ArticlePDF Available

Effects of Dopamine Transporter and Receptor Polymorphisms on Smoking Cessation in a Bupropion Clinical Trial

Authors:

Abstract

This study examined the role of dopaminergic genes in prospective smoking cessation and response to bupropion treatment in a placebo-controlled clinical trial. Smokers of European ancestry (N=418) provided blood samples for genetic analysis and received either bupropion or placebo (10 weeks) plus counseling. Assessments included the dopamine D2 receptor (DRD2) genotype, dopamine transporter (SLC6A3) genotype, demographic factors, and nicotine dependence. Smoking status was verified at the end of treatment (EOT) and at 6-month follow-up. The results provided evidence for a significant DRD2 * SLC6A3 interaction effect on prolonged smoking abstinence and time to relapse at EOT, independent of treatment condition. Such effects were no longer significant at 6-month follow-up, however. These results provide the first evidence from a prospective clinical trial that genes that alter dopamine function may influence smoking cessation and relapse during the treatment phase.
Effects of Dopamine Transporter and Receptor Polymorphisms
on Smoking Cessation in a Bupropion Clinical Trial
Caryn Lerman
University of Pennsylvania Peter G. Shields
Georgetown University
E. Paul Wileyto and Janet Audrain
University of Pennsylvania Larry H. Hawk Jr.
State University of New York at Buffalo
Angela Pinto and Susan Kucharski
University of Pennsylvania Shiva Krishnan
Georgetown University
Ray Niaura
Brown University Leonard H. Epstein
State University of New York at Buffalo
This study examined the role of dopaminergic genes in prospective smoking cessation and response to
bupropion treatment in a placebo-controlled clinical trial. Smokers of European ancestry (N418)
provided blood samples for genetic analysis and received either bupropion or placebo (10 weeks) plus
counseling. Assessments included the dopamine D2 receptor (DRD2) genotype, dopamine transporter
(SLC6A3) genotype, demographic factors, and nicotine dependence. Smoking status was verified at the
end of treatment (EOT) and at 6-month follow-up. The results provided evidence for a significant
DRD2 SLC6A3 interaction effect on prolonged smoking abstinence and time to relapse at EOT,
independent of treatment condition. Such effects were no longer significant at 6-month follow-up,
however. These results provide the first evidence from a prospective clinical trial that genes that alter
dopamine function may influence smoking cessation and relapse during the treatment phase.
Key words: tobacco, smoking, treatment, genetics, bupropion, dopamine
To improve treatment of nicotine dependence, researchers are
increasing attention to the genetic basis of smoking behavior.
Although the task of identifying the specific genetic variants that
predispose individuals to smoking behavior has proved daunting,
initial studies are yielding some promising results. Much of this
research has focused on the neurotransmitter dopamine and genetic
variants relevant to dopamine synthesis, reuptake (i.e., removal of
excess dopamine from the neuronal synapse), and receptors. Do-
pamine is considered an important substrate for the reinforcing
effects of nicotine (Henningfield, 1995; Pontieri, Tanda, Orzi, &
Di Chiara, 1996) as well as for the behavioral conditioning pro-
cess in general addiction (Garris et al., 1999; Schultz, 1998).
These processes have obvious implications for nicotine self-
administration, the reinforcing properties of smoking, and smoking
cessation treatment.
In considering the biological evidence relating dopamine to
nicotine reinforcement, one should note that genes affecting do-
pamine function may contribute to individual differences in nico-
tine dependence. To be specific, individuals with lower activity
genetic variants (i.e., those associated with low-endogenous dopa-
mine levels) may be more likely to self-administer nicotine
through smoking to boost dopamine levels. Preliminary support for
the effects of genes in the dopamine pathway is found in two
studies of a polymorphism (variation) in the dopamine transporter
(SLC6A3) gene. In a case control study of smokers and nonsmok-
ers, persons with at least one copy of the 9-repeat allele (variant)
of SLC6A3 were significantly less likely to be smokers than those
with no copies of the 9-repeat allele (mostly SLC6A3-10 geno-
types; Lerman et al., 1999). Although the proportion of variance
accounted for by SLC6A3 alone was modest, there was evidence
for an interaction that was statistically and clinically significant.
Caryn Lerman, E. Paul Wileyto, Janet Audrain, Angela Pinto, and Susan
Kucharski, Department of Psychiatry, Abramson Cancer Center, Univer-
sity of Pennsylvania; Peter G. Shields and Shiva Krishnan, Department of
Oncology, Georgetown University; Larry H. Hawk Jr., Department of
Psychology, State University of New York at Buffalo; Ray Niaura, Brown
Medical School, Brown University; Leonard H. Epstein, Department of
Pediatrics, State University of New York at Buffalo.
This research was supported by National Cancer Institute and National
Institute on Drug Abuse Grants CA/DA P50 84718 and CA63562 to Caryn
Lerman. We acknowledge Jodie Jaroni and Vyga Kaufmann for their
assistance with project coordination and Maryanne Foster for her assis-
tance with manuscript preparation. We thank Colin Feyerabend and Martin
Jarvis for assistance with saliva cotinine determination.
Correspondence concerning this article should be addressed to Caryn
Lerman, Tobacco Use Research Center, University of Pennsylvania, 3535
Market Street, Suite 4100, Philadelphia, Pennsylvania 19104. E-mail:
clerman@mail.med.upenn.edu
Health Psychology Copyright 2003 by the American Psychological Association, Inc.
2003, Vol. 22, No. 5, 541–548 0278-6133/03/$12.00 DOI: 10.1037/0278-6133.22.5.541
541
Among persons with dopamine D2 receptor (DRD2)-A2 geno-
types, 62% of nonsmokers had SLC6A3-9 genotypes compared
with 46% of smokers. The association of SLC6A3 with smoking
cessation history was also significant. Sabol et al. (1999) docu-
mented the effect of SLC6A3 on smoking cessation, but two other
studies examining SLC6A3 found no association with smoking
status (Jorm et al., 2000; Vandenbergh et al., 2002). We should
note, however, that these two studies did not determine DRD2
genotype and, therefore, could not test for the DRD2–SLC6A3
interaction observed in the initial report (Lerman et al., 1999).
Several additional genetic factors in the dopamine pathway have
been examined for associations with smoking. Previous studies
found a higher prevalence of the more rare A1 or B1 allele of the
DRD2 gene among smokers than among control subjects (Com-
ings et al., 1996; Noble et al., 1994; Spitz et al., 1998). However,
a family-based analysis (Bierut et al., 2000) did not provide
evidence for significant linkage of smoking to the DRD2 gene
locus. Genetic variation in dopamine beta hydroxylase, the enzyme
that catalyzes the conversion of dopamine to norepinephrine
(thereby reducing dopamine levels), has been linked with cigarette
smoking rates (McKinney et al., 2000). Studies relating smoking
behavior to other candidate genes in the dopamine pathway (do-
pamine D5 receptor, tyrosine hydroxylase) have reported negative
findings (Lerman et al., 1997; Sullivan et al., 2001).
Emerging research on genetic contributions to smoking behav-
ior has the potential to advance the science of smoking cessation
treatment by generating new knowledge about genetic factors that
influence clinical treatment outcome. This approach, termed phar-
macogenetics, is based on the premise that individual differences
in genotype influence drug metabolism and drug targets, thereby
influencing treatment outcome (Evans & Relling, 1999). The ul-
timate objective is to develop empirically based methods of tai-
loring the type and dose of treatment to an individuals genotype
to improve efficacy and minimize side effects (Roses, 2000). The
potential for this approach to enhance the treatment of drug de-
pendence is supported by evidence that response to bromocryptine
treatment for alcohol dependence is predicted by DRD2 genotype
(Lawford et al., 1995).
Bupropion is an antidepressant medication with proven efficacy
for smoking cessation (Holm & Spencer, 2000). Six-month absti-
nence rates for smokers treated with bupropion are reported to be
25%35%, as compared with 15%20% for placebo (Hurt et al.,
1997; Jorenby et al., 1999). Whereas male gender and lower
smoking rate have been shown to predict abstinence in bupropion
clinical trials (independent of treatment group), studies have yet to
identify pretreatment factors that predict response to bupropion
treatment (Dale et al., 2001; Hayford et al., 1999).
Research has begun to elucidate bupropions pharmacokinetic
and pharmacodynamic effects. With regard to the former, bupro-
pion is metabolized to its primary metabolite, hydroxybupropion,
by the cytochrome P450 2B6 (CYP2B6) enzyme (Faucette et al.,
2000). We recently reported that the CYP2B6 genotype is a sig-
nificant predictor of smoking relapse and that bupropion attenu-
ated this effect in women (Lerman, Shields, et al., 2002). CYP2B6
is also involved in nicotine metabolism, and the genotype effect in
this study appeared to be mediated by craving, rather than by
bupropion metabolism (Lerman, Shields, et al., 2002). With regard
to pharmacodynamic effects, available evidence suggests that bu-
propion is a weak inhibitor of dopamine and norepinephrine re-
uptake (Ascher et al., 1995; Sanchez & Hyttel, 1999). This would
result in greater availability of these neurotransmitters and positive
psychobiological effects. These proposed neurochemical effects of
bupropion are consistent with documented beneficial effects on
postcessation withdrawal and mood symptoms in a placebo-
controlled bupropion trial (Lerman, Roth, et al., 2002).
The present pharmacogenetic study investigated whether the
SLC6A3 and DRD2 genes predicted smoking abstinence and re-
sponse to treatment in a placebo-controlled trial of bupropion for
smoking cessation. On the basis of previous findings relating
smoking to genes involved in dopamine regulation (Lerman et al.,
1999; Sabol et al., 1999) and on evidence suggesting that bupro-
pion inhibits dopamine reuptake (Ascher et al., 1995), we made the
following two predictions. First, smokers with the SLC6A3-9 and
DRD2-A2 genotypes would have higher abstinence rates than other
groups. Second, the effects of bupropion on smoking outcomes
would be most pronounced for smokers with the SLC6A3-10 and
DRD2-A1 genotypes, because these individuals would derive the
greatest benefit from increases in dopaminergic activity.
