence reveals that cessation pharmacotherapy effectiveness is modulated by these biomarkers. These findings strengthen the case for the development and rigorous testing of treatments that target patients with different biological risk profiles based on the chromosome 15q25 region that includes the genes encoding the nicotinic receptor subunits or nmetabolic biomarkers. Additional research on prospective replication of these pharmacogenetics findings in diverse populations is needed before clinical translation.
Smoking cessation pharmacotherapy such as nicotine replacement therapy (NRT), bupropion, and varenicline are moderately effective yet with side effects. Identifying genes related to responsiveness to pharmacologic treatment for nicotine addiction may lead to improved treatment algorithms that further the promise of personalized medicine [118]. Additional research on related topics such as epigenetic markers and personalized risk communication is also needed in the future. To implement precision medicine for smoking cessation treatments, we need collaborative, prospective research on pharmacogenetics, well-designed meta-analyses of diverse populations, cost-effectiveness analyses, and useful polygenetic risk scores linked to clinical indicators such as number needed to treat (NNT) in order to match smokers with treatments of maximal efficacy and minimal side effects.
Highlights.
Genetic research has identified robust genetic influences on nicotine dependence
These same genetic risk factors that influence the development of nicotine dependence are the strongest risk factors for the development of lung cancer and chronic obstructive pulmonary disease.
Genetic research has identified prognostic predictor for cessation failure
Genetic research has identified predictors for response to treatment
Tailored cessation treatments may reduce side effects and increase cessation success
Acknowledgments
The authors thank Nina Smock for the assistance in project coordination and editing/preparing the manuscript.
FUNDING SUPPORT
This research was supported by R01 DA038076, and K08 DA030398 (LSC) from the National Institute on Drug Abuse, and sub-award KL2 RR024994 (LSC) from the National Center for Research Resources. Dr. Bierut (LJB) is supported by National Institute on Drug Abuse grant R01DA036583 and National Cancer Institute grant P30CA091842.
Abbreviations
- AGES
Additive genetic efficacy score
- CAD
Coronary artery disease
- CHRNA5
α5 nicotinic cholinergic receptor
- COMT
Catechol-O-methyltransferase
- COPD
Chronic Obstructive Pulmonary Disease
- CPD
Cigarettes per day
- CYP2A6
Cytochrome P450 2A6
- FTND
Fagerstrom Test for Nicotine Dependence
- GWAS
Genome Wide Association Study
- MI
Myocardial infarction
- nAChRs
Nicotinic Acetylcholine Receptor Genes
- NMR
Nicotine metabolism ratio
- NNT
Number Needed-To Treat
- NNH
The Number Needed To Harm
- NRT
Nicotine replacement therapy
- PNAT
Pharmacogenetics of Nicotine Addiction Treatment
- TTF
Time to First Cigarette
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
DISCLOSURES
Dr. Bierut is listed as an inventor on Issued U.S. Patent 8,080,371,“Markers for Addiction” covering the use of certain SNPs in determining the diagnosis, prognosis, and treatment of addiction.
References
- 1.Knopik VS, Maccani MA, Francazio S, McGeary JE. The epigenetics of maternal cigarette smoking during pregnancy and effects on child development. Dev Psychopathol. 2012;24:1377–1390. doi: 10.1017/S0954579412000776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.McClure JB, Swan GE, St John J, Fauver R, Javitz HS, Bergen AW, Nishita D, Niaura R, Munafo MR, David SP. Pharmacogenetic Smoking Cessation Intervention in a Health Care Setting: A Pilot Feasibility Study. Nicotine & Tobacco Research. 2013;15:518–526. doi: 10.1093/ntr/nts173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jha P, Ramasundarahettige C, Landsman V, Rostron B, Thun M, Anderson RN, McAfee T, Peto R. 21st-century hazards of smoking and benefits of cessation in the United States. N Engl J Med. 2013;368:341–350. doi: 10.1056/NEJMsa1211128. [DOI] [PubMed] [Google Scholar]
- 4.Whiteford HA, Baxter AJ. The Global Burden of Disease 2010 Study: what does it tell us about mental disorders in Latin America? Rev Bras Psiquiatr. 2013;35:111–112. doi: 10.1590/1516-4446-2012-3502. [DOI] [PubMed] [Google Scholar]
- 5.Breslau N, Johnson EO, Hiripi E, Kessler R. Nicotine dependence in the United States: prevalence, trends, and smoking persistence.[see comment] Archives of General Psychiatry. 2001;58:810–816. doi: 10.1001/archpsyc.58.9.810. [DOI] [PubMed] [Google Scholar]
- 6.Breslau N, Johnson EO. Predicting smoking cessation and major depression in nicotine-dependent smokers. Am J Public Health. 2000;90:1122–1127. doi: 10.2105/ajph.90.7.1122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Baker TB, Piper ME, McCarthy DE, Bolt DM, Smith SS, Kim SY, Colby S, Conti D, Giovino GA, Hatsukami D, Hyland A, Krishnan-Sarin S, Niaura R, Perkins KA, Toll BA. Time to first cigarette in the morning as an index of ability to quit smoking: implications for nicotine dependence. Nicotine Tob Res. 2007;9(Suppl 4):S555–S570. doi: 10.1080/14622200701673480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hendricks PS, Prochaska JJ, Humfleet GL, Hall SM. Evaluating the validities of different DSM-IV-based conceptual constructs of tobacco dependence. Addiction. 2008;103:1215–1223. doi: 10.1111/j.1360-0443.2008.02232.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kozlowski LT, Porter CQ, Orleans CT, Pope MA, Heatherton T. Predicting smoking cessation with self-reported measures of nicotine dependence: FTQ, FTND, and HSI. Drug Alcohol Depend. 1994;34:211–216. doi: 10.1016/0376-8716(94)90158-9. [DOI] [PubMed] [Google Scholar]
