Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Sep 1.
Published in final edited form as: Pharmacogenet Genomics. 2012 Sep;22(9):695–708. doi: 10.1097/FPC.0b013e3283540217

PharmGKB Summary - Very Important Pharmacogene Information for Cytochrome P-450, Family 2, Subfamily A, polypeptide 6 (CYP2A6)

Ellen M McDonagh a,*, Catherine Wassenaar b,*, Sean P David a,c, Rachel F Tyndale b, Russ B Altman a,d, Michelle Whirl-Carrillo a, Teri E Klein a
PMCID: PMC3413746  NIHMSID: NIHMS372509  PMID: 22547082

Introduction

Human cytochrome P450 (CYP-450), family 2, subfamily A, polypeptide 6 (CYP2A6) is a monooxygenase enzyme that metabolizes xenobiotic compounds and activates toxins [13]. In this review we focus on the effect of genetic variants on the role of CYP2A6 in drug metabolism, and provide a pharmacogenomic overview of CYP2A6 in humans. This Very Important Pharmacogene summary is available with interactive links to gene variants, haplotypes and drugs on the PharmGKB website http://pharmgkb.org/vip/PA121 [4].

CYP2A6 expression and function

CYP2A6 represents approximately 4% of the total CYP-450 enzyme protein content of adult liver microsomes [5]. CYP2A6 is also expressed in the lung, trachea, nasal mucosa, and sex organs such as breast [2, 6]. Human CYP2A6 enzyme activity is determined by measuring coumarin 7-hydroxylation [710]. CYP2A6 expression, enzyme function and induction vary considerably between individuals [2, 5, 712], and an individual’s CYP2A6 enzyme expression and activity may depend on a combination of environmental factors (such as xenobiotic compounds) and genetic factors (including polymorphic variants) [1, 13]. Modeling human CYP2A6 activity in rodents has limited utility due to species-specific CYP2A6 ortholog expression patterns and activity profiles – for example, in rats little or no coumarin is 7-hydroxylated, and a CYP2B enzyme metabolizes nicotine to cotinine [1419]. The crystal structure of human CYP2A6 bound to coumarin was originally described in 2005, revealing a smaller active site than other CYP2 enzymes C8 and C9, composed of a hydrophobic cavity ideal for the oxidation of small planar compounds such as coumarin [20]. Several structures with the enzyme in complex with different substrates and/or mechanism based inhibitors have been described since (the Protein Data Bank [21]; [1]).

The CYP2A6 gene

CYP2A6 was the first gene of the human CYP2A cytochrome subfamily to be cloned and mapped, and was previously known as CYP2A3 (a name now used for the rat ortholog) [3, 2226]. The CYP2A6 gene sits within a cluster of CYP2 genes on chromosome 19 q13.2, thought to have arisen through duplication events, and shares extensive homology with subfamily members CYP2A7 and CYP2A13 [13]. The gene is composed of 9 exons spanning around 6kb, encoding a protein of 494 amino acids [1, 7].

Genetic variation of CYP2A6

The CYP2A6 gene is highly polymorphic, with over 35 different CYP2A6 alleles described thus far, with additional subgroups (see [27, 28]) [3]. Star (*) nomenclature is used to describe different CYP2A6 alleles, with the ‘wildtype’ reference allele defined as *1 [27, 28] [8]. Genetic variants in the CYP2A6 locus include alleles with single nucleotide polymorphisms (SNPs) (for example *2, *9), whole and partial gene deletions (alleles *4A-H), a gene hybrid with CYP2A7 (for example *12) and gene conversions (for example *1B) [28]. The complex genetic architecture of CYP2A6 coupled with its significant homology with other CYP2A genes makes genotyping challenging, particularly when using SNP based arrays [3, 29]. Since single amplifications often cannot discriminate between the different CYP2A genes, special amplification and sequencing techniques have been developed for CYP2A6 genotyping, revealing errors in allele characterization in older studies [3, 2932].

CYP2A6 genotypes are often classified into predicted phenotype groups, describing the effect on enzyme activity, for example ‘poor’ metabolizer (no active CYP2A6 alleles, homozygous for inactive alleles), ‘slow’ (one inactive or two decreased activity alleles), ‘intermediate’ metabolizer (heterozygous with one decreased activity allele and one active allele), ‘normal’/ ‘extensive’ (2 active alleles), or ‘ultrarapid’/ ‘fast’ (>2 active alleles) [29, 3336]. Studies may also compare reduced (one or more inactive or decreased activity allele) to normal (two active alleles) metabolizers [37, 38]. These categories often overlap or differ between studies. In addition, the same polymorphism can have a different effect on the metabolism of different substrates. For example, CYP2A6*18 activity against nicotine is similar to wildtype enzyme, but is reduced for coumarin and tegafur metabolism, in vitro [39], and conversely *17 activity against coumarin is similar to wildtype but significantly reduced for nicotine metabolism [40]. People with CYP2A6*12 are slow metabolizers of letrozole, but intermediate metabolizers of nicotine [41]. Therefore the effect of CYP2A6 polymorphisms on metabolism should be investigated and interpreted in the context of individual substrates [29].

Genetic variants in the CYP2A6 gene can result in reduced expression by affecting transcriptional or translational processes [1]. For example, a SNP within the TATA box of the CYP2A6 promoter (allele *9) reduces gene transcription [42]. The CYP2A6*1B allele differs from *1A by a CYP2A7 gene conversion in the 3’ untranslated region (UTR), and correlates with increased CYP2A6 protein expression and activity, likely through increased mRNA stability [43]. Individuals homozygous for the *4 gene deletion allele lack detectable CYP2A6 mRNA expression and enzyme activity [44, 45].

Wide variation in the frequency of CYP2A6 alleles across ancestral groups is observed. For example, the frequency of CYP2A6*4 alleles ranges from 0–4% in White, 0–2% in Black, 5–15% in Chinese and 17–24% in Japanese populations [30, 36, 4554]. The frequency of the CYP2A6*9 allele ranges from around 5–8% in White, 6–9% in Black, up to 16% in Chinese and 19–21% in Japanese populations [36, 42, 4651, 54, 55]. Other alleles are found predominantly in one ethnic group, for example CYP2A6*17 (defined by the variant rs28399454, V365M) is found at a frequency of around 10% in Black individuals, but not identified in White, Korean or Japanese individuals, whereas *7 is found in Asian subjects at a frequency of around 10%, but not White or Black individuals [49, 56].

CYP2A6 as an important pharmacogene

Around 3% of the drugs metabolized by CYP-450 enzymes involve CYP2A6 (reviewed in [1]). In the sections below, we describe known pharmacogenetic associations between CYP2A6 variants and drugs, further detailed in Table 1 and 2. When examining these associations, it should be taken into account that environmental factors, such as compounds found in food, cigarettes, hormones or therapeutic drugs, can affect CYP2A6 expression, which can therefore influence drug pharmacokinetics and responses [1, 13]. Xenobiotics in our diet, such as the flavonoid biochanin A (found in plants), can upregulate CYP2A6 expression in vitro [57]. The hormone estrodiol induces CYP2A6 expression via direct binding of the transcription factor estrogen receptor 1 (ESR1) to a promoter element upstream of the CYP2A6 gene in vitro [58], and may explain why CYP2A6 activity is higher in women than men, and higher in women taking oral contraceptives compared to those not taking them [59]. The anti-inflammatory drug dexamethasone, via the glucorticoid receptor (NR3C1), induces CYP2A6 transcription in human hepatocytes in vitroby augmentation of Hepatocyte Nuclear Factor 4, alpha (HNF4A) [60], likely explaining the enhanced CYP2A6 activity seen with dexamethasone treatment [9]. The anticonvulsant phenobarbital also enhances CYP2A6 enzyme activity in vitro [9, 10]. Inhibitors of CYP2A6 enzyme activity include the antibacterial and antifungal agents isoniazid and ketoconazole [1, 13, 61], and traditional Chinese medicine [62]. Identifying environmental and therapeutic compounds that regulate CYP2A6 activity, as well as genetic polymorphisms, may be important for optimal therapeutic efficacy and avoiding adverse drug reactions [1].

Table 1.

