Abstract
Background
Substance-related disorders (SRDs) are a major cause of morbidity and mortality worldwide. Family, twin, and adoption studies have demonstrated the substantial heritability of SRDs. To determine the impact of genetic variation on risk for SRD and the response to treatment, researchers have conducted a number of secondary data analyses and quasi-experimental studies that target one or more candidate gene variants.
Methods
This review examines studies in which candidate polymorphisms were examined as mediator variables to identify pharmacogenetic effects on subjective responses to drug administration or cues or outcomes of medication trials for SRDs. Efforts to use a meta-analytic approach to quantify these effects are premature because the number of available studies using similar methods and outcomes is limited, so the present review is qualitative.
Results
Findings from these studies provide preliminary evidence of clinically relevant pharmacogenetic effects. However, independent replication of these findings has been sparse.
Conclusions
Although this growing body of literature has produced conflicting results, improved statistical controls may help to clarify the findings. Additionally, the use of empirically derived sub-phenotypes (i.e., which serve to differentiate distinct groups of affected individuals) may also help to identify genetic mediators of pharmacologic response in relation to SRDs. The identification of genetic mediators can inform clinical care both by identifying risk factors for SRDs and predicting adverse events and therapeutic outcomes associated with specific pharmacotherapies.
Keywords: Genetics, Pharmacogenetics, Pharmacology, Medications Development, Clinical Trial, Drug Abuse
1. BACKGROUND
1.1. Drug Use and Addiction
Addiction is a chronic disease characterized by compulsive drug seeking and use [National Institute on Drug Abuse (NIDA), 2014]. Substance-related disorders (SRDs) cause and contribute to the deaths of millions of people each year by worsening comorbid psychiatric symptoms, such as depression, and medical conditions, such as cirrhosis of the liver, while also aiding in the spread of infectious diseases such as HIV, hepatitis B and hepatitis C. Substance-related disorders are also linked to crime and disability [United Nations Office on Drugs and Crime (UNODC), 2014].
Regular tobacco use contributes to many of the world's leading causes of death, including heart disease, stroke, and cancer (WHO, 2011). Cannabis is the world’s most widely used illicit substance, with upwards of 177 million regular users worldwide (UNODC, 2014). Although the long-term effects of chronic cannabis use are debated, adverse effects on cognition and mental health have been demonstrated (Fergusson et al., 2008; Hall and Degenhardt, 2009). While not commonly considered to be “addictive,” the world-wide use of the mild psychostimulant caffeine attests to its ability to serve as a reinforcer (Grigg, 2002).
More robust psychostimulants, such as cocaine and amphetamine-type drugs, are the second most widely used illegal drugs, with ~75 million estimated global users (UNODC, 2014). A number of serious medical complications are associated with cocaine use, including disturbances in heart rhythm, myocardial infarction, and neurological impairments (NIDA, 2009). Amphetamines may have a variety of neurotoxic, cardiotoxic and adverse neuropsychological effects as well (Scott et al., 2007; Shrem and Halkitis, 2008; Yu et al., 2003).
The abuse liability of naturally occurring opiates (e.g., morphine, codeine) and synthetic opioids (e.g., heroin, oxycodone, buprenorphine) is well known (Comer et al., 2008; Moratti et al., 2010; see also Meyer et al., 2014 and Trigo et al., 2010 for reviews). An estimated 9.2 million people worldwide are regular users of heroin (UNODC, 2014). The abuse of other opioids, including analgesics like oxycodone, is also widespread (Cicero, 2005; Darke et al., 1996; Gilson et al., 2004; Kintz et al., 2001; Substance Abuse and Mental Health Services Administration (SAMSHA), 2013). Opioids can significantly depress respiration, making overdose the most common cause of death among heroin users (Degenhardt et al., 2011; White and Irvine, 1999).
Although SRDs are a global public health concern, we have just begun to understand how genetics affect the initiation, course, and recovery from these disorders. A clearer understanding of the genetic contributions to these phenomena would inform the prevention, identification, and treatment of SRDs. In the qualitative review that follows, we focus on genetic aspects of the more common drugs of abuse that contribute substantially to morbidity and mortality around the world.
1.2. Genetic Involvement in Substance-related Disorders
Twin and adoption studies provided the first strong evidence for a genetic contribution to the susceptibility to develop SRDs (See Goldman et al., 2005 for a review). These investigations revealed that the heritability, the proportion of observable differences in a trait between individuals that is due to additive genetic effects, of substance abuse is quite significant. Depending on the specific drug, it has been estimated that genetic factors contribute 40–80% of the vulnerability to addiction (Agrawal et al., 2012; Crabe et al., 2002; Kendler et al., 2000; Tsuang et al., 1996, 2001; Uhl et al., 1999).
Genetic studies have examined a variety of individual genes that could contribute to the development and maintenance of SRDs. Genetic linkage studies are family-based studies that aim to establish a link between a region of a specific chromosome and the expression of a behavior or trait of interest. Linkage analysis uses panels of markers to identify the chromosomal region that harbors a gene of interest. If a marker consistently segregates in families with the trait under investigation (in this case, drug abuse), it is likely that the gene of interest is located in the chromosomal region identified by that marker. Similarly, association analyses test for a correlation between disease status and genetic variation to identify candidate variants that either contribute to a specific disease, or are in linkage disequilibrium with a causative variant.
Genome-wide association (GWA) and whole exome/genome sequencing techniques have been extremely successful in identifying genetic contributors to a number of complex human traits and diseases (Hindorff et al., 2009). These “agnostic” studies (i.e., those without a prior hypothesis as to function or location) have identified multiple genes and polymorphisms for more targeted investigations into how they mediate vulnerability to abuse. Alternatively, candidate-gene association studies take a hypothesis-driven approach to identifying potential mediator genes. Positional candidate genes are identified through linkage analysis and fine mapping based upon their approximate chromosomal location. More commonly however, functional candidate genes are identified based on their known (or presumed), relevant biological function. Linkage, association, and candidate gene studies have identified a number of specific chromosomal regions, genes, and alleles for further investigation.
2. GOALS AND METHODOLOGY OF THE REVIEW
Scientists have begun to perform studies where the presence or absence of target gene variants is used as an independent variable in laboratory studies measuring the subjective effects produced by a drug or clinical trials measuring treatment outcomes. The present review focuses on this method, known as pharmacogenetics, of identifying how genetic variation contributes to the susceptibility and maintenance of SRD’s. In this review, preclinical, linkage and association studies are often cited to describe how a particular genetic variant could alter gene expression or neurobiological function. However, the primary aim of this paper is to provide an overview of clinical pharmacogenetic studies investigating genetic mediators of the drug’s subjective effects or pharmacotherapy treatment outcome.
Using PubMed and PsychINFO, we searched for English-language articles published between 1970 and 2013. We included various combinations of the following search terms: genetics, polymorphism, SNP, pharmacogenetics, caffeine, opioids, heroin, nicotine, cocaine, and amphetamine. Using this method, we identified over 500 publications. After removing duplicates, we reviewed the titles and/or abstracts to ensure relevance. Data from approximately 150 articles are included here. Due to the extensive literature concerning the pharmacogenetics of alcohol, this drug could not be included in the current review and remain within the length requirements of the journal. Over 90 relevant publications related to the pharmacogenetics of alcohol and treatment of alcohol use disorder were identified. As such, we felt this body of work necessitated a review of its own (see Jones et al., in press).
3. GENETIC AND DOPAMINERGIC INTERACTIONS
Although the various classes of commonly abused drugs act upon a multitude of neurobiological systems, prevailing theory implicates the effects of drugs on mesolimbic dopamine (DA) transmission as the basis for their rewarding and euphoric effects. All major drugs of abuse have been shown to enhance dopaminergic transmission in the “reward centers” of the brain, including the ventral tegmental area, nucleus accumbens, and prefrontal cortex (Di Chiara and Imperato, 1988; Sulzer, 2011; Tanda et al., 1997). As such, we begin this review with a focus on genetic and dopaminergic interactions. Because dopamine plays a key role in reward-related behavior, it has been the most extensively studied neurotransmitter systems. Variation in a number of genes involving dopaminergic signaling (synthesis, transmission, and metabolism) has been studied with respect to their contribution to SRDs.
The five dopamine receptor subtypes are typically divided into two groups, D1-like (D1 and D5) and D2-like (D2, D3 and D4; Girault et al., 2004). Collectively, the D2 dopaminergic pathway appears to be a key neural substrate for drug-related reward (see Di Chiara et al., 2004 for a review). Accordingly, several studies have focused on the connection between the D2 receptor gene (DRD2, which is on chromosome 11) and various drugs of abuse (Li et al., 2011). For example, many studies have shown an association between drug abuse and the presence of a polymorphism in the ankyrin repeat and kinase domain containing 1 (ANKK1) gene adjacent to DRD2 (see Li et al., 2011 for a review; Neville et al., 2004). This Taq1A polymorphism (rs1800497) is a C>T substitution located in a non-coding region of ANKK1. It was originally believed to map to DRD2, though it is 10 kb downstream of the gene. Rs1800497 has been associated with dependence on cocaine and amphetamines (Noble et al., 1993; Persico et al., 1996), nicotine (Noble et al., 1994; Comings et al., 1996; Spitz et al., 1998), cannabis (Nacak et al., 2012) and various drug combinations (Comings et al., 1994; O’Hara et al., 1993). However, the findings have not been consistent; other reports show no association of the Taq1A polymorphism with drug abuse (Heinz et al., 1996; Miginini et al., 2012).
The Taq1A polymorphism has been shown to be associated with altered dopamine transmission. Several studies have shown that the polymorphism is associated with an up to 30% reduction in D2 receptor density, decreased dopamine receptor sensitivity, and elevated dopamine transporter (DAT) density (Jonsson et al., 1999; Laine et al., 2001; Pohjalainen et al., 1998; Schellekens et al., 2012). Although less studied than the ANKK1 single nucleotide polymorphism (SNP), a non-synonymous DRD2 polymorphism (C957T polymorphism; rs6277) has been shown to be functional. In healthy subjects, the rs6277*C allele has been associated with lower striatal D2 binding than the T-allele (Hirvonen et al., 2004). In contrast, some in vitro studies have shown the C allele to be associated with greater stability of DRD2 mRNA and protein (Duan et al., 2003). Two other functionally relevant DRD2 intronic SNPs (rs1076560, rs2283265) have been demonstrated to influence gene splicing leading to increased expression of the short (D2S) vs. long (D2L) receptor isoform (Moyer et al., 2011; Zhang et al., 2007 B). These SNPs have also been reliably associated with cocaine, alcohol and heroin addiction (Clarke et al., 2014 B; Moyer et al., 2011).
Above are just a few examples of the functional influence of various polymorphisms affecting dopamine. Their potentially significant influence has led to several prospective pharmacogenetic studies. Because genetic mediators of dopamine function have been the most extensively investigated, this is where we begin this review. However, exciting pharmacogenetics research with other neurobiological systems (e.g., serotonergic and opioidergic) are introduced and discussed later.
4. PHARMACOGENETICS OF DRUG EFFECTS AND TREATMENT OUTCOME
4.1. Nicotine
To date, pharmacogenetic research on nicotine has focused mainly on the impact of genetic variation on the subjective response to the drug and on treatment outcome in smoking cessation trials. In a randomized, placebo-controlled laboratory study, 72 smokers were assigned to smoke normal and de-nicotinized cigarettes during positive and negative mood induction (Perkins et al., 2008a). In addition to rating the positive affective value of the cigarettes, participants were given the option of smoking the test cigarettes ad libitum (ad lib). During negative mood states, Taq1A T-allele carriers [i.e., heterozygotes (A1/A2) or homozygotes (A2/A2)] reported greater “liking” of nicotine cigarettes and took more puffs than C-allele homozygotes. DRD2 C957T*C allele homozygotes also smoked more nicotine cigarettes during negative mood states than during positive mood states. No such effect was seen among individuals with the other C957T genotypes. In another study by Perkins et al. (2008b), the subjective effects of intranasal nicotine (0, 5, or 10 µg/kg) were assessed among 101 non-smokers. Men who were 957T homozygotes reported greater dose-dependent subjective effects of nicotine, including “feel effect,” “anger,” and “reduced fatigue.” No significant associations were seen with the Taq1A polymorphism. Although both studies showed a possible mediating effect of the two polymorphisms on the subjective and reinforcing effects of nicotine, their results were contradictory.
Wang et al. (2008) examined the interaction of C957T genotype, mood, smoking behavior and nicotine withdrawal. They recruited 13 smokers to undergo two MRI scans separated by 1–3 weeks following either smoking as usual or after 12–14 hours of nicotine abstinence. During abstinence, DRD2 957T homozygotes exhibited less change in regional cerebral blood flow in brain areas previously associated with cigarette craving than C957 allele carriers. Gilbert et al. (2009) observed differences in electroencephalographic activity in Taq1A1 allele carriers during nicotine abstinence, and concluded that mood during nicotine withdrawal may be mediated by the Taq1A SNP.
Although variation in DRD2 may mediate nicotine withdrawal, one prospective outcome trial in women (N = 593) found no association between smoking cessation and variation in a number of genes involved in dopamine synthesis, receptor activation, dopamine transporter reuptake activity and metabolism, including SNPs in COMT (rs4680), DRD2 (rs1799732 and rs1800497), and DRD3 (rs6280), and variable number of tandem repeat polymorphisms (VNTR) in DRD4, SLC6A3, and TH (Ton et al., 2009). The COMT Val158Met SNP (rs4680) is a well-studied G to A substitution that produces a valine (Val) to methionine (Met) exchange in the gene that encodes the enzyme catechol-O-methyltransferase, which degrades catecholamines (dopamine, epinephrine and norepinephrine). The Met allele is associated with a 40% reduction in enzyme activity (Diamond, 2007; Lotta et al., 1995; Mattay et al., 2003).
Other investigators used a similar methodology to investigate the effects of variation in ANKK1 on pharmacological treatment outcome. Cinciripini and colleagues (2004) found that the Taq1A polymorphism mediated the response to the dopaminergic antidepressant venlafaxine in 134 participants in a smoking cessation trial. During the period following smoking cessation, smokers without the A1 allele (A2/A2) treated with venlafaxine showed a substantial reduction in negative affect. A1 allele carriers also quit significantly less often than A2 homozygotes. Interestingly, however, no genotype by treatment interaction was found on abstinence rates. Although, these data suggest that carriers of the Taq1 A1 allele may have more difficulty quitting smoking, the hypothesis was not confirmed in a subsequent clinical trial. In 116 smokers, Robinson and colleagues (2007) found that the Taq1 A1 allele was not associated with scores on the Wisconsin Smoking Withdrawal Scale (WSWS) or the Minnesota Withdrawal Symptom Checklist (MWSC), or with daily smoking abstinence rates during the 6-week post-quit observation period.