Method
Participants
Participants included 193 men and 225 women of European Caucasian
ancestry who reported smoking 10 or more cigarettes a day. They were
recruited using flyers and newspaper advertisements for a free smoking
cessation research program from May 1999 to September 2001. Potential
participants were first screened by telephone for eligibility and again
during an in-person medical screening session. Exclusion criteria included
pregnancy, a history of psychiatric disorder (Diagnostic and Statistical
Manual of Mental Disorders, 4th ed.; DSM-IV; 1994), seizure disorder,
and current use of psychotropic medications. Analyses were limited to
Caucasians of European ancestry to reduce bias due to racial admixture
(N418). Data from participants in this clinical trial were included in a
previous report of the psychological mechanisms of bupropion treatment
(Lerman, Roth, et al., 2002) and the effects of CYP2B6 genotype on
treatment outcome (Lerman, Shields, et al., 2002).
The sample included 418 participants of European Caucasian decent;
227 participants received bupropion and 191 received placebo. Of the
participants, 54% were women and 46% of the total group completed a
college education. The average age of participants was 44 11 years, and
the average smoking rate was 25 12 cigarettes per day.
Procedure
Participants were enrolled at Georgetown University in Washington,
DC, and the State University of New York at Buffalo in Buffalo, NY.
Participants at both sites were ascertained in the same manner and received
identical assessments and treatments, and all participants provided written
consent for genotyping and treatment. At an initial visit to the smoking
clinic, participants provided a 40-ml blood sample and completed a set of
standardized self-report questionnaires (see Measures section). All partic-
ipants received 10 weeks of either placebo (n191) or bupropion (n
227), as determined by randomization. Bupropion treatment was delivered
according to the standard therapeutic dose (150 mg/day for the first 3 days,
followed by 300 mg/day). Participants also received standardized behav-
ioral counseling, focusing on self-monitoring and behavioral-modification
approaches. Trained health educators delivered this counseling during
seven sessions over a 10-week period in a group format. Weekly supervi-
sion and videotaping of sessions were performed to maintain the integrity
of the structured intervention protocol. All participants were instructed to
542 LERMAN ET AL.
quit smoking on a target quit date (TQD) 2 weeks after initiating medica-
tion, which coincided with Session 3 of behavioral counseling.
Self-reported data on smoking status were recorded weekly, at the end of
treatment (EOT; 8 weeks post-TQD) and at a 6-month follow-up using a
validated timeline follow-back method (Brown, Burgess, Sales, & White-
ley, 1998). To biochemically verify self-report data, we performed saliva
cotinine testing for participants who reported abstinence at EOT and at
6-month follow-up using a gasliquid chromatography method (Feyera-
bend & Russell, 1990). Weekly pill counts were also recorded using pill
bottles returned on a weekly basis by participants. Monetary incentives
were provided for study participation but not for successful quitting.
Measures
Smoking outcomes. Consistent with the recommendations of the Soci-
ety for Research on Nicotine and Tobacco (Hughes et al., 2003), we
considered prolonged abstinence to be the primary outcome and 7-day
point prevalence as the secondary outcome. Prolonged abstinence is de-
fined as sustained abstinence after a 2-week grace period, with 7 consec-
utive days of smoking constituting a treatment failure (Hughes et al., 2003).
Seven-day point prevalence reflects smoking status for the prior 7 days and
was biochemically verified on the basis of cotinine levels. Only those
participants with cotinine levels less than or equal to 15 ng/ml were
classified as abstinent for the point-prevalence outcome. Participants lost to
follow-up (5% at EOT, 7% at 6-month follow-up) were classified as
smokers for all outcome analyses.
Genotype. Consistent with previous reports (Lerman et al., 1999; Sabol
et al., 1999), the SLC6A3 genotype was classified as the presence or
absence of the 9-repeat allele. Because the most common alleles are the
9-repeat and 10-repeat alleles and other variants are very rare (3%), we
refer to these groups as 9/9 or 9/10 versus 10/10. The DRD2 genotype was
classified as the presence or absence of the A1 allele (A1/A1 or A1/A2 vs.
A2/A2), consistent with previous reports (Comings et al., 1996; Lerman et
al., 1999). The assays were validated by confirming a polymorphic inher-
itance pattern in seven human family lines that encompassed three gener-
ations (National Institute of General Medical Studies, n.d.). Quality control
procedures included positive and negative controls with each assay and
independent repeat genotyping for 20% of the results.
Demographic factors. Gender, education, marital status, age, and eth-
nic ancestry of grandparents were assessed by self-report during the pre-
treatment assessment visit.
Nicotine dependence. The Fagerstrom Test for Nicotine Dependence
(FTND) is a 6-item self-report measure of nicotine dependence derived
from the Fagerstrom Tolerance Questionnaire (Heatherton, Kozlowski,
Frecker, & Fagerstrom, 1991). The FTND was administered during the
pretreatment assessment visit.
Statistical Considerations
An association (case-control) study design was used to evaluate geno-
type effects on treatment outcome. This approach was chosen over a
family-based analysis (Spielman & Ewens, 1996) both because of the
limited feasibility of recruiting family members of adults in a clinical trial,
and because the case-control design is more powerful. Intent-to-treat meth-
ods were used to evaluate the trial outcomes. Chi-square tests of associa-
tion and ttests were used to identify factors associated with the smoking
outcomes. All statistical tests were two-sided. Variables with significant
(p.10) associations with smoking status were entered into logistic
regression models to identify predictors of abstinence at EOT and at
6-month follow-up. Main effects of genotype and covariates were tested in
a first step, and interaction terms were included in a second step. All
models controlled for study site. Cox regression was used to examine the
main and interacting effects of genotype on time to relapse (the first day of
a prolonged abstinence failure). The time to event for participants lost to
follow-up was the last date of contact.
Results
Descriptive Data
Participants were classified according to variation in the
SLC6A3 and DRD2 genes. The frequencies for the different
SLC6A3 genotype groups are as follows: 210 participants (50.2%)
had the 10/10 genotype, 162 (38.8%) had the 9/10 genotype, 34
(8.1%) had the 9/9 genotype, and 12 (2.9%) had genotypes con-
taining other allelic combinations (10/11, 10/6, 10/8, 9/11, 9/6, or
9/3). Thus, 200 (47.9%) participants were classified as SLC6A3-9
(9/9, 9/10 or 9/*, in which * refers to alleles other than 9 or 10) and
218 (52.1%) as SLC6A3-10 (10/10 or 10/*). For the DRD2 gene,
genotype frequencies were as follows: 28 participants (6.7%) had
A1/A1 genotypes, 152 (36.4%) had A1/A2 genotypes, and 238
(56.9%) had A2/A2 genotypes. Thus, 180 (43.1%) participants
were classified as DRD2-A1 (i.e., A1/A1 or A1/A2), and 238
(56.9%) were classified as DRD2-A2 (i.e., A2/A2). Genotypes for
both genes were in HardyWeinberg equilibrium, and there were
no differences by study site. There were no significant differences
between the bupropion and placebo groups by genotype or the
controlling variables, or between participants retained and those
lost to follow-up (ps.10). Pill counts indicated that, on average,
participants consumed four pills less than the total recommended
number of pills for the entire course of treatment. Pill counts did
not differ significantly by treatment or genotype (ps.10).
Bivariate Analysis of the Effects of Genotype and
Treatment on Smoking Abstinence
Predictors of prolonged abstinence are shown in Table 1. There
was a significant effect of treatment condition at EOT,
2
(1, N
418) 9.69, p.01, and at 6-month follow-up,
2
(1, N
418) 4.40, p.04. The main effects of SLC6A3 and DRD2
genotypes were not significant at either timepoint (ps.10).
Additional factors associated significantly with prolonged absti-
nence included nicotine dependence and gender. The results of
univariate analyses of point-prevalence abstinence were similar;
treatment condition was significant at the end of treatment (p
.01) and at 6-month follow-up (p.03), but the individual
genotype variables were not.
Figure 1 shows the combined effects of the two genotypes on
prolonged abstinence rates. A significant effect of SLC6A3 on
abstinence at EOT was found among participants with A2 geno-
types (odds ratio [OR] 1.74; confidence interval [CI]
1.03, 2.93; p.03), but not among those with A1 genotypes
(OR 0.67, CI 0.37, 1.21; p.18). The MantelHaenszel test
for homogeneity of ORs indicated that these values are signifi-
cantly different,
2
(1, N418) 5.74, p.02. A similar trend
for an effect of SLC6A3 on abstinence in the A2 genotype group
was observed for point-prevalence abstinence at EOT, but the
results were not significant ( p.11). No significant effects for the
two genotypes combined were found for either abstinence measure
at 6-month follow-up.
Figure 2 shows the effect of treatment on prolonged abstinence
by SLC6A3 and DRD2 genotypes. These descriptive data suggest
that the largest effect of treatment occurs in the DRD2-A2/
SLC6A3-10 group (OR 2.80; CI 1.30, 6.00; p.01), because
of a lower abstinence rate on placebo. By contrast, the A1/10
543
SMOKING CESSATION IN A BUPROPION CLINICAL TRIAL
group has the smallest treatment effect (OR 0.87;
CI 0.37, 2.00; p.74), because of a high abstinence rate on
placebo. The treatment effects in the A1/9 (OR 2.23;
CI 0.92, 5.41; p.07) and A2/9 (OR 1.91; CI 0.88, 4.14;
p.10) groups were intermediate. The MantelHaenszel statistic
indicated that these values are not significantly different,
2
(3,
N418) 4.53, p.21.
Multivariate Models of Abstinence
Variables with significant (p.10) associations with absti-
nence were entered into logistic regression models to test the main
and interacting effects of genotypes and treatment. The best fitting
model for prolonged abstinence at the end of treatment is shown in
Table 2. The final model included significant effects for treatment
condition, gender, nicotine dependence level, DRD2 genotype, and
the DRD2 SLC6A3 interaction. Participants treated with bupro-
pion, women, and those with lower baseline levels of nicotine
dependence were more likely to be abstinent at the end of treat-
ment. The genegene interaction (see Figure 1) indicates that,
among participants with DRD2-A2 genotypes, SLC6A3-9 repeat
genotype is associated with a higher abstinence rate (p.03).
Among participants with DRD2-A1 genotypes, the effect of
SLC6A3 is not significant (p.18). The two-way and three-way
interactions for Treatment Genotype were not significant in the
logistic regression model.