CYP2A6 polymorphisms and nicotine associations

CYP2A6 allele Effect on nicotine metabolism Association with smoking behavior and response to
nicotine replacement therapy
CYP2A6*1B Greater nicotine clearance in individuals
with alleles *1B1-15 compared to wildtype
homozygotes *1A/*1A [158].
Genotype *1B/*1B genotype is associated with
increased cpd compared to *1A/*1A, but not
significantly associated with smoking status or ability
to quit [82]. Increased likelihood of being a smoker
[81].
CYP2A6*1X2 1X2A: Higher levels of exhaled carbon
monoxide compared to *1/*2, *2/*2, *1/*4,
and higher cotinine plasma levels compared
to *1/*1 or *1/*2, *2/*2, *1/*4 [31] #.
1X2B: increased nicotine metabolism
compared to *1/*1 (ns), measured by
cotinine/nicotine ratios [159].
1X2A: Greater smoking intensity (CO per cpd, COT
per cpd) compared to *1/*1 or *1/*2, *2/*2, *1/*4
[31] #.
CYP2A6*2 Longer half-life of nicotine and cotinine,
and reduced nicotine metabolism [49],
[34]#.
In adolescents, increased risk of becoming nicotine
dependent, but slower progression and lower cigarette
consumption once dependent [74], [47]#. Fewer
compared to normal nicotine metabolizers [31, 36, 74]
#, [160]. Fewer cpd and lower FTND [38]#. Increased
ability to quit smoking (ns) [161]#. Better response to
extended transdermal nicotine replacement therapy
[37]#.
CYP2A6*4 Reduced nicotine metabolism and altered
metabolite profile [49, 50,162164].
In adolescents, increased risk of becoming nicotine
dependent, but slower progression and lower cigarette
consumption once dependent [74], [47]#. Reduced risk
of being a smoker [81], reduced cpd [31, 36, 54, 74,
152]#, [165, 166], and increased ability to quit smoking
(ns) [161]#. Though in other studies, not significantly
associated with smoker status, age started smoking, cpd
or ability to quit [82, 167169]. Fewer cpd and lower
FTND [38]#. Better response to extended transdermal
nicotine replacement therapy [37]#.
CYP2A6*7 Reduced nicotine metabolism [39, 45, 49,
50, 164]. SNP T1412C (rs5031016):
Reduced nicotine metabolism [170].
Fewer cpd, later onset of smoking, shorter smoking
duration, but reduced likelihood of smoking cessation
[54, 152]#.
CYP2A6*9 Reduced nicotine metabolism and clearance
[44, 49], [34]#.
Fewer cpd [36, 38, 54]#, lower FTND [38]#, later onset
of smoking, shorter smoking duration, but reduced
likelihood of smoking cessation [54]#. Better response
to extended transdermal nicotine replacement therapy
[37]#.
CYP2A6*10 Reduced nicotine metabolism [45, 49, 164]. Reduced cpd [54, 152]#.
CYP2A6*12 *1/*12 genotype is associated with normal
nicotine metabolism, but *9/*12 is
associated with reduced metabolism [34]#.
Fewer cpd [36, 38]#, lower FTND [38]#. Better
response to extended transdermal nicotine replacement
therapy [37]#.
CYP2A6*17 Reduced nicotine metabolism and clearance
[49, 56].
Increased ability to quit smoking (ns) [161]#.
CYP2A6*35 Reduced nicotine metabolism [171]. Increased ability to quit smoking (ns) [161]#.

Table Key:

#

= studies that analyze combined genotypes, or analyze this allele combined with other alleles in a phenotype category, e.g. reduced activity alleles.

CO = carbon monoxide

COT = cotinine

cpd = Cigarettes per day

FTND: Fagerström Test for Nicotine Dependence

ns = not significant

Table 2.

CYP2A6 polymorphisms and association with therapeutic drug response

CYP2A6 Allele or
Genotype
Details of genotyping Drug Association Reference and study
details
*1/*4 or

*4/*4 or

*4/*9 or

*1/*9
§ Caffeine Significantly reduced
metabolism of paraxanthine
compared to *1/*1 (not
detectable in *4/*4
samples)
[86] in vitro study, n=42
human liver microsomes
*7 or