In contrast to results with the Taq1A SNP, Jonsson et al. (1999) found that the less studied Taq1B SNP (rs1079597) was associated with a lower D2 receptor density (Hauge et al., 1991). Robinson et al. (2007) found that smokers with the ANKK1 Taq1 B1 allele reported significantly more daily nicotine withdrawal symptoms than those homozygous for the Taq1 B2 allele whose withdrawal symptoms decreased significantly over the same period. The study also showed that smokers carrying the Taq1 B1 allele were significantly less likely to be abstinent on a given day than those homozygous for the B2 allele.
Pharmacogenetic treatment trials of ANKK1 have provided mixed results. A study of bupropion (a weak dopamine receptor antagonist) for smoking cessation in 577 heavy smokers showed that the medication was particularly effective among Taq1 A1 homozygotes (Breitling et al., 2010). Specifically, A1 homozygotes treated with bupropion had a 28% greater likelihood of smoking cessation, compared to a 12% greater likelihood of cessation among A2 allele carriers (i.e., A1/A2, or A2/A2 genotypes). In contrast, in a study of 30 smokers randomized to treatment with bupropion or placebo, the active medication reduced nicotine withdrawal symptoms (craving, irritability and anxiety) only among A2 homozygotes (David et al., 2003), a finding that was later replicated in a larger, two-site study (David et al., 2007a). In that study, 722 smokers were randomized to treatment with bupropion or placebo. In A2 homozygotes, those treated with bupropion were more than three times as likely as placebo-treated individuals to be abstinent at the end of treatment and at 6-month follow-up. There was no significant effect of bupropion on smoking cessation outcomes in Taq1 A1 carriers (A1/A1 or A1/A2). The Taq1A polymorphism was also shown to mediate smoking cessation outcomes using the medication rimonabant (an inverse agonist for the cannabinoid receptor CB1; Fong and Heymsfield, 2009). Wilcok and colleagues (2011) found that A2 homozygote smokers were significantly more successful in completely abstaining from smoking while on rimonabant (total N = 76).
Multiple studies have also evaluated the mediating effect of the Taq1A polymorphism on the effectiveness of nicotine replacement therapy (NRT). Yudkin et al. (2004) found that A1 allele carriers showed less therapeutic benefit from the nicotine patch than A2 allele homozygotes. Similarly, Johnstone et al. (2004) genotyped 755 heavy smokers for Taq1A alleles following completion of a randomized trial of the nicotine patch. They concluded that, at Week 1, heterozygous individuals for the ANKK1 SNP (CT/TT or A1/A2) showed a better response to NRT than to placebo. However, the pharmacogenetic effect on abstinence did not persist at 12 weeks. There was significantly greater abstinence at 12 weeks among individuals with at least one A1 allele and an A allele of rs77905 in the dopamine beta hydroxylase gene (DβH). Dopamine beta hydroxylase is an enzyme that catalyzes the synthesis of norepinephrine from dopamine. Individuals with the DβH A allele tend to have lower dopamine levels, and smokers with the allele tend to smoke more cigarettes (McKinney et al., 2000; Zabetian et al., 2001; though see Shiels et al., 2008). Munafo et al. (2009) later failed to replicate these findings in a larger cohort. Similar prospective trials have also failed to find a significant mediating effect of the Taq1A genotype on the efficacy of NRT in smoking cessation (Berlin et al., 2005; De Ruyck et al., 2010; Stapleton et al., 2011).
Although Lerman et al. (2006) failed to find a relationship between the Taq1A SNP and response to pharmacotherapy for smoking cessation, using data pooled from two clinical trials they found a significant mediating effect of other DRD2 polymorphisms. The first study was a double-blind, placebo-controlled trial of bupropion 300 mg/day (N = 414) and the second was an open-label trial that compared transdermal nicotine with nicotine nasal spray (N = 368). In addition to ANKK1, these investigators examined the mediating effect of an insertion/deletion polymorphism in the DRD2 promoter region (−141C Ins/Del, rs1799732). The more common 141C Ins allele was previously shown to confer greater transcriptional efficiency than the 141C Del allele (Arinami et al., 1997). The possible influence of the aforementioned DRD2 C957T SNP was also assessed. At the end of the 6-month treatment phase, 141C Ins allele homozygotes showed a more favorable response to bupropion. In contrast, smokers carrying the 141C Del allele had significantly higher quit rates while on NRT, independent of NRT type. Individuals homozygous for the C957T T allele also demonstrated increased abstinence following NRT. Based on these data, the investigators concluded that bupropion may be a more effective pharmacologic treatment for smokers homozygous for the DRD2 14C1 Ins allele, while NRT may be more beneficial for those who carry the Del allele. Their data also suggest that T-allele homozygotes for the C957T variant may, in general, be better able to maintain abstinence. This is an interesting finding when considered in the context of the study by Wang et al. (2008), which found that individuals with this genotype may experience less nicotine withdrawal.
Lerman and colleagues replicated their findings with DRD2-141C Ins/Del in another 6-month NRT trial (N = 363; Dahl et al. 2006), in which they observed an interaction between DRD2 141C and variation in the gene encoding the neuronal calcium sensor-1 protein (FREQ), the protein product that regulates D2 receptor desensitization (Burgoyne et al. 2004; Kabbani et al., 2002). They found that 62% of the smokers with at least one copy of the DRD2 141C Del allele and two copies of the FREQ A allele (rs1054879) were abstinent from smoking, compared to 29–38% abstinence among other smokers.
Variants in the serotonin transporter gene (SLC6A4) have also been shown to influence the effectiveness of bupropion and nortriptyline on smoking cessation rates (Quaak et al., 2012). SLC6A4 encodes the serotonin transporter, a membrane protein that takes serotonin back up into pre-synaptic neurons. In this study, 214 daily smokers were randomized and genotyped for the 5-HTTLPR variant (rs25531) that can result in a short (S) allele (14 repeats, associated with lower gene transcription) or a longer one (16 repeats, higher transcription, Nakamura et al., 2000). Primary outcome measures were prolonged abstinence from weeks 4–12, 4–26 and 4–52. The researchers concluded that bupropion and nortriptyline seem to be more effective in smoking cessation among SLC6A4 high-activity variant carriers. See Table 11 for a list of nicotine pharmacogenetic treatment outcome trials.
Table 1.
Gene | Polymorphism | Treatment Medication |
Sample Size |
Population | Treatment × Genotype Finding* ↑ More effective ↓ Less effective ↔ No interaction |
Citation |
---|---|---|---|---|---|---|
ANKK1 | rs1800497 | Bupropion | 577 | German | ↑ CC genotype | Breitling et al., 2010 |
rs1800497 | Transdermal Nicotine | 755 | Primarily Caucasian (98%) | ↑ CT/TT genotypes | Johnstone et al. (2004) | |
rs1800497 | Transdermal Nicotine | 752 | European | ↑ CT/TT genotypes (women only) | Yudkin et al. (2004) | |
rs1800497 | Bupropion | 722 | Caucasian /European Ancestry |
↑A2/A2 (CC) genotype | David et al. (2007b) | |
rs1800497 | Transdermal Nicotine | 908 | European or Unknown Ancestry |
↓ CT/TT genotypes | Munafo et al. (2009) | |
rs1800497 | Transdermal Nicotine | 419 | Primarily Caucasian/European |
↔ | Stapleton et al. (2011) | |
rs1800497 | Rimonabant | 76 | European Decent | ↑A2/A2 (CC) genotype | Wilcox et al. (2011) | |
ARRB2 | rs1045280 rs2036657 |
NRT (transdermal, nasal spray) | 374 | European Ancestry | ↔ | Ray et al. (2007) |
CHRNA4 | rs2236196 | NRT (all formulations) | 316 | European Ancestry | ↔ | Hutchison et al. (2007) |
CHRNB2 | rs2072661 | Bupropion | 412 | European | ↔ | Conti et al. (2008) |
rs2072661 | Transdermal Nicotine | 156 | European | ↑ GG genotype | Perkins et al. (2009) | |
COMT | Val158Met: rs4680 | NRT (transdermal nicotine, nicotine spray) | 290 | Females of European and African ancestry | ↑ Met/Met | Colilla et al. (2005) |
rs4680 | Transdermal Nicotine | 749 | European | ↑ Met/Met | Johnstone et al. (2007) | |
rs737865 rs165599 |
Bupropion | 511 | Caucasian- and African-American |
↑ A/G allele of rs165599 ↑ GG at both SNPs (AA participants only) |
Berrettini et al. (2007) | |
CYP2A6 | Bupropion | 414 | European | ↑ Fast metabolizers | Patterson et al. (2008) | |
Transdermal Nicotine | 568 | European | ↓ Fast metabolizers | Schnoll et al. (2009) | ||
CYP2A6 CHRNA5: rs1696998, rs680244 |
Bupropion NRT (lozenge, transdermal) Bupropion + NRT |
709 | Caucasian -American | ↑ Fast metabolizers (NRT) ↑ CYP2A6: Fast metabolizers + CHRNA5: GG/TT,GA/CT, AA/CC (NRT) |
Chen et al. (2014) | |
CYP2B6 | C1459T: rs3211371 |
Bupropion | 426 | Caucasian-European | ↑Female CT/TT | Lerman et al. (2002) |
CYP2B6: rs3211371 ANKK1: rs1800497 SLC6A3 3’ VNTR: rs27072 |
Bupropion | 291 | European | ↑ CYP2B6 CT/TT variant + ANKK1 A2/A2 |
David et al. (2007b) | |
*6 | Bupropion | 326 | European Ancestry | ↑ CYP2B6*6 carriers | Lee et al. (2007a) | |
*1, *4, *6 | NRT (transdermal, nasal spray) | 369 | European Ancestry | ↔ | Lee et al. (2007b) | |
DβH |
DβH: rs77905 ANKK1: rs1800497 |
Transdermal Nicotine | 755 | Primarily Caucasian (98%) | ↑ Taq1A CT/TT + DβH GA/AA genotypes | Johnstone et al. (2004) |
rs77905 | Bupropion | 577 | German (98%) | ↔ | Breitling et al. (2010) | |
DRD2 | −141C Ins/Del: rs1799732 |
NRT (transdermal, nasal spray) | 368 | European | ↑ Smokers with at least one copy of the Del C allele | Lerman et al. (2006) |
rs1799732 FREQ: rs1054879 |
NRT (transdermal, nasal spray) | 363 | European | ↑ Smokers with at least one copy of −141 Del allele and two copies of FREQ | Dahl et al. (2006) | |
rs1799732 | Bupropion | 414 | European | ↑ Smokers homozygous for the Ins C allele | Lerman et al. (2006) | |
C957T: rs6277 |
NRT (transdermal, nasal spray) | 368 | European | ↑ C allele | Lerman et al. (2006) | |
rs6277 | Bupropion | 414 | European | ↔ | Lerman et al. (2006) | |
Exon 8 A/G: rs6276 | Transdermal Nicotine | 755 | ↔ | Johnstone et al. (2004) | ||
DRD4 | VNTR | Transdermal Nicotine | 720 | European Ancestry | ↔ | David et al. (2008) |
C-521T: rs1800955 | Transdermal Nicotine | 720 | European | ↔ | David et al. (2008) | |
HINT1 | rs3852209 rs2278060 |
NRT (transdermal, nasal spray) | 374 | European Ancestry | ↑ rs3852209 TT genotype | Ray et al. (2007) |
OPRM1 |
Asn40Asp (A118G): rs1799971 |
NRT (transdermal, nasal spray) | 320 | European | ↑ G carriers w/transdermal nicotine (no interaction with nasal nicotine) | Lerman et al. (2004) |
rs1799971 | Transdermal Nicotine | 710 | European Ancestry | ↑ AA carriers | Munafo et al. (2007) | |
rs1799971 | NRT (transdermal, nasal spray) | 374 | European | ↑ G carriers | Ray et al. (2007) | |
rs1799971 | Transdermal Nicotine | 598 | European Ancestry | ↔ | Munafo et al. (2013) | |
SLC6A3 ANKK1 |
3′ UTR VNTR Taq1A: rs1800497 |
Bupropion | 418 | European | ↔ | Lerman et al. (2003) |
3′ UTR VNTR Taq1A: rs1800497 |
Bupropion | 416 | European | ↓ ANKK1 A1 and SLC6A3-9 | Swan et al. (2007) | |
SLC6A3 | 3′ UTR VNTR | NRT (all formulations) Bupropion | 583 | European | ↔ | O'Gara et al. (2007) |
3′ UTR VNTR | Bupropion | 291 | European | ↔ | David et al. (2007b) | |
SLC6A4 | 5-HTTLPR | Nicotine Replacement Therapy (NRT): (transdermal patch, nasal spray) | 397 | European | ↔ | Munafo et al. (2006) |
5-HTTLPR | Transdermal patch | 741 | European | ↔ | David et al. (2007c) | |
5-HTTLPR | Bupropion Nortriptyline |
214 | Caucasian | ↑ carriers L-variant (rs25531) Bupropion ↑ carriers of 3 combined high-activity variants Bupropion & Nortriptyline |
Quaak et al. (2012) |
Relationship between genotype and pharmacotherapy effectiveness outcome
4.2. Opioids
A number of studies have examined the effect of genetic variation on features of opioid dependence. One study investigated the mediating effect of the dopamine receptor gene (DRD4) 48-bp VNTR polymorphism (2, 4 and 7 repeat variants) on cueinduced craving in 420 heroin users (Shao et al., 2006). This polymorphism was selected based on its prior association with addictive disorders (Benjamin et al., 1996) and because of evidence that it is functional (Asghari et al., 1995; Van Tol et al., 1992). Following exposure to neutral and heroin-related cues, such as syringes, lighters, and tinfoil, participants completed a 5-item craving questionnaire. The study showed significantly greater cue-elicited heroin craving among individuals carrying the DRD4 VNTR 7 (longer) allele than in non-carriers. A similar study showed significantly greater cue-elicited heroin craving among Taq1 A1 allele carriers (A1/A1 or A1/A2 genotypes) than in A2 homozygotes (Li et al., 2006). These variants were identified as potential genetic risk factors for cue-induced drug craving and subsequent relapse to substance use. Though other findings relating DRD4 VNTR allele and cue reactivity have been contradictory (MacKillop et al., 2007; van den Wildenberg et al., 2007), Hutchison et al. (2002) found an association between this allele with greater cue-induced craving and arousal among smokers.
The effects of a number of opioid receptor gene polymorphisms have been implicated in subjective response to the first use of opioids. One such study assessed the association of several OPRM1 variants with positive vs. negative subjective responses to initial heroin use (Zhang et al., 2007a). Heroin users (N = 336) were divided into two groups based on their retrospective recall concerning their first experience with heroin (i.e., positive vs. negative groups). Association analyses were performed between these two groups and nine OPRM1 SNPs. An initially positive response upon first use of heroin was found to be associated with three OPRM1 SNPs (rs696522, rs1381376, and rs3778151), although the functional significance of these SNPs has yet to be determined. These data highlight the need for more studies into the mediation of subjective response to drugs by genes and how this consequently affects susceptibility to abuse.