- 10.West R. Defining and Assessing Nicotine Dependence in Humans. In: Bock G, Goode J, editors. Understanding Nicotine and Tobacco Addiction: Novartis Foundation Symposium. Vol. 275. Chichester, UK: John Wiley & Sons, Ltd; 2006. pp. 36–58. [Google Scholar]
- 11.Green ED, Guyer MS. Charting a course for genomic medicine from base pairs to bedside. Nature. 2011;470:204–213. doi: 10.1038/nature09764. [DOI] [PubMed] [Google Scholar]
- 12.Visvanathan K, Hurley P, Bantug E, Brown P, Col NF, Cuzick J, Davidson NE, Decensi A, Fabian C, Ford L, Garber J, Katapodi M, Kramer B, Morrow M, Parker B, Runowicz C, Vogel VG, 3rd, Wade JL, Lippman SM. Use of Pharmacologic Interventions for Breast Cancer Risk Reduction: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2013 doi: 10.1200/JCO.2013.49.3122. [DOI] [PubMed] [Google Scholar]
- 13.Chen LS, Bierut LJ. Genomics and personalized medicine: and smoking cessation treatment. J Food Drug Anal. 2013;21:S87–S90. doi: 10.1016/j.jfda.2013.09.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kortmann GL, Dobler CJ, Bizarro L, Bau CH. Pharmacogenetics of smoking cessation therapy. Am J Med Genet B Neuropsychiatr Genet. 2010;153B:17–28. doi: 10.1002/ajmg.b.30978. [DOI] [PubMed] [Google Scholar]
- 15.Bough KJ, Amur S, Lao G, Hemby SE, Tannu NS, Kampman KM, Schmitz JM, Martinez D, Merchant KM, Green C, Sharma J, Dougherty AH, Moeller FG. Biomarkers for the development of new medications for cocaine dependence. Neuropsychopharmacology. 2014;39:202–219. doi: 10.1038/npp.2013.210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Epstein RS. What's needed for personalized therapy in smoking cessation. Clin Pharmacol Ther. 2008;84:309–310. doi: 10.1038/clpt.2008.125. [DOI] [PubMed] [Google Scholar]
- 17.Tanner JA, Chenoweth MJ, Tyndale RF. Pharmacogenetics of nicotine and associated smoking behaviors. Curr Top Behav Neurosci. 2015;23:37–86. doi: 10.1007/978-3-319-13665-3_3. [DOI] [PubMed] [Google Scholar]
- 18.Gold AB, Lerman C. Pharmacogenetics of smoking cessation: role of nicotine target and metabolism genes. Hum Genet. 2012 doi: 10.1007/s00439-012-1143-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Liu JZ, Tozzi F, Waterworth DM, Pillai SG, Muglia P, Middleton L, Berrettini W, Knouff CW, Yuan X, Waeber G, Vollenweider P, Preisig M, Wareham NJ, Zhao JH, Loos RJ, Barroso I, Khaw KT, Grundy S, Barter P, Mahley R, Kesaniemi A, McPherson R, Vincent JB, Strauss J, Kennedy JL, Farmer A, McGuffin P, Day R, Matthews K, Bakke P, Gulsvik A, Lucae S, Ising M, Brueckl T, Horstmann S, Wichmann HE, Rawal R, Dahmen N, Lamina C, Polasek O, Zgaga L, Huffman J, Campbell S, Kooner J, Chambers JC, Burnett MS, Devaney JM, Pichard AD, Kent KM, Satler L, Lindsay JM, Waksman R, Epstein S, Wilson JF, Wild SH, Campbell H, Vitart V, Reilly MP, Li M, Qu L, Wilensky R, Matthai W, Hakonarson HH, Rader DJ, Franke A, Wittig M, Schafer A, Uda M, Terracciano A, Xiao X, Busonero F, Scheet P, Schlessinger D, St Clair D, Rujescu D, Abecasis GR, Grabe HJ, Teumer A, Volzke H, Petersmann A, John U, Rudan I, Hayward C, Wright AF, Kolcic I, Wright BJ, Thompson JR, Balmforth AJ, Hall AS, Samani NJ, Anderson CA, Ahmad T, Mathew CG, Parkes M, Satsangi J, Caulfield M, Munroe PB, Farrall M, Dominiczak A, Worthington J, Thomson W, Eyre S, Barton A, Mooser V, Francks C, Marchini J. Meta-analysis and imputation refines the association of 15q25 with smoking quantity. Nat Genet. 2010;42:436–440. doi: 10.1038/ng.572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Saccone NL, Culverhouse RC, Schwantes-An TH, Cannon DS, Chen X, Cichon S, Giegling I, Han S, Han Y, Keskitalo-Vuokko K, Kong X, Landi MT, Ma JZ, Short SE, Stephens SH, Stevens VL, Sun L, Wang Y, Wenzlaff AS, Aggen SH, Breslau N, Broderick P, Chatterjee N, Chen J, Heath AC, Heliovaara M, Hoft NR, Hunter DJ, Jensen MK, Martin NG, Montgomery GW, Niu T, Payne TJ, Peltonen L, Pergadia ML, Rice JP, Sherva R, Spitz MR, Sun J, Wang JC, Weiss RB, Wheeler W, Witt SH, Yang BZ, Caporaso NE, Ehringer MA, Eisen T, Gapstur SM, Gelernter J, Houlston R, Kaprio J, Kendler KS, Kraft P, Leppert MF, Li MD, Madden PA, Nothen MM, Pillai S, Rietschel M, Rujescu D, Schwartz A, Amos CI, Bierut LJ. Multiple independent loci at chromosome 15q25.1 affect smoking quantity: a meta-analysis and comparison with lung cancer and COPD. PLoS Genet. 2010;6:1–16. doi: 10.1371/journal.pgen.1001053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.TAG. Genome-wide meta-analyses identify multiple loci associated with smoking behavior. Nat Genet. 2010;42:441–447. doi: 10.1038/ng.571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Thorgeirsson TE, Gudbjartsson DF, Surakka I, Vink JM, Amin N, Geller F, Sulem P, Rafnar T, Esko T, Walter S, Gieger C, Rawal R, Mangino M, Prokopenko I, Magi R, Keskitalo K, Gudjonsdottir IH, Gretarsdottir S, Stefansson H, Thompson JR, Aulchenko YS, Nelis M, Aben KK, den Heijer M, Dirksen A, Ashraf H, Soranzo N, Valdes AM, Steves C, Uitterlinden AG, Hofman A, Tonjes A, Kovacs P, Hottenga JJ, Willemsen G, Vogelzangs N, Doring A, Dahmen N, Nitz B, Pergadia ML, Saez B, De Diego V, Lezcano V, Garcia-Prats MD, Ripatti S, Perola M, Kettunen J, Hartikainen AL, Pouta A, Laitinen J, Isohanni M, Huei-Yi S, Allen M, Krestyaninova M, Hall AS, Jones GT, van Rij AM, Mueller T, Dieplinger B, Haltmayer M, Jonsson S, Matthiasson SE, Oskarsson H, Tyrfingsson T, Kiemeney LA, Mayordomo JI, Lindholt JS, Pedersen JH, Franklin WA, Wolf H, Montgomery GW, Heath AC, Martin NG, Madden PA, Giegling I, Rujescu D, Jarvelin MR, Salomaa V, Stumvoll M, Spector TD, Wichmann HE, Metspalu A, Samani NJ, Penninx BW, Oostra BA, Boomsma DI, Tiemeier H, van Duijn CM, Kaprio J, Gulcher JR, McCarthy MI, Peltonen L, Thorsteinsdottir U, Stefansson K. Sequence variants at CHRNB3-CHRNA6 and CYP2A6 affect smoking behavior. Nat Genet. 