*10 or

*11
§ Caffeine Reduced activity against
paraxanthine
[86] in vitro study,
CYP2A6 transfected into
E. coli, fractions then
incubated with
paraxanthine
Intermediate
metabolizers:
*1A/*4, *1A/*9,
*1B1/*4, *1B1/*9
Poor metabolizers:
*4/*9, *9/*9
*4: gene deletion
*9: g.-48T>G
(rs28399433)
*1x2 gene duplication
*1B1 gene conversion in
3’ region
Caffeine Reduced metabolism of
paraxanthine into 17U
compared to extensive
metabolizers (*1A/*1A,
*1A/*1B1, *1A1/*1B1x2,
*1B1/*1B1).
[88] n=100 Serbian,
healthy volunteers.
rs8192725
Genotype CC
g.1620T>C
Intron 2
Method: direct gene
sequencing, provide
details of primers and §.
Reference sequences
used: NG_00008.7 and
NP_000753.3
Tegafur Significantly increased
mRNA expression and a
trend for increased rate of 5
FU formation (ns),
compared to genotype TT
[55]
In vitro, (n=45) Chinese
HLM and liver tissue
samples
rs8192720
Genotype C/T or
TT
c.22C>T
(NM_000762.5)
Leu8Leu
exon 1
Method: direct gene
sequencing, provide
details of primers and §.
Reference sequences
used: NG_00008.7 and
NP_000753.3
Tegafur Increased CYP2A6 mRNA
expression and an increased
rate of 5 FU formation,
compared to genotype CC.
[55]
In vitro, (n=45) Chinese
HLM and liver tissue
samples
rs28399433
Genotype G/T or
GG
g. -48T>G
Method: direct gene
sequencing, provide
details of primers and §.
Reference sequences
used: NG_00008.7 and
NP_000753.3
Tegafur A trend for reduced
CYP2A6 mRNA and
protein expression, and
reduced rate of 5 FU
formation (ns), compared to
genotype TT.
[55]
In vitro, (n=45) Chinese
HLM and liver tissue
samples
*4 Allele defined by:
CYP2A6 gene deletion.
Method: direct gene
sequencing, provide
details of primers and §.
Reference sequences
used: NG_00008.7 and
NP_000753.3
Tegafur Significantly reduced
CYP2A6 mRNA and
protein expression, and
significantly reduced rate
of 5 FU formation,
compared to those without
the *4 allele.
[55]
In vitro, (n=45) Chinese
HLM and liver tissue
samples
Two alleles with a
gene conversion in
the 3’ UTR
Method: direct gene
sequencing, provide
details of primers and §.
Reference sequences
used: NG_00008.7 and
NP_000753.3
Tegafur Associated with a trend for
reduced CYP2A6
expression and a trend for
reduced rate of 5 FU
formation, compared to
those with no alleles with a
gene conversion in the 3’
UTR.
[55]
In vitro, (n=45) Chinese
HLM and liver tissue
samples
*1B (haplotype 14) Haplotype defined by:
gene conversion in the
CYP2A6 3’ UTR, and the
SNPs 22C>T (rs8192720)
and 1620T>C
(rs8192725)
Method: direct gene
sequencing, provide
details of primers and §.
Reference sequences
used: NG_00008.7 and
NP_000753.3
Tegafur significantly increased
CYP2A6 mRNA
expression, and
significantly increased rates
of 5 FU formation,
compared to those without
the allele.
[55]
In vitro, (n=45) Chinese
HLM and liver tissue
samples
Genotype *1/*1 Sequenced for *9 at
-48T>G (rs28399433),
*10 at 6600G>T)
rs28399468), *7 at
6558T>C (rs5031016).
*4 gene deletion §.
Therefore *1 was defined
as wildtype with none of
the above variants.
S-1 and
oxaliplatin
Associated with increased
tegafur metabolism and a
trend for higher 5 FU
plasma concentrations
compared to individuals
with one or two variant
alleles (*4, *7, *9, *10
combined) (p values were
not given), but not
associated with increased
likelihood of diarrhea or
neutropenia.
[98]
Study: Biliary tract
cancer patients, (n=48).
*4C and *11
combined
Alleles defined by: *4C:
Gene deletion (identical
to *4A according to [27,
28]). *11: c.670T>C,
g.3391T>C, Ser244Pro
(rs111033610)
Method: §, amplification
of exon 8 to 3’ UTR,
restriction digest analysis.
Amplification and
sequencing of exon 5.
Tegafur Reduced metabolism
compared to four other
patients.
[95]
A case study of a gastric
cancer patient
subsequently found to
have both alleles, and
follow-up in vitro
experiments.
Allele *7 g.6558T>C (Ile471Thr)
(rs5031016)
and gene conversion with
CYP2A7 in the 3’ UTR
§
Tegafur Reduced 5 FU formation
rate compared to *1A/*1A
[39]
In vitro kinetic assays,
using transformed E.
coli membrane
preparations (n=3)
Allele *18 Single SNP
rs1809810 (g. 5668A>T,
Tyr392Phe). Allele
specific primers used for
genotyping.
Tegafur Slightly reduced 5 FU
formation rate compared to
*1A/*1A
[39]
In vitro kinetic assays,
using transformed E.
coli membrane
preparations (n=3)
Allele *19 g.5668A>T (Tyr392Phe)
(rs1809810) and
g.6354T>C (intron 8),
6558T>C (Ile471Thr)
(rs5031016)
and gene conversion with
CYP2A7 in the 3’ UTR.
Allele specific primers
used for genotyping.
Tegafur Reduced 5 FU formation
rate compared to *1A/*1A
[39] In vitro kinetic
assays, using
transformed E. coli
membrane preparations
(n=3)
Individuals with
two variant alleles
(*4/*4, *7/*7 or
*4/*7)
*4A: §
*7: § with some
modifications,
c.1412T>C, Ile471Thr
(rs5031016)
S-1 Significantly reduced oral
clearance of tegafur,
compared to *1/*1
[100]
n=54 Japanese patients
*4C Alleles defined by: *4C,
*7, *9. Method: §
amplification
S-1 Significantly reduced
metabolism of tegafur,
compared to individuals
without the allele.
[99]
n=46 Japanese patients
with non-small-cell lung
cancer.
Individuals with
two variant alleles,
(combined
genotypes):
*4/*4
*4/*7
*4/*9
*7/*9
*9/*9
Alleles defined by: *4:
entire gene deletion §,
Positions genotyped: g.-
48T>G (for *9)
(rs28399433), g.
6558T>C (rs5031016
Ile471Thr) (for *7, *10),
and g.6600G>T
(rs28399468,
Arg485Leu) (for *10)
S-1 Reduced treatment efficacy
compared to individuals
with one or two wildtype
*1 alleles. Increased risk of
disease progression and
reduced progression-free
survival, as measured by
significantly reduced
probability of tumor
response,
[102]
n= 50 Korean patients
with metastatic gastric
cancer
Two variant
alleles, (combined
genotypes):
*4A/*4A
*4A/*7
*4A/*9
*7/*7
*7/*9
*9/*9
§
(introduction describes *7
as c.1412T>C (rs5031016
Ile471Thr), and *9 as g.-
48T>G (rs28399433),
and *4 as complete lack
of activity.)
S-1 Associated with
significantly reduced
tegafur clearance,
compared to *1/*1 or
heterozygotes with one
variant allele (tegafur
plasma concentrations did
not correlate with 5 FU
concentrations)
[97]
n=57 Japanese patients
with solid tumors.
Individuals with
two variant alleles,
(combined
genotypes):
*4A/*4A
*4A/*9
*4A/*10
*7/*9
*9/*9
*9/*10
Alleles defined by:
*4 gene deletion
*7: c.1412T>C ,
Ile471Thr (rs5031016)
*9: g.-48T>G
(rs28399433)
S-1 Associated with higher
tegafur and significantly
lower 5 FU plasma
concentrations compared to
heterozygote patients
(*1/*4A
*1/*7
*1/*9
*1/*11 combined
genotypes).
No significant association
with adverse effects or
treatment response were
found.
[103]
n=34 patients with solid
tumors.
Individuals with
one or two variant
alleles, (combined
genotypes):
*4/*7
*4/*9
*4/*10
*9/*9
*1/*4
Alleles defined by:
*4 entire gene deletion,
positions genotyped: g.-
48T>G (rs28399433) (for
*9), g.6558T>C
(rs5031016 Ile471Thr)
(for *7, *10), and
g.6600G>T (rs28399468,
Arg485Leu) (for *10).
§
S-1 and
cisplatin
Significantly associated
with lower response rate,
increased risk of disease
progression and reduced
overall survival time.
[101]
n=106, Korean patients,
with metastatic gastric
cancer
*1/*4 or

*4/*4 or

*4/*9 or

*1/*7
§ Letrozole Significantly associated
with reduced metabolism
compared with *1/*1
wildtype
[106] In vitro study
using Japanese HLM
samples (n=31)
*1A/*1A
*1A/*1B
*1B/*1B
§ Letrozole Increased clearance
compared to heterozyogous
or homozygous individuals
with variant alleles (*4, *9,
*7)
[105] n=22 healthy
Japanese
postmenopausal women
Slow metabolizers
=2 copies of
decrease-of-
function alleles (*9,
*12), or 1 or 2
copies of loss-of-
function alleles (*2,
*4, *7, *10, *17,
*20, *23-*27, *35),
or 1 decrease-of-
function allele and 1
loss-of-function
allele.
§ *2: 1799 T>A; *4E:
gene deletion (intron 7);
*7: 6558 T>C; *9: -48
T>G; *10: 6558 T>C,
6600 G>T; *12: exon 1–2
CYP2A7, exon 3-9
CYP2A6; *17: 5065
G>A; *23: 2161 C>T;
*24: 594 G>C; (if *25 is
positive then perform *26
& *27 assays) *26: 1711
T>G; *27: 2162–2163
GC>A frameshift; *35:
6458 A>T.
Letrozole Significantly higher plasma
levels of drug compared to
normal metabolizers
(genotype *1/*1)
[41]
n=259 mixed
population,
postmenopausal women
with hormone receptor
positive breast cancer
Intermediate
metabolizer = one
copy of decrease-of-
function alleles *9,
*12
§ *2: 1799 T>A; *4E:
gene deletion (intron 7);
*7: 6558 T>C; *9: -48
T>G; *10: 6558 T>C,
6600 G>T; *12: exon 1–2
CYP2A7, exon 3–9
CYP2A6; *17: 5065
G>A; *23: 2161 C>T;
*24: 594 G>C; (if *25 is
positive then perform *26
& *27 assays) *26: 1711
T>G; *27: 2162–2163
GC>A frameshift; *35:
6458 A>T.
Letrozole Significantly higher plasma
levels of drug compared to
normal metabolizers
(genotype *1/*1)
[41]
n=259 mixed
population,
postmenopausal women
with hormone receptor
positive breast cancer
Two loss-of-
function alleles
(*2, *4A-F, *5,
*34) or two
reduced function
alleles (*1H, *1J,
*7, *9, *10, *12,
*13, *15, *17, *19)
or one of each
: for novel variants.
*1X2A, *1X2B: gene
duplication, *1H and *1J:
rs61663607 g.-745A>G,
*2: rs1801272
g.1799T>A
*4A – *4F intron 8 gene
conversion with CYP2A7
- gene deletion. *5:
rs5031017 g.6582G>T,
*7, *10, *19: rs5031016
g.6558T>C, *9, *13, *15:
rs28399433 g.-48T>G,
*17: rs28399454
g.5065G>A, *12: intron 2
gene conversion with
CYP2A7, *34: intron 4,
gene conversion with
CYP2A7.
Efavirenz Reduced EFV metabolism
– significantly higher EFV
plasma AUC compared to
those without variant
alleles (in individuals with
CYP2B6 reference alleles –
please note reference
alleles were not stated in
the study).
[118]
n=169 mixed
population, HIV
-infected individuals, The
Swiss HIV Cohort
Study.
(Please note, the
phenotype categories
included different allele
groupings from those
described in [121]).
loss-of-function
alleles (*2, *4) and/
or diminished
function alleles
(*1H,*1J, *5, *7,
*9,*10,*12,*13,
*15, *17, *19, *34)
Efavirenz Reduced clearance of the
drug irrespective of
CYP2B6 status, however
more pronounced in
homozygotes with CYP2B6
loss-of-function alleles.
[121] n=169 mixed
population, HIV-
infected individuals, The
Swiss HIV Cohort
Study.
(Please note, the
phenotype categories
included different allele
groupings from those
described in [118]).
*9B allele rs8192726 g.1836G>T
(genotyped only
rs8192726 and no other
SNPs which make up the
*9B allele) §
Efavirenz Slow metabolism of EFV
(significantly increased
plasma concentrations)
compared to those without
the allele. CYP2B6
genotype status was not
accounted for in the initial
analysis.
[123]
n=94 Ghanaian patients
with HIV-infection,
some also with TB
coinfection. Please note;
this was the same study
cohort as [122] but more
patients
*17 rs28399454 genotype
AA+GA
g. 5065G>A, Val365Met
(genotyped only
rs28399454 and no other
SNPs which make up the
*17 allele)
Efavirenz Not associated with slow
metabolism of EFV (not
statistically significant
higher plasma
concentrations) compared
to those without the allele.
CYP2B6 genotype status
was not accounted for in
the initial analysis.
[123]
n=94 Ghanaian patients
with HIV-infection,
some also with TB
coinfection. Please note;
this was the same study
cohort as [122] but more
patients
*9B and/ or *17 rs8192726 or rs28399454
(genotyped only
rs8192726 and
rs28399454, and no other
SNPs which make up the
*9B and *17 alleles)
Efavirenz Slow metabolism of EFV
(significantly increased
plasma concentrations)
compared to individuals
without the alleles.
CYP2B6 genotype status
was not accounted for in
the initial analysis but in
multiple regression
analysis, CYP2A6 genotype
status independently
contributed to EFV
variation, along with
CYP2B6 and UGT2B7
genotype.
[123]
n=94 Ghanaian patients
with HIV-infection,
some also with TB
coinfection. Please note;
this was the same study
cohort as [122] but more
patients
*9B
genotype TG vs GG
rs8192726 g.1836G>T
(genotyped only this
position and no other
SNPs which make up the
*9B allele)
Efavirenz Not associated with EFV
plasma or cell
concentrations (CYP2B6
status not considered in this
initial analysis)
[119]
n=50 a mixed
population of patients
with HIV-infection.
*17
Genotype GA vs
GG
rs28399454 g.5065 G>A,
Val365 Met
(genotyped only this
position and no other
SNPs which make up the
*17 allele)
Efavirenz Not associated with EFV
plasma concentration,
peripheral blood
mononuclear cell
concentrations or
accumulation ratio.
(CYP2B6 status was not
considered in this initial
analysis)
[119] n=50 a mixed
population of patients
with HIV-infection.
*9 rs28399433
Genotype T/G
Efavirenz A trend for higher EFV
plasma concentrations
compared to genotype T/T
(not considering CYP2B6
status), although this was
not statistically significant
after Bonferroni correction
for multiple comparisons.
[115]
n=45, Haitians of
African decent, with
HIV-infection.
*4 § Valproic acid Significantly higher steady
state plasma concentrations
(reduced metabolism) in
individuals with the *4
allele compared to those
without the allele.
[137]
n=179 Northern Han
Chinese epilepsy
patients
Genotype *7/*9 or