Additional research has shown an association between gene variants and treatment outcome among opioid users. Methadone and buprenorphine are two common and effective maintenance medications for opioid use disorder (Ball and Ross, 1991; Joseph et al., 2000; Mattick et al., 2008). Lawford and colleagues (2000) reported a four-fold increase in the frequency of the Taq1 A1 allele among a sample of 95 treatment-seeking opioid users who responded poorly to methadone treatment compared to those with successful treatment outcomes. However, the association of the Taq1 A1 allele with the treatment effectiveness of opioid substitution therapy was not confirmed by a later study (Barrat et al., 2006). No significant differences in A1 allele frequency were observed between methadone- and buprenorphine-treated patients with poor and successful treatment outcomes (N = 116). Interestingly, the researchers observed significantly less opioid withdrawal among carriers of the A1 allele who had a more successful treatment outcome on methadone. Crettol et al. (2008) also failed to find an association between the presence of the Taq1 A1 allele and response to methadone treatment in a sample of 238 methadone-maintained patients. They found that the Taq1 A1 allele was not associated with response to methadone treatment, as measured by use of heroin or cocaine, withdrawal symptoms, program attendance and required daily methadone dose. They also found a lack of an association with the common variants of the D1 dopamine (Ddel, −48A>G) and mu opioid Asn40Asp (A118G) receptor gene, though they did find an association with the other DRD2 polymorphism C957T: patients with the C/C genotype were more frequently nonresponders to methadone treatment.
A later study failed to find an association between OPRM1 alleles and methadone treatment response, but did report significant interactions with other genes implicated in the pharmacodynamics of methadone (Fonseca et al., 2010). In addition to OPRM1, researchers assessed the mediating effects of genes encoding the metabotropic glutamate receptors (GRM6, GRM8), the nuclear receptor (NR4A2), the photolyase enzyme cryptochrome 1 (CRY1), and the transcription factor myocardin (MYOCD), which have previously been associated with the risk of opioid abuse (Nielsen et al., 2008). A sample of 116 opioid-dependent patients were classified as methadone responders (n = 83) and nonresponders (n = 33) according to illicit opioid use detected with urinalysis. Participants carrying the CRY1 AA genotype at (rs1861591) had a higher risk of being nonresponders. Participants carrying the MYOCD A allele (rs1714984) had an increased risk of being nonresponders but only if they were also carriers of the GRM6 AG genotype (rs953741).
Subsequent research has shown pharmacogenetic interactions between methadone pharmacokinetics and dosing requirements in a sample of 312 opioid-dependent patients (Hung et al., 2011). This study examined SNPs (G516T, A785G) in CYP2B6, which encodes the liver enzyme that is partially responsible for methadone metabolism and the activity of which is known to alter methadone plasma levels (Crettol et al., 2006). Also assessed were a common SNP (C3435T) of the highly polymorphic ABCB1 gene (rs1045642) that has been associated with disease severity and treatment response (Coller et al., 2006; Levran et al., 2008a) and the A118G SNP in of the gene encoding the mu opioid receptor (OPRM1). Investigators also assessed several polymorphisms associated with D2 expression or function: DRD2 C957T, DRD2 C939T, DRD2 A214G, ANKK1 C2137T (Crettol et al., 2008; Doehring et al., 2009; Lawford et al., 2000). Carriers of the variant ABCB1 3435C>T and CYP2B6 516G>T alleles were significantly more likely to require higher methadone doses than non-carriers. In contrast, carriers of the DRD2 -214A>G and 939C>T alleles required a significantly lower methadone dosage than non-carriers (Hung et al., 2011, see also Levran et al., 2013 and Barratt et al., 2012). A recent meta-analysis of the literature concerning a number of ABCB1 and CYP2B6 failed to find an overall effect of allelic variation and treatment response to methadone (Dennis et al., 2014). Their analysis did find that the CYP2B6*6 haplotype did significantly influence methadone metabolism. Specifically, methadone metabolism was significantly slower in *6 homozygous carriers.
Although variation in OPRM1 has been more extensively studied than the genes encoding other opioid receptors, variation in the delta opioid receptor gene (OPRD1) has been associated with the risk of opioid abuse and dependence (Mayer et al., 1997; Zhang et al 2008). Accordingly, researchers have attempted to assess whether OPRD1 variants may also mediate the response to opioid substitution therapy. In one such trial, patients (N = 643) were randomized to receive buprenorphine/naloxone or methadone maintenance treatment over the course of 24 weeks (Crist et al., 2013 a). The researchers’ primary measure of treatment effectiveness was illicit opioid use, measured by weekly urinalysis. Six OPRD1 SNPs were genotyped as predictor variables. Among African-American participants (N = 77), an intronic OPRD1 SNP (rs678849) predicted treatment outcome for both pharmacotherapies. Methadone patients with the C/C genotype were less likely to have opioid-positive urine tests than T-allele carriers (C/T and T/T genotypes). The opposite outcome was observed with respect to buprenorphine/naloxone treatment. Patients with the C/C genotype were more likely to have positive opioid drug screens than T-allele carriers. Though it is unclear what effect this SNP has on OPRD1 transcription or delta receptor function, its association with both methadone and buprenorphine/naloxone treatment outcome suggests that it either directly affects or is in linkage disequilibrium with a functional variant that mediates the effects of these medications. Interestingly, this group of investigators found similar associations between OPRD1 SNPs and cocaine abuse that were specific to African-Americans, highlighting the possible influence of population genetics in this type of research (Crist et al., 2013 b).
In a more recent study, investigators found that two other intronic OPRD1 SNPs (rs581111 and rs529520) predicted buprenorphine treatment outcomes. However, this interaction was found only in females. Women with the AA or AG genotypes at rs581111 had significantly worse outcomes compared to women with the GG genotype, while women with rs529520 AA genotype had a significantly worse outcome compared to women with the CC genotype (Clarke et al., 2014 A). These studies highlight the potential of pharmacogenetic studies to provide important information for the selection of treatment when different options are available. Table 2 lists opioid use disorder pharmacogenetic treatment outcome trials, and the nature of their findings.
Table 2.
Gene | Polymorphism | Treatment Medication |
Drug of Abuse |
Sample Size |
Population | Treatment × Genotype Finding* ↑ More effective ↓ Less effective ↔ No interaction |
Citation |
---|---|---|---|---|---|---|---|
ANKK1 | rs1800497 | Methadone | Heroin | 95 | Caucasian | ↓ T allele | Lawford et al. (2000) |
rs1800497 | Buprenorphine | Heroin | 25 | Primarily Caucasian (88%) | ↔ | Barrat et al. (2006) | |
rs1800497 | Methadone | Heroin | 46 | Primarily Caucasian (89%) | ↔ | Barrat et al. (2006) | |
rs1800497 | Methadone | Heroin | 238 | Caucasian | ↔ | Crettol et al. (2008) | |
CYP3A4 | *1B | Methadone | Opioids | 245 | Caucasian-European | ↔ | Crettol et al. (2006) |
CYP2B6 | *4, *5, *6, *9 | Methadone | Opioids | 245 | Caucasian-European | ↔ | Crettol et al. (2006) |
DRD1 | rs4532 | Methadone | Heroin | 238 | Caucasian | ↔ | Crettol et al. (2008) |
DRD2 | rs6277 | Methadone | Heroin | 238 | Caucasian | ↑ CC genotype | Crettol et al. (2008) |
Multiple Variants |
MYOCD: rs1714984, GRM8: rs1034576, CRY1: rs1861591, GRM6: rs953741, OPRM1: rs1074287, NR4A2: rs1405735, intergenic variants: rs965972, rs1867898 |
Methadone | Opioids | 169 | Caucasian | ↓CRY1 AA ↓ MYOCD A allele + GRM6 AG |
Fonseca et al. (2010) |
OPRD1 | rs2234918 | Methadone | Heroin | 238 | Caucasian | ↔ | Crettol et al. (2008) |
rs678849 rs1042114 rs10753331 rs59520 rs581111 rs2234918 |
Buprenorphine | Opioids | 566 Caucasian 77 African-American |
Caucasian- and African-American |
↓ African-American rs678849 CC genotype |
Crist et al. (2013 A) | |
rs678849 rs1042114 rs10753331 rs59520 rs581111 rs2234918 |
Methadone | Opioids | 566 Caucasian 77 African-American |
Caucasian- and African-American |
↑ African-American rs678849 CC genotype |
Crist et al. (2013 A) | |
rs678849 rs1042114 rs10753331 rs59520 rs581111 rs2234918 |
Buprenorphine Methadone |
Opioids | 582 | European-American | ↓ Female rs581111 AA/AG genotype ↓ Female rs529520 AA genotype |
Clarke et al. (2014 A) | |
OPRM1 | rs1799971 | Methadone | Heroin | 238 | Caucasian | ↔ | Crettol et al. (2008) |
G-172T: rs6912029 G-1510A: rs12205732 |
Oral Naltrexone | Opioids | 138 | Arab | ↓ 172GG and 1510GG | Al-Eitan et al. (2012) |
4.3. Cocaine
Cocaine dependence has a strong genetic component, with an estimated heritability of 72%, but unlike the other substances discussed here, lacks an FDA-approved pharmacotherapy (Goldman et al., 2005). Researchers have tested over 60 medications, and though some have shown promise, none so convincingly as to have received FDA approval (Grabowski et al. 2004; Vocci and Ling 2005). Pharmacogenetics may help to identify subsets of users who respond to a particular pharmacotherapy, which could then be developed and marketed to a patient population with a specific genetic profile.
The acetaldehyde dehydrogenase-inhibiting drug disulfiram (Antabuse, Antabus), has been used to treat alcoholism since the 1940’s (Hald et al., 1948). Disulfiram has also shown some potential in treating cocaine abuse, and research has begun to examine whether genetic variation mediates treatment response (Carroll et al., 1998; 2004). Some researchers have proposed that by inhibiting dopamine β-hydrolylase (DβH), disulfiram alters cocaine signaling and/or availability (Bourdelat-Parks et al., 2005; Schank et al., 2006; Vaccari et al., 1996). Consequently, genetic variants affecting the DβH gene were among the first to be examined as genetic mediators of the efficacy of disulfiram in treating cocaine dependence.
A functional DβH gene polymorphism that alters transcription and decreases DβH plasma levels was studied with respect to its interaction with disulfiram (Bhaduri et al., 2008; Zabetian et al., 2001). The C-1021T polymorphism (rs1611115) accounts for up to 52% of the variation in DβH enzyme levels, with individuals homozygous for the T allele having the lowest levels (Khonke et al., 2002; Deinum et al., 2004). In their assessment of the influence of this polymorphism, Kosten and colleagues (2013) randomized 72 cocaine- and opioid-dependent patients to receive disulfiram (250 mg/day) or placebo for 10 weeks. Overall, disulfiram treatment significantly reduced the number of cocaine-positive urines but the effect was only seen among subjects with the DβH gene variant that is associated with normal DβH levels.
Spellicy et al. (2013) also found that the aforementioned ANKK1 (rs1800497) and DRD2 (rs2283265) polymorphisms interacted with disulfiram treatment response. Patients homozygous (T/T) or heterozygous (C/T) for the minor alleles of both SNPs had a significantly lower number of cocaine-positive urines when maintained on disulfiram (250 mg/day), while their homozygous counterparts showed a marginal (rs2283265*CC) or no (rs1800497*CC) treatment effect (N = 68).
Other analyses of genetic mediators of disulfiram treatment have focused on a SNP in the gene that encodes 5, 10-methylene tetrahydrofolate reductase (MTHFR). In vitro studies have shown that the minor T allele of the C677T SNP (rs1801133) results in an enzyme with reduced activity and increased thermolability (Frosst et al., 1995). Because this enzyme affects a number of developmental and biochemical pathways, Spellicy and colleagues (2013) hypothesized that the polymorphism would be associated with the response to disulfiram treatment in cocaine users. The primary treatment outcome measure was the number of cocaine-positive urines. The percentage of cocaine-positive urines dropped from 73% to 52% for the combined C/T and T/T group pre- to post-disulfiram treatment. Subjects with the C/C genotype had a smaller reduction in cocaine-positive urines (from 81% to 69%) while on disulfiram, a small but significant treatment effect when compared to placebo.
Investigations into the interaction between disulfiram and a functional variant of the gene that encodes the α-1 adrenergic receptor (ADRA1A) are based on disulfiram’s effect on norepinephrine production (Shorter et al., 2013). This polymorphism (rs1048101) in exon 2 results in a substitution of arginine for a cytosine within the C-terminus of the protein (Cys347Arg). Data suggest that this single amino acid change alters the activity of the receptor and may impact cognition (Lei et al., 2005). Using the clinical trial methodology described above (disulfiram 250 mg/day, 69 cocaine and opioid co-dependent participants; Shorter et al., 2013), the investigators found a significant interaction between genotype and treatment, where the patients who were carriers of the minor T allele had a significantly lower percentage of cocaine-positive urines, while those homozygous for the major C allele showed no treatment effect.
Genetic mediation of treatment response has also been found for a novel immunotherapy that produces selective anti-cocaine antibodies (cocaine vaccine (TA-CD); Carrera et al., 1995). Nielsen et al. (2013) genotyped 66 of 114 cocaine- and opioid-dependent patients enrolled in a 16-week trial of the vaccine for a variant of the kappa subtype of opioid receptor, OPRK1 gene (rs6473797), an A to G transition, with the G allele previously shown to be a risk allele for opioid dependence (Levran et al., 2008b). Participants homozygous for the major (A) allele had a significantly better vaccine treatment response (i.e., decrease in cocaine-positive urines) than carriers of the G allele. A similar investigation by this group (Kosten et al., 2013) identified the C-1021T DβH SNP as a mediator of cocaine vaccine response (See Table 3 for pharmacogenetic medication trials for cocaine abuse trials).
Table 3.
