You are on page 1of 14

Review

For reprint orders, please contact: reprints@futuremedicine.com

Pharmacogenetics of nicotine addiction:


role of dopamine

The neurotransmitter dopamine (DA) plays a central role in addictive disorders, including nicotine addiction.
Specific DA-related gene variants have been studied to identify responsiveness to treatment for nicotine
addiction. Genetic variants in DRD2, DRD4, ANKK1, DAT1, COMT and DBH genes show some promise in
informing personalized prescribing of smoking cessation pharmacotherapies. However, many trials studying
these variants had small samples, used retrospective design or were composed of mainly self-identified
Caucasian individuals. Furthermore, many of these studies lacked a comprehensive measurement of
nicotine metabolism rate, did not assess the roles of sex or the menstrual cycle, and did not investigate
the role of rare variants and/or epigenetic factors. Future work should be conducted addressing these
limitations to more effectively utilize DA genetic information to unlock the potential of smoking cessation
pharmacogenetics.

KEYWORDS: addiction n dopamine n genetic n nicotine n pharmacogenetic n smoking Aryeh I Herman*1,


cessation Elise E DeVito1,
Kevin P Jensen1 &
Mehmet Sofuoglu1
Nicotine addiction continues to be the chief pre- CNS and the mechanism of action of the first- 1
Yale University, School of Medicine,
ventable cause of death in developed countries, line treatments for smoking cessation. Next, Department of Psychiatry & VA
Connecticut Healthcare System,
causing an estimated 435,000 premature deaths we review the genetic variations that directly VA Medical Center, 950 Campbell
in the USA and 5 million deaths worldwide [1,2]. or indirectly influence brain DA as they relate Avenue, West Haven, CT 06516, USA
Quitting smoking is associated with immediate to smoking cessation pharmacogenetics. This *Author for correspondence:
Tel.: +1 203 932 3486 ext. 7432
health benefits, irrespective of age or presence manuscript aims to extend the excellent review Fax: +1 203 937 3472
of smoking-related diseases [3]. Current first-line by David and Munafò in 2008 on DA genetics aryeh.herman@yale.edu
pharmacological treatments for smoking cessa- and smoking cessation [8] by including studies
tion (i.e., nicotine replacement therapy [NRT], that have been published subsequently and by
bupropion and varenicline) increase the chances providing discussion for future directions. Sev-
of quitting smoking two- to three-fold. However, eral recent reviews are available for a broader per-
even when smokers utilize evidence-based cessa- spective of DA neurobiology [9], genetics [10–12]
tion treatments, only 15–25% of those who quit and pharmacogenetics of nicotine addiction [13].
maintain tobacco abstinence for at least 1 year
[201]. Thus, there is a great need to develop more Neurobiology of nicotine addiction
effective treatments for nicotine addiction. The Nicotine, the main addictive chemical in tobacco
development of new treatments requires a better smoke, is essential to continued and compul-
understanding of the individual factors contrib- sive tobacco use [14]. Following a cigarette puff,
uting to the different stages of nicotine addiction. nicotine enters cerebral circulation and binds
Genetic factors are significant contributors to ini- to neuronal nicotinic acetylcholine receptors
tiation and severity of nicotine addiction, as well (nAChRs). nAChRs are ligand-gated ion chan-
as response to treatment [4]. An estimated 50% nels that are permeable to sodium, potassium
of variance in nicotine addiction is explained by and calcium ions. Most nAChRs in the CNS
genetic factors, although the specific genes con- are presynaptically located and modulate the
tributing to this genetic component remain to release of acetylcholine (ACh), DA, serotonin,
be identified [5,6]. glutamate, g-aminobutyric acid (GABA) and
This review summarizes the pharmaco­ norepinephrine [15]. nAChRs can also be post-
genetics of smoking cessation, with a focus on synaptically located, such as on the DA neurons
dopamine (DA), a neurotransmitter that plays in the ventral tegmental area (VTA). The two
a central role in addictive disorders, including most commonly expressed nAChRs in the brain
nicotine addiction [7]. First, we briefly review are a4b2 or a7nAChR [15]. Stimulation of the
the neurobiology of nicotine’s effects in the a4b2 nAChRs located on the DA cell bodies part of

10.2217/PGS.13.246 © 2014 Future Medicine Ltd Pharmacogenomics (2014) 15(2), 221–234 ISSN 1462-2416 221
Review Herman, DeVito, Jensen & Sofuoglu

in the VTA shifts these cells from tonic to pha- changes vary across nAChR subtypes (Figure 1).
sic firing mode, which results in increased DA For example, the nAChR subtypes controlling
release in both the nucleus accumbens and the GABA release (mainly non-a7 subtypes), desensi-
prefrontal cortex. As with other drugs of abuse, tize faster than those controlling glutamate release
this increased DA release in the nucleus accum- (mainly the a7 subtype) [16]. This unequal desen-
bens is thought to mediate the rewarding and sitization may result in reduced GABA release,
pleasurable effects of nicotine, and is thought relative to glutamate, following prolonged nico-
to be a critical mechanism for the initiation and tine exposure, such as in regular cigarette smokers.
maintenance of nicotine addiction. Increased A relative deficiency of GABA over glutamate may
DA release in the prefrontal cortex is thought to lead to an enhanced DA release in the nucleus
be critical in mediating the cognitive-enhancing accumbens and this may be a crucial mechanism
effects of nicotine. that perpetuates nicotine addiction [16].
Besides DA receptors, the nucleus accumbens Another mechanism that contributes to nicotine
also contains GABA and glutamate synapses, reward occurs through nicotine-induced release of
which inhibit and stimulate nicotine-induced the endogenous opioid peptide, b-endorphin [17].
DA release, respectively. Prolonged exposure to b-endorphin stimulates the µ-opioid receptors
nicotine is associated with desensitization and that are located on the GABA interneurons in the
upregulation of nAChRs and the rates of these VTA, resulting in reduced GABA release. This

D2D3D4
D1D5

DA

D2 DAT

MAO
DA DOPAC
α4 2nAChR
TH COMT
Tyrosine HVA

DA
projection
neuron

Glutamatergic
projection
GABAergic α7nAChR neuron
projection
MOR neuron VTA DA Glu
neuron

α4 2nAChR
α4 2nAChR

Figure 1. A simplified illustration of the dopamine neuron in the ventral tegmental area
and the hypothesized effects of nicotinic acetylcholine receptors on the regulation of
dopamine, glutamate and GABA release in the ventral tegmental area and nucleus
accumbens.
DA: Dopamine; DAT: Dopamine transporter; DOPAC: 3,4-dihydroxyphenylacetic acid;
HVA: Homovanillic acid; MOR: µ-opioid receptor; nAChR: Nicotinic acetylcholine receptor;
VTA: Ventral tegmental area.