*4A/*7 or

*4A/*9 or

*4A/*10
§
*4A (entire gene deletion)
*7: Ile471Thr
(rs5031016)
*8: Arg485Leu
(rs28399468)
*9: g.-48T>G
(rs28399433)
*10: Ile471Thr
(rs5031016) and
Arg485Leu (rs28399468)
Pilocarpine Poor metabolism and low
clearance.
[139] study 1 n=20,
study 2 n=8, healthy
Japanese individuals
administered with a
single dose of
pilocarpine
hydrochloride.

Table Key:

5 FU = Fluorouracil

AUC = Area Under the Curve

EFV = efavirenz

HLM = Human Liver Microsomes

ns = not statistically significant

SNP = Single Nucleotide Polymorphism

§

= give reference to other studies for the method of genotyping.

= refer to genotyping alleles in concordance with the CYP Allele Nomenclature Committee (www.cypalleles.ki.se) [27, 28].

g

= gene nucleotide position according to NG_000008.7 (unless otherwise stated) as given by the CYP Allele Nomenclature Committee [27, 28] Please note that this NCBI reference sequence has been removed.

c

= cDNA nucleotide position according to NM_000762.4 (unless otherwise stated) as given by the CYP Allele Nomenclature Committee [27, 28]. Please note that this NCBI reference sequence has been updated.

Where possible, dbSNP rsID have been provided for variants, according to links from [27, 28] or from the cited journal. Please note, due to the reference sequences on the NCBI having been updated from those given by the CYP Allele Nomenclature Committee (www.cypalleles.ki.se) [27, 28], the gene and cDNA nucleotide positions on dbSNP may differ, despite being consistent in amino acid position.

More details on these associations can be found at www.pharmgkb.org. Please note that the alleles reported throughout this summary are those on the minus chromosomal strand, where the CYP2A6 gene is found. On the PharmGKB all alleles are standardized to the plus strand, and therefore will differ in a complementary manner from those given here.

Another factor is CYP2A6 genotyping. Most drug-response studies group CYP2A6 genotypes into predicted enzyme activity phenotype groups and assess associations compared to *1 homozygotes, because there are many variant alleles found at low frequencies. Some studies use the * allele name without screening for all variants in the allele as defined by the CYP-450 allele nomenclature committee [27, 28]. Therefore individuals may have variants not screened for, or not have all variants conferring an allele. As described previously, the complexity of CYP2A6 in terms of polymorphisms and homology to other genes means that genotyping errors can occur; as knowledge about the gene and its variants increases, genotyping assays should improve, as was seen with CYP2D6. We therefore provide genotyping details in Table 2.

CYP2A6 polymorphisms, nicotine metabolism and cigarette smoking behavior

The vast majority of published work describing the phenotypic effects of CYP2A6 polymorphisms on enzyme activity has been carried out using nicotine as a substrate (Table 1). These studies have revealed important mechanistic consequences of CYP2A6 alleles on nicotine metabolic inactivation and related smoking behaviors, and by characterizing the relationship between CYP2A6 genotypes and enzyme phenotypes provide a starting point for how these polymorphisms may contribute to the observed inter-individual variability in the PK of other CYP2A6 substrates.

Nicotine is extensively metabolized and has a short plasma half-life of around 2 hours [63]. Approximately 80% of nicotine is inactivated in vivo into cotinine in a two-step process (see the PharmGKB Nicotine Pathway, Pharmacokinetics https://www.pharmgkb.org/pathway/PA2011) [6365]. CYP2A6 has a predominant role in the oxidation of nicotine to form a nicotine iminium ion, which is subsequently converted to cotinine (COT) by aldehyde oxidase (AOX) [63, 6567]. The majority of COT is metabolized to trans-3’-hydroxy-cotinine (3HC) in a reaction exclusively mediated by CYP2A6 [6769]. The ratio of 3HC/COT is often used as a phenotypic marker of CYP2A6 metabolic activity among smokers due to the long half-life of COT and the in vivo formation dependent kinetics of 3HC [63, 65, 70].

Nicotine metabolism is subject to large inter-individual variation [67, 71], and seven CYP2A6 polymorphisms explain most of the inter-individual variation in nicotine to COT metabolism, in European-Americans [72]. Nicotine dependence and cigarette smoking behaviors are closely related to the pharmacokinetics of nicotine, for example cigarette craving negatively correlates with blood levels of nicotine [73]. Polymorphisms in CYP2A6 which effect nicotine metabolism have therefore been associated with smoking behaviors (see Table 1) [31, 53, 74], and are an important consideration in the efficacy of nicotine replacement based smoking cessation treatments [7577]. For example, slow metabolizers (as determined by CYP2A6*2, *4, *9 and *12 alleles), are less likely to be smokers, smoke fewer cigarettes per day, take smaller puff volumes, have lower levels of dependence, are more able to quit, and benefit more from regular and extended nicotine patch replacement therapy compared to normal metabolizers [31, 36, 37, 46, 7881].

Evidence suggests that CYP2A6 polymorphisms that confer decreased CYP2A6 enzyme activity result in reduced or deficient nicotine metabolism [31, 56, 71], and this is thought to lead to lower cigarette consumption [47]. On the other hand, alleles conferring increased enzyme activity (e.g. duplication or *1B) result in enhanced rates of nicotine metabolism and thus are associated with increased cigarette consumption and depth of inhalation [29, 31, 47, 71, 82]. Inhibition of CYP2A6 has therefore been investigated as a smoking cessation treatment [29]. In Chronic Obstructive Pulmonary Disease (COPD) patients, CYP2A6 is associated with number of cigarettes smoked per day (cpd) and age of initiation of smoking [83]. As smoking cessation is important in preventing COPD progression, identifying patients with risk genotypes for particular smoking behaviors may aid in treatments to help patients reduce smoking [83].