Gene | Polymorphism | Treatment Medication |
Sample Size |
Population | Treatment × Genotype Finding* ↑ More effective ↓ Less effective ↔ No interaction |
Citation |
---|---|---|---|---|---|---|
ADRA1A | rs1048101 | Disulfiram | 69 | Caucasian (>50%); others unidentified | ↑ carriers of at least one T allele | Shorter et al. (2013) |
rs1611115 | Cocaine Vaccine | 71 | Primarily Caucasian (79% and greater) | ↑ CT/TT genotype | Kosten et al. (2013) | |
rs1611115 | Disulfiram | 74 | Primarily Caucasian (67% and greater) | ↑ CC/TT genotype | Kosten et al. (2013) | |
ANKK1 | rs1800497 | Disulfiram | 68 | Caucasian-, African- and Latino -American | ↑ CT/TT genotypes | Spellicy et al. (2013) |
DRD2 | rs2283265 | Disulfiram | 68 | Caucasian, African-American, & Latino | ↑ GT or TT genotypes | Spellicy et al. (2013) |
MTHFR | rs1801133 | Disulfiram | 67 | Between 43% and 90% Caucasian, others unidentified | ↑ CT or TT genotypes | Spellicy et al. (2013) |
OPRK1 | rs6473797 | Cocaine Vaccine | 114 | Primarily Caucasian (80% and greater) | ↑ AA allele carriers | Nielsen et al., (2013) |
Cocaine-induced paranoia is another interesting area of pharmacogenetic research. Association studies have failed to consistently demonstrate heritability and genetic influence in the occurrence of this drug effect (Gelernter et al., 2005; Kalayasiri et al., 2006). However, candidate gene studies have found evidence of allelic association with cocaine-induced paranoia. Variants that have been implicated as possible mediators include: SLC6A3 3’ VNTR (Gelernter et al., 1994), COMT variants: rs737866, rs4680, rs174696, rs4680, rs933271, rs993883, rs740603 (Ittiwut et al., 2011), DβH rs1611115 (Kalayasiri et al., 2007), CNR1 rs806368 (Zuo et al., 2009) and MANEA rs9387522 (Farrer et al., 2009).
4.4. Amphetamines and Caffeine
Pharmacogenetic research on methamphetamine and caffeine is very limited. However, there is research to suggest that OPRM1 variation mediates subjective response to amphetamine. One hundred and sixty-two Caucasian participants completed three sessions receiving either placebo or d-amphetamine (10 and 20 mg). Several OPRM1 SNPs (rs510769, rs2281617, rs1799971, rs510769, rs1918760, rs2281617 and rs1998220) were associated with significantly higher subjective ratings of “Euphoria,” “Energy” and “Stimulation” after 10 mg amphetamine. These data imply that genetic variability in the μ-opioid receptor gene influences the subjective effects of amphetamine, though the investigators were later unable to replicate these findings (Duglos et al., 2011).
With respect to methamphetamine, another area under investigation is possible genetic involvement in the occurrence of methamphetamine-induced psychosis. The majority of this research has focused on D2 receptor gene variants. A number of publications have identified DRD2 alleles as possible risk genotypes for methamphetamine psychosis (Harano et al., 2004; Ujike et al., 2009) with the body of literature expanding each year (Chanasong et al., 2013; Kishi et al., 2011; Kobayashi et al., 2004; Matsuzawa et al., 2007; Ujike et al., 2003).
Similarly, the aversive effects of caffeine consumption may have a genetic link. Caffeine produces mild psychostimulant and anxiogenic effects by antagonizing adenosine receptors and increasing dopaminergic transmission (Daly and Fredholm, 1998). Childs et al. (2008) examined functional polymorphisms in the genes for adenosine and dopamine receptors as possible mediators of caffeine-induced anxiety. They examined associations between self-reported anxiety following caffeine administration and variation in the genes for the A2A receptor (ADORA2A) and D2 (DRD2) receptor. Healthy male and female individuals (N = 102), who consumed less than 300 mg caffeine per week, ingested capsules containing caffeine 0, 50, 150, or 450 mg under double-blind conditions, in four separate experimental sessions. Subjective anxiety was measured before and multiple times after ingestion of the capsules. They found a significant association between caffeine-induced anxiety and ADORA2A alleles: rs5751876, rs2298383, rs4822492, and DRD2 allele: rs1110976. This study provides evidence of mediation of aversive drug effects by genetic variation and suggests a mechanism by which these alleles may influence caffeine intake.
5. CONCLUSIONS
One cautionary tale concerning the reproducibility of pharmacogenetic research comes from one of the most prolific groups in this field, the Department of Human Genetics at the University of Chicago. Investigators there assessed acute response to d-amphetamine in 398 individuals with no history of drug abuse. Interim pharmacogenetic analyses using 99–162 participants from this data set found associations between a number of gene variants and amphetamine response: ADORA2A (Hohoff et al., 2005), BDNF (Flanagin et al., 2006), CSNK1E (Veenstra-VanderWeele et al., 2006), COMT (Hamidovic et al., 2010a), DRD2 (Hamidovic et al., 2009), FAAH (Dlugos et al., 2010), OPRM1 (Dlugos et al., 2011), SLC6A3 (Hamidovic et al., 2010b; Lott et al., 2005), SLC6A4 (Lott et al., 2006), and SLC6A2 (Dlugos et al., 2007). The promising data from these publications were cited throughout the psychiatric genetics literature. However, these researchers recently conducted a study with an additional 200 participants, in which they were unable to replicate the aforementioned findings (Hart et al., 2013).
The authors acknowledged that their failure to replicate indicated that the previous findings may have been false positives. The paper also cites insufficient correction for multiple testing (within each study and across the various studies) and publication bias as contributors to their erroneous findings. The influence of these factors requires that appropriate caution be used in pharmacogenetics research, which may warrant novel statistical or methodological approaches to control for their influence (Bosker et al., 2011; Munafó et al, 2007).
Though we have reviewed several studies showing a pharmacogenetic effect, the literature is often inconsistent about whether a particular gene alteration is related to the treatment outcome under observation. In addition to statistical sources of discrepant findings, other methodological differences in these studies may contribute to inconsistencies in the literature. Therefore, as the field of pharmacogenetics continues to develop, it would greatly benefit from standardization in measures such as the dosage of the pharmacological treatment being studied and how relapse is defined.
It has also been suggested that the source of this problem lies in the fact that our classification system for SRDs lacks etiological (causative) justification. Following this argument, the new DSM-V classification system, which combines substance abuse and dependence into an overarching disorder (Substance Use Disorder), does little to improve upon this issue. Some argue that the classification of patients with SRDs based upon endophenotypes related to disease pathogenesis may identify more distinct groups of affected individuals, which would be more suitable for experimental genetic approaches (see Ball et al., 1995; Basu et al., 2004; Chan et al., 2011; Kranzler et al., 2008 and Moss et al., 2007). For example, two possible endophenotypes of heroin users may be differentiated based their motivation to use the drug. The ability of heroin to activate the opioid and dopaminergic systems and produce pleasurable effects is undoubtedly an important contributor to its abuse liability. However, multiple studies have suggested that some individuals abuse opioids to self-medicate anxiety, depression, mania and psychosis (Fatseas et al., 2009). As such, the etiology of heroin abuse in some patient populations (initially at least) may be attributed to the positive subjective effects of the drug; these individuals may be genetically distinct from those whose abuse can be attributed to the negative reinforcing properties of the drug. As such, studies that examine pharmacogenetics of heroin abuse treatment response may yield different results, depending on the endophenotype of the sample that was tested. In addition to controlling for sample heterogeneity, employing larger samples sizes could improve replicability, leading to more clinically meaningful findings. In this respect, the field of substance abuse could learn from genetic studies investigating response to antidepressants (which utilize large sample sizes, control for population stratification, and employ sound statistical methodology) as a model for future studies (see Hunter et al., 2013 for an example).
The data presented in this review indicate that genetic variation as minimal as a single nucleotide change may alter drug effects, craving, withdrawal and abstinence. Because DA may constitute a pathway to addiction common to all drugs, the majority of this research has focused on genetic alteration in the dopaminergic pathway. However, the gene targets of this type of research are expanding each year. When combined with polygenetic approaches examining the combined effects of multiple variants, we continue to gain a more comprehensive view of the multitude of genetic factors involved in drug abuse. Despite the obstacles in this novel area of research, once refined, pharmacogenetics may allow for the identification of a subset of treatment-seeking patients most likely to respond to a particular medication and those who may be at an increased risk of more severe complications related to drug abuse. The development and course of drug addiction is a highly individualized experience. Genetics may allow us to determine why there is such variability in susceptibility, response, and prognosis. In the future, understanding the influence of genetic factors may change standard clinical practices and improve the lives of those suffering from drug abuse worldwide.
Supplementary Material
Highlights.
We reviewed pharmacogenetic studies on drug subjective effects and medication trials
These studies provide evidence of clinically relevant pharmacogenetic effects
This body of literature has produced conflicting results that require resolution
More rigorous procedural and statistical controls may reduce false positives
Understanding genetic modulation may advance individualized Tx of drug use disorders
Acknowledgements
The authors would like to thank Henry R. Kranzler for his mentorship and contributions this manuscript, along with the National Institute on Drug Abuse (NIDA) for their generous research support. Financial support for the preparation of this manuscript was provided by the NIDA grants: DA030446 to JDJ, and DA016759 and DA09236 to SDC
Role of the Funding Source
The funding sources played no role in the collection, analysis and interpretation of data; in the writing of the article; and in the decision to submit it for publication.
Conflicts of Interest
Over the past three years, SDC has received compensation (in the form of partial salary support) from investigator-initiated studies supported by Reckitt-Benckiser Pharmaceuticals, Schering-Plough Corporation, Johnson & Johnson Pharmaceutical Research & Development, Endo Pharmaceuticals, and MediciNova and served as a consultant to the following companies: AstraZeneca, Camarus, Grunenthal USA, Guidepoint Global, Janssen, Mallinckrodt, Neuromed, Orexo, Pfizer, and Salix.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Supplementary material can be found by accessing the online version of this paper at http://dx.doi.org and by entering doi:…
The studies referenced in Tables 1–3 can be found grouped as a function of their target variant(s) in the online Supplementary Materials, which can be found by can be found by accessing the online version of this paper at http://dx.doi.org and by entering doi:…
Contributions
Only the authors listed are responsible for the content and preparation of this manuscript. JDJ conducted the literature review and produced the first version of the manuscript. SDC participated in filling any gaps in the literature and helped finalize draft versions of review of the manuscript. All authors have approved this manuscript for publication.
REFERENCES
- Agrawal A, Verweij KJ, Gillespie NA, Heath AC, Lessov-Schlaggar CN, Martin NG, Nelson EC, Slutske WS, Whitfield JB, Lynskey MT. The genetics of addiction a translational perspective. Transl. Psychiatry. 2012;2:e140. doi: 10.1038/tp.2012.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al-Eitan LN, Jaradat SA, Su SY, Tay GK, Hulse GK. Mu opioid receptor (OPRM1) as a predictor of treatment outcome in opiate-dependent individuals of Arab descent. Pharmgenomics Pers. Med. 2012;5:99–111. doi: 10.2147/PGPM.S33351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arinami T, Gao M, Hamaguchi H, Toru M. A functional polymorphism in the promoter region of the dopamine D2 receptor gene is associated with schizophrenia. Hum. Mol. Genet. 1997;6:577–582. doi: 10.1093/hmg/6.4.577. [DOI] [PubMed] [Google Scholar]