2010;42:448–453. doi: 10.1038/ng.573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Amos CI, Wu X, Broderick P, Gorlov IP, Gu J, Eisen T, Dong Q, Zhang Q, Gu X, Vijayakrishnan J, Sullivan K, Matakidou A, Wang Y, Mills G, Doheny K, Tsai YY, Chen WV, Shete S, Spitz MR, Houlston RS. Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q25.1. Nature Genetics. 2008;40:616–622. doi: 10.1038/ng.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Thorgeirsson TE, Geller F, Sulem P, Rafnar T, Wiste A, Magnusson KP, Manolescu A, Thorleifsson G, Stefansson H, Ingason A, Stacey SN, Bergthorsson JT, Thorlacius S, Gudmundsson J, Jonsson T, Jakobsdottir M, Saemundsdottir J, Olafsdottir O, Gudmundsson LJ, Bjornsdottir G, Kristjansson K, Skuladottir H, Isaksson HJ, Gudbjartsson T, Jones GT, Mueller T, Gottsater A, Flex A, Aben KKH, de Vegt F, Mulders PFA, Isla D, Vidal MJ, Asin L, Saez B, Murillo L, Blondal T, Kolbeinsson H, Stefansson JG, Hansdottir I, Runarsdottir V, Pola R, Lindblad B, van Rij AM, Dieplinger B, Haltmayer M, Mayordomo JI, Kiemeney LA, Matthiasson SE, Oskarsson H, Tyrfingsson T, Gudbjartsson DF, Gulcher JR, Jonsson S, Thorsteinsdottir U, Kong A, Stefansson K. A variant associated with nicotine dependence, lung cancer and peripheral arterial disease. Nature. 2008;452:638–642. doi: 10.1038/nature06846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Pillai SG, Ge D, Zhu G, Kong X, Shianna KV, Need AC, Feng S, Hersh CP, Bakke P, Gulsvik A, Ruppert A, Lodrup Carlsen KC, Roses A, Anderson W, Rennard SI, Lomas DA, Silverman EK, Goldstein DB. A genome-wide association study in chronic obstructive pulmonary disease (COPD): identification of two major susceptibility loci. PLoS Genet. 2009;5:e1000421. doi: 10.1371/journal.pgen.1000421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chen LS, Hung RJ, Baker T, Horton A, Culverhouse R, Saccone N, Cheng I, Deng B, Han Y, Hansen HM, Horsman J, Kim C, Lutz S, Rosenberger A, Aben KK, Andrew AS, Breslau N, Chang SC, Dieffenbach AK, Dienemann H, Frederiksen B, Han J, Hatsukami DK, Johnson EO, Pande M, Wrensch MR, McLaughlin J, Skaug V, van der Heijden HF, Wampfler J, Wenzlaff A, Woll P, Zienolddiny S, Bickeboller H, Brenner H, Duell EJ, Haugen A, Heinrich J, Hokanson JE, Hunter DJ, Kiemeney LA, Lazarus P, Le Marchand L, Liu G, Mayordomo J, Risch A, Schwartz AG, Teare D, Wu X, Wiencke JK, Yang P, Zhang ZF, Spitz MR, Kraft P, Amos CI, Bierut LJ. CHRNA5 Risk Variant Predicts Delayed Smoking Cessation and Earlier Lung Cancer Diagnosis-A Meta-Analysis. J Natl Cancer Inst. 2015;107 doi: 10.1093/jnci/djv100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chen LS, Baker TB, Piper ME, Breslau N, Cannon DS, Doheny KF, Gogarten SM, Johnson EO, Saccone NL, Wang JC, Weiss RB, Goate AM, Bierut LJ. Interplay of genetic risk factors (CHRNA5-CHRNA3-CHRNB4) and cessation treatments in smoking cessation success. Am J Psychiatry. 2012;169:735–742. doi: 10.1176/appi.ajp.2012.11101545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bergen AW, Javitz HS, Krasnow R, Nishita D, Michel M, Conti DV, Liu J, Lee W, Edlund CK, Hall S, Kwok PY, Benowitz NL, Baker TB, Tyndale RF, Lerman C, Swan GE. Nicotinic acetylcholine receptor variation and response to smoking cessation therapies. Pharmacogenet Genomics. 2013;23:94–103. doi: 10.1097/FPC.0b013e32835cdabd. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.David SP. Commentary on Chen et al. (2014): another step on the road to clinical utility of pharmacogenetics for smoking cessation? Addiction. 2014;109:138–139. doi: 10.1111/add.12390. [DOI] [PubMed] [Google Scholar]
- 30.Bough KJ, Lerman C, Rose JE, McClernon FJ, Kenny PJ, Tyndale RF, David SP, Stein EA, Uhl GR, Conti DV, Green C, Amur S. Biomarkers for Smoking Cessation. Clin Pharmacol Ther. 2013 doi: 10.1038/clpt.2013.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sturgess JE, George TP, Kennedy JL, Heinz A, Muller DJ. Pharmacogenetics of alcohol, nicotine and drug addiction treatments. Addict Biol. 2011;16:357–376. doi: 10.1111/j.1369-1600.2010.00287.x. [DOI] [PubMed] [Google Scholar]
- 32.Furberg H, Ostroff J, Lerman C, Sullivan PF. The public health utility of genome-wide association study results for smoking behavior. Genome Medicine. 2010;2 doi: 10.1186/gm147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Weiss RB, Baker TB, Cannon DS, von Niederhausern A, Dunn DM, Matsunami N, Singh NA, Baird L, Coon H, McMahon WM, Piper ME, Fiore MC, Scholand MB, Connett JE, Kanner RE, Gahring LC, Rogers SW, Hoidal JR, Leppert MF. A candidate gene approach identifies the CHRNA5-A3-B4 region as a risk factor for age-dependent nicotine addiction. PLoS Genet. 2008;4:e1000125. doi: 10.1371/journal.pgen.1000125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bierut LJ, Stitzel JA, Wang JC, Hinrichs AL, Grucza RA, Xuei X, Saccone NL, Saccone SF, Bertelsen S, Fox L, Horton WJ, Breslau N, Budde J, Cloninger CR, Dick DM, Foroud T, Hatsukami D, Hesselbrock V, Johnson EO, Kramer J, Kuperman S, Madden PA, Mayo K, Nurnberger J, Jr, Pomerleau O, Porjesz B, Reyes O, Schuckit M, Swan G, Tischfield JA, Edenberg HJ, Rice JP, Goate AM. Variants in nicotinic receptors and risk for nicotine dependence. Am J Psychiatry. 2008;165:1163–1171. doi: 10.1176/appi.ajp.2008.07111711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kuryatov A, Berrettini W, Lindstrom J. Acetylcholine receptor (AChR) alpha5 subunit variant associated with risk for nicotine dependence and lung cancer reduces (alpha4beta2)alpha5 AChR function. Mol Pharmacol. 2011;79:119–125. doi: 10.1124/mol.110.066357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wang JC, Cruchaga C, Saccone NL, Bertelsen S, Liu P, Budde JP, Duan W, Fox L, Grucza RA, Kern J, Mayo K, Reyes O, Rice J, Saccone SF, Spiegel N, Steinbach JH, Stitzel JA, Anderson MW, You M, Stevens VL, Bierut LJ, Goate AM. Risk for nicotine dependence and lung cancer is conferred by mRNA expression levels and amino acid change in CHRNA5. Hum Mol Genet. 2009;18:3125–3135. doi: 10.1093/hmg/ddp231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Salas R, Sturm R, Boulter J, De Biasi M. Nicotinic receptors in the habenulo-interpeduncular system are necessary for nicotine withdrawal in mice. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2009;29:3014–3018. doi: 10.1523/JNEUROSCI.4934-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Fowler CD, Lu Q, Johnson PM, Marks MJ, Kenny PJ. Habenular alpha5 nicotinic receptor subunit signalling controls nicotine intake. Nature. 2011;471:597–601. doi: 10.1038/nature09797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Robinson TE, Berridge KC. The psychology and neurobiology of addiction: an incentive-sensitization view. Addiction. 2000;95(Suppl 2):S91–S117. doi: 10.1080/09652140050111681. [DOI] [PubMed] [Google Scholar]