The CYP2A6 and CYP2B6 genes are closely localized within the CYP2 cluster on chromosome 19, suggesting potential linkage disequilibrium [3, 84]. Human liver CYP2A6 and CYP2B6 enzyme expression is correlated and they share some inducers and substrates (as discussed in [85]). Examining the contribution of CYP2B6 genotype on a potentially common substrate (i.e. nicotine, efavirenz) should take into consideration CYP2A6 status, and vice-versa. For example, the association seen between CYP2B6 and nicotine C-oxidation in vitro is abrogated after controlling for CYP2A6 protein levels [85].

CYP2A6 and caffeine

CYP2A6 plays a part in caffeine metabolism, as the major enzyme required to convert paraxanthine (1,7-dimethylxanthine, 17X) into 1,7-dimethyluric acid (17U) via 8-hydroxylation [86] (see the PharmGKB Caffeine Pathway, Pharmacokinetics https://www.pharmgkb.org/pathway/PA165884757) [87]. Human liver microsomes (HLMs) with the CYP2A6 genotype *1/*4, *4/*9 or *1/*9 display significantly reduced 8-hydroxylase enzyme activity against paraxanthine, and *4/*4 samples have undetectable activity, compared to *1/*1 wildtype samples [86]. Kinetic assays with CYP2A6 protein fractions demonstrate *7, *10 and *11 alleles confer reduced 8-hydroxylation activity [86]. Amongst non-smokers, CYP2A6 intermediate and poor metabolizer genotypes have lower paraxanthine metabolism compared to ‘normal metabolizers’ (Table 2) [88]. In addition to CYP2A6 genotype, cigarette smoking significantly reduces paraxanthine metabolism (17U/ 17X ratio in urine), and may be due to competition between paraxanthine and nicotine [88], or via the impact of current smoking status which decreases CYP2A6 activity [89]. Therefore, both smoking and CYP2A6 genotype influences CYP2A6 8-hydroxylation activity against caffeine, contributing to the inter-individual variability observed [88]. By measuring 17U/17X ratios, caffeine has been proposed as a potentially more suitable substrate for studying the functional effects of CYP2A6 polymorphisms in vivo than nicotine or coumarin [86], although further characterization of the timing, dose and association with genotype is required.

CYP2A6 and cancer therapeutics

CYP2A6 has a key role in the metabolism of several drugs involved in cancer treatment. Associations between CYP2A6 genotype, rate of drug metabolism and treatment efficacy are detailed below and in Table 2. CYP2A6 genotype is also associated with risk of developing cancer [90], see the ‘CYP2A6 and toxicology’ section.

Tegafur

The prodrug tegafur is initially metabolized into 5’ hydroxytegafur, which rapidly breaks down into 5-fluorouracil (5 FU) and is further processed into active metabolites with anticancer properties (see the PharmGKB Fluoropyrimidine Pathway, Pharmacokinetics https://www.pharmgkb.org/pathway/PA150653776 and the Fluoropyrimidine (PD) Pathway https://www.pharmgkb.org/pathway/PA165291507) [91]. Although several CYP-450 proteins have a role in the biotransformation of tegafur into 5 FU, including CYP1A2, CYP2C8, CYP2C9 and CYP1A1, CYP2A6 has a principle role [92, 93]. In human liver microsomes, 5 FU formation correlates significantly with coumarin 7-hydroxylation and with CYP2A6 expression [92, 93]. Selective inhibition of CYP2A6 activity drastically attenuates 5 FU formation [9294]. Genetic variants that affect CYP2A6 expression and function are associated with altered metabolism of tegafur and clinical outcome. The CYP2A6*4 gene deletion allele significantly reduces CYP2A6 mRNA and protein levels in human liver samples, which correlates with a reduced rate of tegafur metabolism in vitro [55] and in vivo the *4C and *11 alleles confer poor tegafur metabolism [95]. Conversely, a novel *1B allele is associated with increased CYP2A6 protein expression and significantly higher rates of 5 FU formation in human liver microsome (HLM) samples, compared to samples without the haplotype (Table 2) [55].

To optimize 5 FU efficacy and reduce the toxicity of side effects, tegafur is combined with 5-chloro-2,4-dihydroxypyridine (CDHP) (inhibits degradation of 5 FU) and potassium oxonate (prevents gastrointestinal toxicity), to form the oral drug S-1 [96]. In cancer patients treated with S-1, genotypes containing CYP2A6*4, *7, *9 and *10 alleles are associated with reduced metabolism of tegafur compared to wildtype CYP2A6(Table 2) [97100]. Those with two variant CYP2A6 alleles (*4/*4, *4/*7 or *7/*7) have significantly lower oral clearance of tegafur compared to wildtype homozygotes [100]. Those without the *4C allele have significantly lower tegafur and higher 5 FU plasma concentrations compared to those with the allele [99]. However, other studies find no association between 5 FU plasma concentrations and CYP2A6 genotype, with 5 FU levels correlating instead with CDHP concentrations [97, 100].

Examining clinical outcome rather than 5FU blood concentrations suggests that the influence of CYP2A6 genetic variants on tegafur’s pharmacokinetics (PK) may have clinical importance. S-1 and cisplatin-treated patients with two CYP2A6 variant alleles *4, *7, *9, *10, or the *1/*4 genotype, have significantly lower treatment response rates, increased risk of disease progression and increased likelihood of reduced overall survival time than other genotypes [101]. Similar findings are observed in a study treating patients with S-1 and docetaxel; those with two CYP2A6 variant alleles (*4, *7, *9 or *10) display a 5-fold increased risk of cancer progression [102]. However, in this study overall survival is not significantly associated with genotype, possibly due to switching to alternative chemotherapy upon tumor progression (as discussed in [102]). Whether the association between CYP2A6 variants and reduced tegafur treatment efficacy is due to reduced formation of 5 FU cannot be concluded due to a lack of parallel PK studies [101, 102]. Significantly higher 5 FU plasma concentrations are found in responders to S-1 treatment compared to non-responders, and although no direct significant association between CYP2A6 genotype and treatment response is found in this study, those with one variant CYP2A6 allele have significantly higher 5 FU and significantly lower tegafur plasma concentrations compared to those with two variant alleles [103].

Combining these results suggests that a poor-metabolizer CYP2A6 genotype is associated with reduced tegafur metabolism to 5 FU and thus reduced anti-tumor efficacy [101103]. CYP2A6 genotype does not seem to influence treatment side effects, such as hematological toxicity [101103]. Although these associations remain to be investigated further in studies that combine PK and clinical outcome in large sample sizes, current findings suggest that CYP2A6 genotype may be a useful addition to tegafur dosing guidelines to increase treatment efficacy.

Letrozole

Letrozole is a daily oral treatment for estrogen- or progesterone-receptor positive breast cancer in postmenopausal women, and suppresses estrogen synthesis by inhibiting the aromatase enzyme [104]. Letrozole plasma levels, elimination rate and clearance show high inter-individual variation, and may contribute to adverse drug reactions or differences in treatment efficacy [41, 105]. CYP2A6 has a major role in the breakdown of letrozole into its inactive carbinol metabolite [106, 107] (see also the letrozole (Femara) tablet drug label, Novartis Pharmaceuticals Corporation) [108]. In vitro, HLM samples from individuals with CYP2A6 genotypes *1/*4, *4/*4, *4/*9 and *1/*7 have significantly reduced letrozole oxidation compared to *1/*1 wildtype samples [106]. Clinical studies in healthy postmenopausal women show that clearance of letrozole is lower in individuals with a CYP2A6 variant allele (*4, *9, *7) compared to *1 (see Table 2) [105]. In a cohort of breast cancer patients, CYP2A6 genotype is significantly related to letrozole plasma concentrations, explaining around 26% of the inter-individual variability observed [41]. CYP2A6 genotypes defined as conferring slow and intermediate metabolism are associated with significantly higher plasma concentrations of letrozole compared to the wildtype genotype (*1/*1) [41] (see Table 2). The ability to predict letrozole plasma concentration using CYP2A6 genotype is further improved by integrating age and body mass index (BMI) variables, explaining 32.3% of inter-individual variation [41]. As both CYP2A6 genotype and body weight influence letrozole levels, together they may explain some of the differences in letrozole PK parameters seen between Asian and Caucasian individuals [105]. Overall, these studies provide evidence to suggest that CYP2A6 genotyping, along with BMI and age, could be useful for predicting exposure to letrozole in patients [41, 105, 106].