222 Pharmacogenomics (2014) 15(2) future science group


Nicotine pharmacogenetics & dopamine Review
reduction in GABA levels in the VTA reduces reduces D2 expression, resulting in enhanced
the inhibition on the DA neurons, resulting in DA release, possibly due to decreased inhibitory
enhanced DA release in nucleus accumbens [18]. autoregulation, which is controlled by D2 recep-
tors [25]. Lerman et al. studied -141C Ins/Del in
Mechanism of action of first-line 368 smokers during an open-label 8-week trial
treatments for smoking cessation comparing two forms of NRT (transdermal
There are currently three first-line pharmaco- nicotine vs nicotine nasal spray) and observed
logic treatments for smoking cessation: NRT that the -141C insertion allele improves cessation
(including patch, gum, nasal spray, inhaler and rates independent of NRT type [26]. Dahl et al.
lozenge), bupropion and varenicline [19,201]. [27] extended the findings of Lerman et al. [26]
NRTs, by stimulating nAChRs, release DA in using the same open-label trial with 363 smok-
the mesolimbic pathways, including the nucleus ers, and observed an interactive effect between
accumbens [14]. This increased DA release dur- the -141C allele and having two copies of a neu-
ing smoking abstinence may relieve tobacco ronal calcium sensor-1 protein SNP A allele,
withdrawal, making it easier for smokers trying rs1054879. Among 414 smokers participating
to quit smoking. Bupropion, an atypical anti- in an 8-week double-blind placebo-­controlled
depressant, is effective for smoking cessation. trial of bupropion, those with the -141C inser-
Bupropion blocks the reuptake of norepineph- tion had higher abstinence than those with the
rine and DA in the mesolimbic pathway, and -141C deletion [26]. These findings suggest that
also blocks nAChRs [20]. These effects may genetic variations associated with greater synap-
contribute to attenuation of nicotine’s reward- tic DA availability may predict higher rates of
ing effects and tobacco withdrawal symptoms smoking cessation irrespective of treatment type.
by bupropion. Varenicline is a partial agonist at Lerman et al. also examined the influence of
the a4b2 and a full antagonist at the a7nAChR. DRD2 C957T (rs2283265) on smoking cessa-
Both of these nAChR subtypes are critical in tion outcomes [26]. DRD2 C957T is a synony-
controlling DA release in the brain reward path- mous SNP that modulates striatal D2 binding
ways [21]. Varenicline attenuates nicotine reward in vivo and mRNA stability in vitro, wherein the
when the nAChR are stimulated by nicotine and T allele is associated with higher D2 availability
improves tobacco withdrawal symptoms during [28,29]. Smokers homozygous for the T allele of
abstinence from nicotine [22]. Thus, because of the C957T SNP who were on NRT experienced
its critical role for nicotine reward and with- better smoking outcomes on NRT [26], while
drawal symptoms, DA is thought to play a cen- no genotype effect was observed in the group
tral role in the therapeutic effects of smoking receiving bupropion responses.
cessation pharmacotherapies. The ANKK1 gene contains a functional SNP
previously known as DRD2TaqI (rs1800497).
Dopamine receptors Although this SNP is located 10 kb downstream
DA acts through f ive receptor subtypes from DRD2, individuals who carry the ANKK1
(D1–D5) [23]. The DA receptors are also clas- A1 allele have a 30–40% reduction of striatal D2
sified under two receptor families: D1-like (D1 receptor density compared with those who are
and D5 receptors) and D2-like (D2, D3 and D4 homozygous for the A2 allele [30]. Several stud-
receptors). These receptor families have oppos- ies suggest that ANKK1 A1 and A2 alleles may
ing effects on signal transduction. While stimu- influence smoking cessation outcomes. John-
lation of D1-like receptors activates adenylate stone et al. reported that among 755 heavy smok-
cyclase, stimulation of D2-like receptors inhib- ers who participated in a 12-week double-blind,
its adenylate cyclase. The D2 receptor family randomized controlled trial of NRT, the nico-
also functions as an autoreceptor that reduces tine patch was more effective for smokers who
DA release. While the D1 and D2 receptors are carried at least one ANKK1 A1 allele at week 1
the most commonly expressed DA receptors in [31]. At week 12, smokers who carried both the
the CNS, the D2 and D4 receptors have been ANKK1 A1 allele and DBH 1368A (rs77905)
the foci of studies of DA pharmacogenetics of allele had both the highest cessation rates on
nicotine addiction. NRT and the lowest quit rates on placebo. In a
reana­lysis of a sample that overlapped with John-
„„ DRD2 stone et al.’s. [31] sample, Yudkin et al. observed
In DRD2, a single nucleotide insertion/deletion that women with at least one copy of the ANKK1
in the 5´ promoter region (-141C Ins/Del) alters A1 allele had better response to NRT compared
D2 expression [24]. As such, the deletion variant with women without the A1 allele [32].

future science group www.futuremedicine.com 223


Review Herman, DeVito, Jensen & Sofuoglu

Many smoking cessation pharmacogenetics Collectively, these studies report that DRD2
studies have investigated interactions between may be an important moderator of smoking ces-
bupropion and ANKK1. David et al. studied sation, especially in smokers who have the A2/A2
whether ANKK1 moderated the effects of genotype on bupropion, while those who carry
bupropion in a randomized, double-blind, the A1 allele seem to report more side effects
placebo-­c ontrolled smoking cessation study from the medication [33,34,36,38]. DRD2 and the
with 29 heavy smokers [33]. The greatest reduc- proximal ANKK1 remain potential pharmaco-
tions in craving, anxiety and irritability were genetic targets. The studies conducted seem to
observed in smokers carrying the A2/A2 geno- implicate this region as moderating smoking
type who were on bupropion [33]. This type cessation pharmacotherapies; however, these
of pharmaco­genetic effect was also observed reports contain issues that need to be addressed
in a sample of 218 smokers during a random- in future work. For example, the samples col-
ized, double-blinded, placebo-controlled trial lected were primarily from Caucasian individuals
of bupropion for smoking cessation [34]. Smok- and race was not usually confirmed through the
ers carrying the A2/A2 genotype experienced measurement of genetic ancestry. Additionally,
larger reductions in craving and greater likeli- more stringent correction for multiple testing
hood of abstinence on bupropion. Addition- should be employed. At least three reports on
ally, those who carried the A2/A2 allele plus the pharmacogenetics of smoking cessation and
the T allele from a functional CYP2B6 SNP DRD2 utilize nearly the exact same sample and
(rs3211371) achieved more abstinence [34]. The it is unclear whether stringent statistical methods
same research group also pooled their data [34] were employed to guard against Type I errors.
with pharmacogenetic work from Lerman et al.
[35] for a combined sample of 722 smokers to „„ DRD4
determine whether the ANKK1 SNP rs1800497 The structure and pharmacology of the D4 recep-
moderated treatment response to bupropion for tor is similar to the D2 receptor. The D4 recep-
smoking cessation [36]. The results from the tor is predominantly expressed in the prefrontal
combined sample indicated that those who car- cortex and has been widely examined in relation
ried the A2/A2 genotype and were given bupro- to psychiatric disorders, including nicotine addic-
pion were more likely to be abstinent at the tion. DRD4 contains two polymorphisms that
end of treatment compared with placebo [37]. In have been studied in relation to nicotine phar-
an 8-week open-label study of treatment with macogenetics. The first polymorphism of inter-
bupropion sustained-release with 451 smok- est is located in exon 3, where a 48 bp variable
ers in the pharmacogenetic component of number tandem repeat (VNTR) encodes the
the study, Swan et al. [38] found that smok- third intracellular loop of the receptor [40,41]. The
ers who carried the A1 allele were more likely 7-bp repeat variant displays decreased sensitivity
to report discontinuing bupropion because of to DA and differences in DA binding potential
adverse effects [38]. One clinical trial evaluat- compared with other common variants (2-bp or
ing the effects of an antidepressant medication 4-bp repeats) [42].
(venlafaxine or placebo) plus standard care in In a randomized placebo-controlled trial of
134 smokers observed that those carrying the NRT with 720 smokers, David et al. found
ANKK1 A2/A2 genotype quit significantly that those who had at least one copy of ≥7-bp
more often and experienced a significant reduc- repeats were less successful in quitting smok-
tion in reduced negative mood symptoms com- ing at 12 weeks than those with no copies of
pared with those with the A1 allele, regardless ≥7-bp repeats, irrespective of treatment assign-
of medication condition [39]. The mechanisms ment [43]. However, this genetic effect on out-
by which ANKK1 moderates differential treat- comes dissipated at 26 weeks. Leventhal et al.
ment response to NRT and bupropion have not studied 331 smokers in a double-blind placebo-
been elucidated. The reduced D2 receptor den- controlled 12-week randomized trial of treat-
sity associated with the A1 allele may enhance ment with bupropion [44]. Smokers who were
DA release by the reduction of D2 autorecep- treated with bupropion and had at least one copy
tors, potentially alleviating tobacco withdrawal of ≥7-bp repeats, compared with those with-
severity among those who are treated with out any ≥7-bp repeat copies, were more likely
NRT. By contrast, the A1 allele may enhance to quit smoking at 2, 6 and 12 months after
bupropion’s DA-enhancing effects and lead to treatment ended [44]. In another study, Bergen
increased adverse effects. These possibilities et al. [45] utilized data from the Lerman et al.
remain to be examined in future studies. [35] double-blind pharmacogenetic efficacy trial,