Similar modeling approaches were used to examine prolonged
abstinence at 6-month follow-up. In this model, only treatment
(p.05) and gender (p.01) were significant predictors. To
Table 1
Predictors of Prolonged Abstinence
Variable and level
End of treatment 6-month follow-up
% abstinent p% abstinent p
Treatment
Placebo 39.8 22.5
Bupropion 55.1 .01 31.7 .04
SLC6A3
9/9, 9/10 50.0 29.0
10/10 46.3 .45 26.1 .51
DRD2
A1 51.7 26.1
A2 45.9 .20 28.6 .58
Gender
Female 41.8 22.2
Male 55.4 .01 33.7 .01
Site
Washington, DC 49.4 24.7
Buffalo, NY 46.1 .51 31.7 .11
Variable and
abstinence status
End of treatment 6-month follow-up
MSD p MSDp
Nicotine dependence
Abstinent 4.8 2.1 4.8 2.2
Nonabstinent 5.4 2.1 .01 5.2 2.1 .06
Age
Abstinent 44.1 10.9 44.5 10.6
Nonabstinent 44.6 11.6 .61 44.3 11.5 .90
Figure 1. Interacting effects of SLC6A3 and DRD2 genotypes on pro-
longed abstinence at the end of treatment.
Figure 2. Effects of bupropion treatment on prolonged abstinence at the
end of treatment, by SLC6A3 and DRD2 genotypes.
Table 2
Logistic Regression Model of Prolonged Abstinence
at the End of Treatment
Variable OR CI p
Drug 1.87 1.24, 2.82 .01
Site 0.67 0.43, 1.03 .07
Gender 0.51 0.34, 0.78 .01
Nicotine dependence 0.83 0.75, 0.92 .00
SLC6A3 0.67 0.36, 1.20 .20
DRD2 0.54 0.30, 0.96 .04
SLCA3 DRD2 2.40 1.07, 5.48 .03
Note. OR odds ratio; CI confidence interval.
544 LERMAN ET AL.
determine whether the effects of genotype were significantly dif-
ferent at EOT and 6-month follow-up, we performed logistic
regression using general estimating equations. There was a signif-
icant effect of time point (p.01) and a significant Time
DRD2 SLC6A3 interaction, indicating that the genotype effect
was significantly different at the two time points.
We also examined the effects of treatment and genotype on our
secondary outcome, point-prevalence abstinence. At EOT, only
treatment (p.01), gender (p.03), and nicotine dependence
(p.03) were significant predictors of abstinence. At 6-month
follow-up, the only significant predictor was treatment (p.03).
Survival Analysis
Cox regression was used to model the time to prolonged absti-
nence failure by EOT as a function of individual genotype effects
and Genotype Treatment interactions, controlling for study site,
gender, and nicotine dependence. Significant effects on days to
relapse were found for the following variables: treatment (hazard
ratio [HR] 0.64; CI 0.46, 0.80; p.01); site (HR 1.36;
CI 1.01, 1.80; p.04), gender (HR 1.46; CI 1.10, 1.92;
p.01), nicotine dependence (HR 1.14; CI 1.07, 1.22; p
.01), DRD2 (HR 1.50; CI 1.02, 2.23; p.04), and the
DRD2 SLC6A3 interaction (HR 0.50; CI 0.29, 0.87; p
.01). Among participants with DRD2-A2 genotypes, the average
number of days to relapse during the treatment phase for the
SLC6A3-9 group was 28.2, compared with 21.2 for the SLC6A3-10
group, t(416) 2.56, p.01. The pattern of results was similar
for days to relapse by 6-month follow-up; however, the Cox
regression results were not statistically significant.
Examination of Effects of Ethnic Ancestry
We directly explored the potential impact of ethnic admixture in
our European Caucasian sample on bias in the ORs for SLC6A3
and DRD2, by adjusting for self-reported ethnic heritage (beyond
White, non-Hispanic). Heritage was clustered into categories for
Northern European (e.g., English, German), Southern European
(e.g., Spanish, Italian), and Slavic or Eastern European. Partici-
pants were excluded if no one category was predominant (i.e., at
least 3 of 4 grandparents). In this reduced sample (n283), the
MantelHaenszel statistic showed that there was no evidence of
heterogeneity: for SLC6A3,
2
(2, N283) 0.32, p.85; and
for DRD2,
2
(2, N283) 2.00, p.36. Thus, it appears that
in a European Caucasian sample, any bias due to ethnic ancestry
would be minimal.
Discussion
We investigated the effects of two dopaminergic genes, SLC6A3
and DRD2, on smoking abstinence and response to treatment in a
placebo-controlled bupropion clinical trial. The results revealed a
significant genegene interaction effect on prolonged abstinence,
independent of treatment condition. Among smokers with
DRD2-A2 genotypes, those with SLC6A3-9 genotypes (compared
with SLC6A3-10 genotypes) had significantly higher abstinence
rates at the end of treatment (53% vs. 39%) and a longer latency
to relapse at EOT (28 vs. 21 days) and at 6-month follow-up (83
vs. 65 days). These effect sizes are comparable with those
achieved in placebo-controlled pharmacotherapy trials for smok-
ing cessation (Silagy, Mant, Fowler, & Lodge, 1994). By contrast,
among smokers with DRD2-A1 genotypes, the effect of SLC6A3
on abstinence rates and time to relapse was not significant.
These results provide new evidence for dopaminergic genetic
effects on prospective smoking cessation and extend previous
reports of associations of smoking status with SLC6A3 and DRD2
(Comings et al., 1996; Lerman et al., 1999; Sabol et al., 1999;
Spitz et al., 1998). The presence of a genegene interaction,
mirroring results from a previous study of smoking status (Lerman
et al., 1999), underscores the importance of not limiting genetic
investigations of smoking behavior to single-gene effects (Lerman
& Swan, 2002). In fact, the lack of attention to multigenic effects
on a complex trait such as smoking may explain, in part, the failure
of some studies to replicate genetic effects on smoking behavior
(Bierut et al., 2000; Jorm et al., 2000; Vandenbergh et al., 2002).
On the basis of existing biological and epidemiological data on
DRD2 and SLC6A3, one can postulate a biobehavioral mechanism
for the observed findings. Available neurobiological correlative
data suggest that individuals with DRD2-A1 genotypes exhibit
altered receptor density and binding characteristics and therefore
may have lower levels of neuronal dopaminergic activity com-
pared with individuals with DRD2-A2 genotypes (Noble, Blum,
Ritchie, Montgomery, & Sheridan, 1991; Ritchie & Noble, 1996;
Thompson et al., 1997). This is consistent with epidemiological
evidence linking DRD2-A1 with altered neurocognitive function
(Anokhin, Todorov, Madden, Grant, & Heath, 1999; Blum,
Braverman, Dinardo, Wood, & Sheridan, 1994) as well as with a
variety of addictive behaviors (Noble, 2000). The 9-repeat
SLC6A3 genotype has been associated with lower levels of dopa-
mine transporter protein expression (Fuke et al., 2001) and lower
brain protein levels (Heinz et al., 2000). This would be expected to
result in less neuronal dopamine reuptake and higher levels of
dopamine, possibly resulting in less reinforcement from dopamine-
stimulating drugs such as nicotine (Heinz et al., 2000). This
assertion is consistent with evidence relating SLC6A3-9 genotypes
with cocaine-induced paranoia, a state attributed to excess dopa-
mine levels (Gelernter, Kranzler, Satel, & Rao, 1994), and with
smoking cessation history in a cross-sectional study (Sabol et al.,
1999). Thus, one could speculate that, in the presence of normal
receptor function (i.e., DRD2-A2), lower dopamine transporter
levels and higher levels of dopamine (i.e., SLC6A3-9) would
minimize the phasic effects of nicotine on dopamine release,
thereby reducing positive reinforcement from smoking. This, in
turn, would make it easier for smokers with DRD2-A2/SLC6A3-9
genotypes to maintain smoking abstinence. By contrast, low do-
pamine levels (i.e., SLC6A3-10) and normal receptor density (i.e.,
DRD2-A2) might produce the greatest need for and reinforcement
from nicotine. To provide support for this hypothesis, we con-
ducted a post hoc analysis of levels of nicotine dependence by the
DRD2 and SLC6A3 genotypes. Consistent with the findings for
smoking abstinence, there was no effect of SLC6A3 in the
DRD2-A1 group and a trend for an effect in the DRD2-A2 group
(FTND scores of 5.0 2.1 vs. 5.3 2.2 for the SLC6A3-9 vs.
SLC6A3-10 groups, respectively; p.12).
This interpretation, although speculative, may also explain why
the DRD2 SLC6A3 interaction was significant for the prolonged
abstinence measure but weaker for the point-prevalence measure at
the end of treatment. The prolonged abstinence measure allows for
545
SMOKING CESSATION IN A BUPROPION CLINICAL TRIAL
slips, provided the participant does not engage in 7 days of
consecutive smoking (Hughes et al., 2003). By contrast, the point-
prevalence measure requires that the participant does not smoke
even a puff of a single cigarette during the 7 days prior to
assessment. Smokers who carry the putative protective genotypes
may be able to smoke intermittently without experiencing a full-
blown relapse. By contrast, other smokers who experience more
reinforcement following a slip may be more likely to return to
smoking. If this is true, then the DRD2 SLC6A3 effect could be
stronger for the outcome that allows for intermittent slips than for
the outcome that requires continuous abstinence. The weaker ef-
fect for the point-prevalence outcome alternatively may relate to
the requirement for biochemical verification. A recent analysis
from a large number of smoking trials indicated that it is not
uncommon for abstinence rates to differ when biochemical veri-
fication is used (Society for Research on Nicotine & Tobacco
Subcommittee on Biochemical Verification, 2002). In the present
study, the rate of misreporting of abstinence was 20%, consistent
with previous reports (Gariti, Alterman, Ehrman, Mulvaney, &
OBrien, 2002). However, because the rates of misreporting in this
study did not differ by genotype or treatment group, inaccurate
self-reporting is unlikely to bias the study results (Society for
Research on Nicotine & Tobacco Subcommittee on Biochemical
Verification, 2002).