CYP2A6 and treatment of infectious diseases

CYP2A6 has a role in the metabolism of several drugs involved in the treatment of infectious diseases, as outlined below and detailed in Table 2. When treating individuals for co-infections, such as HIV and malaria infection, the added complication of drug-drug interactions and induction or inhibition of CYP-450 enzymes by these drugs should be considered when assessing the pharmacogenetic effect [109, 110].

Efavirenz

Efavirenz (EFV) is a non-nucleoside reverse transcriptase inhibitor (NNRTI) that suppresses viral replication and is used as a component in highly active anti-retroviral therapy (HAART) regimens for HIV-infected patients [111]. High inter-individual variability of EFV plasma levels exists between patients receiving a fixed daily dose, and this has clinical implications; higher EFV plasma levels are associated with increased risk of central nervous system (CNS) side effects, whereas significantly lower levels are associated with failure to suppress viral replication [112115]. Demographic factors sex, age, body mass index, or co-medication, cannot explain this variability [113115], and instead genetic variants underlie a high proportion of the inter-individual variation in EFV plasma concentrations (discussed below). Genotyping may therefore aid EFV dosing, help avoid virolic failure and CNS related adverse reactions.

CYP2B6 has a key role in EFV metabolism, predominantly forming the major metabolite 8-hydroxyefavirenz (the product of over 90% of EFV oxidation) [116118], and a high percentage of inter-individual variation in EFV PK is attributed to CYP2B6 genetic variation [115, 118123]. CYP2B6 and CYP2A6 contribute to the 7-hydroxylation of EFV (represents less than 10% of EFV oxidation) [116118]. CYP2A6 genetic variation therefore also plays a role in the variability of EFV PK seen in patients, the effects of which are particularly prominent in CYP2B6 slow metabolizers [118, 121, 122]. These studies are detailed below and in Table 2.

In small studies of 50–65 individuals, there is no statistically significant association between EFV PK parameters and the SNPs rs8192726 (1836 G>T, *9b) or rs28399454 (5065 G>A, *17) [119, 122], though rs8192726 is associated with higher EFV plasma levels when the study size is increased to 94 individuals [123]. A trend for higher EFV plasma concentrations in rs28399433 T/G (CYP2A6*1/*9) heterozygotes compared to TT homozygotes does not reach significance after correcting for multiple comparisons in a small study of 45 individuals [115]. The lack of significant association in these studies may be due to low allele frequencies of the variants examined, small sample sizes, the relatively smaller contribution of CYP2A6 to EFV PK and/ or a weak association; for instance, in Kwara et al. a significant association between EFV PK parameters and CYP2A6 was observed when individuals with one or more copies of CYP2A6*9b (rs8192726) and CYP2A6*17 (rs28399454) were grouped together, but not when the SNPs were analyzed individually [122].

Individuals with two CYP2A6 loss-of-function alleles, two decreased function CYP2A6 alleles, or one of each (Table 2) have significantly higher EFV plasma concentrations, compared to those without these alleles, in individuals carrying CYP2B6 reference alleles [118]. Stratifying for both CYP2A6 and CYP2B6 genotype, a trend for lower 7-hydroxy-EFV metabolite levels is seen in patients with two loss of function CYP2B6 alleles and two CYP2A6 loss or decrease function alleles, and higher levels of the CYP-450 independent metabolite N-glucuronide-EFV [118]. Individuals with CYP2A6 loss-of-function alleles have lower clearance of EFV, and this is more pronounced in those who also have CYP2B6 loss-of-function alleles [121]. In multiple regression analysis, incorporating multiple variants, CYP2A6 rs8192726 and/ or rs28399454 status independently contributes to EFV inter-individual plasma concentrations, accounting for around 10% (8.6–12%), with CYP2B6 rs3745274 genotype TT contributing 36–45.2%, and UGT2B7 *1a genotype also significantly contributing [122, 123]. To conclude, genetic variation in the CYP2B6, CYP2A6, CYP3A4 and UGT2B7 genes contributes to inter-individual variation of EFV clearance [121123], and the effect of carrying CYP2A6 loss-of-function alleles on EFV clearance is more pronounced in people who are also CYP2B6 slow metabolizers [118, 121, 122]. The clinical consequence of CYP2A6 genotypes is not reported in the studies above, although higher plasma concentrations of EFV are associated with both increased viral suppression and increased likelihood of CNS adverse reactions [112114]. The CYP2A6 enzyme may have a role in the metabolism of other HAART drugs prescribed alongside EFV, for example Zidovudine (see the PharmGKB Zidovudine Pathway, PK/PD https://www.pharmgkb.org/pathway/PA165859361) [124], which could affect overall clinical outcome of viral suppression or adverse drug effects.

Artemisinin and derivatives

Artemisinin and its derivatives are drugs used to fight malaria infection [109, 125]. Recombinant CYP2A6 metabolizes artemisinin and CYP2A6 inhibition attenuates the rate of drug disappearance in human microsomes in vitro [126]. However, CYP2B6 and CYP3A4 enzymes are thought to have a greater role in artemisinin metabolism [126] (see the PharmGKB Artemisinin and Derivatives Pathway, Pharmacokinetics https://www.pharmgkb.org/pathway/PA165378192). Artemisinin derivatives (arteether, artemether, artesunate) were developed to enhance drug bioavailability, and are used in combination with a second unrelated slower acting drug, in order to initially rapidly eradicate malaria parasites within red blood cells, and then kill any residual parasites [109,125]. CYP2A6 is the major CYP450 enzyme involved in artesunate metabolism, forming dihydro-artemisinin, which is then inactivated by UGT enzymes (see the PharmGKB Artemisinin and Derivatives Pathway, Pharmacokinetics) [109, 127]. Therefore, CYP2A6 alleles which confer loss-of-function or decreased function may affect metabolism of these anti-malarial drugs. However, studies investigating urinary metabolites after dosing with artemisinin or derivatives, and use concurrent coumarin or nicotine probe drugs, see no clear correlation between CYP2A6 genotype and an effect on PK or enzyme activity [128, 129]. Artemisinin and derivatives induce CYP2B6 expression [130, 131], further complicating CYP2A6 association studies. Studies controlling for CYP2B6 status and with larger numbers are therefore required to investigate the clinical implications of CYP2A6 genotype on the metabolism of artemisinin and its derivatives.

CYP2A6 and other therapeutic drugs

Valproic acid

The antiepileptic drug valproic acid (VPA) is also used to treat migraines and schizophrenia, and could be a potential anti-cancer drug [132]. In vitro studies demonstrate CYP2C9 is the principle enzyme in VPA metabolism, however CYP2A6 contributes to around 50% of VPA 3-hydroxylation in human microsomes [133] and can contribute to 4-ene-VPA formation, a metabolite of VPA thought to cause hepatotoxicity [11, 133135]. CYP2A6 activity against coumarin is inhibited by VPA treatment, therefore VPA may affect responses to drugs metabolized by CYP2A6 taken concurrently [136]. Individuals with the *4 gene deletion allele have significantly increased plasma levels of VPA, likely due to decreased CYP2A6 enzyme metabolic activity, and may result in increased drug exposure [137].

Pilocarpine

Pilocarpine is used for the treatment of glaucoma and xerostomia [138]. CYP2A6 is the principle enzyme involved in 3-hydroxypilocarpine formation from pilocarpine in HLMs [138]. Poor metabolizers with two inactive CYP2A6 alleles have significantly increased pilocarpine plasma concentrations and a trend for higher excretion in the urine, with concurrent reductions of the metabolite 3-hydroxypilocarpine compared to non-poor metabolizers [139]. Despite these significant differences in metabolism, no severe adverse effects are observed, and the authors suggest CYP2A6 poor metabolizers likely use alternative renal clearance pathways [139].