- Asghari V, Sanyal S, Buchwaldt S, Paterson A, Jovanovic V, Van Tol HH. Modulation of intracellular cyclic AMP levels by different human dopamine D4 receptor variants. J. Neurochem. 1995;65:1157–1165. doi: 10.1046/j.1471-4159.1995.65031157.x. [DOI] [PubMed] [Google Scholar]
- Ball JC, Ross A. The Effectiveness Of Methadone Maintenance Treatment. New York: Springer-Verlag; 1991. [Google Scholar]
- Ball SA, Carroll KM, Babor TF, Rounsaville BJ. Subtypes of cocaine abusers: support for a type A-type B distinction. J. Consult. Clin. Psychol. 1995;63:115–124. doi: 10.1037//0022-006x.63.1.115. [DOI] [PubMed] [Google Scholar]
- Barratt DT, Coller JK, Hallinan R, Byrne A, White JM, Foster DJ, Somogyi AA. ABCB1 haplotype and OPRM1 118A > G genotype interaction in methadone maintenance treatment pharmacogenetics. Pharmgenomics Pers. Med. 2012;5:53–62. doi: 10.2147/PGPM.S29272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basu D, Ball SA, Feinn R, Gelernter J, Kranzler HR. Typologies of drug dependence: comparative validity of a multivariate and four univariate models. Drug Alcohol Depend. 2004;73:289–300. doi: 10.1016/j.drugalcdep.2003.11.004. [DOI] [PubMed] [Google Scholar]
- Barratt DT, Coller JK, Somogyi AA. Association between the DRD2 A1 allele and response to methadone and buprenorphine maintenance treatments. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 2006;141B:323–331. doi: 10.1002/ajmg.b.30319. [DOI] [PubMed] [Google Scholar]
- Benjamin J, Li L, Patterson C, Greenberg BD, Murphy DL, Hamer DH. Population and familial association between the D4 dopamine receptor gene and measures of novelty seeking. Nat. Genet. 1996;12:81–84. doi: 10.1038/ng0196-81. [DOI] [PubMed] [Google Scholar]
- Berlin I, Covey LS, Jiang H, Hamer D. Lack of effect of D2 dopamine receptor TaqI A polymorphism on smoking cessation. Nicotine Tob. Res. 2005;7:725–728. doi: 10.1080/14622200500259176. [DOI] [PubMed] [Google Scholar]
- Berrettini WH, Wileyto EP, Epstein L, Restine S, Hawk L, Shields P, Niaura R, Lerman C. Catechol-O-methyltransferase (COMT) gene variants predict response to bupropion therapy for tobacco dependence. Biol. Psychiatry. 2007;61:111–118. doi: 10.1016/j.biopsych.2006.04.030. [DOI] [PubMed] [Google Scholar]
- Bhaduri N, Mukhopadhyay K. Correlation of plasma dopamine beta-hydroxylase activity with polymorphisms in DβH gene: a study on eastern Indian population. Cell. Mol. Neurobiol. 2008;28:343–350. doi: 10.1007/s10571-007-9256-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bourdelat-Parks BN, Anderson GM, Donaldson ZR, Weiss JM, Bonsall RW, Emery MS, Liles LC, Weinshenker D. Effects of dopamine beta-hydroxylase genotype and disulfiram inhibition on catecholamine homeostasis in mice. Psychopharmacol. (Berl.) 2005;183:72–80. doi: 10.1007/s00213-005-0139-8. [DOI] [PubMed] [Google Scholar]
- Bosker FJ, Hartman CA, Nolte IM, Prins BP, Terpstra P, Posthuma D, van Veen T, Willemsen G, DeRijk RH, de Geus EJ, Hoogendijk WJ, Sullivan PF, et al. Poor replication of candidate genes for major depressive disorder using genome-wide association data. Mol. Psychiatry. 2011;16:516–532. doi: 10.1038/mp.2010.38. [DOI] [PubMed] [Google Scholar]
- Breitling LP, Twardella D, Hoffmann MM, Witt SH, Treutlein J, Brenner H. Prospective association of dopamine-related polymorphisms with smoking cessation in general care. Pharmacogenomics. 2010;11:527–536. doi: 10.2217/pgs.10.1. [DOI] [PubMed] [Google Scholar]
- Burgoyne RD, O’Callaghan DW, Hasdemir B, Haynes LP, Tepikin AV. Neuronal Ca2+-sensor proteins: multitalented regulators of neuronal function. Trends Neurosc. 2004;27:203–209. doi: 10.1016/j.tins.2004.01.010. [DOI] [PubMed] [Google Scholar]
- Carrera MR, Ashley JA, Parsons LH, Wirsching P, Koob GF, Janda KD. Suppression of psychoactive effects of cocaine by active immunization. Nature. 1995;378:727–730. doi: 10.1038/378727a0. [DOI] [PubMed] [Google Scholar]
- Carroll KM, Fenton LR, Ball SA, Nich C, Frankforter TL, Shi J, et al. Efficacy of disulfiram and cognitive behavior therapy in cocaine-dependent outpatients: a randomized placebo-controlled trial. Arch. Gen. Psychiatry. 2004;61:264–272. doi: 10.1001/archpsyc.61.3.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carroll KM, Nich C, Ball SA, McCance E, Rounsavile BJ. Treatment of cocaine and alcohol dependence with psychotherapy and disulfiram. Addiction. 1998;93:713–727. doi: 10.1046/j.1360-0443.1998.9357137.x. [DOI] [PubMed] [Google Scholar]
- Chanasong R, Thanoi S, Watiktinkorn P, Reynolds GP, Nudmamud-Thanoi S. Genetic variation of GRIN1 confers vulnerability to methamphetamine-dependent psychosis in a Thai population. Neurosci. Lett. 2013;551:58–61. doi: 10.1016/j.neulet.2013.07.017. [DOI] [PubMed] [Google Scholar]
- Chan G, Gelernter J, Oslin D, Farrer L, Kranzler HR. Empirically derived subtypes of opioid use and related behaviors. Addiction. 2011;106:1146–1154. doi: 10.1111/j.1360-0443.2011.03390.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen LS, Bloom AJ, Baker TB, Smith SS, Piper ME, Martinez M, Saccone N, Hatsukami D, Goate A, Bierut L. Pharmacotherapy effects on smoking cessation vary with nicotine metabolism gene (CYP2A6) Addiction. 2014;109:128–137. doi: 10.1111/add.12353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Childs E, Hohoff C, Deckert J, Xu K, Badner J, de Wit H. Association between ADORA2A and DRD2 polymorphisms and caffeine-induced anxiety. Neuropsychopharmacol. 2008;33:2791–2800. doi: 10.1038/npp.2008.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cicero TJ, Inciardi JA, Munoz A. Trends in abuse of oxycontin and other opioid analgesics in the United States: 2002–2004. J. Pain. 2005;6:662–672. doi: 10.1016/j.jpain.2005.05.004. [DOI] [PubMed] [Google Scholar]
- Cinciripini PM, Wetter DW, Tomlinson GE, Tsoh JY, De Moor CA, Cinciripini LG. The effects of the DRD2 polymorphism on smoking cessation and negative affect: evidence for a pharmacogenetic effect on mood. Nicotine Tob. Res. 2004;6:229–239. doi: 10.1080/14622200410001676396. [DOI] [PubMed] [Google Scholar]
- Clarke TK, Crist RC, Ang A, Ambrose-Lanci LM, Lohoff FW, Saxon AJ, Ling W, Hillhouse MP, Bruce RD, Woody G, Berrettini WH. Genetic variation in OPRD1 and the response to treatment for opioid dependence with buprenorphine in European-American females. Pharmacogenomics J. 2014 a;14:303–308. doi: 10.1038/tpj.2013.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clarke TK, Weiss AR, Ferarro TN, Kampman KM, Dackis CA, Pettinati HM, O'brien CP, Oslin DW, Lohoff FW, Berrettini WH. The dopamine receptor D2 (DRD2) SNP rs1076560 is associated with opioid addiction. Ann. Hum. Genet. 2014 b;78:33–39. doi: 10.1111/ahg.12046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colilla S, Lerman C, Shields PG, Jepson C, Rukstalis M, Berlin J, DeMichele A, Bunin G, Strom BL, Rebbeck TR. Association of catechol-O-methyltransferase with smoking cessation in two independent studies of women. Pharmacogenet. Genomics. 2005;15:393–398. doi: 10.1097/01213011-200506000-00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coller JK, Barratt DT, Dahlen K, Loennechen MH, Somogyi AA. ABCB1 genetic variability and methadone dosage requirements in opioid-dependent individuals. Clin. Pharmacol. Ther. 2006;80:682–690. doi: 10.1016/j.clpt.2006.09.011. [DOI] [PubMed] [Google Scholar]
- Comer SD, Sullivan MA, Whittington RA, Vosburg SK, Kowalczyk WJ. Abuse liability of prescription opioids compared to heroin in morphine-maintained heroin abusers. Neuropsychopharmacol. 2008;33:1179–1191. doi: 10.1038/sj.npp.1301479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Comings DE, Muhleman D, Ahn C, Gysin R, Flanagan SD. The dopamine D2 receptor gene: a genetic risk factor in substance abuse. Drug Alcohol Depend. 1994;34:175–180. doi: 10.1016/0376-8716(94)90154-6. [DOI] [PubMed] [Google Scholar]
- Comings DE, Ferry L, Bradshaw-Robinson S, Burchette R, Chiu C, Muhleman D. The dopamine D2 receptor (DRD2) gene: a genetic risk factor in smoking. Pharmacogenetics. 1996;6:73–79. doi: 10.1097/00008571-199602000-00006. [DOI] [PubMed] [Google Scholar]
- Conti DV, Lee W, Li D, Liu J, Van Den Berg D, Thomas PD, Bergen AW, Swan GE, Tyndale RF, Benowitz NL, Lerman C. Nicotinic acetylcholine receptor beta2 subunit gene implicated in a systems-based candidate gene study of smoking cessation. Hum. Mol. Genet. 2008;17:2834–2848. doi: 10.1093/hmg/ddn181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crabbe JC. Genetic contributions to addiction. Ann. Rev. Psychol. 2002;53:435–462. doi: 10.1146/annurev.psych.53.100901.135142. [DOI] [PubMed] [Google Scholar]
- Crettol S, Besson J, Croquette-Krokar M, Hämmig R, Gothuey I, Monnat M, Déglon JJ, Preisig M, Eap CB. Association of dopamine and opioid receptor genetic polymorphisms with response to methadone maintenance treatment. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2008;32:1722–1727. doi: 10.1016/j.pnpbp.2008.07.009. [DOI] [PubMed] [Google Scholar]
- Crettol S, Deglon JJ, Besson J, Croquette-Krokar M, Hammig R, Gothuey I. ABCB1 and cytochrome P450 genotypes and phenotypes: influence on methadone plasma levels and response to treatment. Clin. Pharmacol. Ther. 2006;80:668–681. doi: 10.1016/j.clpt.2006.09.012. [DOI] [PubMed] [Google Scholar]
- Crist RC, Clarke TK, Ang A, Ambrose-Lanci LM, Lohoff FW, Saxon AJ, Ling W, Hillhouse MP, Bruce RD, Woody G, Berrettini WH. An intronic variant in OPRD1 predicts treatment outcome for opioid dependence in African-Americans. Neuropsychopharmacol. 2013a;38:2003–2010. doi: 10.1038/npp.2013.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crist RC, Clarke TK, Ang A. Case-control association analysis of polymorphisms in the delta-opioid receptor, OPRD1, with cocaine and opioid addicted populations. Drug. Alcohol Depend. 2013b;127:122–128. doi: 10.1016/j.drugalcdep.2012.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dahl JP, Jepson C, Levenson R, Wileyto EP, Patterson F, Berrettini WH, Lerman C. Interaction between variation in the D2 dopamine receptor (DRD2) and the neuronal calcium sensor-1 (FREQ) genes in predicting response to nicotine replacement therapy for tobacco dependence. Pharmacogenomics J. 2006;6:194–199. doi: 10.1038/sj.tpj.6500358. [DOI] [PubMed] [Google Scholar]
- Daly JW, Fredholm BB. Caffeine: an atypical drug of dependence. Drug Alcohol Depend. 1998;51:199–206. doi: 10.1016/s0376-8716(98)00077-5. [DOI] [PubMed] [Google Scholar]
- David SP, Munafò MR, Murphy MF, Proctor M, Walton RT, Johnstone EC. Genetic variation in the dopamine D4 receptor (DRD4) gene and smoking cessation: follow-up of a randomised clinical trial of transdermal nicotine patch. Pharmacogenomics J. 2008;8:122–128. doi: 10.1038/sj.tpj.6500447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- David SP, Niaura R, Papandonatos GD, Shadel WG, Burkholder GJ, Britt DM, Day A, Stumpff J, Hutchison K, Murphy M, Johnstone E, Griffiths SE, Walton RT. Does the DRD2-Taq1 A polymorphism influence treatment response to bupropion hydrochloride for reduction of the nicotine withdrawal syndrome. Nicotine Tob. Res. 2003;5:935–942. doi: 10.1080/14622200310001615295. [DOI] [PubMed] [Google Scholar]
- David SP, Strong DR, Munafò MR, Brown RA, Lloyd-Richardson EE, Wileyto PE, Evins EA, Shields PG, Lerman C, Niaura R. Bupropion efficacy for smoking cessation is influenced by the DRD2 Taq1 A polymorphism: analysis of pooled data from two clinical trials. Nicotine Tob. Res. 2007a;9:1251–1257. doi: 10.1080/14622200701705027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- David SP, Brown RA, Papandonatos GD, Kahler CW, Lloyd-Richardson EE, Munafo MR, Shields PG, Lerman C, Strong D, McCaffery J, Niaura R. Pharmacogenetic clinical trial of sustained-release bupropion for smoking cessation. Nicotine Tob. Res. 2007b;9:821–833. doi: 10.1080/14622200701382033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- David SP, Munafo MR, Murphy MF, Walton RT, Johnstone EC. The serotonin transporter 5-HTTLPR polymorphism and treatment response to nicotine patch: follow-up of a randomized controlled trial. Nicotine Tob. Res. 2007c;9:225–231. doi: 10.1080/14622200601078566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darke SG, Rossn J, Hall W. Overdose among heroin users in Sydney, Australia: I. Prevalence and correlates of non-fatal overdose. Addiction. 1996;91:405–411. [PubMed] [Google Scholar]
- Degenhardt L, Bucello C, Mathers B, Briegleb C, Ali H, Hickman M, McLaren J. Mortality among regular or dependent users of heroin and other opioids: a systematic review and meta-analysis of cohort studies. Addiction. 2011;106:32–51. doi: 10.1111/j.1360-0443.2010.03140.x. [DOI] [PubMed] [Google Scholar]
- De Ruyck K, Nackaerts K, Beels L, Werbrouck J, De Volder A, Meysman M, Salhi B, Van Meerbeeck J, Thierens H. Genetic variation in three candidate genes and nicotine dependence, withdrawal and smoking cessation in hospitalized patients. Pharmacogenomics. 2010;11:1053–1063. doi: 10.2217/pgs.10.75. [DOI] [PubMed] [Google Scholar]