- 40.Chen LS, Saccone NL, Culverhouse RC, Bracci PM, Chen CH, Dueker N, Han Y, Huang H, Jin G, Kohno T, Ma JZ, Przybeck TR, Sanders AR, Smith JA, Sung YJ, Wenzlaff AS, Wu C, Yoon D, Chen YT, Cheng YC, Cho YS, David SP, Duan J, Eaton CB, Furberg H, Goate AM, Gu D, Hansen HM, Hartz S, Hu Z, Kim YJ, Kittner SJ, Levinson DF, Mosley TH, Payne TJ, Rao DC, Rice JP, Rice TK, Schwantes-An TH, Shete SS, Shi J, Spitz MR, Sun YV, Tsai FJ, Wang JC, Wrensch MR, Xian H, Gejman PV, He J, Hunt SC, Kardia SL, Li MD, Lin D, Mitchell BD, Park T, Schwartz AG, Shen H, Wiencke JK, Wu JY, Yokota J, Amos CI, Bierut LJ. Smoking and genetic risk variation across populations of European, Asian, and African American ancestry--a meta-analysis of chromosome 15q25. Genet Epidemiol. 2012;36:340–351. doi: 10.1002/gepi.21627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.David SP, Hamidovic A, Chen GK, Bergen AW, Wessel J, Kasberger JL, Brown WM, Petruzella S, Thacker EL, Kim Y, Nalls MA, Tranah GJ, Sung YJ, Ambrosone CB, Arnett D, Bandera EV, Becker DM, Becker L, Berndt SI, Bernstein L, Blot WJ, Broeckel U, Buxbaum SG, Caporaso N, Casey G, Chanock SJ, Deming SL, Diver WR, Eaton CB, Evans DS, Evans MK, Fornage M, Franceschini N, Harris TB, Henderson BE, Hernandez DG, Hitsman B, Hu JJ, Hunt SC, Ingles SA, John EM, Kittles R, Kolb S, Kolonel LN, Le Marchand L, Liu Y, Lohman KK, McKnight B, Millikan RC, Murphy A, Neslund-Dudas C, Nyante S, Press M, Psaty BM, Rao DC, Redline S, Rodriguez-Gil JL, Rybicki BA, Signorello LB, Singleton AB, Smoller J, Snively B, Spring B, Stanford JL, Strom SS, Swan GE, Taylor KD, Thun MJ, Wilson AF, Witte JS, Yamamura Y, Yanek LR, Yu K, Zheng W, Ziegler RG, Zonderman AB, Jorgenson E, Haiman CA, Furberg H. Genome-wide meta-analyses of smoking behaviors in African Americans. Transl Psychiatry. 2012;2:e119. doi: 10.1038/tp.2012.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hancock DB, Reginsson GW, Gaddis NC, Chen X, Saccone NL, Lutz SM, Qaiser B, Sherva R, Steinberg S, Zink F, Stacey SN, Glasheen C, Chen J, Gu F, Frederiksen BN, Loukola A, Gudbjartsson DF, Bruske I, Landi MT, Bickeboller H, Madden P, Farrer L, Kaprio J, Kranzler HR, Gelernter J, Baker TB, Kraft P, Amos CI, Caporaso NE, Hokanson JE, Bierut LJ, Thorgeirsson TE, Johnson EO, Stefansson K. Genome-wide meta-analysis reveals common splice site acceptor variant in CHRNA4 associated with nicotine dependence. Transl Psychiatry. 2015;5:e651. doi: 10.1038/tp.2015.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bloom AJ, Hartz SM, Baker TB, Chen LS, Piper ME, Fox L, Martinez M, Hatsukami D, Johnson EO, Laurie CC, Saccone NL, Goate A, Bierut LJ. Beyond cigarettes per day. A genome-wide association study of the biomarker carbon monoxide. Ann Am Thorac Soc. 2014;11:1003–1010. doi: 10.1513/AnnalsATS.201401-010OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ware JJ, Chen X, Vink J, Loukola A, Minica C, Pool R, Milaneschi Y, Mangino M, Menni C, Chen J, Peterson RE, Auro K, Lyytikainen LP, Wedenoja J, Stiby AI, Hemani G, Willemsen G, Hottenga JJ, Korhonen T, Heliovaara M, Perola M, Rose RJ, Paternoster L, Timpson N, Wassenaar CA, Zhu AZ, Davey Smith G, O TR, Lehtimaki T, Kahonen M, Koskinen S, Spector T, Penninx BW, Salomaa V, Boomsma DI, Tyndale RF, Kaprio J, Munafo MR. Genome-Wide Meta-Analysis of Cotinine Levels in Cigarette Smokers Identifies Locus at 4q13.2. Sci Rep. 2016;6:20092. doi: 10.1038/srep20092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chen LS, Baker TB, Grucza R, Wang JC, Johnson EO, Breslau N, Hatsukami D, Smith SS, Saccone N, Saccone S, Rice JP, Goate AM, Bierut LJ. Dissection of the phenotypic and genotypic associations with nicotinic dependence. Nicotine Tob Res. 2012;14:425–433. doi: 10.1093/ntr/ntr231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bousman CA, Rivard C, Haese JD, Ambrosone C, Hyland A. Alpha-5 and-3 nicotinic receptor gene variants predict nicotine dependence but not cessation: findings from the COMMIT cohort. Am J Med Genet B Neuropsychiatr Genet. 2012;159B:227–235. doi: 10.1002/ajmg.b.32019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Cannon DS, Mermelstein RJ, Hedeker D, Coon H, Cook EH, McMahon WM, Hamil C, Dunn D, Weiss RB. Effect of neuronal nicotinic acetylcholine receptor genes (CHRN) on longitudinal cigarettes per day in adolescents and young adults. Nicotine Tob Res. 2014;16:137–144. doi: 10.1093/ntr/ntt125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lazary J, Dome P, Csala I, Kovacs G, Faludi G, Kaunisto M, Dome B. Massive withdrawal symptoms and affective vulnerability are associated with variants of the CHRNA4 gene in a subgroup of smokers. PLoS One. 2014;9:e87141. doi: 10.1371/journal.pone.0087141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bergen AW, Michel M, Nishita D, Krasnow R, Javitz HS, Conneely KN, Lessov-Schlaggar CN, Hops H, Zhu AZ, Baurley JW, McClure JB, Hall SM, Baker TB, Conti DV, Benowitz NL, Lerman C, Tyndale RF, Swan T GE. Transdisciplinary Research in Cancer of the Lung Research, Drug Metabolizing Enzyme and Transporter Gene Variation, Nicotine Metabolism, Prospective Abstinence, and Cigarette Consumption. PLoS One. 2015;10:e0126113. doi: 10.1371/journal.pone.0126113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Park SL, Tiirikainen MI, Patel YM, Wilkens LR, Stram DO, Le Marchand L, Murphy SE. Genetic determinants of CYP2A6 activity across racial/ethnic groups with different risks of lung cancer and effect on their smoking intensity. Carcinogenesis. 