224 Pharmacogenomics (2014) 15(2) future science group


Nicotine pharmacogenetics & dopamine Review
which randomized 416 smokers to active or pla- paired with smoking cessation counseling [51],
cebo bupropion [45]. While they did not observe those who carried the 9-repeat allele (compared
statistically significant effects of genotype or with those with other alleles) had poorer smok-
genotype by treatment interactions, they con- ing cessation outcomes at 12 months. A separate
ceded that the lack of pharmacogenetic effects in randomized placebo-controlled clinical trial on
their study may have been owing to insufficient 291 smokers found no effect of DAT1 VNTR on
sample size. bupropion’s efficacy for smoking cessation [34].
A second relevant DRD4 variation is a C-to- The results of the DAT smoking cessation
T transition, located 521-bp upstream from pharmacogenetic studies are equivocal. The
the transcription start site, wherein the T allele sample sizes of these studies are modest, but the
reduces transcriptional efficacy by 40% com- studies are more numerous compared with DA
pared with the C allele. One study investigat- smoking cessation pharmacogenetic studies,
ing this SNP in 720 smokers in a double-blind, looking at other genetic regions (DRD2, COMT
randomized, placebo-controlled trial of NRT and so on). DAT1 remains a good candidate
observed no pharmacogenetic effects [43,46]. gene for moderating the therapeutic effects of
The D4 receptor plays a critical role in the smoking cessation medications and should be
neurobiology of DA transmission and, there- of particular interest in relation to bupropion,
fore, the rationale of studying DRD4 variation given bupropion’s capacity to inhibit DAT. As
as a moderator of smoking cessation pharmaco­ with the rest of this literature on DA pharmaco-
therapies is sound. However, the results from genetics for tobacco, the majority of the available
existing clinical trials generally do not support literature is in Caucasian individuals, so it is not
roles for these most well-characterized DRD4 currently known whether these effects generalize
polymorphisms in response to smoking cessation to non-Caucasians smokers.
pharmacotherapy (Table 1).
DA synthesis & inactivation: TH,
„„ DAT1 (SLC6A3) DBH, MAO & COMT genes
The DA transporter (DAT) terminates DA Tyrosine hydroxylase converts tyrosine to
neuro­ t ransmission in subcortical regions dihydroxy­phenylalanine, the rate-limiting step
through reuptake of synaptic DA to the pre- for DA biosynthesis. MAO and COMT con-
synaptic neurons. DAT is the site of action for vert DA into homovanillic acid. Both MAO and
stimulant drugs such as cocaine, amphetamine COMT are found in monoamine neurons and
and methylphenidate. DAT1, composed of 15 glial cells. DBH converts DA to norepineph-
exons on chromosome 5p15.3, has few coding rine in the synaptic vesicles of the noradrener-
region variants. Therefore, most DAT1 studies gic neurons [52], thereby causing synaptic DA
have focused on noncoding SNPs and VNTRs levels to increase, relative to norepinephrine.
[47,48]. The 3´-UTR contains a VNTR varying Among these genes important for DA synthesis
between 3- and 12-bp repeats, with the most fre- and inactivation, only COMT and DBH have
quently observed alleles containing 9- and 10-bp been examined in pharmacogenetic studies for
repeats. The 10-bp repeat allele has been associ- nicotine addiction.
ated with an abnormally active DAT, eliciting
increased reuptake of DA and DA degradation, „„ COMT
leading to reduced DA transmission [49]. COMT contains a well-studied missense
In a placebo-controlled trial, this VNTR vari- SNP (Val158Met) that results in the presence
ation did not moderate bupropion’s smoking ces- of methionine (Met) or valine (Val) at codon
sation effects in 418 smokers [35]. However, the 158 in the membrane-bound COMT. The Val
same study reported that smokers who carried allele is three- to four-times more active than
both the 9-repeat allele and the ANKK1 A2 allele the Met-coded allele, resulting in reduced DA
had longer successful abstinence before relaps- levels in the synapse. In a clinical trial of 290
ing to smoking, regardless of treatment condi- Caucasian and black female smokers, those
tion [35]. O’Gara et al. conducted an open-label with the Met/Met genotype were more likely
trial of bupropion versus NRT treatment, that have prolonged abstinence in response to NRT
both included behavioral support, in 583 smok- treatment compared with women who carried
ers and observed that the 9-repeat VNTR was the Val/Val genotype [53]. These results were
associated with higher rates of quitting smok- replicated in a randomized placebo-controlled
ing with either treatment [50]. By contrast, in an NRT trial, where (n = 749) Caucasian smokers
open-label study of 416 smokers using bupropion with the Met/Met allele were more likely to be

future science group www.futuremedicine.com 225


Table 1. Pharmacogenetic and genetic effects of dopamine-related genes on smoking-cessation outcomes.

226
DA system Gene Allelic variant Genetic Medication Pharmaco- Summary
genetic
Review

D2 receptor DRD2 -141C + NRT - Genetic effects were found for -141C and C957T in the context of NRT and bupropion trials.
Insertion (vs deletion) variant of -141C had better smoking outcomes [26] , but in a second
Bupropion - study this effect was only observed when -141C insertion was paired with rs1054879A [27] .
The TT (vs CT or CC) variant of C957T had better smoking outcomes in individuals receiving
C957T + NRT -
NRT but not among those receiving bupropion [26] . Since no formal medication by genotype
Bupropion - interactions were observed, direct evidence for pharmacogenetic effects of these DRD2-
related allelic variants for NRT or bupropion is lacking
ANKK1 rs1800497 + NRT + Differential pharmacogenetic effects of rs1800497 have been observed for NRT and
bupropion. NRT smoking outcomes were better for smokers with A1 alleles in mixed-gender
Bupropion + or female-only analyses of an overlapping sample [31,32] . By contrast, bupropion smoking
outcomes were better for smokers with A2/A2 alleles [33,34,36] . A study of venlafaxine found
Venlafaxine -
no pharmacogenetic effects, yet found genetic effects favoring smoking outcomes in A2/A2
smokers [39]
D4 receptor DRD4 VNTR + NRT - Differential genetic effects of DRD4 VNTR were observed for NRT and bupropion. In an NRT
trial [43] , smokers with <7 bp had better smoking outcomes. However, in the context of
Bupropion - bupropion trials, smokers with ≥7 bp had better [43] or a nonsignificant tendency towards
Herman, DeVito, Jensen & Sofuoglu

better [44] outcomes. No evidence of genetic or pharmacogenetic effects of DRD4 C-to-T


C-to-T - NRT -
variant was observed on smoking outcomes with NRT [43,46] . No formal medication by
genotype interactions were reported, so evidence of DRD4-related pharmacogenetic effects is
limited
Dopamine DAT1 VNTR + NRT - Although one study found better outcomes for smokers with 9 bp VNTR in an NRT trial [50] ,
transporter the findings in bupropion trials are mixed. Bupropion trials have found evidence for 9 bp
(DAT) carriers having better [50] , worse [51] or equivalent [34,35] smoking outcomes compared with

Pharmacogenomics (2014) 15(2)


other VNTRs, or better outcomes when smokers are also carriers of the ANKK1 A2 allele [35] .
Bupropion -
Inclusion of variable control conditions (e.g., open-label, placebo, counselling) may have
contributed to this variation in findings. No formal medication by genotype interactions were
reported in support of DAT1-related pharmacogenomic effects on NRT or bupropion
Dopamine COMT Val158Met + NRT + Findings are mixed regarding which variants are beneficial for smoking outcomes. Two studies
inactivation including a mixed-race female only sample [53] or Caucasian-only mixed-sex sample [54] found
Bupropion - better outcomes for Met/Met carriers on NRT. However, in a Chinese-only sample, Val/Val
was associated with better NRT outcomes [55] . Similarly, a trend towards better outcomes for
rs165599 - Bupropion + Val/Val carriers on bupropion was reported in a Korean-only sample [59] . In a mixed-race,
mixed-sex sample, genetic, but not pharmacogenetic, effects were found with Val/Val
rs737865 - Bupropion - experiencing worse withdrawal symptoms [56] . For rs165599, A carriers had better outcomes
on bupropion [58] . A subset of findings support pharmacogenetic effects of COMT on NRT
and bupropion. The variable findings may be partially attributable to race/ethnicity and sex
differences across the samples
DBH DBH1368 - NRT + The only study assessing DBH1368 found better outcomes on NRT for A (versus G) carriers,
but this effect was conditional on the presence of the ANKK1 Taq1A allele [31]