Although the present findings supported our hypothesis about
genetic effects on smoking abstinence at the end of treatment, the
results were negative for genetic effects at 6-month follow-up. In
fact, at 6-month follow-up, abstinence rates were substantially
lower for all groups, and the treatment effect was also substantially
weakened. Previous research has documented the key role of
environmental and psychological factors in smoking relapse, par-
ticularly after active treatment has ended (Brandon, Zelman, &
Baker, 1987; Hall, Munoz, Reus, & Sees, 1993; Mermelstein,
Cohen, Lichtenstein, Baer, & Kamarck, 1986). Thus, although
genotype may play an important role in maintaining abstinence
while smokers are receiving behavioral counseling or pharmaco-
therapy, environmental factors may be relatively more important
after active treatment has ended. The role of psychological and
environmental factors in promoting relapse may also depend, in
part, on genetic predisposition. Thus, future smoking outcome
studies should incorporate data on these influences, making it
possible to test hypotheses about geneenvironment interactions in
smoking cessation.
Contrary to predictions, we did not find evidence that DRD2 and
SLC6A3 significantly modify response to bupropion treatment.
Inspection of the descriptive data (see Figure 2) shows a relatively
equivalent response to bupropion in all four genotype groups, but
a differential response to placebo. To be specific, the DRD2-A2/
SLC6A3-10 group had the lowest quit rate on placebo (27%),
resulting in a highly significant 24% difference in abstinence rates
for the treatment groups. This is consistent with our hypothesis that
less endogenous dopamine (SLC6A3-10) in the presence of normal
receptor density (DRD2-A2) would produce the greatest need for
reinforcement from nicotine. Although bupropion appears to com-
pensate for a possible genetic predisposition to relapse in this
group, our sample size of 418 may not have been sufficient to
detect a Treatment Gene Gene interaction effect.
Dopaminergic processes are but one mechanism postulated for
the effect of bupropion on smoking cessation (Sanchez & Hyttel,
1999). Additional data suggest that bupropion influences serotonin
and norepinephrine activity (Cooper et al., 1994; Dong & Blier,
2001) and that it acts as a neuronal nicotinic receptor antagonist
(i.e., reduces nicotine binding in the brain; Slemmer, Martin, &
Damaj, 2000). Given the lack of specificity of the neuropharma-
cologic effects of this medication, any individual genetic effect on
treatment outcome would be expected to be quite small. Thus, the
greatest promise of pharmacogenetic research on smoking cessa-
tion may be realized in larger collaborative studies that examine
multiple genetic polymorphisms across different neurotransmitter
and receptor pathways.
As an investigation in the emerging area of behavioral genetics
and smoking, the present study has both strengths and weaknesses.
The strengths include the collection of DNA samples from all
study participants, thereby limiting self-selection bias as well as
the use of more refined longitudinal smoking phenotypes. One
limitation of the study is that we did not collect posttreatment
blood samples to determine levels of bupropion and to biochem-
ically verify medication compliance; however, genotype and treat-
ment effects on pill count were not found, suggesting that medi-
cation compliance is unlikely to exert a substantial bias. An
additional issue to consider is the use of an intent-to-treat ap-
proach, in which participants lost to follow-up were assumed to be
smokers. We selected this approach to intervention evaluation
because of concerns about reduced power and, more important, the
potential for biasing the study outcomes by excluding noncompli-
ers (Hall et al., 2001). Because only 5% of participants were lost
to follow-up, any bias due to our choice of analytic design is likely
to be minimal. In fact, when the final model was rerun excluding
participants lost to follow-up, the results were the same, and the
DRD2 SLC6A3 interaction remained significant.
Another potential limitation of this study is that the results may
be biased by ethnic admixture in the study population. In other
words, ethnic heterogeneity in the European Caucasian population
may be associated with both allele frequencies and smoking prac-
tices. Although population stratification can be eliminated in
family-based genetic analyses (Spielman & Ewens, 1996), it is not
very feasible to accrue family members of adults participating in a
randomized trial. Moreover, the significance of population strati-
fication remains controversial. Wacholder, Rothman, and Capo-
raso (2000) used the confounding risk ratio (the ratio of crude and
adjusted relative risks) as a measure of bias in the estimation of
relative risk and determined by simulation that ignoring genetic
admixtures of non-Hispanic European descent would produce a
bias of less than 10%. Millikan (2001) found a similar result using
confounding ORs in a study of genetic markers for breast cancer
and extended the results to include African Americans in the
admixture. We have addressed the potential impact of admixture in
our sample by comparing the crude ORs for genotype effects with
that adjusted for ethnic ancestry. These estimates differed by less
than 2%, indicating that accounting for ethnic ancestry within our
European Caucasian sample is not important.
Despite these potential limitations, the present study provides a
first step toward identifying genetic determinants of smoking
relapse in the context of smoking treatment. Although our inter-
pretations of the data remain speculative until the biobehavioral
mechanisms of genetic influences can be empirically tested, these
results provide directions for future studies. Such studies could
include controlled laboratory investigations to elucidate genetic
546 LERMAN ET AL.
influences on the pharmacodynamic effects of bupropion as well
as larger scale collaborative clinical trials that are amply powered
to detect complex multigenetic and treatment interactions and to
adjust for ethnic ancestry. Although the collection of genetic data
in intervention trials does present a number of unique challenges
(e.g., increased cost, expanded discussion of confidentiality, and
potential risks in the informed consent process), this line of re-
search could enhance our knowledge of nicotine addiction and lead
to interventions that are tailored to individual smokersneeds.
References
American Psychiatric Association. (1994). Diagnostic and statistical man-
ual of mental disorders (4th ed.). Washington, DC: Author.
Anokhin, A. P., Todorov, A. A., Madden, P. A., Grant, J. D., & Heath,
A. C. (1999). Brain event-related potentials, dopamine D2 receptor gene
polymorphism, and smoking. Genetic Epidemiology, 17(Suppl. 1), S37
S42.
Ascher, J. A., Cole, J. O., Colin, J. N., Feighner, J. P., Ferris, R. M.,
Fibiger, H. C., et al. (1995). Bupropion: A review of its mechanism of
antidepressant activity. Journal of Clinical Psychiatry, 56, 395401.
Bierut, L., Rice, J., Edenberg, H., Goate, A., Foroud, T., & Cloninger, C.
(2000). Family-based study of the association of the dopamine D2
receptor gene (DRD2) with habitual smoking. American Journal of
Medical Genetics, 90, 299302.
Blum, K., Braverman, E. R., Dinardo, M. J., Wood, R. C., & Sheridan, P. J.
(1994). Prolonged P300 latency in a neuropsychiatric population with
the D2 dopamine receptor A1 allele. Pharmacogenetics, 4, 313322.
Brandon, T. H., Zelman, D. C., & Baker, T. B. (1987). Effects of main-
tenance sessions on smoking relapse: Delaying the inevitable? Journal
of Consulting and Clinical Psychology, 55, 780782.
Brown, R., Burgess, E., Sales, S., & Whiteley, J. (1998). Reliability and
validity of a smoking timeline follow-back interview. Psychology of
Addictive Behaviors, 12, 101112.
Comings, D., Ferry, L., Bradshaw-Robinson, S., Burchette, R., Chiu, C., &
Muhleman, D. (1996). The dopamine D2 receptor (DRD2) gene: A
genetic risk factor in smoking. Pharmacogenetics, 6, 7379.
Cooper, B. R., Wang, C. M., Cox, R. F., Norton, R., Shea, V., & Ferris,
R. M. (1994). Evidence that the acute behavioral and electrophysiolog-
ical effects of bupropion (Wellbutrin) are mediated by a noradrenergic
mechanism. Neuropsychopharmacology, 11, 133141.
Dale, L. C., Glover, E. D., Sachs, D. P., Schroeder, D. R., Offord, K. P.,
Croghan, I. T., & Hurt, R. D. (2001). Bupropion for smoking cessation:
Predictors of successful outcome. Chest, 119, 13571364.
Dong, J., & Blier, P. (2001, April 1). Modification of norepinephrine and
serotonin, but not dopamine, neuron firing by sustained bupropion
treatment. Psychopharmacology (Berl), 155, 5257.
Evans, W. E., & Relling, M. V. (1999, October 15). Pharmacogenomics:
Translating functional genomics into rational therapeutics. Science, 286,
487491.
Faucette, S. R., Hawke, R. L., Lecluyse, E. L., Shord, S. S., Yan, B.,
Laethem, R. M., et al. (2000). Validation of bupropion hydroxylation as
a selective marker of human cytochrome P450 2B6 catalytic activity.
Drug Metabolism and Disposition, 28, 12221230.
Feyerabend, C., & Russell, M. A. (1990). A rapid gas-liquid chromato-
graphic method for the determination of cotinine and nicotine in bio-
logical fluids. Journal of Pharmacy and Pharmacology, 42, 450452.
Fuke, S., Suo, S., Takahashi, N., Koike, H., Sasagawa, N., & Ishiura, S.
(2001). The VNTR polymorphism of the human dopamine transporter
(DAT1) gene affects gene expression. Pharmacogenomics Journal, 1,
152156.
Gariti, P., Alterman, A. I., Ehrman, R., Mulvaney, F. D., & OBrien, C. P.
(2002). Detecting smoking following smoking cessation treatment. Drug
and Alcohol Dependence, 65, 191196.
Garris, P. A., Kilpatrick, M., Bunin, M. A., Michael, D., Walker, Q. D., &
Wightman, R. M. (1999, March 4). Dissociation of dopamine release in
the nucleus accumbens from intracranial self-stimulation. Nature, 398,
6769.
Gelernter, J., Kranzler, H. R., Satel, S. L., & Rao, P. A. (1994). Genetic
association between dopamine transporter protein alleles and cocaine-
induced paranoia. Neuropsychopharmacology, 11, 195200.
Hall, S. M., Delucchi, K., Velicer, W., Kahler, C., Ranger-Moore, J.,
Hedeker, D., et al. (2001). Statistical analysis of randomized trials in
tobacco treatment: Longitudinal designs with dichotomous outcome.
Nicotine & Tobacco Research, 3, 193202.
Hall, S. M., Munoz, R. F., Reus, V. L., & Sees, K. L. (1993). Nicotine,
negative affect, and depression. Journal of Consulting and Clinical
Psychology, 61, 761767.
Hayford, K. E., Patten, C. A., Rummans, T. A., Schroeder, D. R., Offord,
K. P., Croghan, I. T., et al. (1999). Efficacy of bupropion for smoking
cessation in smokers with a former history of major depression or
alcoholism. British Journal of Clinical Pharmacology, 174, 173178.
Heatherton, T., Kozlowski, L., Frecker, R., & Fagerstrom, K. (1991). The
Fagerstrom Test for Nicotine Dependence: A revision of the Fagerstrom
Tolerance Questionnaire. British Journal of Addiction, 86, 11191127.