2016;37:269–279. doi: 10.1093/carcin/bgw012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wassenaar CA, Ye Y, Cai Q, Aldrich MC, Knight J, Spitz MR, Wu X, Blot WJ, Tyndale RF. CYP2A6 reduced activity gene variants confer reduction in lung cancer risk in African American smokers--findings from two independent populations. Carcinogenesis. 2015;36:99–103. doi: 10.1093/carcin/bgu235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lutz SM, Cho MH, Young K, Hersh CP, Castaldi PJ, McDonald ML, Regan E, Mattheisen M, DeMeo DL, Parker M, Foreman M, Make BJ, Jensen RL, Casaburi R, Lomas DA, Bhatt SP, Bakke P, Gulsvik A, Crapo JD, Beaty TH, Laird NM, Lange C, Hokanson JE, Silverman EK, Investigators E, Investigators CO. A genome-wide association study identifies risk loci for spirometric measures among smokers of European and African ancestry. BMC Genet. 2015;16:138. doi: 10.1186/s12863-015-0299-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lerman C, Jepson C, Wileyto EP, Patterson F, Schnoll R, Mroziewicz M, Benowitz N, Tyndale RF. Genetic variation in nicotine metabolism predicts the efficacy of extended-duration transdermal nicotine therapy. Clin Pharmacol Ther. 2010;87:553–557. doi: 10.1038/clpt.2010.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Patterson F, Schnoll RA, Wileyto EP, Pinto A, Epstein LH, Shields PG, Hawk LW, Tyndale RF, Benowitz N, Lerman C. Toward personalized therapy for smoking cessation: a randomized placebo-controlled trial of bupropion. Clin Pharmacol Ther. 2008;84:320–325. doi: 10.1038/clpt.2008.57. [DOI] [PubMed] [Google Scholar]
- 55.Schnoll RA, Patterson F, Wileyto EP, Tyndale RF, Benowitz N, Lerman C. Nicotine metabolic rate predicts successful smoking cessation with transdermal nicotine: a validation study. Pharmacol Biochem Behav. 2009;92:6–11. doi: 10.1016/j.pbb.2008.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Chen LS, Bloom AJ, Baker TB, Smith SS, Piper ME, Martinez M, Saccone N, Hatsukami D, Goate A, Bierut L. Pharmacotherapy effects on smoking cessation vary with nicotine metabolism gene (CYP2A6) Addiction. 2014;109:128–137. doi: 10.1111/add.12353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Falcone M, Jepson C, Benowitz N, Bergen AW, Pinto A, Wileyto EP, Baldwin D, Tyndale RF, Lerman C, Ray R. Association of the nicotine metabolite ratio and CHRNA5/CHRNA3 polymorphisms with smoking rate among treatment-seeking smokers. Nicotine Tob Res. 2011;13:498–503. doi: 10.1093/ntr/ntr012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Loukola A, Buchwald J, Gupta R, Palviainen T, Hallfors J, Tikkanen E, Korhonen T, Ollikainen M, Sarin AP, Ripatti S, Lehtimaki T, Raitakari O, Salomaa V, Rose RJ, Tyndale RF, Kaprio J. A Genome-Wide Association Study of a Biomarker of Nicotine Metabolism. PLoS Genet. 2015;11:e1005498. doi: 10.1371/journal.pgen.1005498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Baker TB, Weiss RB, Bolt D, von Niederhausern A, Fiore MC, Dunn DM, Piper ME, Matsunami N, Smith SS, Coon H, McMahon WM, Scholand MB, Singh N, Hoidal JR, Kim SY, Leppert MF, Cannon DS. Human neuronal acetylcholine receptor A5-A3-B4 haplotypes are associated with multiple nicotine dependence phenotypes. Nicotine Tob Res. 2009;11:785–796. doi: 10.1093/ntr/ntp064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Freathy RM, Ring SM, Shields B, Galobardes B, Knight B, Weedon MN, Smith GD, Frayling TM, Hattersley AT. A common genetic variant in the 15q24 nicotinic acetylcholine receptor gene cluster (CHRNA5-CHRNA3-CHRNB4) is associated with a reduced ability of women to quit smoking in pregnancy. Hum Mol Genet. 2009;18:2922–2927. doi: 10.1093/hmg/ddp216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Munafo MR, Johnstone EC, Walther D, Uhl GR, Murphy MF, Aveyard P. CHRNA3 rs1051730 Genotype and Short-Term Smoking Cessation. Nicotine Tob Res. 2011;13:982–988. doi: 10.1093/ntr/ntr106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sarginson JE, Killen JD, Lazzeroni LC, Fortmann SP, Ryan HS, Schatzberg AF, Murphy GM., Jr Markers in the 15q24 nicotinic receptor subunit gene cluster (CHRNA5-A3-B4) predict severity of nicotine addiction and response to smoking cessation therapy. Am J Med Genet B Neuropsychiatr Genet. 2011;156B:275–284. doi: 10.1002/ajmg.b.31155. [DOI] [PubMed] [Google Scholar]
- 63.King DP, Paciga S, Pickering E, Benowitz NL, Bierut LJ, Conti DV, Kaprio J, Lerman C, Park PW. Smoking cessation pharmacogenetics: analysis of varenicline and bupropion in placebo-controlled clinical trials. Neuropsychopharmacology. 2012;37:641–650. doi: 10.1038/npp.2011.232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhu AZ, Zhou Q, Cox LS, David SP, Ahluwalia JS, Benowitz NL, Tyndale RF. Association of CHRNA5-A3-B4 SNP rs2036527 With Smoking Cessation Therapy Response in African-American Smokers. Clin Pharmacol Ther. 2014 doi: 10.1038/clpt.2014.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Breitling LP, Twardella D, Hoffmann MM, Witt SH, Treutlein J, Brenner H. Prospective association of dopamine-related polymorphisms with smoking cessation in general care. Pharmacogenomics. 2010;11:527–536. doi: 10.2217/pgs.10.1. [DOI] [PubMed] [Google Scholar]