future science group


Pharmacogenetic effects represent gene variant by medication interactions on better smoking outcomes; genetic effects represent gene variant main effects on better smoking outcomes. C957T is also known as
rs2283265. rs1800497 is also known as Taq1. DAT1 is also known as SLC6A3. ‘+’ indicates evidence in support of an effect; ‘-‘ indicates that despite studies testing this relationship, no effect was indicated.
DA: Dopamine; NRT: Nicotine replacement therapy; VNTR: Variable number tandem repeats.
Nicotine pharmacogenetics & dopamine Review
abstinent than smokers carrying at least one Val the 1368 DBHA allele, combined with ANKK1
allele, regardless of treatment condition [54]. By Taq 1A, experienced better NRT efficacy after
contrast, in a double-blind, placebo-controlled, 12 weeks of treatment [31].
8-week trial of NRT with 250 Chinese smok- Genes that control DA synthesis and inac-
ers, those with the Val/Val genotype, compared tivation remain particularly compelling as
with those who carried at least one Met allele, pharmaco­genetic moderators of smoking cessa-
had better abstinence rates with NRT [55]. tion. Studying the pharmacogenetics of COMT
Recently, we examined whether COMT mod- in a female-only sample [53] is important given
erates behavioral responses to intravenously that COMT exhibits sexual dimorphisms regu-
administered nicotine in 124 black and Cau- lated by estrogens [61] and yet the study did not
casian smokers. Smokers who carried the Val/ go far enough. Because key hormone levels, such
Val genotype experienced more negative subjec- as estrogen and progesterone, differ between
tive effects from nicotine and had higher with- males and females, as well as within females
drawal severity following overnight abstinence throughout their menstrual cycle, future work
from smoking [56]. These findings are consistent will require controlling for sex and menstrual
with other human studies suggesting that the cycle effects. Additionally, COMT effects are
Val/Val genotype, by reducing the tonic DA differentially regulated by ethnicity, and there-
levels, may lead to greater withdrawal severity fore collecting representative samples from dis-
[53]. By contrast, greater phasic DA release asso- tinct genetic ancestries may further disentangle
ciated with the Val/Val genotype may lead to this literature.
greater subjective drug effects from nicotine [57]. To our knowledge, there have been no smok-
The combination of greater withdrawal sever- ing cessation pharmacogenetic studies that have
ity and enhanced drug effects from nicotine investigated MAOA, MAOB or TH variations,
may provide mechanisms by which COMT may and there is scant data from DBH. Perhaps even a
modulate nicotine dependence phenotypes. retrospective ana­lysis in some of the larger smok-
COMT variation has been associated with ing cessation trials where DNA was collected,
bupropion efficacy for smoking cessation. Ber- along with appropriate correction for multiple
rittini et al. studied the Val/Met polymorphism testing, could clarify whether polymorphisms
and two additional SNPs (rs737865 [located in in these genetic regions are worthwhile pursuing
the first intron of the MB-COMT transcript] as clinically meaningful moderators of smoking
and rs165599 [near the 3´-UTR]) that have cessation pharmacotherapies.
shown evidence for differential allelic expres-
sion during a double-blind, placebo-controlled, Race & ethnicity
10-week trial of bupropion in 430 black sub- The majority of smoking cessation pharmaco­
jects and Caucasian smokers. Quit rates were genetic studies include predominantly Cau-
better for smokers who carried the rs165599 casian samples, despite race and ethnicity
GG genotype on placebo, but were better for A appearing to be related to multiple aspects of
genotype carriers on bupropion [58]. In a sample nicotine addiction. For example, in the USA,
of 250 Korean smokers who participated in an black subjects smoke fewer cigarettes, compared
open-label bupropion trial, Han et al. found the with Caucasians, but have higher levels of nico-
Val/Val genotype was more commonly associ- tine dependence. Compared with Caucasians,
ated with smoking abstinence [59]. The authors black smokers are more likely to attempt to,
cautioned that the pharmacogenetic effect did but less likely to succeed in, quitting smok-
not withstand correction for multiple testing. ing [62,63]. Asian individuals from China have
smoking prevalence rates of 28.1%, yet along
„„ DBH with Latino individuals, have the lowest rates
DBH contains multiple well-characterized poly- of lung cancer [64]. Genetic variation could, in
morphisms that explain significant proportions part, help explain why individuals from different
of DBH activity. DBH1368 is in tight linkage races and ethnicities have significantly different
disequilibrium to a G-to-A substitution associ- rates of smoking and health-related outcomes.
ated with higher DBH levels [60]. Increased DBH Importantly, many pharmacogenetic studies
activity results in a greater amount of DA being rely on self-reported race as a proxy for ances-
converted to norepinephrine. Johnstone et al. try, which may not be a reliable approach. To
tested whether DBH1368 variants moderate improve the accuracy of ancestry determination,
responses to transdermal NRT [31]. The results self-reported race should be verified with ances-
of this study indicated that smokers who carried try informative markers [65]. Failure to account

future science group www.futuremedicine.com 227


Review Herman, DeVito, Jensen & Sofuoglu

for population structure in a sample of mixed In a recent study, we observed that female
ancestry can result in a high false-discovery rate smokers (n = 45), relative to males (n = 115) had
or reduce the power of the study [66,67]. greater physiological responses to, yet dimin-
Research as early as the 1950s reported ished subjective drug effects from intravenous
racial/ethnic differences in drug response. nicotine administration. Among women, smok-
One of the most well-studied examples comes ers who were in the luteal phase (progesterone
from research in cardiovascular disease. In the dominant), showed attenuated subjective drug
early 1980s, clinical observations reported that effects and better cognitive performance rela-
response to antihypertensive drugs differed by tive to women who were in the follicular phase
race such that self-identified Caucasian individu- (estradiol dominant) [74]. Our findings are con-
als experienced higher response to b-adrenergic sistent with the activation and inhibition of the
receptor antagonists (i.e., b-blockers) compared DA reward pathway by estradiol and progester-
with self-identified black subjects [68]. Subse- one, respectively [75]. Our findings also suggest
quently, researchers have observed functional that cyclic changes in estradiol and progester-
genetic variations in the genes encoding the one levels that occur across the menstrual cycle
b-adrenergic receptors, which are the targets of may contribute to the observed sex differences
this class of drugs [69]. The same variants often in nicotine’s effects. These findings offer several
show marked differences in frequency among candidate mechanisms through which the luteal
world populations, such that individuals of phase, compared to the follicular phase, may
primarily Caucasian ancestry are more likely be protective against vulnerability to smoking.
to carry the alleles that are more responsive Other studies have also reported that the phase
to b-adrenergic receptor antagonists at these of the menstrual cycle may influence smoking
sites in comparison with individuals of black cessation outcomes [76]. Most of the smoking ces-
ancestry [70]. sation pharmacogenetic studies published to date
have not included sex hormones as covariates in
Sex & the menstrual cycle their analyses. Introducing these biological mea-
Women respond less favorably to smoking sures in subsequent trials could further clarify
cessation treatments [71], even though they the role of sex differences in pharmacogenetic
maintain their nicotine addiction with lower smoking cessation studies.
levels of nicotine intake than men [72]. Data
from multiple smoking cessation clinical trials Nicotine metabolism
have suggested that sex may critically modu- Nicotine is inactivated in the liver to its main
late pharmacogenetic effects. For example, metabolite, cotinine, primarily by the CYP2A6
women, but not men, with at least one A1 allele microsomal enzyme [77,78]. The same enzyme
experienced greater benefits from transdermal oxidizes cotinine to 3´-hydroxycotinine. The
NRT, compared with women homozygous for gene encoding the CYP2A6 enzyme is highly
the A2 allele [32]. In another study, Swan et al. polymorphic, and multiple functional allelic
evaluated the role of ANKK1 in an open-label, variations and SNPs have been described [79].
randomized effectiveness trial comparing low Many studies have demonstrated that the ratio
and high doses of bupropion [38]. Compared of plasma 3´-hydroxycotinine /cotinine corre-
with women homozygous for the A2 allele, lates with nicotine clearance [80,81]. Because this
women with at least one A1 allele were less nicotine metabolite ratio correlates with nico-
likely to quit smoking with bupropion treat- tine clearance and CYP2A6 genotype [79,82], it is
ment [38,53]. However, this genotype effect on considered to be a marker of CYP2A6 enzyme
treatment response was not observed in men. activity [81].
Furthermore, in a smoking cessation trial of The nicotine metabolite ratio may also be pre-
transdermal nicotine and nicotine nasal spray, dictive of smoking cessation outcomes. Smokers
the presence of the Met allele of COMT facili- in the lowest ratio quartile (slower metaboliz-
tated smoking cessation in women [53]. Addi- ers), compared with those in higher quartiles,
tionally, females with the Val/Val genotype show better smoking cessation outcomes with
experienced greater difficulty in concentrat- transdermal NRT [83]. In another clinical trial
ing and reported more irritability than men that tested the efficacy of bupropion for smok-
who were in either Val/Val or Met genotype ing cessation, smokers in the fourth quartile
groups [56]. In contrast to these findings, other (faster metabolizers) had poor smoking ces-
DA pharmacogenetic studies did not report sex sation rates if they were assigned the placebo
differences [44,54,73]. treatment relative to slower metabolizers;