Heinz, A., Goldman, D., Jones, D. W., Palmour, R., Hommer, D., Gorey,
J. G., et al. (2000). Genotype influences in vivo dopamine transporter
availability in human striatum. Neuropsychopharmacology, 22, 133
139.
Henningfield, J. (1995). Nicotine medications for smoking cessation. New
England Journal of Medicine, 333, 11961203.
Holm, K. J., & Spencer, C. M. (2000). Bupropion: A review of its use in
the management of smoking cessation. Drugs, 59, 10071024.
Hughes, J. R., Keely, J. P., Niaura, R. S., Ossip-Klein, D. J., Richmond,
R. L., & Swan, G. E. (2003). Measures of abstinence in clinical trials:
Issues and recommendations. Nicotine & Tobacco Research, 5, 1325.
Hurt, R., Sachs, D., Glover, E., Offord, K., Johnston, J., Dale, L., et al.
(1997). A comparison of sustained-release bupropion and placebo for
smoking cessation. New England Journal of Medicine, 337, 11951202.
Jorenby, D. E., Leischow, S. J., Nides, M. A., Rennard, S. I., Johnston,
J. A., Hughes, A. R., et al. (1999). A controlled trial of sustained-release
bupropion, a nicotine patch, or both for smoking cessation. New England
Journal of Medicine, 340, 685691.
Jorm, A., Henderson, A., Jacomb, P., Christensen, H., Korten, A., Rodgers,
B., et al. (2000). Association of smoking and personality with a poly-
morphism of the dopamine transporter gene: Results from a community
survey. American Journal of Medical Genetics, 96, 331334.
Lawford, B. R., Young, R. M., Rowell, J. A., Qualichefski, J., Fletcher,
B. H., Syndulko, K., et al. (1995). Bromocriptine in the treatment of
alcoholics with the D2 dopamine receptor A1 allele. Nature Medicine, 1,
337341.
Lerman, C., Audrain, J., Main, D., Boyd, N., Caporaso, N., Bowman, E.,
et al. (1999). Evidence suggesting the role of specific genetic factors in
cigarette smoking. Health Psychology, 18, 1420.
Lerman, C., Roth, D., Kaufmann, V., Audrain, J., Hawk, L., Liu, A., et al.
(2002). Mediating mechanisms for the impact of bupropion in smoking
cessation treatment. Drug and Alcohol Dependence, 67, 219223.
Lerman, C., Shields, P. G., Main, D., Audrain, J., Roth, J., Boyd, N. R., et
al. (1997). Lack of association of tyrosine hydroxylase genetic polymor-
phism with cigarette smoking. Pharmacogenetics, 7, 521524.
Lerman, C., Shields, P. G., Wileyto, E., Audrain, J., Pinto, A., Hawk, L.,
et al. (2002). Pharmacogenetic investigation of smoking cessation treat-
ment. Pharmacogenetics, 12, 627634.
Lerman, C., & Swan, G. (2002). Non-replication of genetic association
studies: Is DAT all folks? Nicotine & Tobacco Research, 4, 247249.
McKinney, E., Walton, R., Yudkin, P., Fuller, A., Haldar, N., Mant, D., et
al. (2000). Association between polymorphisms in dopamine metabolic
547
SMOKING CESSATION IN A BUPROPION CLINICAL TRIAL
enzymes and tobacco consumption in smokers. Pharmacogenetics, 10,
483491.
Mermelstein, R., Cohen, S., Lichtenstein, E., Baer, J. S., & Kamarck, T.
(1986). Social support and smoking cessation and maintenance. Journal
of Consulting and Clinical Psychology, 54, 447453.
Millikan, R. C. (2001). Re: Population stratification in epidemiologic
studies of common genetic variants and cancer: Quantification of bias.
Journal of the National Cancer Institute, 93, 156158.
National Institute of General Medical Sciences. (n.d.). Human genetic
mutant cell repository. Camden, NJ: Coriell Institute. Retrieved from
locus.umdnj.edu/nigms/overview.html
Noble, E. P. (2000). The DRD2 gene in psychiatric and neurological
disorders and its phenotypes. Pharmacogenomics, 1, 309333.
Noble, E. P., Blum, K., Ritchie, T., Montgomery, A., & Sheridan, P. J.
(1991). Allelic association of the D2 dopamine receptor gene with
receptor-binding characteristics in alcoholism. Archives of General Psy-
chiatry, 48, 648654.
Noble, E. P., St. Jeor, S. T., Ritchie, T., Syndulko, K., St. Jeor, S. C., Fitch,
R., et al. (1994). D2 dopamine receptor gene and cigarette smoking: A
reward gene? Medical Hypotheses, 42, 257260.
Pontieri, F., Tanda, G., Orzi, F., & Di Chiara, G. (1996, July 18). Effects
of nicotine on the nucleus accumbens and similarity to those of addictive
drugs. Nature, 382, 255257.
Ritchie, T., & Noble, E. P. (1996, July 18). [
3
H]naloxone binding in the
human brain: Alcoholism and the TaqI A D2 dopamine receptor poly-
morphism. Brain Research, 718, 193197.
Roses, A. D. (2000, June 15). Pharmacogenetics and the practice of
medicine. Nature, 405, 857865.
Sabol, S. Z., Nelson, M. L., Fisher, C., Gunzerath, L., Brody, C. L., Hu, S.,
et al. (1999). A genetic association for cigarette smoking behavior.
Health Psychology, 18, 713.
Sanchez, C., & Hyttel, J. (1999). Comparison of the effects of antidepres-
sants and their metabolites on reuptake of biogenic amines and on
receptor binding. Cell and Molecular Neurobiology, 19, 467489.
Schultz, W. (1998). Predictive reward signal of dopamine neurons. Journal
of Neurophysiology, 80, 127.
Silagy, C., Mant, D., Fowler, G., & Lodge, M. (1994). Meta-analysis on
efficacy of nicotine replacement therapies in smoking cessation. Lancet,
343, 139142.
Slemmer, J. E., Martin, B. R., & Damaj, M. I. (2000). Bupropion is a
nicotinic antagonist. Journal of Pharmacology and Experimental Ther-
apeutics, 295, 321327.
Society for Research on Nicotine & Tobacco Subcommittee on Biochem-
ical Verification. (2002). Biochemical verification of tobacco use and
cessation. Nicotine & Tobacco Research, 4, 149159.
Spielman, R., & Ewens, W. (1996). The TDT and other family-based tests
for linkage disequilibrium and association. American Journal of Human
Genetics, 59, 983989.
Spitz, M., Shi, H., Yang, F., Hudmon, K., Jiang, H., & Chanberlain, R.
(1998). Case-control study of the D2 dopamine receptor gene and
smoking status in lung cancer patients. Journal of the National Cancer
Institute, 90, 358363.
Sullivan, P. F., Neale, M. C., Silverman, M. A., Harris-Kerr, C., Myaki-
shev, M. V., Wormley, B., et al. (2001). An association study of DRD5
with smoking initiation and progression to nicotine dependence. Amer-
ican Journal of Medical Genetics, 105, 259265.
Thompson, J., Thomas, N., Singleton, A., Piggott, M., Lloyd, S., Perry,
E. K., et al. (1997). D2 dopamine receptor gene (DRD2) Taq1 A
polymorphism: Reduced dopamine D2 receptor binding in the human
striatum associated with the A1 allele. Pharmacogenetics, 7, 479484.
Vandenbergh, D., Bennett, C., Grant, M., Strasser, A., OConnor, R.,
Stauffer, R., et al. (2002). Smoking status and the human dopamine
transporter variable number of tandem repeats (VNTR) polymorphism:
Failure to replicate and finding that never-smokers may be different.
Nicotine & Tobacco Research, 4, 333340.
Wacholder, S., Rothman, N., & Caporaso, N. (2000). Population stratifi-
cation in epidemiologic studies of common genetic variants and cancer:
Quantification of bias. Journal of the National Cancer Institute, 92,
11511158.
548 LERMAN ET AL.
... The 10-repeats allele is associated with a higher rate of gene transcription and, therefore, with higher levels of the carrier protein 88 . Studies have shown that individuals with the 9-repeats allele are less likely to start smoking before the age of 16, have a shorter smoking time, longer periods of abstinence, and are more likely to quit smoking [54][55][56][57][58] . However, controversial results 89 and a lack of significant association 90 demonstrate the need for further studies on this subject. ...
... A meta-analysis study showed that, although the genetic variations of SLC6A3 are related to dopamine regulation, there is a lack of evidence on their influence on smoking cessation, given the multifactorial nature of smoking 88 . However, this study reinforced the importance of gene interaction in susceptibility to smoking and showed that the interaction between the DRD2 Taq1A and SLC6A3 genes prolongs abstinence time and influences smoking cessation with the use of bupropion 55 . The results showed the role of gene-gene interaction in the probability of relapse: smokers possessing the A2 allele of DRD2 Taq1A and SLC6A3-9 had significantly higher rates of abstinence at the end of treatment and a longer latency period for relapse 55 . ...
... However, this study reinforced the importance of gene interaction in susceptibility to smoking and showed that the interaction between the DRD2 Taq1A and SLC6A3 genes prolongs abstinence time and influences smoking cessation with the use of bupropion 55 . The results showed the role of gene-gene interaction in the probability of relapse: smokers possessing the A2 allele of DRD2 Taq1A and SLC6A3-9 had significantly higher rates of abstinence at the end of treatment and a longer latency period for relapse 55 . ...
... Given bupropion's mechanism of action within the brain, individuals with genotypes that predispose for higher dopamine availability have better responses to bupropion. For example, genetic variation in the promoter region (−141 C Ins) of the dopamine D2 receptor (DRD2) translates to higher transcriptional efficiency, and subjects with the variant alleles how an association with higher quit rates when using bupropion vs NRT [80][81][82]. In addition, the dopamine transporter gene SLC6A3/DAT1 presents a variable number of tandem repeats (VNTRs), with the presence of 9 vs 10 VTNRs associated with an increased ability to stop smoking using either NRT or bupropion [81,83]. ...