- 66.Conti DV, Lee W, Li D, Liu J, Van Den Berg D, Thomas PD, Bergen AW, Swan GE, Tyndale RF, Benowitz NL, Lerman C. Nicotinic acetylcholine receptor beta2 subunit gene implicated in a systems-based candidate gene study of smoking cessation. Hum Mol Genet. 2008;17:2834–2848. doi: 10.1093/hmg/ddn181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Uhl GR, Liu QR, Drgon T, Johnson C, Walther D, Rose JE, David SP, Niaura R, Lerman C. Molecular genetics of successful smoking cessation: convergent genome-wide association study results. Arch Gen Psychiatry. 2008;65:683–693. doi: 10.1001/archpsyc.65.6.683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Tyndale RF, Zhu AZ, George TP, Cinciripini P, Hawk LW, Jr, Schnoll RA, Swan GE, Benowitz NL, Heitjan DF, Lerman C. Lack of Associations of CHRNA5-A3-B4 Genetic Variants with Smoking Cessation Treatment Outcomes in Caucasian Smokers despite Associations with Baseline Smoking. PLoS One. 2015;10:e0128109. doi: 10.1371/journal.pone.0128109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Spitz MR, Amos CI, Dong Q, Lin J, Wu X. The CHRNA5-A3 region on chromosome 15q24-25.1 is a risk factor both for nicotine dependence and for lung cancer. J Natl Cancer Inst. 2008;100:1552–1556. doi: 10.1093/jnci/djn363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Tseng TS, Park JY, Zabaleta J, Moody-Thomas S, Sothern MS, Chen T, Evans DE, Lin HY. Role of nicotine dependence on the relationship between variants in the nicotinic receptor genes and risk of lung adenocarcinoma. PLoS One. 2014;9:e107268. doi: 10.1371/journal.pone.0107268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lips EH, Gaborieau V, McKay JD, Chabrier A, Hung RJ, Boffetta P, Hashibe M, Zaridze D, Szeszenia-Dabrowska N, Lissowska J, Rudnai P, Fabianova E, Mates D, Bencko V, Foretova L, Janout V, Field JK, Liloglou T, Xinarianos G, McLaughlin J, Liu G, Skorpen F, Elvestad MB, Hveem K, Vatten L, Study E, Benhamou S, Lagiou P, Holcatova I, Merletti F, Kjaerheim K, Agudo A, Castellsague X, Macfarlane TV, Barzan L, Canova C, Lowry R, Conway DI, Znaor A, Healy C, Curado MP, Koifman S, Eluf-Neto J, Matos E, Menezes A, Fernandez L, Metspalu A, Heath S, Lathrop M, Brennan P. Association between a 15q25 gene variant, smoking quantity and tobacco-related cancers among 17000 individuals. Int J Epidemiol. 2010;39:563–577. doi: 10.1093/ije/dyp288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Cho MH, McDonald ML, Zhou X, Mattheisen M, Castaldi PJ, Hersh CP, Demeo DL, Sylvia JS, Ziniti J, Laird NM, Lange C, Litonjua AA, Sparrow D, Casaburi R, Barr RG, Regan EA, Make BJ, Hokanson JE, Lutz S, Dudenkov TM, Farzadegan H, Hetmanski JB, Tal-Singer R, Lomas DA, Bakke P, Gulsvik A, Crapo JD, Silverman EK, Beaty TH. Risk loci for chronic obstructive pulmonary disease: a genome-wide association study and meta-analysis. Lancet Respir Med. 2014;2:214–225. doi: 10.1016/S2213-2600(14)70002-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.A genome-wide association study in Europeans and South Asians identifies five new loci for coronary artery disease. Nat Genet. 2011;43:339–344. doi: 10.1038/ng.782. [DOI] [PubMed] [Google Scholar]
- 74.Schunkert H, Konig IR, Kathiresan S, Reilly MP, Assimes TL, Holm H, Preuss M, Stewart AF, Barbalic M, Gieger C, Absher D, Aherrahrou Z, Allayee H, Altshuler D, Anand SS, Andersen K, Anderson JL, Ardissino D, Ball SG, Balmforth AJ, Barnes TA, Becker DM, Becker LC, Berger K, Bis JC, Boekholdt SM, Boerwinkle E, Braund PS, Brown MJ, Burnett MS, Buysschaert I, Carlquist JF, Chen L, Cichon S, Codd V, Davies RW, Dedoussis G, Dehghan A, Demissie S, Devaney JM, Diemert P, Do R, Doering A, Eifert S, Mokhtari NE, Ellis SG, Elosua R, Engert JC, Epstein SE, de Faire U, Fischer M, Folsom AR, Freyer J, Gigante B, Girelli D, Gretarsdottir S, Gudnason V, Gulcher JR, Halperin E, Hammond N, Hazen SL, Hofman A, Horne BD, Illig T, Iribarren C, Jones GT, Jukema JW, Kaiser MA, Kaplan LM, Kastelein JJ, Khaw KT, Knowles JW, Kolovou G, Kong A, Laaksonen R, Lambrechts D, Leander K, Lettre G, Li M, Lieb W, Loley C, Lotery AJ, Mannucci PM, Maouche S, Martinelli N, McKeown PP, Meisinger C, Meitinger T, Melander O, Merlini PA, Mooser V, Morgan T, Muhleisen TW, Muhlestein JB, Munzel T, Musunuru K, Nahrstaedt J, Nelson CP, Nothen MM, Olivieri O, Patel RS, Patterson CC, Peters A, Peyvandi F, Qu L, Quyyumi AA, Rader DJ, Rallidis LS, Rice C, Rosendaal FR, Rubin D, Salomaa V, Sampietro ML, Sandhu MS, Schadt E, Schafer A, Schillert A, Schreiber S, Schrezenmeir J, Schwartz SM, Siscovick DS, Sivananthan M, Sivapalaratnam S, Smith A, Smith TB, Snoep JD, Soranzo N, Spertus JA, Stark K, Stirrups K, Stoll M, Tang WH, Tennstedt S, Thorgeirsson G, Thorleifsson G, Tomaszewski M, Uitterlinden AG, van Rij AM, Voight BF, Wareham NJ, Wells GA, Wichmann HE, Wild PS, Willenborg C, Witteman JC, Wright BJ, Ye S, Zeller T, Ziegler A, Cambien F, Goodall AH, Cupples LA, Quertermous T, Marz W, Hengstenberg C, Blankenberg S, Ouwehand WH, Hall AS, Deloukas P, Thompson JR, Stefansson K, Roberts R, Thorsteinsdottir U, O'Donnell CJ, McPherson R, Erdmann J, Samani NJ. Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nat Genet. 2011;43:333–338. doi: 10.1038/ng.784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kupiainen H, Kuokkanen M, Kontto J, Virtamo J, Salomaa V, Lindqvist A, Kilpelainen M, Laitinen T. CHRNA5/CHRNA3 Locus Associates with Increased Mortality among Smokers. Copd. 2016:1–7. doi: 10.3109/15412555.2015.1049260. [DOI] [PubMed] [Google Scholar]