- Deinum J, Steenbergen-Spanjers GC, Jansen M, Boomsma F, Lenders JW, van Ittersum FJ, Huck N, van den Heuvel LP, Wevers RA. DβH gene variants that cause low plasma dopamine beta hydroxylase with or without a severe orthostatic syndrome. J. Med. Genet. 2004;41:e38. doi: 10.1136/jmg.2003.009282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dennis BB, Bawor M, Thabane L, Sohani Z, Samaan Z. Impact of ABCB1 and CYP2B6 genetic polymorphisms on methadone metabolism, dose and treatment response in patients with opioid addiction: a systematic review and meta-analysis. PLoS One. 2014 doi: 10.1371/journal.pone.0086114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Chiara G, Imperato A. Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc. Natl. Acad. Sci. 1988;85:5274–5278. doi: 10.1073/pnas.85.14.5274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Chiara G, Bassareo V, Fenu S, De Luca M, Spina L, Cadoni C, Acquas E, Carboni E, Valentini V, Lecca D. Dopamine and drug addiction: the nucleus accumbens shell connection. Neuropharmacol. 2004;47:227–241. doi: 10.1016/j.neuropharm.2004.06.032. [DOI] [PubMed] [Google Scholar]
- Diamond A. Consequences of variations in genes that affect dopamine in prefrontal cortex. Cereb. Cortex. 2007;17:i161–i170. doi: 10.1093/cercor/bhm082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doehring A, Hentig N, Graff J, Salamat S, Schmidt M, Geisslinger G. Genetic variants altering dopamine D2 receptor expression or function modulate the risk of opiate addiction and the dosage requirements of methadone substitution. Pharmacogenet. Genomics. 2009;19:407–414. doi: 10.1097/FPC.0b013e328320a3fd. [DOI] [PubMed] [Google Scholar]
- Duan J, Wainwright MS, Comeron JM, Saitou N, Sanders AR, Gelernter J, Gejman PV. Synonymous mutations in the human dopamine receptor D2 (DRD2) affect mRNA stability and synthesis of the receptor. Hum. Mol. Genet. 2003;12:205–216. doi: 10.1093/hmg/ddg055. [DOI] [PubMed] [Google Scholar]
- Dlugos A, Freitag C, Hohoff C, McDonald J, Cook E, Deckert J, de Wit H. Norepinephrine transporter gene variation modulates acute response to D-amphetamine. Biol. Psychiatry. 2007;61:1296–1305. doi: 10.1016/j.biopsych.2006.09.031. [DOI] [PubMed] [Google Scholar]
- Dlugos AM, Hamidovic A, Hodgkinson CA, Goldman D, Palmer AA, de Wit H. More aroused, less fatigued: fatty acid amide hydrolase gene polymorphisms influence acute response to amphetamine. Neuropsychopharmacol. 2010;35:613–622. doi: 10.1038/npp.2009.166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dlugos AM, Hamidovic A, Hodgkinson C, Shen PH, Goldman D, Palmer AA, de Wit H. OPRM1 gene variants modulate amphetamine-induced euphoria in humans. Genes Brain Behav. 2011;10:199–209. doi: 10.1111/j.1601-183X.2010.00655.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farrer LA, Kranzler HR, Yu Y, Weiss RD, Brady KT, Anton R, Cubells JF, Gelernter J. Association of variants in MANEA with cocaine-related behaviors. Arch. Gen. Psychiatry. 2009;66:267–274. doi: 10.1001/archgenpsychiatry.2008.538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fatseas M, Lavie E, Denis C, Auriacombe M. Self-perceived motivation for benzodiazepine use and behavior related to benzodiazepine use among opiate-dependent patients. J. Subst. Abuse Treat. 2009;37:407–411. doi: 10.1016/j.jsat.2009.03.006. [DOI] [PubMed] [Google Scholar]
- Fergusson DM, Boden JM. Cannabis use and later life outcomes. Addiction. 2008;103:969–976. doi: 10.1111/j.1360-0443.2008.02221.x. [DOI] [PubMed] [Google Scholar]
- Flanagin B, Cook E, Jr, de Wit H. An association study of the brain-derived neurotrophic factor Val66Met polymorphism and amphetamine response. Am. J. Med. Genet. 2006;141:576–583. doi: 10.1002/ajmg.b.30327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fong TM, Heymsfield SB. Cannabinoid-1 receptor inverse agonists: current understanding of mechanism of action and unanswered questions. Int. J. Obes. (Lond.) 2009;33:947–955. doi: 10.1038/ijo.2009.132. [DOI] [PubMed] [Google Scholar]
- Fonseca F, Gratacòs M, Escaramís G, De Cid R, Martín-Santos R, Fernández-Espejo E, Estivill X, Torrens M. Response to methadone maintenance treatment is associated with the MYOCD and GRM6 genes. Mol. Diagn. Ther. 2010;14:171–178. doi: 10.1007/BF03256370. [DOI] [PubMed] [Google Scholar]
- Frosst P, Blom H, Milos R, Govette P, Sheppard C, Matthews R, Boers GJH, den Heijer M, Kluijtmans LAJ, van den Heuve LP, Rozen R. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat. Genet. 1995;10:111–113. doi: 10.1038/ng0595-111. [DOI] [PubMed] [Google Scholar]
- Grabowski JA, Shearer J, Merrill J, Negus SS. Agonist-like, replacement pharmacotherapy for stimulant abuse and dependence. Addict. Behav. 2004;29:1439–1464. doi: 10.1016/j.addbeh.2004.06.018. [DOI] [PubMed] [Google Scholar]
- Gelernter J, Panhuysen C, Weiss R, Brady K, Hesselbrock V, Rounsaville B, Poling J, Wilcox M, Farrer L, Kranzler HR. Genomewide linkage scan for cocaine dependence and related traits: significant linkages for a cocaine-related trait and cocaine-induced paranoia. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 2005;136B:45–52. doi: 10.1002/ajmg.b.30189. [DOI] [PubMed] [Google Scholar]
- Gelernter J, Kranzler HR, Satel SL, Rao PA. Genetic association between dopamine transporter protein alleles and cocaine-induced paranoia. Neuropsychopharmacol. 1994;11:195–200. doi: 10.1038/sj.npp.1380106. [DOI] [PubMed] [Google Scholar]
- Gilbert DG, Zuo Y, Rabinovich NE, Riise H, Needham R, Huggenvik JI. Neurotransmission-related genetic polymorphisms, negative affectivity traits, and gender predict tobacco abstinence symptoms across 44 days with and without nicotine patch. J. Abnorm. Psychol. 2009;118:322–334. doi: 10.1037/a0015382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilson AM, Ryan KM, Joranson DE, Dahl JL. A reassessment of trends in the medical use and abuse of opioid analgesics and implications for diversion control: 1997–2002. J. Pain Symptom Manage. 2004;28:176–188. doi: 10.1016/j.jpainsymman.2004.01.003. [DOI] [PubMed] [Google Scholar]
- Girault JA, Greengard P. The neurobiology of dopamine signaling. Arch. Neurol. 2004;61:641–644. doi: 10.1001/archneur.61.5.641. [DOI] [PubMed] [Google Scholar]
- Goldman G, Oroszi F, Ducci F. The genetics of addictions: uncovering the genes. Nat. Rev. Genet. 2005;6:521–532. doi: 10.1038/nrg1635. [DOI] [PubMed] [Google Scholar]
- Grigg D. The worlds of tea and coffee: patterns of consumption. Geojournal. 2002;57:283. [Google Scholar]
- Hald J, Jacobsen E, Larsen V. The sensitizing effect of tetraethylthiuramdisulphide (antabuse) to ethylalcohol. Acta. Pharmacol. Toxicol. (Copenh) 1948;4:285–296. [Google Scholar]
- Hall W, Degenhardt L. Adverse health effects of non-medical cannabis use. Lancet. 2009;374:1383–1391. doi: 10.1016/S0140-6736(09)61037-0. [DOI] [PubMed] [Google Scholar]
- Hamidovic A, Dlugos A, Palmer AA, de Wit H. Catechol-O-methyltransferase val158met genotype modulates sustained attention in both the drug-free state and in response to amphetamine. Psychiatr. Genet. 2010a;20:85. doi: 10.1097/YPG.0b013e32833a1f3c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamidovic A, Dlugos A, Palmer AA, de Wit H. Polymorphisms in dopamine transporter (SLC6A3) are associated with stimulant effects of d-amphetamine: an exploratory pharmacogenetic study using healthy volunteers. Behav. Genet. 2010b;40:255–261. doi: 10.1007/s10519-009-9331-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamidovic A, Dlugos A, Skol A, Palmer AA, de Wit H. Evaluation of genetic variability in the dopamine receptor D2 in relation to behavioral inhibition and impulsivity/sensation seeking: an exploratory study with d-amphetamine in healthy participants. Exp. Clin. Psychopharmacol. 2009;17:374–383. doi: 10.1037/a0017840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harano M, Uchimura N, Abe H, Ishibashi M, Iida N, Yanagimoto K, Tanaka T, Maeda H, Sora I, Iyo M, Komiyama T, Yamada M, Sekine Y, Inada T, Ozaki N, Ujike H. A polymorphism of DRD2 gene and brain atrophy in methamphetamine psychosis. Ann. N.Y. Acad. Sci. 2004;1025:307–315. doi: 10.1196/annals.1316.038. [DOI] [PubMed] [Google Scholar]
- Hart AB, de Wit H, Palmer AA. Candidate gene studies of a promising intermediate phenotype: failure to replicate. Neuropsychopharmacol. 2013;38:802–816. doi: 10.1038/npp.2012.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hauge XY, Grandy DK, Eubanks JH, Evans GA, Civelli O, Litt M. Detection and characterization of additional DNA polymorphisms in the dopamine D2 receptor gene. Genomics. 1991;10:527–530. doi: 10.1016/0888-7543(91)90431-d. [DOI] [PubMed] [Google Scholar]
- Heinz A, Sander T, Harms H, Finckh U, Kuhn S, Dufeu P, Dettling M, Gräf K, Rolfs A, Rommelspacher H, Schmidy LG. Lack of allelic association of dopamine D1 and D2 (TaqIA) receptor gene polymorphisms with reduced dopaminergic sensitivity to alcoholism. Alcohol. Clin. Exp. Res. 2006;20:1109–1113. doi: 10.1111/j.1530-0277.1996.tb01954.x. [DOI] [PubMed] [Google Scholar]
- Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS, Manolio TA. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc. Natl. Acad. Sci. 2009;106:9362–9367. doi: 10.1073/pnas.0903103106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirvonen M, Laakso A, Nagren K, Rinne JO, Pohjalainen T, Hietala J. C957T polymorphism of the dopamine D2 receptor (DRD2) gene affects striatal DRD2 availability in vivo. Mol. Psychiatry. 2004;9:1060–1061. doi: 10.1038/sj.mp.4001561. [DOI] [PubMed] [Google Scholar]
- Hohoff C, Mcdonald JM, Baune BT, Cook EH, Deckert J, de Wit H. Interindividual variation in anxiety response to amphetamine: possible role for adenosine A 2Areceptor gene variants. Am. J. Med. Genet. 2005;139B:42–44. doi: 10.1002/ajmg.b.30228. [DOI] [PubMed] [Google Scholar]
- Hung CC, Chiou MH, Huang BH, Hsieh YW, Hsieh TJ, Huang CL, Lane HY. Impact of genetic polymorphisms in ABCB1, CYP2B6, OPRM1, ANKK1 and DRD2 genes on methadone therapy in Han Chinese patients. Pharmacogenomics. 2011;12:1525–1533. doi: 10.2217/pgs.11.96. [DOI] [PubMed] [Google Scholar]
- Hunter AM, Leuchter AF, Power RA, Muthén B, McGrath PJ, Lewis CM, Cook IA, Garriock HA, McGuffin P, Uher R, Hamilton SP. A genome-wide association study of a sustained pattern of antidepressant response. J. Psychiatr. Res. 2013;47:1157–1165. doi: 10.1016/j.jpsychires.2013.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hutchison KE, Allen DL, Filbey FM, Jepson C, Lerman C, Benowitz NL, Stitzel J, Bryan A, McGeary J, Haughey HM. CHRNA4 and tobacco dependence: from gene regulation to treatment outcome. Arch. Gen. Psychiatry. 2007;64:1078–1086. doi: 10.1001/archpsyc.64.9.1078. [DOI] [PubMed] [Google Scholar]
- Hutchison KE, Lachance H, Niaura R, Bryan A, Smolen A. The DRD4 VNTR polymorphism influences reactivity to smoking cues. J. Abnorm. Psychol. 2002;111:134–143. doi: 10.1037//0021-843x.111.1.134. [DOI] [PubMed] [Google Scholar]
- Ittiwut R, Listman JB, Ittiwut C, Cubells JF, Weiss RD, Brady K, Oslin D, Farrer LA, Kranzler HR, Gelernter J. Association between polymorphisms in catechol-O-methyltransferase (COMT) and cocaine-induced paranoia in European-American and African-American populations. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2011;156:651–660. doi: 10.1002/ajmg.b.31205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnstone EC, Elliot KM, David SP, Murphy MF, Walton RT, Munafo MR. Association of COMT Val108/158Met genotype with smoking cessation in a nicotine replacement therapy randomized trial. Cancer Epidemiol. Biomarkers Prev. 2007;16:1065–1069. doi: 10.1158/1055-9965.EPI-06-0936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnstone EC, Yudkin PL, Hey K, Roberts SJ, Welch SJ, Murphy MF. Genetic variation in dopaminergic pathways and short-term effectiveness of the nicotine patch. Pharmacogenetics. 2004;14:83–90. doi: 10.1097/00008571-200402000-00002. [DOI] [PubMed] [Google Scholar]
- Jones JD, Comer SD, Kranzler HR. The pharmacogenetics of alcohol use disorder. Alcohol. Clin. Exp. Res. 2014 doi: 10.1111/acer.12643. (In Press). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jönsson EG, Nöthen MM, Grünhage F, Farde L, Nakashima Y, Propping P, Sedvall GC. Polymorphisms in the dopamine D2 receptor gene and their relationships to striatal dopamine receptor density of healthy volunteers. Mol. Psychiatry. 1999;3:290–296. doi: 10.1038/sj.mp.4000532. [DOI] [PubMed] [Google Scholar]
- Joseph H, Stancliff S, Langrod J. methadone maintenance treatment (mmt): a review of historical and clinical issues. Mount Sinai Med. J. 2000;5–6:347–364. [PubMed] [Google Scholar]
- Kabbani N, Negyessy L, Lin R, Goldman-Rakic P, Levenson R. Interaction with neuronal calcium sensor NCS-1 mediates desensitization of the D2 dopamine receptor. J. Neurosci. 2002;22:8476–8486. doi: 10.1523/JNEUROSCI.22-19-08476.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalayasiri R, Kranzler HR, Weiss R, Brady K, Gueorguieva R, Panhuysen C, Yang BZ, Farrer L, Gelernter J, Malison RT. Risk factors for cocaine-induced paranoia in cocaine-dependent sibling pairs. Drug Alcohol Depend. 2006;84:77–84. doi: 10.1016/j.drugalcdep.2005.12.002. [DOI] [PubMed] [Google Scholar]