... For example, genetic variation in the promoter region (−141 C Ins) of the dopamine D2 receptor (DRD2) translates to higher transcriptional efficiency, and subjects with the variant alleles how an association with higher quit rates when using bupropion vs NRT [80][81][82]. In addition, the dopamine transporter gene SLC6A3/DAT1 presents a variable number of tandem repeats (VNTRs), with the presence of 9 vs 10 VTNRs associated with an increased ability to stop smoking using either NRT or bupropion [81,83]. Also, the GG haplotype (rs737865 and rs165599) in the enzyme catechol-O-methyltransferase (COMT), which is involved in the metabolic inactivation of dopamine, has been associated with a favorable outcome when using bupropion for smoking cessation in a Caucasian population [84]. ...
Article
Introduction: Smoking remains a worldwide epidemic, and despite an increase in public acceptance of the harms of tobacco use, it remains the leading cause of preventable death. It is estimated that up to 70% of all smokers express a desire to quit, but only 3-5% of them are successful. Areas covered: The goal of this review was to evaluate the current status of smoking cessation treatments and the feasibility of implementing personalized-medicine approaches to these pharmacotherapies. We evaluated the genetics associated with higher levels of nicotine addiction and follow with an analysis of the genetic variants that affect the nicotine metabolic ratio (NMR) and the FDA approved treatments for smoking cessation. We also highlighted the gaps in the process of translating current laboratory understanding into clinical practice, and the benefits of personalized treatment approaches for a successful smoking cessation strategy. Expert opinion: Evidence supports the use of tailored therapies to ensure that the most efficient treatments are utilized in an individual’s smoking cessation efforts. An understanding of the genetic effects on the efficacy of individualized smoking cessation pharmacotherapies is key to smoking cessation, ideally utilizing a polygenetic risk score that considers all genetic variation.
... However, we report these novel SNPs in Supplementary table 1. Published reports that used the same data from the same study population and tested the same SNPs as another published report were also excluded. One of these excluded studies utilized data from a placebo-controlled bupropion trial [25]; however, this trial data was pooled with a second bupropion trial for use in another study [26] and the latter study was included in this review. Two other studies utilized the same study population to test associations between Taq1A and cessation [27]; the study with a primary aim of testing for the Taq1A interaction with a serotonin genetic variant was excluded [28]. ...
Article
Full-text available
Purpose of Review This systematic review summarizes evidence of single nucleotide polymorphism (SNPs) associations with smoking cessation focusing on dopamine receptor or dopamine metabolism genes. Summary odds ratios (ORs) of SNP associations were calculated and stratified by ancestry, pharmacotherapy, and sex where feasible. Recent Findings The 30 included articles reported the results of 32 studies. The minor allele of DRD2/ANKK1 SNP rs1800497 was associated with lower odds of cessation among people of European ancestry [OR = 0.88, 95% CI 0.82 – 0.95, n= 16 studies]; this association was not observed among people of non-European ancestry. Heterogeneity by sex in rs1800497 associations was present [female: OR = 0.91, 95% CI 0.85 – 0.97, n = 9 studies; male: OR = 1.16, 95% CI 0.85 – 1.55, n = 7 studies]. Recipients of nicotine replacement therapy (NRT) with the minor allele of DRD2 SNP rs6277 [OR = 1.43, 95% CI 1.06 – 1.92, n = 2 studies] or COMT SNP rs4680 [OR = 1.61, 95% CI 1.11 – 2.35, n = 3 studies] had increased odds of cessation. Heterogeneity by sex in rs4680 associations was observed [combined sex: OR = 0.73, 95% CI 0.57 – 0.93, n = 3 studies; male: OR = 0.74, 95% CI 0.53 – 1.02, n = 1 study; female: OR = 1.01, 95% CI 0.77 – 1.32, n = 5 studies]. Summary Associations between rs1800497 and rs4680 and cessation may differ by biological sex. Limited evidence suggests some genetic associations may differ by ancestry and that rs4680 or rs6277 genotype may influence NRT efficacy.
... Seven studies were removed during full article review because incorrect study designs (e.g., a focus on the use of mouse models, meta-analysis, or outcome not focused on smoking abstinence). Results from a final total of 79 articles are summarized (Figure 1)) (Bergen et al., 2014;Breitling et al., 2010;Chen et al., 2012;Cameli et al., 2018;Chen et al., 2014;Chen et al., 2015;Cinciripini et al., 2004;Dahl et al., 2006;David et al., 2007b;David et al., 2007a;David et al., 2008b;David et al., 2008a;David et al., 2013;El-Boraie et al., 2020;Glatard et al., 2017;Gold et al., 2012;Guo & Heitjan, 2010;Heitjan et al., 2008b;Ho et al., 2009;Hutchison et al., 2007;Johnstone et al., 2004;Johnstone et al., 2007;Lee et al., 2007;Lee et al., 2012;Lerman et al., 2002;Lerman et al., 2003;Lerman et al., 2004;Lerman et al., 2006;Lerman et al., 2010;Leventhal et al., 2012;Munafò et al., 2007;Munafò et al., 2009;Munafò et al., 2013;Quintana & Conti, 2013;Robinson et al., 2007;Rocha Santos et al., 2015;Roche et al., 2019;Rose et al., 2010;Swan et al., 2005;Swan et al., 2007;Swan et al., 2012;Tomaz et al., 2015;Tomaz et al., 2018;Tomaz et al., 2019;Santos et al., 2020;Ton et al., 2007;Tyndale et al., 2015;Uhl et al., 2008;Verde et al., 2014;Ware et al., 2015;Zhu et al., 2012;Zhu et al., 2014a;Zhu et al., 2014b) and 18 (23%) estimated genetic ancestry categories (Hu et al., 2006;Berrettini et al., 2007;Ray et al., 2007;Conti et al., 2008;Uhl et al., 2010;Sarginson et al., 2011;King et al., 2012;Bergen et al., 2013a;Ashare et al., 2013;Bergen et al., 2013b;Bergen et al., 2015;Bress et al., 2015;Chen et al., 2018a;Chenoweth et al., 2018;Chenoweth et al., 2021;Chenoweth et al., 2022;Chen et al., 2020;. ...
Article
Full-text available
Abstinence rates among smokers attempting to quit remain low despite the wide availability and accessibility of pharmacological smoking cessation treatments. In addition, the prevalence of cessation attempts and abstinence differs by individual-level social factors such as race and ethnicity. Clinical treatment of nicotine dependence also continues to be challenged by individual-level variability in effectiveness to promote abstinence. The use of tailored smoking cessation strategies that incorporate information on individual-level social and genetic factors hold promise, although additional pharmacogenomic knowledge is still needed. In particular, genetic variants associated with pharmacological responses to smoking cessation treatment have generally been conducted in populations with participants that self-identify as White race or who are determined to be of European genetic ancestry. These results may not adequately capture the variability across all smokers as a result of understudied differences in allele frequencies across genetic ancestry populations. This suggests that much of the current pharmacogenetic study results for smoking cessation may not apply to all populations. Therefore, clinical application of pharmacogenetic results may exacerbate health inequities by racial and ethnic groups. This scoping review examines the extent to which racial, ethnic, and ancestral groups that experience differences in smoking rates and smoking cessation are represented in the existing body of published pharmacogenetic studies of smoking cessation. We will summarize results by race, ethnicity, and ancestry across pharmacological treatments and study designs. We will also explore current opportunities and challenges in conducting pharmacogenomic research on smoking cessation that encourages greater participant diversity, including practical barriers to clinical utilization of pharmacological smoking cessation treatment and clinical implementation of pharmacogenetic knowledge.
Article
Full-text available
Background: The pharmacological profiles and mechanisms of antidepressants are varied. However, there are common reasons why they might help people to stop smoking tobacco: nicotine withdrawal can produce short-term low mood that antidepressants may relieve; and some antidepressants may have a specific effect on neural pathways or receptors that underlie nicotine addiction. Objectives: To assess the evidence for the efficacy, harms, and tolerability of medications with antidepressant properties in assisting long-term tobacco smoking cessation in people who smoke cigarettes. Search methods: We searched the Cochrane Tobacco Addiction Group Specialised Register, most recently on 29 April 2022. Selection criteria: We included randomised controlled trials (RCTs) in people who smoked, comparing antidepressant medications with placebo or no pharmacological treatment, an alternative pharmacotherapy, or the same medication used differently. We excluded trials with fewer than six months of follow-up from efficacy analyses. We included trials with any follow-up length for our analyses of harms. Data collection and analysis: We extracted data and assessed risk of bias using standard Cochrane methods. Our primary outcome measure was smoking cessation after at least six months' follow-up. We used the most rigorous definition of abstinence available in each trial, and biochemically validated rates if available. Our secondary outcomes were harms and tolerance outcomes, including adverse events (AEs), serious adverse events (SAEs), psychiatric AEs, seizures, overdoses, suicide attempts, death by suicide, all-cause mortality, and trial dropouts due to treatment. We carried out meta-analyses where appropriate. Main results: We included a total of 124 studies (48,832 participants) in this review, with 10 new studies added to this update version. Most studies recruited adults from the community or from smoking cessation clinics; four studies focused on adolescents (with participants between 12 and 21 years old). We judged 34 studies to be at high risk of bias; however, restricting analyses only to studies at low or unclear risk of bias did not change clinical interpretation of the results. There was high-certainty evidence that bupropion increased smoking cessation rates when compared to placebo or no pharmacological treatment (RR 1.60, 95% CI 1.49 to 1.72; I2 = 16%; 50 studies, 18,577 participants). There was moderate-certainty evidence that a combination of bupropion and varenicline may have resulted in superior quit rates to varenicline alone (RR 1.21, 95% CI 0.95 to 1.55; I2 = 15%; 3 studies, 1057 participants). However, there was insufficient evidence to establish whether a combination of bupropion and nicotine replacement therapy (NRT) resulted in superior quit rates to NRT alone (RR 1.17, 95% CI 0.95 to 1.44; I2 = 43%; 15 studies, 4117 participants; low-certainty evidence). There was moderate-certainty evidence that participants taking bupropion were more likely to report SAEs than those taking placebo or no pharmacological treatment. However, results were imprecise and the CI also encompassed no difference (RR 1.16, 95% CI 0.90 to 1.48; I2 = 0%; 23 studies, 10,958 participants). Results were also imprecise when comparing SAEs between people randomised to a combination of bupropion and NRT versus NRT alone (RR 1.52, 95% CI 0.26 to 8.89; I2 = 0%; 4 studies, 657 participants) and randomised to bupropion plus varenicline versus varenicline alone (RR 1.23, 95% CI 0.63 to 2.42; I2 = 0%; 5 studies, 1268 participants). In both cases, we judged evidence to be of low certainty. There was high-certainty evidence that bupropion resulted in more trial dropouts due to AEs than placebo or no pharmacological treatment (RR 1.44, 95% CI 1.27 to 1.65; I2 = 2%; 25 studies, 12,346 participants). However, there was insufficient evidence that bupropion combined with NRT versus NRT alone (RR 1.67, 95% CI 0.95 to 2.92; I2 = 0%; 3 studies, 737 participants) or bupropion combined with varenicline versus varenicline alone (RR 0.80, 95% CI 0.45 to 1.45; I2 = 0%; 4 studies, 1230 participants) had an impact on the number of dropouts due to treatment. In both cases, imprecision was substantial (we judged the evidence to be of low certainty for both comparisons). Bupropion resulted in inferior smoking cessation rates to varenicline (RR 0.73, 95% CI 0.67 to 0.80; I2 = 0%; 9 studies, 7564 participants), and to combination NRT (RR 0.74, 95% CI 0.55 to 0.98; I2 = 0%; 2 studies; 720 participants). However, there was no clear evidence of a difference in efficacy between bupropion and single-form NRT (RR 1.03, 95% CI 0.93 to 1.13; I2 = 0%; 10 studies, 7613 participants). We also found evidence that nortriptyline aided smoking cessation when compared with placebo (RR 2.03, 95% CI 1.48 to 2.78; I2 = 16%; 6 studies, 975 participants), and some evidence that bupropion resulted in superior quit rates to nortriptyline (RR 1.30, 95% CI 0.93 to 1.82; I2 = 0%; 3 studies, 417 participants), although this result was subject to imprecision. Findings were sparse and inconsistent as to whether antidepressants, primarily bupropion and nortriptyline, had a particular benefit for people with current or previous depression. Authors' conclusions: There is high-certainty evidence that bupropion can aid long-term smoking cessation. However, bupropion may increase SAEs (moderate-certainty evidence when compared to placebo/no pharmacological treatment). There is high-certainty evidence that people taking bupropion are more likely to discontinue treatment compared with people receiving placebo or no pharmacological treatment. Nortriptyline also appears to have a beneficial effect on smoking quit rates relative to placebo, although bupropion may be more effective. Evidence also suggests that bupropion may be as successful as single-form NRT in helping people to quit smoking, but less effective than combination NRT and varenicline. In most cases, a paucity of data made it difficult to draw conclusions regarding harms and tolerability. Further studies investigating the efficacy of bupropion versus placebo are unlikely to change our interpretation of the effect, providing no clear justification for pursuing bupropion for smoking cessation over other licensed smoking cessation treatments; namely, NRT and varenicline. However, it is important that future studies of antidepressants for smoking cessation measure and report on harms and tolerability.