228 Pharmacogenomics (2014) 15(2) future science group


Nicotine pharmacogenetics & dopamine Review
however, bupropion lowered the relapse liability a small portion of phenotypic variance (~1%)
among ‘fast’ metabolizing smokers [84]. Overall, [92,93]. Rare variants, by conferring a moderate
these findings are consistent with the idea that but readily detectable, increase in relative risk
smokers who rapidly metabolize nicotine might may help to account for the unexplained varia-
be more dependent and have greater difficulty tion in gene function and nicotine dependence
with smoking cessation [85]. The underlying phenotypes, including response to pharma-
mechanisms that associate nicotine metabolite cological treatments. Identification of the rare
ratio with treatment outcomes have not been variants are expected to accelerate in the near
fully elucidated. In a recent study, we have future at pace with enhancement of whole-exome
shown that higher nicotine metabolite ratio and whole-genome sequencing techniques [74,75].
(i.e., ‘faster’ metabolizing) was associated with Several recent publications addressed the possible
a greater craving for cigarettes and a greater contribution of rare variants in addictive disor-
reward from intravenous nicotine following ders [94–96]. Wang et al. elegantly describes how
overnight abstinence in smokers [86]. These to integrate rare variants and deep sequencing in
results suggest that enhanced nicotine reward the context of drug addiction research [97].
and cigarette craving may contribute to the poor
treatment response in smokers with a high nico- „„ Epigenetic regulatory effects
tine metabolite ratio. Studies are underway to The effects of DA gene variants might be
test the utility of the nicotine metabolite ratio modulated by epigenetic mechanisms. Under-
for smoking cessation. standing these important effects might help
to clarify the role of dopamine in the pharma-
„„ Genome-wide association studies cogenetics of nicotine addiction and lead to
Genome-wide association studies (GWAS) better treatments. Epigenetic effects involve
have proven to be a useful technique in inves- changes to chromatin structure caused by
tigating the role of genetic variations across direct covalent modifications of DNA or fac-
the genome for multiple phenotypes associated tors that interact with DNA, such as histones.
with nicotine dependence phenotypes, includ- These changes to DNA structure might influ-
ing vulnerability to initiation and maintenance ence the spatial or temporal pattern of gene
of smoking, smoking-related diseases, and expression. Exposure to addictive drugs [98,99]
ability to quit smoking [87]. GWAS approach and early life stress [100] have effects on chroma-
has also been instrumental in discovering tin structure, including some effects that are
novel variants that had not previously been stable with long-term consequences on brain
linked with nicotine dependence phenotypes. function and behavior. Exposure to nicotine,
For example, several studies have implicated specifically, may influence vulnerability to the
a cluster of three nAChR genes in smoking development of drug dependence through epi-
behaviors, such as the number of cigarettes genetic mechanisms. In rodent studies, nico-
smoked per day [88] and nicotine dependence tine acts as a chromatin modifier by reducing
[89] . One of the SNPs located in CHRNA5 the expression of mRNA from multiple his-
D398N (rs16969968) is a particularly attrac- tone methyltransferase genes and the level of
tive pharmacogenetic target because it may dimethylated histone H3 lysine 9 (H3K9me2)
regulate DA-mediated reward signaling [90]. A [101]. Gozen et al. showed that a 15‑day nicotine
large study reporting on a meta-analyses of sev- exposure induced DRD1 mRNA expression
eral smoking phenotypes within cohorts of the in the prefrontal cortex [102]. The increase in
Tobacco and Genetics Consortium observed DRD1 mRNA expression occurred with a con-
that a SNP located near DBH on chromosome comitant increase in the fraction of acetylated
9q34 was significantly associated with smoking histone H4 at the DRD1 gene promoter [102]. In
cessation [91]. GWAS approach has not been a rodent model, Levine et al. investigated the
widely used for the pharmacogenetics of smok- effects of nicotine pretreatment on response to
ing cessation and remains an area of potential cocaine and found that nicotine increased the
focus in the future. level of acetylated histone H3 and H4 [103]. The
increased levels of histone acetylation corre-
„„ Rare variants lated with increased sensitivity to the addictive
The candidate gene variants reviewed in this properties of cocaine. These findings may have
article, and the ones identified by newer GWAS important clinical implications given the fre-
approaches, are considered to be ‘common vari- quent comorbidity of nicotine with substance
ants’. Common genetic variation explains only dependence.

future science group www.futuremedicine.com 229


Review Herman, DeVito, Jensen & Sofuoglu

Conclusion findings could not be replicated by other research-


Many studies have examined the contribution of ers. Issues related to multiple testing [104], identi-
DA-related genes to smoking cessation outcomes. fying better endophenotypes and using standard-
The genes that have been evaluated include ized outcome assessments have been discussed
DRD2, DRD4, COMT and DBH. Generally, elsewhere [105]. It is important to emphasize that
the studies mentioned in this review had small nicotine addiction is a complex multifactorial
samples (ranging from 29 to 755). Many initial disorder. Drug treatment for smoking cessation

Executive summary
Clinical relevance of pharmacogenetic effects of treatments for nicotine dependence tobacco
ƒƒ Despite dire health consequences of nicotine addiction and sizeable immediate health benefits of quitting smoking, the majority of
cigarette smokers who attempt to quit are unsuccessful.
ƒƒ Several pharmacotherapies for nicotine dependence have shown promise, but are not effective for all individuals.
ƒƒ Consideration of the effects of genetic variants on the efficacy of nicotine pharmacotherapies may provide clinical guidance on which
of the available pharmacotherapies are most likely to aid a given individual’s attempt to quit.
ƒƒ Understanding these mechanisms of individual variation in treatment efficacy may guide development of novel pharmacotherapies.
Dopaminergic genes mechanistically important to nicotine addiction
ƒƒ Dopamine plays a central role in nicotine addiction.
ƒƒ When nicotine binds to neuronal nicotinic acetylcholine receptors (e.g., subtype a4b2), dopamine is released, along with other
neurotransmitters.
ƒƒ Nicotine-induced dopamine release, in regions including the nucleus accumbens, is thought to be a critical mechanism for initiation and
maintenance of nicotine addiction.
ƒƒ Given the centrality of dopamine to nicotine addiction, we review pharmacogenetic studies focusing on genes with known impact on
the dopaminergic system, including gene variants influencing the function of subtypes (e.g., D2, D4) of dopamine receptors (DR) or
gene variants affecting the inactivation of dopamine.
Key dopamine-related polymorphisms that may moderate nicotine addiction pharmacotherapies
ƒƒ DRD2 C957T (rs2283265), a synonymous SNP, modulates striatal dopamine D2 receptor binding and mRNA stability.
ƒƒ DRD2 (-141C Ins/Del) a single nucleotide insertion/deletion in the 5´ gene promoter region alters D2 expression. The deletion variant
reduces D2 expression, leading to greater dopamine release.
ƒƒ ANKK1 (rs1800497) A1 allele reduces D2 receptor density 40% compared with presence of the homozygous A2 allele.
ƒƒ COMT (rs4680) Val158Met polymorphism influences dopamine inactivation, with the valine (Val) variant catabolizing dopamine at up
to four-times the rate of its methionine (Met) variant.
Evidence indicating clinical meaningful nicotine addiction pharmacogenetic effects
ƒƒ Multiple studies report that smokers who carry the DRD2 A2 allele had better smoking cessation outcomes and fewer side effects
while on bupropion, relative to those who carry the DRD2 A1 allele.
ƒƒ Reports show pharmacogenetic effects between nicotine replacement therapy and COMT genotype. However, two nicotine
replacement trials found better outcomes with smokers who carried the Met/Met genotype and two found better outcomes for
Val/Val.
ƒƒ Several other dopamine-related genetic polymorphisms (DRD4 VNTR, DAT1 VNTR and so on) have been investigated in the context of
nicotine addiction pharmacogenetic trials with equivocal findings.
Considerations during future nicotine addiction pharmacogenetic trials
ƒƒ Future work of innovating personalized medicine approaches for nicotine addiction will probably require more nuanced research than
simply studying gene variation and drug together during a clinical trial.
ƒƒ Hidden genetic admixture among study participants could facilitate Type I and Type II errors. As such, future work should incorporate
ancestral genotype when working with genetically heterogeneous groups of people.
ƒƒ Given the neuroactive effects of estradiol and progesterone on systems relevant to nicotine addiction, future nicotine addiction
pharmacogenetic studies may consider stratifying groups by sex and menstrual cycle phase.
ƒƒ Nicotine metabolite ratio may also be predictive of smoking cessation outcomes and should be a covariate in subsequent analyses.
ƒƒ Genome-wide association studies and rare variants have not been widely studied in the pharmacogenetics of smoking cessation and
remain areas of potential focus in the future.
ƒƒ Dopamine gene variants might be modulated by epigenetic mechanisms, which are only beginning to be understood.
Conclusion
ƒƒ Gene variants in the dopamine system remain extremely relevant in the search to personalize nicotine pharmacotherapies.
ƒƒ Unlocking the full potential of personalized treatment is possible, but will probably require the careful consideration of additional
stratifying variables such as sex, menstrual cycle phase, ethnicity, nicotine metabolite ratio and epigenetic regulatory effects.