- Kalayasiri R, Sughondhabirom A, Gueorguieva R, Coric V, Lynch WJ, Lappalainen J, Gelernter J, Cubells JF, Malison RT. Dopamine beta-hydroxylase gene (DbetaH)-1021C-T influences self-reported paranoia during cocaine self-administration. Biol. Psychiatry. 2007;61:1310–1313. doi: 10.1016/j.biopsych.2006.08.012. [DOI] [PubMed] [Google Scholar]
- Kendler KS, Karkowski LM, Neale MC, Prescott CA. Illicit psychoactive substance use, heavy use, abuse, and dependence in a US population-based sample of male twins. Arch. Gen. Psychiatry. 2000;57:261–269. doi: 10.1001/archpsyc.57.3.261. [DOI] [PubMed] [Google Scholar]
- Kintz P. Deaths involving buprenorphine: a compendium of French cases. Forensic Sci. Int. 2001;121:65–69. doi: 10.1016/s0379-0738(01)00454-6. [DOI] [PubMed] [Google Scholar]
- Kishi T, Fukuo Y, Okochi T, Kitajima T, Kawashima K, Naitoh H, Ujike H, Inada T, Yamada M, Uchimura N, Sora I, Iyo M, Ozaki N, Iwata N. Serotonin 6 receptor gene is associated with methamphetamine-induced psychosis in a Japanese population. Drug Alcohol Depend. 2011;113:1–7. doi: 10.1016/j.drugalcdep.2010.06.021. [DOI] [PubMed] [Google Scholar]
- Kobayashi H, Ide S, Hasegawa J, Ujike H, Sekine Y, Ozaki N, Inada T, Harano M, Komiyama T, Yamada M, Iyo M, Shen HW, Ikeda K, Sora I. Study of association between alpha-synuclein gene polymorphism and methamphetamine psychosis/dependence. Ann. N. Y. Acad. Sci. 2004;1025:325–334. doi: 10.1196/annals.1316.040. [DOI] [PubMed] [Google Scholar]
- Köhnke MD, Zabetian CP, Anderson GM, Kolb W, Gaertner I, Buchkremer G, Vonthein R, Schick S, Lutz U, Köhnke AM, Cubells JF. A genotype-controlled analysis of plasma dopamine beta-hydroxylase in healthy and alcoholic subjects: evidence for alcohol-related differences in noradrenergic function. Biol. Psychiatry. 2002;52:1151–1158. doi: 10.1016/s0006-3223(02)01427-0. [DOI] [PubMed] [Google Scholar]
- Kosten TR, Wu G, Huang W, Harding MJ, Hamon SC, Lappalainen J, Nielsen DA. Pharmacogenetic randomized trial for cocaine abuse: disulfiram and dopamine β-hydroxylase. Biol. Psychiatry. 2013;73:219–224. doi: 10.1016/j.biopsych.2012.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kranzler HR, Wilcox M, Weiss RD, Brady K, Hesselbrock V, Rounsaville B, Farrer L, Gelernter J. The validity of cocaine dependence subtypes. Addict. Behav. 2008;33:41–53. doi: 10.1016/j.addbeh.2007.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laine TP, Ahonen A, Räsänen P, Pohjalainen T, Tiihonen J, Hietala J. The A1 allele of the D2 dopamine receptor gene is associated with high dopamine transporter density in detoxified alcoholics. Alcohol Alcohol. 2001;36:262–265. doi: 10.1093/alcalc/36.3.262. [DOI] [PubMed] [Google Scholar]
- Lawford BR, Young RM, Noble EP, Sargent J, Rowell J, Shadforth S, Zhang X, Ritchie T. The D(2) dopamine receptor A(1) allele and opioid dependence: association with heroin use and response to methadone treatment. Am. J. Med. Genet. 2000;96:592–598. doi: 10.1002/1096-8628(20001009)96:5<592::aid-ajmg3>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
- Lee AM, Jepson C, Hoffmann E, Epstein L, Hawk LW, Lerman C, Tyndale RF. CYP2B6 genotype alters abstinence rates in a bupropion smoking cessation trial. Biol. Psychiatry. 2007a;62:635–641. doi: 10.1016/j.biopsych.2006.10.005. [DOI] [PubMed] [Google Scholar]
- Lee AM, Jepson C, Shields PG, Benowitz N, Lerman C, Tyndale RF. CYP2B6 genotype does not alter nicotine metabolism, plasma levels, or abstinence with nicotine replacement therapy. Cancer Epidemiol. Biomarkers Prev. 2007b;16:1312–1314. doi: 10.1158/1055-9965.EPI-07-0188. [DOI] [PubMed] [Google Scholar]
- Lei B, Morris DP, Smith MP, Svetkey LP, Newman MF, Rotter JI, Buchanan TA, Beckstrom-Sternberg SM, Green ED, Schwinn DA. Novel human alpha-1a-adrenocepter single nucleotide polymorphisms alter receptor pharmacology and biological function. Naunyn Schmiedebergs Arch. Pharmacol. 2005;371:229–239. doi: 10.1007/s00210-005-1019-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lerman C, Jepson C, Wileyto EP, Epstein LH, Rukstalis M, Patterson F. Role of functional genetic variation in the dopamine D2 receptor (DRD2) in response to bupropion and nicotine replacement therapy for tobacco dependence: results of two randomized clinical trials. Neuropsychopharmacol. 2006;6:47–64. doi: 10.1038/sj.npp.1300861. [DOI] [PubMed] [Google Scholar]
- Lerman C, Shields PG, Wileyto EP, Audrain J, Pinto A, Hawk L, Krishnan S, Niaura R, Epstein L. Pharmacogenetic investigation of smoking cessation treatment. Pharmacogenetics. 2002;12:627–634. doi: 10.1097/00008571-200211000-00007. [DOI] [PubMed] [Google Scholar]
- Lerman C, Shields PG, Wileyto EP, Audrain J, Pinto A, Hawk L, Jr, Pinto A, Kucharski S, Krishnan S, Niaura R, Epstein LH. Effects of dopamine transporter and receptor polymorphisms on smoking cessation in a bupropion clinical trial. Health Psychol. 2003;22:541–548. doi: 10.1037/0278-6133.22.5.541. [DOI] [PubMed] [Google Scholar]
- Lerman C, Wileyto EP, Patterson F, Rukstalis M, Audrain-McGovern J, Restine S, Shields PG, Kaufmann V, Redden D, Benowitz N, Berrettini WH. The functional mu opioid receptor (OPRM1) Asn40Asp variant predicts short-term response to nicotine replacement therapy in a clinical trial. Pharmacogenomics J. 2004;4:184–192. doi: 10.1038/sj.tpj.6500238. [DOI] [PubMed] [Google Scholar]
- Levran O, O'Hara K, Peles E, Li D, Barral S, Ray B. ABCB1 (MDR1) genetic variants are associated with methadone doses required for effective treatment of heroin dependence. Hum. Mol. Genet. 2008a;17:2219–2227. doi: 10.1093/hmg/ddn122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levran O, Londono D, O’Hara K, Nielsen DA, Peles E, Rotrosen J, Casadonte P, Linzy S, Randesi M, Ott J, Adelson M, Kreek MJ. Genetic susceptibility to heroin addiction: a candidate gene association study. Genes Brain Behav. 2008b;7:720–729. doi: 10.1111/j.1601-183X.2008.00410.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levran O, Peles E, Randesi M, Shu X, Ott J, Shen PH, Adelson M, Kreek MJ. Association of genetic variation in pharmacodynamic factors with methadone dose required for effective treatment of opioid addiction. Pharmacogenomics. 2013;14:755–768. doi: 10.2217/pgs.13.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Shao C, Zhang D, Zhao M, Lin L, Yan P, Xie Y, Jiang K, Jin L. The effect of dopamine D2, D5 receptor and transporter (SLC6A3) polymorphisms on the cue-elicited heroin craving in Chinese. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 2006;141:269–273. doi: 10.1002/ajmg.b.30264. [DOI] [PubMed] [Google Scholar]
- Li CY, Zhou WZ, Zhang PW, Johnson C, Wei L, Uhl GR. Meta-analysis and genome-wide interpretation of genetic susceptibility to drug addiction. BMC Genomics. 2011;12:508. doi: 10.1186/1471-2164-12-508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lott D, Kim S, Cook E, Jr, de Wit H. Dopamine transporter gene associated with diminished subjective response to amphetamine. Neuropsychopharmacol. 2005;30:602–609. doi: 10.1038/sj.npp.1300637. [DOI] [PubMed] [Google Scholar]
- Lott D, Kim S, Cook E, Jr, de Wit H. Serotonin transporter genotype and acute subjective response to amphetamine. Am. J. Addict. 2006;15:327–335. doi: 10.1080/10550490600859868. [DOI] [PubMed] [Google Scholar]
- Lotta T, Vidgren J, Tilgmann C, Ulmanen I, Melen K, Julkunen I, Taskinen J. Kinetics of human soluble and membrane-bound catechol O-methyltransferase: a revised mechanism and description of the thermolabile variant of the enzyme. Biochem. 1995;34:4202–4210. doi: 10.1021/bi00013a008. [DOI] [PubMed] [Google Scholar]
- Mackillop J, Menges DP, McGeary JE, Lisman SA. Effects of craving and DRD4 VNTR genotype on the relative value of alcohol: an initial human laboratory study. Behav. Brain Funct. 2007;3:11. doi: 10.1186/1744-9081-3-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsuzawa D, Hashimoto K, Miyatake R, Shirayama Y, Shimizu E, Maeda K, Suzuki Y, Mashimo Y, Sekine Y, Inada T, Ozaki N, Iwata N, Harano M, Komiyama T, Yamada M, Sora I, Ujike H, Hata A, Sawa A, Iyo M. Identification of functional polymorphisms in the promoter region of the human PICK1 gene and their association with methamphetamine psychosis. Am. J. Psychiatry. 2007;164:1105–1114. doi: 10.1176/ajp.2007.164.7.1105. [DOI] [PubMed] [Google Scholar]
- Mattick RP, Kimber J, Breen C, Davoli M. Buprenorphine maintenance versus placebo or methadone maintenance for opioid dependence. Cochrane Database Syst. Rev. 2008;16:CD002207. doi: 10.1002/14651858.CD002207.pub3. [DOI] [PubMed] [Google Scholar]
- Mattay VS, Goldberg TE, Fera F, Hariri AR, Tessitore A, Egan MF, Kolachana B, Callicott JH, Weinberger DR. Catechol O-methyltransferase val158-met genotype and individual variation in the brain response to amphetamine. Proc. Natl. Acad. Sci. U.S.A. 2003;100:6186–6191. doi: 10.1073/pnas.0931309100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mayer P, Rochlitz H, Rauch E, Rommelspacher H, Hasse HE, Schmidt S, Hollt V. Association between a delta opioid receptor gene polymorphism and heroin dependence in man. Neuroreport. 1997;8:2547–2550. doi: 10.1097/00001756-199707280-00025. [DOI] [PubMed] [Google Scholar]
- McKinney EF, Walton RT, Yudkin P, Fuller A, Haldar NA, Mant D. Association between polymorphisms in dopamine metabolic enzymes and tobacco consumption in smokers. Pharmacogenetics. 2000;10:483–491. doi: 10.1097/00008571-200008000-00001. [DOI] [PubMed] [Google Scholar]
- Meyer R, Patel AM, Rattana SK, Quock TP, Mody SH. Prescription opioid abuse: a literature review of the clinical and economic burden in the United States. Popul. Health Manag. EPub Ahead of Print. 2014 doi: 10.1089/pop.2013.0098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mignini F, Napolioni V, Codazzo C, Carpi FM, Vitali M, Romeo M, Ceccanti M. DRD2/ANKK1 TaqIA and SLC6A3 VNTR polymorphisms in alcohol dependence: association and gene-gene interaction study in a population of Central Italy. Neurosci. Lett. 2012;522:103–107. doi: 10.1016/j.neulet.2012.06.008. [DOI] [PubMed] [Google Scholar]
- Moratti E, Kashanpour H, Lombardelli T, Maisto M. Intravenous misuse of buprenorphine: characteristics and extent among patients undergoing drug maintenance therapy. Clin. Drug Investig. 2010;30:3–11. doi: 10.2165/11536020-000000000-00000. [DOI] [PubMed] [Google Scholar]
- Moss HB, Chen CM, Yi HY. Subtypes of alcohol dependence in a nationally representative sample. Drug Alcohol Depend. 2007;91:149–158. doi: 10.1016/j.drugalcdep.2007.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moyer RA, Wang D, Papp AC, Smith RM, Duque L, Mash DC, Sadee W. Intronic polymorphisms affecting alternative splicing of human dopamine D2 receptor are associated with cocaine abuse. Neuropsychopharmacol. 2011;36:753–762. doi: 10.1038/npp.2010.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Munafò MR, Elliot KM, Murphy MF, Walton RT, Johnstone EC. Association of the mu-opioid receptor gene with smoking cessation. Pharmacogenomics J. 2007;7:353–361. doi: 10.1038/sj.tpj.6500432. [DOI] [PubMed] [Google Scholar]
- Munafò MR, Johnstone EC, Aveyard P, Marteau T. Lack of association of OPRM1 genotype and smoking cessation. Nicotine Tob. Res. 2013;15:739–744. doi: 10.1093/ntr/nts174. [DOI] [PubMed] [Google Scholar]
- Munafo MR, Johnstone EC, Wileyto EP, Shields PG, Elliot KM, Lerman C. Lack of association of 5-HTTLPR genotype with smoking cessation in a nicotine replacement therapy randomized trial. Cancer Epidemiol. Biomarkers Prev. 2006;15:398–400. doi: 10.1158/1055-9965.EPI-05-0648. [DOI] [PubMed] [Google Scholar]
- Munafò MR, Timpson NJ, David SP, Ebrahim S, Lawlor DA. Association of the DRD2 gene Taq1 A polymorphism and smoking behaviour: a meta-analysis and new data. Nicotine Tob. Res. 2009;11:64–76. doi: 10.1093/ntr/ntn012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakamura M, Ueno S, Sano A, Tanabe H. The human serotonin transporter gene linked polymorphism (5-HTTLPR) shows ten novel allelic variants. Mol. Psychiatry. 2000;5:32–38. doi: 10.1038/sj.mp.4000698. [DOI] [PubMed] [Google Scholar]
- Nacak M, Isir AB, Balci SO, Pehlivan S, Benlier N, Aynacioglu S. Analysis of dopamine D2 receptor (DRD2) gene polymorphisms in cannabinoid addicts. J. Forensic Sci. 2012;57:1621–1624. doi: 10.1111/j.1556-4029.2012.02169.x. [DOI] [PubMed] [Google Scholar]
- Neville MJ, Johnstone EC, Walton RT. Identification and characterization of ANKK1: a novel kinase gene closely linked to DRD2 on chromosome band 11q23.1. Hum. Mutat. 2004;23:540–545. doi: 10.1002/humu.20039. [DOI] [PubMed] [Google Scholar]
- National Institute on Drug Abuse (NIDA) Research Report Series: Cocaine Abuse and Addiction. [Last Accessed, Nov. 14th 2013];2009 http://www.drugabuse.gov/sites/default/files/rrcocaine.pdf.
- National Institute on Drug Abuse (NIDA) The Science of Drug Use and Addiction: The Basics. [Last Accessed, Dec. 15th 2014];2014 http://www.drugabuse.gov/sites/default/files/mediaguide_web_3_0.pdf.