Article
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.
Chapter
Approximately one in six American adults is a current smoker, and smoking accounts for one-third of all cancer deaths. In general, patients with cancer have a higher dependence on nicotine and are more likely to be smokers or ex-smokers. Several instruments measure nicotine dependence among cigarette smokers. However, the evaluation and final recommendation are clinical, and the treatment plan must be individualized. Nonpharmacologic treatments include behavioral counseling, quitlines, and self-help material, but yield relatively low quit rates if used alone. Pharmacologic treatments approved by the US Food and Drug Administration include nicotine replacement therapies, bupropion, and varenicline. Patients with cancer who use tobacco should be treated according to evidence-based treatment guidelines, with particular attention to tailoring education about their disease-tobacco link, pharmacotherapy, comorbid medical and psychiatric disorders, and family and household tobacco use. Health care providers have limited time and expertise to address smoking among patients with cancer, and patients may have comorbid substance use or dependence or other emotional and mental disorders that undermine their ability to quit smoking. Systems-level challenges and tailored treatment approaches are needed to identify all tobacco users, lower the rate of persistent tobacco use, and reduce recidivism among patients with cancer.
Chapter
Despite the reality that smoking remains the most important preventable cause of death and disability, most clinicians underperform in helping smokers quit. Nearly 70 per cent of smokers want to quit, and 42.5 per cent attempt to quit each year. The most effective smoking cessation programmes involve a combination of pharmacotherapy and behavioural and/or cognitive counselling to improve abstinence rates. Ways to counter clinicians’ pessimism about cessation include the knowledge that most smokers require multiple attempts before they succeed in quitting.
Article
Background: Whilst the pharmacological profiles and mechanisms of antidepressants are varied, there are common reasons why they might help people to stop smoking tobacco. Firstly, nicotine withdrawal may produce depressive symptoms and antidepressants may relieve these. Additionally, some antidepressants may have a specific effect on neural pathways or receptors that underlie nicotine addiction. Objectives: To assess the evidence for the efficacy, safety and tolerability of medications with antidepressant properties in assisting long-term tobacco smoking cessation in people who smoke cigarettes. Search methods: We searched the Cochrane Tobacco Addiction Specialized Register, which includes reports of trials indexed in the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, and PsycINFO, clinicaltrials.gov, the ICTRP, and other reviews and meeting abstracts, in May 2019. Selection criteria: We included randomized controlled trials (RCTs) that recruited smokers, and compared antidepressant medications with placebo or no treatment, an alternative pharmacotherapy, or the same medication used in a different way. We excluded trials with less than six months follow-up from efficacy analyses. We included trials with any follow-up length in safety analyses. Data collection and analysis: We extracted data and assessed risk of bias using standard Cochrane methods. We also used GRADE to assess the certainty of the evidence. The primary outcome measure was smoking cessation after at least six months follow-up, expressed as a risk ratio (RR) and 95% confidence intervals (CIs). We used the most rigorous definition of abstinence available in each trial, and biochemically validated rates if available. Where appropriate, we performed meta-analysis using a fixed-effect model. Similarly, we presented incidence of safety and tolerance outcomes, including adverse events (AEs), serious adverse events (SAEs), psychiatric AEs, seizures, overdoses, suicide attempts, death by suicide, all-cause mortality, and trial dropout due to drug, as RRs (95% CIs). Main results: We included 115 studies (33 new to this update) in this review; most recruited adult participants from the community or from smoking cessation clinics. We judged 28 of the studies to be at high risk of bias; however, restricting analyses only to studies at low or unclear risk did not change clinical interpretation of the results. There was high-certainty evidence that bupropion increased long-term smoking cessation rates (RR 1.64, 95% CI 1.52 to 1.77; I2 = 15%; 45 studies, 17,866 participants). There was insufficient evidence to establish whether participants taking bupropion were more likely to report SAEs compared to those taking placebo. Results were imprecise and CIs encompassed no difference (RR 1.16, 95% CI 0.90 to 1.48; I2 = 0%; 21 studies, 10,625 participants; moderate-certainty evidence, downgraded one level due to imprecision). We found high-certainty evidence that use of bupropion resulted in more trial dropouts due to adverse events of the drug than placebo (RR 1.37, 95% CI 1.21 to 1.56; I2 = 19%; 25 studies, 12,340 participants). Participants randomized to bupropion were also more likely to report psychiatric AEs compared with those randomized to placebo (RR 1.25, 95% CI 1.15 to 1.37; I2 = 15%; 6 studies, 4439 participants). We also looked at the safety and efficacy of bupropion when combined with other non-antidepressant smoking cessation therapies. There was insufficient evidence to establish whether combination bupropion and nicotine replacement therapy (NRT) resulted in superior quit rates to NRT alone (RR 1.19, 95% CI 0.94 to 1.51; I2 = 52%; 12 studies, 3487 participants), or whether combination bupropion and varenicline resulted in superior quit rates to varenicline alone (RR 1.21, 95% CI 0.95 to 1.55; I2 = 15%; 3 studies, 1057 participants). We judged the certainty of evidence to be low and moderate, respectively; in both cases due to imprecision, and also due to inconsistency in the former. Safety data were sparse for these comparisons, making it difficult to draw clear conclusions. A meta-analysis of six studies provided evidence that bupropion resulted in inferior smoking cessation rates to varenicline (RR 0.71, 95% CI 0.64 to 0.79; I2 = 0%; 6 studies, 6286 participants), whilst there was no evidence of a difference in efficacy between bupropion and NRT (RR 0.99, 95% CI 0.91 to 1.09; I2 = 18%; 10 studies, 8230 participants). We also found some evidence that nortriptyline aided smoking cessation when compared with placebo (RR 2.03, 95% CI 1.48 to 2.78; I2 = 16%; 6 studies, 975 participants), whilst there was insufficient evidence to determine whether bupropion or nortriptyline were more effective when compared with one another (RR 1.30 (favouring bupropion), 95% CI 0.93 to 1.82; I2 = 0%; 3 studies, 417 participants). There was no evidence that any of the other antidepressants tested (including St John's Wort, selective serotonin reuptake inhibitors (SSRIs), monoamine oxidase inhibitors (MAOIs)) had a beneficial effect on smoking cessation. Findings were sparse and inconsistent as to whether antidepressants, primarily bupropion and nortriptyline, had a particular benefit for people with current or previous depression. Authors' conclusions: There is high-certainty evidence that bupropion can aid long-term smoking cessation. However, bupropion also increases the number of adverse events, including psychiatric AEs, and there is high-certainty evidence that people taking bupropion are more likely to discontinue treatment compared with placebo. However, there is no clear evidence to suggest whether people taking bupropion experience more or fewer SAEs than those taking placebo (moderate certainty). Nortriptyline also appears to have a beneficial effect on smoking quit rates relative to placebo. Evidence suggests that bupropion may be as successful as NRT and nortriptyline in helping people to quit smoking, but that it is less effective than varenicline. There is insufficient evidence to determine whether the other antidepressants tested, such as SSRIs, aid smoking cessation, and when looking at safety and tolerance outcomes, in most cases, paucity of data made it difficult to draw conclusions. Due to the high-certainty evidence, further studies investigating the efficacy of bupropion versus placebo are unlikely to change our interpretation of the effect, providing no clear justification for pursuing bupropion for smoking cessation over front-line smoking cessation aids already available. However, it is important that where studies of antidepressants for smoking cessation are carried out they measure and report safety and tolerability clearly.