230 Pharmacogenomics (2014) 15(2) future science group


Nicotine pharmacogenetics & dopamine Review
aims to reduce the symptoms of nicotine absti- Future perspective
nence to make it easier for a smoker to quit smok- Cumulating evidence suggests that race/ethnicity,
ing. Current pharmacological smoking cessation sex, menstrual cycle phase, nicotine metabolism
treatments have limited efficacy and there is room rate, rare genetic variants and epigenetic regula-
for improvement. Pharmacogenetic smoking tion may all contribute to individual differences in
cessation studies targeting the DA system have nicotine addiction and may need to be considered
a high potential to help increase an individual’s in nicotine pharmacogenetic studies.
ability to quit smoking, and reduce the likelihood
of adverse effects from medications. Smoking ces- Financial & competing interests disclosure
sation pharmacogenetic data published thus far This research was supported by the Veterans Administration
has not been robust enough to translate directly Mental Illness Research, Education and Clinical Center
to clinical treatment settings. Unlocking the full (MIRECC) and NIH grants R03 DA027474, and
potential of personalized treatment is possible, but K12-DA-031050 (EE DeVito). M Sofuoglu served as an
will probably require the careful consideration of expert witness on behalf of Pfizer in lawsuits related to
additional stratifying variables such as sex, men- varenicline. The authors have no other relevant affiliations
strual cycle phase, ethnicity, nicotine metabolite or financial involvement with any organization or entity
ratio and epigenetic regulatory effects. Once these with a financial interest in or financial conflict with the
effects are successfully harmonized with the avail- subject matter or materials discussed in the manuscript
able data from well-established pharmacogenetic apart from those disclosed.
clinical trials, cigarette smoking’s societal burden No writing assistance was utilized in the production of
may be drastically reduced. this manuscript.

8 David SP, Munafo MR. Genetic variation in circuits. Semin. Cell Dev. Biol. 20(4),
References the dopamine pathway and smoking 432–440 (2009).
1 Centers for Disease Control and Prevention cessation. Pharmacogenomics 9(9), 1307–1321 17 Hadjiconstantinou M, Neff NH. Nicotine
(CDC). Smoking-Attributable Mortality, (2008). and endogenous opioids: neurochemical and
Years of Potential Life Lost, and Productivity 9 Tritsch NX, Sabatini BL. Dopaminergic pharmacological evidence. Neuropharmacology
Losses – United States, 2000–2004. MMWR modulation of synaptic transmission in cortex 60(7–8), 1209–1220 (2011).
Morb. Mortal. Wkly Rep. 57(45), 1226–1228 and striatum. Neuron 76(1), 33–50 (2012). 18 Walters CL, Cleck JN, Kuo YC, Blendy JA.
(2008).
10 Ho MK, Tyndale RF. Overview of the Mu-opioid receptor and CREB activation are
2 Thorne SL, Malarcher A, Maurice E, pharmacogenomics of cigarette smoking. required for nicotine reward. Neuron 46(6),
Caraballo R. Cigarette smoking among adults- Pharmacogenomics J. 7(2), 81–98 (2007). 933–943 (2005).
United States, 2007 (Reprinted from
11 Kendler KS, Neale MC, Sullivan P, Corey 19 Herman AI, Sofuoglu M. Comparison of
MMWR, 57, 1221–1226, (2008)). JAMA
301(4), 373–375 (2009). LA, Gardner CO, Prescott CA. A population- available treatments for tobacco addiction.
based twin study in women of smoking Curr. Psychiatry Rep. 12(5), 433–440 (2010).
3 Godtfredsen NS, Holst C, Prescott E, Vestbo initiation and nicotine dependence. Psychol. 20 Mooney ME, Sofuoglu M. Bupropion for the
J, Osler M. Smoking reduction, smoking Med. 29(2), 299–308 (1999).
cessation, and mortality: a 16‑year follow-up of treatment of nicotine withdrawal and craving.
12 Maes HH, Sullivan PF, Bulik CM et al. Expert Rev. Neurother. 6(7), 965–981 (2006).
19,732 men and women from the Copenhagen
Centre for Prospective Population Studies. Am. A twin study of genetic and environmental 21 Coe JW, Brooks PR, Vetelino MG et al.
J. Epidemiol. 156(11), 994–1001 (2002). influences on tobacco initiation, regular Varenicline: an alpha4beta2 nicotinic receptor
tobacco use and nicotine dependence. Psychol. partial agonist for smoking cessation. J. Med.
4 Li MD, Cheng R, Ma JZ, Swan GE. Med. 34(7), 1251–1261 (2004).
A meta-ana­lysis of estimated genetic and Chem. 48(10), 3474–3477 (2005).
13 Schnoll RA, Johnson TA, Lerman C. 22 Sofuoglu M, Herman AI, Mooney M, Waters
environmental effects on smoking behavior in
male and female adult twins. Addiction 98(1), Genetics and smoking behavior. Curr. AJ. Varenicline attenuates some of the
23–31 (2003). Psychiatry Rep. 9(5), 349–357 (2007). subjective and physiological effects of
14 Benowitz NL. Pharmacology of nicotine: intravenous nicotine in humans.
5 Sullivan PF, Kendler KS. The genetic
epidemiology of smoking. Nicotine Tob. Res. addiction, smoking-induced disease, and Psychopharmacology 207(1), 153–162 (2009).
1(Suppl. 2), S51–S57; discussion S69–S70 therapeutics. Annu. Rev. Pharmacol. Toxicol. 23 Zhu H, Clemens S, Sawchuk M, Hochman S.
(1999). 49, 57–71 (2009). Unaltered D1, D2, D4, and D5 dopamine
15 Dani JA, Bertrand D. Nicotinic acetylcholine receptor mRNA expression and distribution
6 Hoekstra RA, Bartels M, Boomsma DI.
Longitudinal genetic study of verbal and receptors and nicotinic cholinergic in the spinal cord of the D3 receptor
nonverbal IQ from early childhood to young mechanisms of the central nervous system. knockout mouse. J. Comp. Physiol. A
adulthood. Learn. Individ. Differ. 17(2), Annu. Rev. Pharmacol. Toxicol. 47, 699–729 Neuroethol. Sens. Neural Behav. Physiol.
97–114 (2007). (2007). 194(11), 957–962 (2008).
16 Mansvelder HD, Mertz M, Role LW. 24 Arinami T, Gao M, Hamaguchi H, Toru M.
7 Dani JA. Roles of dopamine signaling in
nicotine addiction. Mol. Psychiatry 8(3), Nicotinic modulation of synaptic A functional polymorphism in the promoter
255–256 (2003). transmission and plasticity in cortico-limbic region of the dopamine D2 receptor gene is