SM-12502

SM-12502 is a platelet activating factor (PAF) receptor antagonist and has potential for use in asthma therapy [140]. CYP2A6 is the major metabolizer enzyme of SM-12502 in human microsomes in vitro [141], and the compound has been used to identify CYP2A6 polymorphisms that confer poor and extensive metabolizer phenotypes in vivo [142].

CYP2A6 is involved to a varying extent in the metabolism of numerous other therapeutic drugs (see [1] for an extensive list of substrates). These include the antitumour drugs ifosfamide (see the PharmGKB Ifosfamide Pathway, Pharmacokinetics https://www.pharmgkb.org/pathway/PA2037) and cyclophosphamide (see the PharmGKB cyclophosphamide Pathway, Pharmacokinetics https://www.pharmgkb.org/pathway/PA2034), the epilepsy treatments phenytoin (see the PharmGKB Phenytoin Pathway, Pharmacokinetics https://www.pharmgkb.org/pathway/PA145011115) [143], carbamazepine (see the PharmGKB Carbamazepine Pathway, Pharmacokinetics https://www.pharmgkb.org/pathway/PA165817070) [144, 145], and losigamone [146,147]. CYP2A6 also has a secondary role in metabolism of the anaesthesia halothane [148]. Therefore, CYP2A6 polymorphisms may affect additional pharmaceuticals, however the extent of CYP2A6’s role in the metabolism/ clearance of these drugs may be minor and/ or redundant, and other genetic variants including those in CYP-450 genes may play a more prominent role in the overall outcome.

CYP2A6 and toxicology

CYP2A6 polymorphisms have not only been associated with extent of nicotine metabolism and effect on smoking behaviors (as discussed previously and outlined in Table 1), but also with activation of carcinogens from tobacco and xenobiotics [1, 149]. Tobacco-specific nitrosamines, including nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and N-nitrosonornicotine (NNN), are present in tobacco and cigarette smoke, and hydroxlylation by CYP2A6, CYP2A13 and other CYPs activates NNK and NNN into metabolites that can then react with DNA to form adducts [1, 149]. CYP2A6 is involved in the activation of herbicides and pollutants such as hexamethylphosphoramide, dichlorobenzonitrile, aflatoxin B1, naphthalene, methyl tert-butyl ether, and N-nitro-sobenzylmethylamine (NBzMA) [1, 12], which are also procarcinogens.

CYP2A6 polymorphisms conferring lower enzyme activity are associated with reduced risk of lung, oral, head and neck, and upper aerodigestive cancers, particularly in tobacco users [150156]. The association between CYP2A6 and lung cancer is especially prominent among those who smoke 20 or fewer cigarettes per day [38]. Higher CYP2A6 enzyme activity is associated with increased risk of pancreatic cancer (adjusted for smoking status) [157]. The results of different association studies are mixed, often due to a lack of statistical power or failure to adjust for smoking status and behaviors [90, 149]. The relative contribution of CYP2A6 polymorphisms to cancer risk through smoking behavior versus carcinogen activation is difficult to define [29, 90, 149], though studies controlling for the amount of cigarette exposure suggest that carcinogen bioactivation is a significant contributor [38, 152]. Inhibition of CYP2A6 could potentially reduce cancer risk [29, 90, 149].

Conclusions

CYP2A6 has a major role in the metabolism of nicotine and several commonly prescribed drugs, as well as a lesser role in the metabolism of a large variety of other pharmaceuticals and xenobiotics. The CYP2A6 gene is highly polymorphic, and this has been associated with the high level of inter-individual and inter-ethnic variability in CYP2A6 enzyme activity and to variation in drug responses. CYP2A6 alleles conferring reduced enzyme activity have been associated with reduced metabolism and drug clearance, and thus linked with altered treatment response, as well as protection from cancer. CYP2A6 genotype may be important in determining the dosage for several treatments important in cancer therapy and HIV, in order to optimize efficacy and reduce the likelihood of adverse drug reactions. Further studies are required to confirm the role of individual genotypes in drug responses in larger cohorts and new technologies may help define CYP2A6 alleles more accurately.

Acknowledgments

This work is supported by the NIH/NIGMS (R24 GM61374), a CAMH, CRC (RFT), CIHR grant MOP86471, NIH grant UO1 DA020830, and an Ashley scholarship (CW).

Abbreviations

17U

1,7-dimethyluric acid

17X

1,7-dimethylxanthine, paraxanthine

3HC

trans-3’-hydroxy-cotinine

5FU

5-fluorouracil

AOX

aldehyde oxidase

CDHP

chloro-2,4-dihydroxypyridine

CNS

central nervous system

COPD

Chronic Obstructive Pulmonary Disease

COT

cotinine

Cpd

cigarettes per day

CYP-450

cytochrome P450

EFV

efavirenz

HAART

highly active anti-retroviral therapy

HLM

human liver microsomes

NBzMA

N-nitro-sobenzylmethylamine

NNK

nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone

NNN

N-nitrosonornicotine

PAF

platelet activating factor

PD

pharmacodynamic

PK

pharmacokinetic

SNP

single nucleotide polymorphism

VPA

valproic acid

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.

Conflicts of Interest

Dr. Tyndale owns shares and participates in Nicogen Research Inc., a company focused on novel smoking cessation treatment approaches. No Nicogen funds were used in this work and no other Nicogen participants reviewed the manuscript. Dr. Tyndale has also participated in one day Novartis and McNeil meetings. S.P.D. is a consultant for Genophen and has participated in a one day Pfizer meeting. R.B.A. serves as a founder and consultant for Personalis.