Article
Full-text available
Dopaminergic genes are likely candidates for heritable influences on cigarette smoking. In an accompanying article, Lerman et al. (1999) report associations between allele 9 of a dopamine transporter gene polymorphism (SLC6A3-9) and lack of smoking, late initiation of smoking, and length of quitting attempts. The present investigation extended their study by examining both smoking behavior and personality traits in a diverse population of nonsmokers, current smokers, and former smokers (N = 1,107). A significant association between SLC6A3-9 and smoking status was confirmed and was due to an effect on cessation rather than initiation. The SLC6A3-9 polymorphism was also associated with low scores for novelty seeking, which was the most significant personality correlate of smoking cessation. It is hypothesized that individuals carrying the SLC6A3-9 polymorphism have altered dopamine transmission, which reduces their need for novelty and reward by external stimuli, including cigarettes.
Article
Depression, whether conceptualized as a trait, symptom, or as a diagnosable disorder, is overrepresented among smokers. Depressed smokers appear to experience more withdrawal symptoms on quitting, are less likely to be successful at quitting, and are more likely to relapse. This article documents these relationships and explores several potential links between smoking and depression. The potential efficacy of antidepressant therapy, cognitive-behavioral therapy, and nicotine replacement therapy for smokers with depressive disorders or traits is discussed. Clinical implications and the role of patient treatment matching are also discussed.
Article
Sustained release bupropion (amfebutamone) is a non-nicotine agent that is indicated as an aid to smoking cessation. In 2 large well designed clinical trials, sustained release bupropion 300 mg/day (the recommended dose) for 7 or 9 weeks was associated with considerably and significantly higher smoking abstinence rates (continuous abstinence and 7-day point prevalence rates) than placebo during treatment and at follow-up at 6 and 12 months. Point prevalence rates at 12 months in 2 studies were 23.1 and 30.3% with bupropion, whereas values for placebo were 12.4 and 15.6%. Continuous abstinence rates at 12 months, available from 1 trial, were 18.4% with bupropion and 5.6% with placebo. Furthermore, bupropion was associated with significantly higher quitting rates than nicotine patch in a comparative study. Combination therapy with bupropion and nicotine patch provided slightly higher abstinence rates than bupropion alone, although differences were not statistically significant. The combination was superior to nicotine patch alone. Data from a preliminary report of long term bupropion treatment (52 weeks) showed that the drug was associated with significantly higher continuous abstinence rates than placebo only to 6 months. However, point prevalence abstinence rates were significantly higher with bupropion than placebo to 18 months. Bupropion 300 mg/day recipients reported nicotine withdrawal symptoms during treatment; however, the symptoms were significantly less severe with bupropion than placebo. Patients receiving bupropion 300 mg/day or bupropion in combination with nicotine patch for smoking cessation generally gained less body weight than placebo recipients. The benefits of bupropion for preventing weight gain persisted after the completion of long term, but not short term therapy. Bupropion was well tolerated in clinical trials, and the only adverse events that were significantly more common with bupropion than placebo were insomnia and dry mouth. Data published so far suggest that sustained release bupropion has a low potential for inducing seizures (seizure rate ≈0.1% in patients with depression). Conclusions: Bupropion is an effective and well tolerated smoking cessation intervention. Further studies with long term follow-up will be useful in determining whether abstinence rates are maintained with bupropion. In addition, clarification of its efficacy in comparison with other therapies used for smoking cessation would help to establish its clinical value. The reduced potential for weight gain with bupropion and the ability to use bupropion in combination with nicotine replacement therapy make the drug a useful treatment option for smoking cessation. Pharmacology The mechanism by which bupropion (amfebutamone) acts as an aid in smoking cessation is unknown. However, bupropion is thought to produce its therapeutic antidepressant effects via the inhibition of noradrenaline and/or dopamine reuptake. Bupropion does not affect serotonin reuptake. Bupropion showed dependence potential in animal models, but not at therapeutic dosages in individuals who abuse drugs or in healthy volunteers. Post-marketing surveillance data have shown that bupropion has a very low abuse potential. Maximum plasma concentrations of sustained release bupropion are reached approximately 3 hours after an oral 150mg dose. Bupropion is highly plasma protein bound, and is extensively metabolised to 3 active metabolites. A single 150mg dose of sustained release bupropion has a mean elimination half-life of 18 to 19 hours. Around 84% and 9% of an oral dose of bupropion was recovered in the urine and faeces, respectively, within 72 hours after administration. There is little available data on the effects of the concomitant administration of bupropion and other drugs on the metabolism of each drug. However, there is potential for interactions between bupropion and drugs that affect the cytochrome P450 (CYP) 2B6 isoenzyme. In addition, bupropion inhibits the activity of the CYP2D6 isoenzyme, which metabolises certain antidepressants (including tricyclic antidepressants and selective serotonin reuptake inhibitors), β-blockers, antiarrhythmics and antipsychotics. It is recommended that coadministration of bupropion and such drugs is approached with caution. There are no significant differences in the pharmacokinetics of sustained release bupropion between smokers and nonsmokers. Sustained release bupropion is bioequivalent to the immediate release formulation in humans. Therapeutic Efficacy Sustained release bupropion 300 mg/day for 7 or 9 weeks significantly increased smoking cessation rates (continuous abstinence and 7-day point prevalence rates) during treatment and at follow-up at 6 and 12 months versus placebo in 2 large well designed studies. Point prevalence rates at 12 months were ≤30.3% with bupropion, whereas values for placebo were ≤15.6%. In 1 trial, continuous abstinence rates at 12 months were 18.4% with bupropion and 5.6% with placebo. Furthermore, bupropion was associated with significantly higher quitting rates than nicotine patch in the only comparison. Combination therapy with bupropion and nicotine patch provided slightly higher abstinence rates than bupropion alone, although differences were not statistically significant. The combination was superior to nicotine patch alone. Data from a preliminary report of long term bupropion treatment (52 weeks) showed that the drug was associated with significantly higher continuous abstinence rates than placebo only to 6 months. However, point prevalence abstinence rates were significantly higher with bupropion than placebo to 18 months. Bupropion 300 mg/day recipients reported significant withdrawal symptoms during treatment; however, the symptoms were significantly less with bupropion than placebo. In the preliminary report of a long term (52 weeks’ treatment) study, bupropion recipients had significantly less craving for cigarettes than placebo recipients and craving was less likely to be the reason for relapse with bupropion than placebo. Bodyweight gain was generally less in patients receiving bupropion 300 mg/day or bupropion in addition to nicotine patch for smoking cessation than in placebo recipients. The benefits of bupropion for preventing weight gain persisted after the completion of long term, but not short term therapy. Tolerability Short term treatment with sustained release bupropion 300 mg/day was well tolerated in clinical trials of the drug for smoking cessation. The only adverse events that were significantly more common with bupropion than placebo were insomnia and dry mouth. Sustained release bupropion appears to have a lower propensity to cause seizures than the immediate release formulation (≈0.1 vs 0.4% for therapeutic dosages); however, no direct comparison of seizure rates between the formulations has been made. Immediate release bupropion was generally well tolerated in patients with pre-existing heart disease. The cardiovascular effects of bupropion have not been assessed in patients with unstable heart disease or recent myocardial infarction, although studies are ongoing. Dosage and Administration It is recommended that sustained release bupropion 300 mg/day (twice daily) is given for 7 to 12 weeks for smoking cessation in adults. A target quitting date should generally be set for within the first 2 weeks of treatment. Patients are able to continue smoking while they take bupropion. In patients requiring continuous treatment, bupropion can be continued for up to 6 months (US) or a year (Canada). Bupropion can be given with transdermal nicotine. Patients with hepatic or renal disease should be treated with reduced dosages of bupropion. Bupropion is contraindicated in patients with bulimia or anorexia nervosa and in patients with seizure disorders. In addition, bupropion should be given with caution to patients with risk factors for seizures. Mothers should not continue breastfeeding infants while taking bupropion.
Article
Background: Some critics argue that bias from population stratification (the mixture of individuals from heterogeneous genetic backgrounds) undermines the credibility of epidemiologic studies designed to estimate the association between a genotype and the risk of disease. We investigated the degree of bias likely from population stratification in U.S. studies of cancer among non-Hispanic Caucasians of European origin. Methods: An expression of the confounding risk ratio-the ratio of the effect of the genetic factor on risk of disease with and without adjustment for ethnicity-is used to measure the potential relative bias from population stratification. We first use empirical data on the frequency of the N-acetyltransferase (NAT2) slow acetylation genotype and incidence rates of male bladder cancer and female breast cancer in non-Hispanic U.S. Caucasians with ancestries from eight European countries to assess the bias in a hypothetical population-based U.S. study that does not take ethnicity into consideration. Then, we provide theoretical calculations of the bias over a large range of allele frequencies and disease rates. Results: Ignoring ethnicity leads to a bias of 1% or less in our empirical studies of NAT2. Furthermore, evaluation of a wide range of allele frequencies and representative ranges of cancer rates that exist across European populations shows that the risk ratio is biased by less than 10% in U.S. studies except under extreme conditions. We note that the bias decreases as the number of ethnic strata increases. Conclusions: There will be only a small bias from population stratification in a well-designed case-control study of genetic factors that ignores ethnicity among non-Hispanic U.S. Caucasians of European origin. Further work is needed to estimate the effect of population stratification within other populations.
Article
• The allelic association of the human D2 dopamine receptor gene with the binding characteristics of the D2 dopamine receptor was determined in 66 brains of alcoholic and nonalcoholic subjects. In a blinded experiment, DNA from the cerebral cortex was treated with the restriction endonuclease Taql and probed with a 1.5-kilobase (kb) digest of a clone (XhD2G1) of the human D2 dopamine receptor gene. The binding characteristics (Kd [binding affinity] and Bmax [number of binding sites]) of the D2 dopamine receptor were determined in the caudate nuclei of these brains using tritiated spiperone as the ligand. The adjusted Kd was significantly lower in alcoholic than in nonalcoholic subjects. In subjects with the A1 allele, in whom a high association with alcoholism was found, the Bmax was significantly reduced compared with the Bmax of subjects with the A2 allele. Moreover, a progressively reduced Bmax was found in subjects with A2/A2, A1/A2, and A1/A1 alleles, with subjects with A2/A2 having the highest mean values, and subjects with A1/A1, the lowest. The polymorphic pattern of the D2 dopamine receptor gene and its differential expression of receptors suggests the involvement of the dopaminergic system in conferring susceptibility to at least one subtype of severe alcoholism.