- 76.Hallden S, Sjogren M, Hedblad B, Engstrom G, Hamrefors V, Manjer J, Melander O. Gene variance in the nicotinic receptor cluster (CHRNA5-CHRNA3-CHRNB4) predicts death from cardiopulmonary disease and cancer in smokers. Journal of internal medicine. 2015 doi: 10.1111/joim.12454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Chen LS, Bach RG, Lenzini PA, Spertus JA, Bierut LJ, Cresci S. CHRNA5 Variant Predicts Smoking Cessation in Patients With Acute Myocardial Infarction. Nicotine Tob Res. 2014 doi: 10.1093/ntr/ntu059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bloom AJ, Harari O, Martinez M, Madden PA, Martin NG, Montgomery GW, Rice JP, Murphy SE, Bierut LJ, Goate A. Use of a predictive model derived from in vivo endophenotype measurements to demonstrate associations with a complex locus, CYP2A6. Hum Mol Genet. 2012;21:3050–3062. doi: 10.1093/hmg/dds114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Fujieda M, Yamazaki H, Saito T, Kiyotani K, Gyamfi MA, Sakurai M, Dosaka-Akita H, Sawamura Y, Yokota J, Kunitoh H, Kamataki T. Evaluation of CYP2A6 genetic polymorphisms as determinants of smoking behavior and tobacco-related lung cancer risk in male Japanese smokers. Carcinogenesis. 2004;25:2451–2458. doi: 10.1093/carcin/bgh258. [DOI] [PubMed] [Google Scholar]
- 80.Schoedel KA, Hoffmann EB, Rao Y, Sellers EM, Tyndale RF. Ethnic variation in CYP2A6 and association of genetically slow nicotine metabolism and smoking in adult Caucasians. Pharmacogenetics. 2004;14:615–626. doi: 10.1097/00008571-200409000-00006. [DOI] [PubMed] [Google Scholar]
- 81.Ho MK, Mwenifumbo JC, Al Koudsi N, Okuyemi KS, Ahluwalia JS, Benowitz NL, Tyndale RF. Association of nicotine metabolite ratio and CYP2A6 genotype with smoking cessation treatment in African-American light smokers. Clin Pharmacol Ther. 2009;85:635–643. doi: 10.1038/clpt.2009.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kaufmann A, Hitsman B, Goelz PM, Veluz-Wilkins A, Blazekovic S, Powers L, Leone FT, Gariti P, Tyndale RF, Schnoll RA. Rate of nicotine metabolism and smoking cessation outcomes in a community-based sample of treatment-seeking smokers. Addict Behav. 2015;51:93–99. doi: 10.1016/j.addbeh.2015.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Chen LS, Baker TB, Piper ME, Smith SS, Gu C, Grucza RA, Smith GD, Munafo M, Bierut LJ. Interplay of genetic risk (CHRNA5) and environmental risk (partner smoking) on cigarette smoking reduction. Drug Alcohol Depend. 2014;143:36–43. doi: 10.1016/j.drugalcdep.2014.06.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Sarginson JE, Killen JD, Lazzeroni LC, Fortmann SP, Ryan HS, Ameli N, Schatzberg AF, Murphy GM., Jr Response to Transdermal Selegiline Smoking Cessation Therapy and Markers in the 15q24 Chromosomal Region. Nicotine Tob Res. 2015 doi: 10.1093/ntr/ntu273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Chen LS, Baker TB, Bierut LJ. Response to Kaufman and Harper letter. Am J Psychiatry. 2012;169:1118–1119. doi: 10.1176/appi.ajp.2012.12060800r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Matsuzaki M, Yokoyama M, Saito Y, Origasa H, Ishikawa Y, Oikawa S, Sasaki J, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata T, Shimada K, Shirato K, Matsuzawa Y. Incremental effects of eicosapentaenoic acid on cardiovascular events in statin-treated patients with coronary artery disease. Circ J. 2009;73:1283–1290. doi: 10.1253/circj.cj-08-1197. [DOI] [PubMed] [Google Scholar]
- 87.Suissa S. Number needed to treat in COPD: exacerbations versus pneumonias. Thorax. 2013;68:540–543. doi: 10.1136/thoraxjnl-2012-202709. [DOI] [PubMed] [Google Scholar]
- 88.Young J, De Sutter A, Merenstein D, van Essen GA, Kaiser L, Varonen H, Williamson I, Bucher HC. Antibiotics for adults with clinically diagnosed acute rhinosinusitis: a meta-analysis of individual patient data. Lancet. 2008;371:908–914. doi: 10.1016/S0140-6736(08)60416-X. [DOI] [PubMed] [Google Scholar]
- 89.Leung T, Bergen A, Munafo MR, De Ruyck K, Selby P, De Luca V. Effect of the rs1051730-rs16969968 variant and smoking cessation treatment: a meta-analysis. Pharmacogenomics. 2015;16:713–720. doi: 10.2217/pgs.15.34. [DOI] [PubMed] [Google Scholar]
- 90.Chen LS, Baker TB, Jorenby D, Piper M, Saccone N, Johnson E, Breslau N, Hatsukami D, Carney RM, Bierut LJ. Genetic variation (CHRNA5), medication (combination nicotine replacement therapy vs. varenicline), and smoking cessation. Drug Alcohol Depend. 2015;154:278–282. doi: 10.1016/j.drugalcdep.2015.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Chen LS, Baker TB, Bierut LJ. The value of control conditions for evaluating pharmacogenetic effects. Pharmacogenomics. 2015;16:2005–2006. doi: 10.2217/pgs.15.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ware JJ, Aveyard P, Broderick P, Houlston RS, Eisen T, Munafo MR. The association of rs1051730 genotype on adherence to and consumption of prescribed nicotine replacement therapy dose during a smoking cessation attempt. Drug Alcohol Depend. 2015;151:236–240. doi: 10.1016/j.drugalcdep.2015.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Benowitz NL, Pomerleau OF, Pomerleau CS, Jacob P., 3rd Nicotine metabolite ratio as a predictor of cigarette consumption. Nicotine Tob Res. 2003;5:621–624. doi: 10.1080/1462220031000158717. [DOI] [PubMed] [Google Scholar]
- 94.International Schizophrenia C, Purcell SM, Wray NR, Stone JL, Visscher PM, O'Donovan MC, Sullivan PF, Sklar P. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature. 2009;460:748–752. doi: 10.1038/nature08185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Haberl M, Anwald B, Klein K, Weil R, Fuss C, Gepdiremen A, Zanger UM, Meyer UA, Wojnowski L. Three haplotypes associated with CYP2A6 phenotypes in Caucasians. Pharmacogenet Genomics. 2005;15:609–624. doi: 10.1097/01.fpc.0000171517.22258.f1. [DOI] [PubMed] [Google Scholar]
- 96.Bloom J, Hinrichs AL, Wang JC, von Weymarn LB, Kharasch ED, Bierut LJ, Goate A, Murphy SE. The contribution of common CYP2A6 alleles to variation in nicotine metabolism among European-Americans. Pharmacogenet Genomics. 2011;21:403–416. doi: 10.1097/FPC.0b013e328346e8c0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lerman C, Schnoll RA, Hawk LW, Jr, Cinciripini P, George TP, Wileyto EP, Swan GE, Benowitz NL, Heitjan DF, Tyndale RF. Use of the nicotine metabolite ratio as a genetically informed biomarker of response to nicotine patch or varenicline for smoking cessation: a randomised, double-blind placebo-controlled trial. Lancet Respir Med. 2015;3:131–138. doi: 10.1016/S2213-2600(14)70294-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Tomaz PR, Santos JR, Issa JS, Abe TO, Gaya PV, Krieger JE, Pereira AC, Santos PC. CYP2B6 rs2279343 polymorphism is associated with smoking cessation success in bupropion therapy. Eur J Clin Pharmacol. 2015;71:1067–1073. doi: 10.1007/s00228-015-1896-x. [DOI] [PubMed] [Google Scholar]