future science group www.futuremedicine.com 231


Review Herman, DeVito, Jensen & Sofuoglu

associated with schizophrenia. Hum. Mol. bupropion clinical trial. Health Psychol. 22(5), mechanism. Eur. J. Pharmacol. 554(2–3),
Genet. 6(4), 577–582 (1997). 541–548 (2003). 128–136 (2007).
25 Sweitzer MM, Donny EC, Hariri AR. 36 David SP, Strong DR, Munafo MR et al. 48 Zhu J, Apparsundaram S, Dwoskin LP.
Imaging genetics and the neurobiological Bupropion efficacy for smoking cessation is Nicotinic receptor activation increases [3H]
basis of individual differences in vulnerability influenced by the DRD2 Taq1A dopamine uptake and cell surface expression
to addiction. Drug Alcohol Depend. polymorphism: ana­lysis of pooled data from of dopamine transporters in rat prefrontal
123(Suppl. 1), S59–S71 (2012). two clinical trials. Nicotine Tob. Res. 9(12), cortex. J. Pharmacol. Exp. Ther. 328(3),
26 Lerman C, Jepson C, Wileyto EP et al. Role 1251–1257 (2007). 931–939 (2009).
of functional genetic variation in the 37 David SP, Johnstone EC, Murphy MF et al. 49 Mill J, Asherson P, Browes C, D’Souza U,
dopamine D2 receptor (DRD2) in response Genetic variation in the serotonin pathway Craig I. Expression of the dopamine
to bupropion and nicotine replacement and smoking cessation with nicotine transporter gene is regulated by the 3´ UTR
therapy for tobacco dependence: results of replacement therapy: new data from the patch VNTR: evidence from brain and lymphocytes
two randomized clinical trials. in practice trial and pooled analyses. Drug using quantitative RT-PCR. Am. J. Med.
Neuropsychopharmacol. 31(1), 231–242 Alcohol Depend. 98(1–2), 77–85 (2008). Genet. 114(8), 975–979 (2002).
(2006). 38 Swan GE, Valdes AM, Ring HZ et al. 50 O’Gara C, Stapleton J, Sutherland G et al.
27 Dahl JP, Jepson C, Levenson R et al. Dopamine receptor DRD2 genotype and Dopamine transporter polymorphisms are
Interaction between variation in the D2 smoking cessation outcome following associated with short-term response to
dopamine receptor (DRD2) and the neuronal treatment with bupropion SR. smoking cessation treatment. Pharmacogenet.
calcium sensor-1 (FREQ) genes in predicting Pharmacogenomics J. 5(1), 21–29 (2005). Genomics 17(1), 61–67 (2007).
response to nicotine replacement therapy for 39 Cinciripini P, Wetter D, Tomlinson G et al. 51 Swan GE, Jack LM, Valdes AM et al. Joint
tobacco dependence. Pharmacogenomics J. The effects of the DRD2 polymorphism on effect of dopaminergic genes on likelihood of
6(3), 194–199 (2006). smoking cessation and negative affect: smoking following treatment with bupropion
28 Duan J, Wainwright MS, Comeron JM et al. evidence for a pharmacogenetic effect on SR. Health Psychol. 26(3), 361–368 (2007).
Synonymous mutations in the human mood. Nicotine Tob. Res. 6(2), 229–239 52 Cubells JF. Disulfiram for cocaine
dopamine receptor D2 (DRD2) affect mRNA (2004). dependence. Curr. Psychiatry Rep. 8(2),
stability and synthesis of the receptor. Hum. 40 Ding YC, Chi HC, Grady DL et al. Evidence 131–132 (2006).
Mol. Genet. 12(3), 205–216 (2003). of positive selection acting at the human 53 Colilla S, Lerman C, Shields PG et al.
29 Hirvonen M, Laakso A, Nagren K, Rinne JO, dopamine receptor D4 gene locus. Proc. Natl Association of catechol-O-methyltransferase
Pohjalainen T, Hietala J. C957T Acad. Sci. USA 99(1), 309–314 (2002). with smoking cessation in two independent
polymorphism of the dopamine D2 receptor 41 Wang E, Ding YC, Flodman P et al. The studies of women. Pharmacogenet. Genomics
(DRD2) gene affects striatal DRD2 genetic architecture of selection at the human 15(6), 393–398 (2005).
availability in vivo. Mol. Psychiatry 9(12), dopamine receptor D4 (DRD4 ) gene locus. 54 Johnstone EC, Elliot KM, David SP, Murphy
1060–1061 (2004). Am. J. Hum. Genet. 74(5), 931–944 (2004). MF, Walton RT, Munafo MR. Association of
30 Young RM, Lawford BR, Feeney GF, Ritchie 42 Brody AL. Functional brain imaging of COMT Val108/158Met genotype with
T, Noble EP. Alcohol-related expectancies are tobacco use and dependence. J. Psychiatr. Res. smoking cessation in a nicotine replacement
associated with the D2 dopamine receptor 40(5), 404–418 (2006). therapy randomized trial. Cancer Epidemiol.
and GABAA receptor beta3 subunit genes. Biomarkers Prev. 16(6), 1065–1069 (2007).
43 David SP, Munafo MR, Murphy MF, Proctor
Psychiatry Res. 127(3), 171–183 (2004).
M, Walton RT, Johnstone EC. Genetic 55 Sun H, Guo S, Chen D et al. Association of
31 Johnstone EC, Yudkin PL, Hey K et al. variation in the dopamine D4 receptor functional COMT Val108/Met polymorphism
Genetic variation in dopaminergic pathways (DRD4 ) gene and smoking cessation: follow- with smoking cessation in a nicotine
and short-term effectiveness of the nicotine up of a randomised clinical trial of replacement therapy. J. Neural. Transm.
patch. Pharmacogenetics 14(2), 83–90 (2004). transdermal nicotine patch. 119(12), 1491–1498 (2012).
32 Yudkin P, Munafo M, Hey K et al. Pharmacogenomics J. 8(2), 122–128 (2008). 56 Herman AI, Jatlow PI, Gelernter J, Listman
Effectiveness of nicotine patches in relation to 44 Leventhal AM, David SP, Brightman M et al. JB, Sofuoglu M. COMT Val158Met
genotype in women versus men: randomised Dopamine D4 receptor gene variation modulates subjective responses to intravenous
controlled trial. BMJ 328(7446), 989–990 moderates the efficacy of bupropion for nicotine and cognitive performance in
(2004). smoking cessation. Pharmacogenomics J. abstinent smokers. Pharmacogenomics J. 13(6),
33 David SP, Niaura R, Papandonatos GD et al. 12(1), 86–92 (2012). 490–497 (2013).
Does the DRD2-Taq1 A polymorphism 45 Bergen AW, Javitz HS, Su L et al. The DRD4 57 Brody AL, Mandelkern MA, Olmstead RE
influence treatment response to bupropion exon III VNTR, bupropion, and associations et al. Gene variants of brain dopamine
hydrochloride for reduction of the nicotine with prospective abstinence. Nicotine Tob. pathways and smoking-induced dopamine
withdrawal syndrome? Nicotine Tob. Res. Res. 15(7), 1190–1200 (2013). release in the ventral caudate/nucleus
5(6), 935–942 (2003). accumbens. Arch. Gen. Psychiatry 63(7),
46 Munafo MR, Murphy MF, Johnstone EC.
34 David SP, Brown RA, Papandonatos GD Smoking cessation, weight gain, and DRD4 808–816 (2006).
et al. Pharmacogenetic clinical trial of -521 genotype. Am. J. Med. Genet. B 58 Berrettini WH, Wileyto EP, Epstein L et al.
sustained-release bupropion for smoking Neuropsychiatr. Genet. 141B(4), 398–402 Catechol-O-methyltransferase (COMT ) gene
cessation. Nicotine Tob. Res. 9(8), 821–833 (2006). variants predict response to bupropion
(2007). therapy for tobacco dependence. Biol.
47 Middleton LS, Apparsundaram S, King-
35 Lerman C, Shields PG, Wileyto EP et al. Pospisil KA, Dwoskin LP. Nicotine increases Psychiatry 61(1), 111–118 (2007).
Effects of dopamine transporter and receptor dopamine transporter function in rat striatum 59 Han DH, Joe KH, Na C, Lee YS. Effect of
polymorphisms on smoking cessation in a through a trafficking-independent genetic polymorphisms on smoking cessation:

232 Pharmacogenomics (2014) 15(2) future science group


Nicotine pharmacogenetics & dopamine Review
a trial of bupropion in Korean male smokers. 72 Zeman MV, Hiraki L, Sellers EM. Gender variation in nicotine-metabolizing enzymes.
Psychiatr. Genet. 18(1), 11–16 (2008). differences in tobacco smoking: higher J. Neurogenet. 23(3), 252–261 (2009).
60 Cubells JF, Van Kammen DP, Kelley ME relative exposure to smoke than nicotine in 86 Sofuoglu M, Herman AI, Nadim H, Jatlow P.
et al. Dopamine beta-hydroxylase: two women. J. Womens Health Gend. Based Med. Rapid nicotine clearance is associated with
polymorphisms in linkage disequilibrium at 11(2), 147–153 (2002). greater reward and heart rate increases from
the structural gene DBH associate with 73 Gold AB, Wileyto EP, Lori A, Conti D, intravenous nicotine. Neuropsychopharmacol.
biochemical phenotypic variation. Hum. Cubells JF, Lerman C. Pharmacogenetic 37(6), 1509–1516 (2012).
Genet. 102(5), 533–540 (1998). association of the galanin receptor (GALR1) 87 Loukola A, Wedenoja J, Keskitalo-Vuokko K
61 Harrison PJ, Tunbridge EM. Catechol-O- SNP rs2717162 with smoking cessation. et al. Genome-wide association study on
methyltransferase (COMT ): a gene Neuropsychopharmacol. 37(7), 1683–1688 detailed profiles of smoking behavior and
contributing to sex differences in brain (2012). nicotine dependence in a twin sample. Mol.
function, and to sexual dimorphism in the 74 DeVito E, Herman A, Waters A, Valentine G. Psychiatry doi:10.1038/mp.2013.72 (2013)
predisposition to psychiatric disorders. Subjective, physiological and cognitive (Epub ahead of print).
Neuropsychopharmacol. 33(13), 3037–3045 responses to intravenous nicotine: effects of 88 Thorgeirsson TE, Geller F, Sulem P et al.
(2008). sex and menstrual cycle phase. A variant associated with nicotine
62 Shiffman S, Brockwell SE, Pillitteri JL, Neuropsychopharmacol. (In Press). dependence, lung cancer and peripheral
Gitchell JG. Individual differences in 75 Anker JJ, Carroll ME. Females are more arterial disease. Nature 452(7187), 638–642
adoption of treatment for smoking cessation: vulnerable to drug abuse than males: evidence (2008).
demographic and smoking history from preclinical studies and the role of 89 Saccone SF, Hinrichs AL, Saccone NL et al.
characteristics. Drug Alcohol Depend. 93(1–2), ovarian hormones. Curr. Top. Behav. Neurosci. Cholinergic nicotinic receptor genes
121–131 (2008). 8, 73–96 (2011). implicated in a nicotine dependence
63 Fu SS, Kodl MM, Joseph AM et al. 76 Allen SS, Bade T, Center B, Finstad D, association study targeting 348 candidate
Racial/Ethnic disparities in the use of Hatsukami D. Menstrual phase effects on genes with 3713 SNPs. Hum. Mol. Genet.
nicotine replacement therapy and quit ratios smoking relapse. Addiction 103(5), 809–821 16(1), 36–49 (2007).
in lifetime smokers ages 25 to 44 years. (2008). 90 Bierut LJ, Stitzel JA, Wang JC et al. Variants
Cancer Epidemiol. Biomarkers Prev. 17(7), 77 Benowitz NL, Hukkanen J, Jacob P 3rd. in nicotinic receptors and risk for nicotine
1640–1647 (2008). Nicotine chemistry, metabolism, kinetics and dependence. Am. J. Psychiatry 165(9),
64 Li Q, Hsia J, Yang G. Prevalence of smoking biomarkers. Handb. Exp. Pharmacol. 192, 1163–1171 (2008).
in China in 2010. N. Engl. J. Med. 364(25), 29–60 (2009). 91 Siedlinski M, Cho MH, Bakke P et al.
2469–2470 (2011). 78 Hukkanen J, Jacob P 3rd, Benowitz NL. Genome-wide association study of smoking
65 Yaeger R, Avila-Bront A, Abdul K et al. Metabolism and disposition kinetics of behaviours in patients with COPD. Thorax
Comparing genetic ancestry and self- nicotine. Pharmacol. Rev. 57(1), 79–115 66(10), 894–902 (2011).
described race in African Americans born in (2005). 92 Flint J, Munafo M. Genetics. Herit-ability.
the United States and in Africa. Cancer 79 Mwenifumbo JC, Tyndale RF. Molecular Science 340(6139), 1416–1417 (2013).
Epidemiol. Biomarkers Prev. 17(6), 1329–1338 genetics of nicotine metabolism. Handb. Exp. 93 Munafo MR, Flint J. Dissecting the genetic
(2008). Pharmacol. 192, 235–259 (2009). architecture of human personality. Trends
66 Clayton DG, Walker NM, Smyth DJ et al. 80 Benowitz NL, Pomerleau OF, Pomerleau CS, Cogn. Sci. 15(9), 395–400 (2011).
Population structure, differential bias and Jacob P 3rd. Nicotine metabolite ratio as a 94 Vrieze SI, Feng S, Miller MB et al. Rare
genomic control in a large-scale, case–control predictor of cigarette consumption. Nicotine nonsynonymous exonic variants in addiction
association study. Nat. Genet. 37(11), Tob. Res. 5(5), 621–624 (2003). and behavioral disinhibition. Biol. Psychiatry
1243–1246 (2005).
81 Dempsey D, Tutka P, Jacob P 3rd et al. doi:10.1016/j.biopsych.2013.08.027 (2013)
67 Marchini J, Cardon LR, Phillips MS, Nicotine metabolite ratio as an index of (Epub ahead of print).
Donnelly P. The effects of human population cytochrome P450 2A6 metabolic activity. 95 Xie P, Kranzler HR, Krauthammer M et al.
structure on large genetic association studies. Clin. Pharmacol. Ther. 76(1), 64–72 (2004). Rare nonsynonymous variants in alpha-4
Nat. Genet. 36(5), 512–517 (2004).
82 Benowitz NL, Swan GE, Jacob P 3rd, Lessov- nicotinic acetylcholine receptor gene protect
68 Johnson JA. Ethnic differences in Schlaggar CN, Tyndale RF. CYP2A6 against nicotine dependence. Biol. Psychiatry
cardiovascular drug response: potential genotype and the metabolism and disposition 70(6), 528–536 (2011).
contribution of pharmacogenetics. Circulation kinetics of nicotine. Clin. Pharmacol. Ther. 96 Xie P, Kranzler HR, Krystal JH, Farrer LA,
118(13), 1383–1393 (2008). 80(5), 457–467 (2006). Zhao H, Gelernter J. Deep resequencing of
69 Muszkat M. Interethnic differences in drug 83 Schnoll RA, Patterson F. Sex heterogeneity in 17 glutamate system genes identifies rare
response: the contribution of genetic pharmacogenetic smoking cessation clinical variants in DISC1 and GRIN2B affecting risk
variability in beta adrenergic receptor and trials. Drug Alcohol Depend. 104(Suppl. 1), of opioid dependence. Addict. Biol.
cytochrome P4502C9. Clin. Pharmacol. Ther. S94–S99 (2009). doi:10.1111/adb.12072 (2013) (Epub ahead
82(2), 215–218 (2007). of print).
84 Patterson F, Schnoll RA, Wileyto EP et al.
70 Urban TJ. Race, ethnicity, ancestry, and Toward personalized therapy for smoking 97 Wang S, Yang Z, Ma JZ, Payne TJ, Li MD.
pharmacogenetics. M. Sinai J. Med. NY cessation: a randomized placebo-controlled Introduction to deep sequencing and its
77(2), 133–139 (2010). trial of bupropion. Clin. Pharmacol. Ther. application to drug addiction research with
71 Perkins KA, Scott J. Sex differences in long- 84(3), 320–325 (2008). a focus on rare variants. Mol. Neurobiol.
term smoking cessation rates due to nicotine 85 Ray R, Tyndale RF, Lerman, C. Nicotine doi:10.1007/s12035-013-8541-4 (2013)
patch. Nicotine Tob. Res. 10(7), 1245–1250 dependence pharmacogenetics: role of genetic (Epub ahead of print).
(2008).

future science group www.futuremedicine.com 233


Review Herman, DeVito, Jensen & Sofuoglu

98 Kumar A, Choi KH, Renthal W et al. the methyltransferases GLP, G9a, Setdb1 and response: a review. Pharmacogenet. Genomics
Chromatin remodeling is a key mechanism levels of H3K9me2. Int. J. 21(5), 243–250 (2011).
underlying cocaine-induced plasticity in Neuropsychopharmacol. 16(5), 1129–1138 105 Agrawal A, Verweij KJ, Gillespie NA et al.
striatum. Neuron 48(2), 303–314 (2005). (2013). The genetics of addiction – a translational
99 Satta R, Maloku E, Zhubi A et al. Nicotine 102 Gozen O, Balkan B, Yildirim E, Koylu EO, perspective. Transl. Psychiatry 2, e140 (2012).
decreases DNA methyltransferase 1 expression Pogun S. The epigenetic effect of nicotine on
and glutamic acid decarboxylase 67 promoter dopamine D1 receptor expression in rat
methylation in GABAergic interneurons. prefrontal cortex. Synapse 67(9), 545–552 Website
„„
Proc. Natl Acad. Sci. USA 105(42), (2013).
201 Fiore MC, Jaen CR, Baker TB. Treating
16356–16361 (2008). 103 Levine A, Huang Y, Drisaldi B et al. tobacco use and dependence: 2008 update.
100 McGowan PO, Sasaki A, D’Alessio AC et al. Molecular mechanism for a gateway drug: US Department of Health and Human
Epigenetic regulation of the glucocorticoid epigenetic changes initiated by nicotine prime Services (USDHHS), US Public Health
receptor in human brain associates with gene expression by cocaine. Sci. Transl. Med. Service, MD, USA (2008).
childhood abuse. Nat. Neurosci. 12(3), 3(107), 107ra109 (2011). www.ahrq.gov/professionals/clinicians-
342–348 (2009). 104 Cobos A, Sanchez P, Aguado J, Carrasco JL. providers/guidelines-recommendations/
101 Chase KA, Sharma RP. Nicotine induces Methodological quality in pharmacogenetic tobacco/index.html
chromatin remodelling through decreases in studies with binary assessment of treatment

234 Pharmacogenomics (2014) 15(2) future science group

You might also like