- Nielsen DA, Hamon SC, Kosten TR. The κ-opioid receptor gene as a predictor of response in a cocaine vaccine clinical trial. Psychiatr. Genet. 2013;23:225–232. doi: 10.1097/YPG.0000000000000008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nielsen DA, Ji F, Yuferov V, Ho A, Chen A, Levran O, Ott J, Kreek MJ. Genotype patterns that contribute to increased risk for or protection from developing heroin addiction. Mol. Psychiatry. 2008;13:417–428. doi: 10.1038/sj.mp.4002147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noble EP, Blum K, Khalsa ME, Ritchie T, Montgomery A, Wood RC, Fitch RJ, Ozkaragoz T, Sheridan PJ, Anglin MD. Allelic association of the D2 dopamine receptor gene with cocaine dependence. Drug Alcohol Depend. 1993;33:271–285. doi: 10.1016/0376-8716(93)90113-5. [DOI] [PubMed] [Google Scholar]
- Noble EP, St. Jeor ST, Ritchie T, Syndulko K, St. Jeor SC, Fitch RJ, Brunner RL, Sparkes RS. D2 dopamine receptor gene and cigarette smoking: a reward gene? Med. Hypotheses. 1994;42:257–260. doi: 10.1016/0306-9877(94)90127-9. [DOI] [PubMed] [Google Scholar]
- O'Gara C, Stapleton J, Sutherland G, Guindalini C, Neale B, Breen G, Ball D. Dopamine transporter polymorphisms are associated with short-term response to smoking cessation treatment. Pharmacogenet. Genomics. 2007;17:61–67. doi: 10.1097/01.fpc.0000236328.18928.4c. [DOI] [PubMed] [Google Scholar]
- O’Hara BF, Smith SS, Bird G, Persico AM, Suarez BK, Cutting GR, Uhl GR. Dopamine D2 receptor RFLPs, haplotypes and their association with substance use in black and Caucasian research volunteers. Hum. Hered. 1993;43:209–218. doi: 10.1159/000154133. [DOI] [PubMed] [Google Scholar]
- Patterson F, Schnoll RA, Wileyto EP, Pinto A, Epstein LH, Shields PG, Hawk LW, Tyndale RF, Benowitz N, Lerman C. Toward personalized therapy for smoking cessation: a randomized placebo-controlled trial of bupropion. Clin. Pharmacol. Ther. 2008;84:320–325. doi: 10.1038/clpt.2008.57. [DOI] [PubMed] [Google Scholar]
- Perkins KA, Lerman C, Coddington S, Jetton C, Karelitz JL, Wilson A, Jennings JR, Ferrell R, Bergen AW, Benowitz NL. Gene and gene by sex associations with initial sensitivity to nicotine in nonsmokers. Behav. Pharmacol. 2008b;19:630–640. doi: 10.1097/FBP.0b013e32830c3621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perkins KA, Lerman C, Grottenthaler A, Ciccocioppo MM, Milanak M, Conklin CA, Bergen AW, Benowitz NL. Dopamine and opioid gene variants are associated with increased smoking reward and reinforcement owing to negative mood. Behav. Pharmacol. 2008a;19:641–649. doi: 10.1097/FBP.0b013e32830c367c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perkins KA, Lerman C, Mercincavage M, Fonte CA, Briski JL. Nicotinic acetylcholine receptor beta2 subunit (CHRNB2) gene and short-term ability to quit smoking in response to nicotine patch. Cancer Epidemiol. Biomarkers Prev. 2009;18:2608–2612. doi: 10.1158/1055-9965.EPI-09-0166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pohjalainen T, Rinne JO, Nagren K, Lehikoinen P, Anttila K, Syvalahti EK. The A1 allele of the human D2 dopamine receptor gene predicts low D2 receptor availability in healthy volunteers. Mol. Psychiatry. 1998;3:256–260. doi: 10.1038/sj.mp.4000350. [DOI] [PubMed] [Google Scholar]
- Quaak M, van Schayck CP, Postma DS, Wagena EJ, van Schooten FJ. Genetic variants in the serotonin transporter influence the efficacy of bupropion and nortriptyline in smoking cessation. Addiction. 2012;107:178–187. doi: 10.1111/j.1360-0443.2011.03534.x. [DOI] [PubMed] [Google Scholar]
- Ray LA, Bujarski S, Chin PF, Miotto K. Pharmacogenetics of naltrexone in asian americans: a randomized placebo-controlled laboratory study. Neuropsychopharmacol. 2012;37:445–455. doi: 10.1038/npp.2011.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ray R, Jepson C, Wileyto EP, Dahl JP, Patterson F, Rukstalis M, Pinto A, Berrettini W, Lerman C. Genetic variation in mu-opioid-receptor-interacting proteins and smoking cessation in a nicotine replacement therapy trial. Nicotine Tob. Res. 2007;9:1237–1241. doi: 10.1080/14622200701648367. [DOI] [PubMed] [Google Scholar]
- Robinson JD, Lam CY, Minnix JA, Wetter DW, Tomlinson GE, Minna JD, Chen TT, Cinciripini PM. The DRD2 TaqI-B polymorphism and its relationship to smoking abstinence and withdrawal symptoms. Pharmacogenomics J. 2007;7:266–274. doi: 10.1038/sj.tpj.6500427. [DOI] [PubMed] [Google Scholar]
- Substance Abuse and Mental Health Services Administration. Results from the 2012 National Survey on Drug Use and Health: Summary of National Findings, NSDUH Series H-46, HHS Publication No. (SMA) 13-4795. Rockville, MD: 2013. [Google Scholar]
- Schank JR, Ventura R, Puglisi-Allegra S, Alcaro A, Cole CD, Liles LC, Seeman P, Weinshenker D. Dopamine beta-hydroxylase knockout mice have alterations in dopamine signaling and are hypersensitive to cocaine. Neuropsychopharmacol. 2006;31:2221–2230. doi: 10.1038/sj.npp.1301000. [DOI] [PubMed] [Google Scholar]
- Schellekens AF, Franke B, Ellenbroek B, Cools A, de Jong CA, Buitelaar JK, Verkes RJ. Reduced dopamine receptor sensitivity as an intermediate phenotype in alcohol dependence and the role of the COMT Val158Met and DRD2 Taq1A genotypes. Arch. Gen. Psychiatry. 2012;69:339–348. doi: 10.1001/archgenpsychiatry.2011.1335. [DOI] [PubMed] [Google Scholar]
- Schnoll RA, Patterson F, Wileyto EP, Tyndale RF, Benowitz N, Lerman C. Nicotine metabolic rate predicts successful smoking cessation with transdermal nicotine: a validation study. Pharmacol. Biochem. Behav. 2009;92:6–11. doi: 10.1016/j.pbb.2008.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scott JC, Woods SP, Matt GE, Meyer RA, Heaton RK, Atkinson JH, Grant I. Neurocognitive effects of methamphetamine: a critical review and meta-analysis. Neuropsychol. Rev. 2007;17:275–297. doi: 10.1007/s11065-007-9031-0. [DOI] [PubMed] [Google Scholar]
- Shao C, Li Y, Jiang K, Zhang D, Xu Y, Lin L, Wang Q, Zhao M, Jin L. Dopamine D4 receptor polymorphism modulates cue-elicited heroin craving in Chinese. Psychopharmacol. (Berl.) 2006;186:185–190. doi: 10.1007/s00213-006-0375-6. [DOI] [PubMed] [Google Scholar]
- Shiels MS, Huang HY, Hoffman SC, Shugart YY, Bolton JH, Platz EA, Helzlsouer KJ, Alberg AJ. A community-based study of cigarette smoking behavior in relation to variation in three genes involved in dopamine metabolism: catechol-O-methyltransferase (COMT), dopamine beta-hydroxylase (DβH) and monoamine oxidase-A (MAO-A) Prev. Med. 2008;47:116–122. doi: 10.1016/j.ypmed.2008.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shorter D, Nielsen DA, Huang W, Harding MJ, Hamon SC, Kosten TR. Pharmacogenetic randomized trial for cocaine abuse: disulfiram and α1A-adrenoceptor gene variation. Eur. Neuropsychopharmacol. 2013;23:1401–1407. doi: 10.1016/j.euroneuro.2013.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shrem MT, Halkitis PN. Methamphetamine abuse in the United States: contextual, psychological and sociological considerations. J. Health. Psychol. 2008;13:669–679. doi: 10.1177/1359105307082461. [DOI] [PubMed] [Google Scholar]
- Spellicy CJ, Kosten TR, Hamon SC, Harding MJ, Nielsen DA. ANKK1 and DRD2 pharmacogenetics of disulfiram treatment for cocaine abuse. Pharmacogenet. Genomics. 2013;23:333–340. doi: 10.1097/FPC.0b013e328361c39d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spitz MR, Shi H, Yang F, Hudmon KS, Jiang H, Chamberlain RM, Amos CI, Wan Y, Cinciripini P, Hong WK, Wu X. Case-control study of the D2 dopamine receptor gene and smoking status in lung cancer patients. J. Natl. Cancer. Inst. 1998;90:358–363. doi: 10.1093/jnci/90.5.358. [DOI] [PubMed] [Google Scholar]
- Stapleton JA, Sutherland G, O'Gara C, Spirling LI, Ball D. Association between DRD2/ANKK1 Taq1A genotypes, depression and smoking cessation with nicotine replacement therapy. Pharmacogenet. Genomics. 2011;21:447–453. doi: 10.1097/FPC.0b013e328347473a. [DOI] [PubMed] [Google Scholar]
- Sulzer D. How addictive drugs disrupt presynaptic dopamine neurotransmission. Neuron. 2011;69:628–649. doi: 10.1016/j.neuron.2011.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swan GE, Jack LM, Valdes AM, Ring HZ, Ton CC, Curry SJ, McAfee T. Joint effect of dopaminergic genes on likelihood of smoking following treatment with bupropion SR. Health Psychol. 2007;26:361–368. doi: 10.1037/0278-6133.26.3.361. [DOI] [PubMed] [Google Scholar]
- Tanda G, Pontieri FE, Di Chiara G. Cannabinoid and heroin activation of mesolimbic dopamine transmission by a common mu1 opioid receptor mechanism. Science. 1997;276:2048–2050. doi: 10.1126/science.276.5321.2048. [DOI] [PubMed] [Google Scholar]
- Ton TG, Rossing MA, Bowen DJ, Srinouanprachan S, Wicklund K, Farin FM. Genetic polymorphisms in dopamine-related genes and smoking cessation in women: a prospective cohort study. Behav. Brain Funct. 2007;3:22. doi: 10.1186/1744-9081-3-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trigo JM, Martin-García E, Berrendero F, Robledo P, Maldonado R. The endogenous opioid system: a common substrate in drug addiction. Drug Alcohol Depend. 2010;108:183–194. doi: 10.1016/j.drugalcdep.2009.10.011. [DOI] [PubMed] [Google Scholar]
- Tsuang MT, Bar JL, Harley RM, Lyons MJ. The Harvard Twin Study of Substance Abuse: what we have learned. Harv. Rev. Psychiatry. 2001;9:267–279. [PubMed] [Google Scholar]
- Tsuang MT, Lyons MJ, Eisen SA, Goldberg J, True W, Lin N, Meyer JM, Toomery R, Faraone SV, Eaves L. Genetic influences on DSM-III–R drug abuse and dependence: a study of 3,372 twin pairs. Am. J. Med. Genet. 1996;67:473–477. doi: 10.1002/(SICI)1096-8628(19960920)67:5<473::AID-AJMG6>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
- United Nations Office on Drugs and Crime. World Drug Report. United Nations publication, Sales No. E.12.XI.1. 2014 [Google Scholar]
- Uhl GR. Molecular genetics of substance abuse vulnerability: a current approach. Neuropsychopharmacol. 1999;20:3–9. doi: 10.1016/S0893-133X(98)00061-X. [DOI] [PubMed] [Google Scholar]
- Ujike H, Harano M, Inada T, Yamada M, Komiyama T, Sekine Y, Sora I, Iyo M, Katsu T, Nomura A, Nakata K, Ozaki N. Nine-or fewer repeat alleles in VNTR polymorphism of the dopamine transporter gene is a strong risk factor for prolonged methamphetamine psychosis. Pharmacogenomics J. 2003;3:242–247. doi: 10.1038/sj.tpj.6500189. [DOI] [PubMed] [Google Scholar]
- Ujike H, Katsu T, Okahisa Y, Takaki M, Kodama M, Inada T, Uchimura N, Yamada M, Iwata N, Sora I, Iyo M, Ozaki N, Kuroda S. Genetic variants of D2 but not D3 or D4 dopamine receptor gene are associated with rapid onset and poor prognosis of methamphetamine psychosis. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2009;33:625–629. doi: 10.1016/j.pnpbp.2009.02.019. [DOI] [PubMed] [Google Scholar]
- Vaccari A, Saba PL, Ruiu S, Collu M, Devoto P. Disulfiram and diethyldithiocarbamate intoxication affects the storage and release of striatal dopamine. Toxicol. Appl. Pharmacol. 1996;139:102–108. doi: 10.1006/taap.1996.0147. [DOI] [PubMed] [Google Scholar]
- Van Tol HH, Wu CM, Guan HC, Ohara K, Bunzow JR, Civelli O, Kennedy J, Seeman P, Niznik HB, Jovanovic V. Multiple dopamine D4 receptor variants in the human population. Nature. 1992;358:149–152. doi: 10.1038/358149a0. [DOI] [PubMed] [Google Scholar]
- van den Wildenberg E, Janssen RG, Hutchison KE, van Breukelen GJ, Wiers RW. Polymorphisms of the dopamine D4 receptor gene (DRD4 VNTR) and cannabinoid CB1 receptor gene (CNR1) are not strongly related to cue-reactivity after alcohol exposure. Addict. Biol. 2007;12:210–220. doi: 10.1111/j.1369-1600.2007.00064.x. [DOI] [PubMed] [Google Scholar]
- Veenstra-VanderWeele J, Qaadir A, Palmer AA, Cook EH, de Wit H. Association between the Casein Kinase 1 Epsilon gene region and subjective response to D-amphetamine. Neuropsychopharmacol. 2006;31:1056–1063. doi: 10.1038/sj.npp.1300936. [DOI] [PubMed] [Google Scholar]
- Vocci FJ, Ling WW. Medications development: successes and challenges. Pharmacol. Ther. 2005;108:94–108. doi: 10.1016/j.pharmthera.2005.06.010. [DOI] [PubMed] [Google Scholar]
- Wang Z, Ray R, Faith M, Tang K, Wileyto EP, Detre JA, Lerman C. Nicotine abstinence-induced cerebral blood flow changes by genotype. Neurosci. Lett. 2008;438:275–280. doi: 10.1016/j.neulet.2008.04.084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White JM, Irvine RJ. Mechanisms of fatal opioid overdose. Addiction. 1999;94:961–972. [PubMed] [Google Scholar]
- Wilcox CS, Noble EP, Oskooilar N. ANKK1/DRD2 locus variants are associated with rimonabant efficacy in aiding smoking cessation: pilot data. J. Investig. Med. 2011;59:1280–1283. doi: 10.2130/JIM.0b013e31823581fa. [DOI] [PubMed] [Google Scholar]
- Yu Q, Larson DF, Watson RR. Heart disease, methamphetamine and AIDS. Life Sci. 2003;73:129–140. doi: 10.1016/s0024-3205(03)00260-1. [DOI] [PubMed] [Google Scholar]
- Yudkin P, Munafò M, Hey K, Roberts S, Welch S, Johnstone E, Murphy M, Griffiths S, Walton R. Effectiveness of nicotine patches in relation to genotype in women versus men: randomised controlled trial. BMJ. 2004;328:989–990. doi: 10.1136/bmj.38050.674826.AE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zabetian CP, Anderson GM, Buxbaum SG, Elston RC, Ichinose H, Nagatsu T. A quantitative-trait analysis of human plasma-dopamine beta-hydroxylase activity: evidence for a major functional polymorphism at the DβH locus. Am. J. Hum. Genet. 2001;68:515–522. doi: 10.1086/318198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang D, Shao C, Shao M, Yan P, Wang Y, Liu Y, Liu W, Lin T, Xie Y, Zhao Y, Lu D, Li Y, Jin L. Effect of mu-opioid receptor gene polymorphisms on heroin-induced subjective responses in a Chinese population. Biol. Psychiatry. 2007 A;61:1244–1251. doi: 10.1016/j.biopsych.2006.07.012. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Bertolino A, Fazio L, Blasi G, Rampino A, Romano R, Lee ML, Xiao T, Papp A, Wang D, Sadee W. Polymorphisms in human dopamine D2 receptor gene affect gene expression, splicing, and neuronal activity during working memory. Proc. Natl. Acad. Sci. U.S.A. 2007 B;104:20552–20557. doi: 10.1073/pnas.0707106104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H, Kranzler HR, Yang BZ, Luo X, Gelernter J. The OPRD1 and OPRK1 loci in alcohol or drug dependence: OPRD1 variation modulates substance dependence risk. Mol. Psychiatry. 2008;13:531–543. doi: 10.1038/sj.mp.4002035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuo L, Kranzler HR, Luo X, Yang BZ, Weiss R, Brady K, Poling J, Farrer L, Gelernter J. Interaction between two independent CNR1 variants increases risk for cocaine dependence in European Americans: a replication study in family-based sample and population-based sample. Neuropsychopharmacol. 2009;34:1504–1513. doi: 10.1038/npp.2008.206. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.