- 99.Choi HD, Shin WG. Association between catechol-O-methyltransferase (COMT) Val/Met genotype and smoking cessation treatment with nicotine: a meta-analysis. Pharmacogenomics. 2015;16:1879–1885. doi: 10.2217/pgs.15.127. [DOI] [PubMed] [Google Scholar]
- 100.Johnstone EC, Yudkin PL, Hey K, Roberts SJ, Welch SJ, Murphy MF, Griffiths SE, Walton RT. Genetic variation in dopaminergic pathways and short-term effectiveness of the nicotine patch. Pharmacogenetics. 2004;14:83–90. doi: 10.1097/00008571-200402000-00002. [DOI] [PubMed] [Google Scholar]
- 101.Berrettini WH, Wileyto EP, Epstein L, Restine S, Hawk L, Shields P, Niaura R, Lerman C. Catechol-O-methyltransferase (COMT) gene variants predict response to bupropion therapy for tobacco dependence. Biol Psychiatry. 2007;61:111–118. doi: 10.1016/j.biopsych.2006.04.030. [DOI] [PubMed] [Google Scholar]
- 102.David SP, Johnstone EC, Churchman M, Aveyard P, Murphy MF, Munafo MR. Pharmacogenetics of smoking cessation in general practice: results from the patch II and patch in practice trials. Nicotine Tob Res. 2011;13:157–167. doi: 10.1093/ntr/ntq246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Leventhal AM, David SP, Brightman M, Strong D, McGeary JE, Brown RA, Lloyd-Richardson EE, Munafo M, Uhl GR, Niaura R. Dopamine D4 receptor gene variation moderates the efficacy of bupropion for smoking cessation. Pharmacogenomics J. 2012;12:86–92. doi: 10.1038/tpj.2010.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Herman AI, DeVito EE, Jensen KP, Sofuoglu M. Pharmacogenetics of nicotine addiction: role of dopamine. Pharmacogenomics. 2014;15:221–234. doi: 10.2217/pgs.13.246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Choi HD, Shin WG. Lack of association between DRD2 Taq1A gene polymorphism and smoking cessation therapy: a meta-analysis. Int J Clin Pharmacol Ther. 2015;53:415–421. doi: 10.5414/CP202214. [DOI] [PubMed] [Google Scholar]
- 106.Verde Z, Santiago C, Rodriguez Gonzalez-Moro JM, de Lucas Ramos P, Lopez Martin S, Bandres F, Lucia A, Gomez-Gallego F. Are serotonergic system genes associated to smoking cessation therapy success in addition to CYP2A6? Pharmacopsychiatry. 2014;47:33–36. doi: 10.1055/s-0033-1358393. [DOI] [PubMed] [Google Scholar]
- 107.David SP, Munafo MR. Genetic variation in the dopamine pathway and smoking cessation. Pharmacogenomics. 2008;9:1307–1321. doi: 10.2217/14622416.9.9.1307. [DOI] [PubMed] [Google Scholar]
- 108.David SP, Strong DR, Leventhal AM, Lancaster MA, McGeary JE, Munafo MR, Bergen AW, Swan GE, Benowitz NL, Tyndale RF, Conti DV, Brown RA, Lerman C, Niaura R. Influence of a dopamine pathway additive genetic efficacy score on smoking cessation: results from two randomized clinical trials of bupropion. Addiction. 2013;108:2202–2211. doi: 10.1111/add.12325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Rose JE, Behm FM, Drgon T, Johnson C, Uhl GR. Personalized smoking cessation: interactions between nicotine dose, dependence and quit-success genotype score. Mol Med. 2010;16:247–253. doi: 10.2119/molmed.2009.00159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Uhl GR, Drgon T, Johnson C, Ramoni MF, Behm FM, Rose JE. Genome-wide association for smoking cessation success in a trial of precessation nicotine replacement. Mol Med. 2010;16:513–526. doi: 10.2119/molmed.2010.00052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Falcone M, Smith RM, Chenoweth MJ, Bhattacharjee AK, Kelsoe JR, Tyndale RF, Lerman C, Pharmacogenomics Research Network Central Nervous System Working G. Neuroimaging in psychiatric pharmacogenetics research: the promise and pitfalls. Neuropsychopharmacology. 2013;38:2327–2337. doi: 10.1038/npp.2013.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Mamoun M, Bergen AW, Shieh J, Wiggins A, Brody AL. Biomarkers of Response to Smoking Cessation Pharmacotherapies: Progress to Date. CNS Drugs. 2015;29:359–369. doi: 10.1007/s40263-015-0243-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Brody AL, Mukhin AG, Mamoun MS, Luu T, Neary M, Liang L, Shieh J, Sugar CA, Rose JE, Mandelkern MA. Brain nicotinic acetylcholine receptor availability and response to smoking cessation treatment: a randomized trial. JAMA Psychiatry. 2014;71:797–805. doi: 10.1001/jamapsychiatry.2014.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wade CH, McBride CM, Kardia SLR, Brody LC. Considerations for Designing a Prototype Genetic Test for Use in Translational Research. Public Health Genomics. 2010;13:155–165. doi: 10.1159/000236061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Walton R, Johnstone E, Munafo M, Neville M, Griffiths S. Genetic clues to the molecular basis of tobacco addiction and progress towards personalized therapy. Trends Mol Med. 2001;7:70–76. doi: 10.1016/s1471-4914(01)01915-3. [DOI] [PubMed] [Google Scholar]
- 116.Carlsten C, Halperin A, Crouch J, Burke W. Personalized medicine and tobacco-related health disparities: is there a role for genetics? Ann Fam Med. 2011;9:366–371. doi: 10.1370/afm.1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Graves KD, Hay JL, O'Neill SC. The promise of using personalized genomic information to promote behavior change: is the debate over, or just beginning? Pers Med. 2014;11:173–185. doi: 10.2217/pme.13.110. [DOI] [PubMed] [Google Scholar]
- 118.Rutter JL. Symbiotic relationship of pharmacogenetics and drugs of abuse. AAPS J. 2006;8:E174–E184. doi: 10.1208/aapsj080121. [DOI] [PMC free article] [PubMed] [Google Scholar]