References

  • 1.Di YM, Chow VD, Yang LP, Zhou SF. Structure, function, regulation and polymorphism of human cytochrome P450 2A6. Curr Drug Metab. 2009;10:754–780. doi: 10.2174/138920009789895507. [DOI] [PubMed] [Google Scholar]
  • 2.Ding X, Kaminsky LS. Human extrahepatic cytochromes P450: function in xenobiotic metabolism and tissue-selective chemical toxicity in the respiratory and gastrointestinal tracts. Annu Rev Pharmacol Toxicol. 2003;43:149–173. doi: 10.1146/annurev.pharmtox.43.100901.140251. [DOI] [PubMed] [Google Scholar]
  • 3.Hoffman SM, Nelson DR, Keeney DS. Organization, structure and evolution of the CYP2 gene cluster on human chromosome 19. Pharmacogenetics. 2001;11:687–698. doi: 10.1097/00008571-200111000-00007. [DOI] [PubMed] [Google Scholar]
  • 4.PharmGKB CYP2A6 VIP: http://www.pharmgkb.org/vip/PA121
  • 5.Shimada T, Yamazaki H, Mimura M, Inui Y, Guengerich FP. Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. J Pharmacol Exp Ther. 1994;270:414–423. [PubMed] [Google Scholar]
  • 6.Nakajima M, Itoh M, Sakai H, Fukami T, Katoh M, Yamazaki H, Kadlubar FF, Imaoka S, Funae Y, Yokoi T. CYP2A13 expressed in human bladder metabolically activates 4-aminobiphenyl. Int J Cancer. 2006;119:2520–2526. doi: 10.1002/ijc.22136. [DOI] [PubMed] [Google Scholar]
  • 7.Miles JS, McLaren AW, Forrester LM, Glancey MJ, Lang MA, Wolf CR. Identification of the human liver cytochrome P-450 responsible for coumarin 7-hydroxylase activity. Biochem J. 1990;267:365–371. doi: 10.1042/bj2670365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yamano S, Tatsuno J, Gonzalez FJ. The CYP2A3 gene product catalyzes coumarin 7-hydroxylation in human liver microsomes. Biochemistry. 1990;29:1322–1329. doi: 10.1021/bi00457a031. [DOI] [PubMed] [Google Scholar]
  • 9.Meunier V, Bourrie M, Julian B, Marti E, Guillou F, Berger Y, Fabre G. Expression and induction of CYP1A1/1A2, CYP2A6 and CYP3A4 in primary cultures of human hepatocytes: a 10-year follow-up. Xenobiotica. 2000;30:589–607. doi: 10.1080/004982500406426. [DOI] [PubMed] [Google Scholar]
  • 10.Donato MT, Viitala P, Rodriguez-Antona C, Lindfors A, Castell JV, Raunio H, Gomez-Lechon MJ, Pelkonen O. CYP2A5/CYP2A6 expression in mouse and human hepatocytes treated with various in vivo inducers. Drug Metab Dispos. 2000;28:1321–1326. [PubMed] [Google Scholar]
  • 11.Sadeque AJ, Fisher MB, Korzekwa KR, Gonzalez FJ, Rettie AE. Human CYP2C9 and CYP2A6 mediate formation of the hepatotoxin 4-ene-valproic acid. J Pharmacol Exp Ther. 1997;283:698–703. [PubMed] [Google Scholar]
  • 12.Le Gal A, Dreano Y, Lucas D, Berthou F. Diversity of selective environmental substrates for human cytochrome P450 2A6: alkoxyethers, nicotine, coumarin, N-nitrosodiethylamine, and N-nitrosobenzylmethylamine. Toxicol Lett. 2003;144:77–91. doi: 10.1016/s0378-4274(03)00229-7. [DOI] [PubMed] [Google Scholar]
  • 13.Satarug S, Tassaneeyakul W, Na-Bangchang K, Cashman JR, Moore MR. Genetic and environmental influences on therapeutic and toxicity outcomes: studies with CYP2A6. Curr Clin Pharmacol. 2006;1:291–309. doi: 10.2174/157488406778249343. [DOI] [PubMed] [Google Scholar]
  • 14.Su T, Ding X. Regulation of the cytochrome P450 2A genes. Toxicol Appl Pharmacol. 2004;199:285–294. doi: 10.1016/j.taap.2003.11.029. [DOI] [PubMed] [Google Scholar]
  • 15.Easterbrook J, Fackett D, Li AP. A comparison of aroclor 1254-induced and uninduced rat liver microsomes to human liver microsomes in phenytoin O-deethylation, coumarin 7-hydroxylation, tolbutamide 4-hydroxylation, S-mephenytoin 4'-hydroxylation, chloroxazone 6-hydroxylation and testosterone 6beta-hydroxylation. Chem Biol Interact. 2001;134:243–249. doi: 10.1016/s0009-2797(01)00159-4. [DOI] [PubMed] [Google Scholar]
  • 16.Kobayashi K, Urashima K, Shimada N, Chiba K. Substrate specificity for rat cytochrome P450 (CYP) isoforms: screening with cDNA-expressed systems of the rat. Biochem Pharmacol. 2002;63:889–896. doi: 10.1016/s0006-2952(01)00843-7. [DOI] [PubMed] [Google Scholar]
  • 17.Born SL, Caudill D, Smith BJ, Lehman-McKeeman LD. In vitro kinetics of coumarin 3,4-epoxidation: application to species differences in toxicity and carcinogenicity. Toxicol Sci. 2000;58:23–31. doi: 10.1093/toxsci/58.1.23. [DOI] [PubMed] [Google Scholar]
  • 18.Lewis DF, Lake BG. Species differences in coumarin metabolism: a molecular modelling evaluation of CYP2A interactions. Xenobiotica. 2002;32:547–561. doi: 10.1080/00498250210128693. [DOI] [PubMed] [Google Scholar]
  • 19.Hammond DK, Bjercke RJ, Langone JJ, Strobel HW. Metabolism of nicotine by rat liver cytochromes P-450. Assessment utilizing monoclonal antibodies to nicotine and cotinine. Drug Metab Dispos. 1991;19:804–808. [PubMed] [Google Scholar]
  • 20.Yano JK, Hsu MH, Griffin KJ, Stout CD, Johnson EF. Structures of human microsomal cytochrome P450 2A6 complexed with coumarin and methoxsalen. Nat Struct Mol Biol. 2005;12:822–823. doi: 10.1038/nsmb971. [DOI] [PubMed] [Google Scholar]
  • 21. [Accessed 21st July 2011];Protein Data Bank http://www.pdb.org/pdb/home/home.do.
  • 22.Phillips IR, Shephard EA, Ashworth A, Rabin BR. Isolation and sequence of a human cytochrome P-450 cDNA clone. Proc Natl Acad Sci U S A. 1985;82:983–987. doi: 10.1073/pnas.82.4.983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Phillips IR, Shephard EA, Povey S, Davis MB, Kelsey G, Monteiro M, West LF, Cowell J. A cytochrome P-450 gene family mapped to human chromosome 19. Ann Hum Genet. 1985;49:267–274. doi: 10.1111/j.1469-1809.1985.tb01702.x. [DOI] [PubMed] [Google Scholar]
  • 24.Nelson DR, Kamataki T, Waxman DJ, Guengerich FP, Estabrook RW, Feyereisen R, Gonzalez FJ, Coon MJ, Gunsalus IC, Gotoh O, et al. The P450 superfamily: update on new sequences, gene mapping, accession numbers, early trivial names of enzymes, and nomenclature. DNA Cell Biol. 1993;12:1–51. doi: 10.1089/dna.1993.12.1. [DOI] [PubMed] [Google Scholar]
  • 25.Miles JS, Bickmore W, Brook JD, McLaren AW, Meehan R, Wolf CR. Close linkage of the human cytochrome P450IIA and P450IIB gene subfamilies: implications for the assignment of substrate specificity. Nucleic Acids Res. 1989;17:2907–2917. doi: 10.1093/nar/17.8.2907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Yamano S, Nagata K, Yamazoe Y, Kato R, Gelboin HV, Gonzalez FJ. cDNA and deduced amino acid sequences of human P450 IIA3 (CYP2A3) Nucleic Acids Res. 1989;17:4888. doi: 10.1093/nar/17.12.4888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Human Cytochrome P450 (CYP) Allele Nomenclature Committee: http://www.cypalleles.ki.se/
  • 28. [accessed on 30th Aug 2011];Human Cytochrome P450 (CYP) Allele Nomenclature Committee CYP2A6: http://www.cypalleles.ki.se/cyp2a6.htm.
  • 29.Tyndale RF, Sellers EM. Genetic variation in CYP2A6-mediated nicotine metabolism alters smoking behavior. Ther Drug Monit. 2002;24:163–171. doi: 10.1097/00007691-200202000-00026. [DOI] [PubMed] [Google Scholar]
  • 30.Oscarson M, McLellan RA, Gullsten H, Agundez JA, Benitez J, Rautio A, Raunio H, Pelkonen O, Ingelman-Sundberg M. Identification and characterisation of novel polymorphisms in the CYP2A locus: implications for nicotine metabolism. FEBS Lett. 1999;460:321–327. doi: 10.1016/s0014-5793(99)01364-2. [DOI] [PubMed] [Google Scholar]
  • 31.Rao Y, Hoffmann E, Zia M, Bodin L, Zeman M, Sellers EM, Tyndale RF. Duplications and defects in the CYP2A6 gene: identification, genotyping, and in vivo effects on smoking. Mol Pharmacol. 2000;58:747–755. doi: 10.1124/mol.58.4.747. [DOI] [PubMed] [Google Scholar]
  • 32.Loriot MA, Rebuissou S, Oscarson M, Cenee S, Miyamoto M, Ariyoshi N, Kamataki T, Hemon D, Beaune P, Stucker I. Genetic polymorphisms of cytochrome P450 2A6 in a case-control study on lung cancer in a French population. Pharmacogenetics. 2001;11:39–44. doi: 10.1097/00008571-200102000-00005. [DOI] [PubMed] [Google Scholar]
  • 33.Johansson I, Ingelman-Sundberg M. Genetic polymorphism and toxicology--with emphasis on cytochrome p450. Toxicol Sci. 2011;120:1–13. doi: 10.1093/toxsci/kfq374. [DOI] [PubMed] [Google Scholar]
  • 34.Benowitz NL, Swan GE, Jacob P, 3rd, Lessov-Schlaggar CN, Tyndale RF. CYP2A6 genotype and the metabolism and disposition kinetics of nicotine. Clin Pharmacol Ther. 2006;80:457–467. doi: 10.1016/j.clpt.2006.08.011. [DOI] [PubMed] [Google Scholar]
  • 35.Malaiyandi V, Goodz SD, Sellers EM, Tyndale RF. CYP2A6 genotype, phenotype, and the use of nicotine metabolites as biomarkers during ad libitum smoking. Cancer Epidemiol Biomarkers Prev. 2006;15:1812–1819. doi: 10.1158/1055-9965.EPI-05-0723. [DOI] [PubMed] [Google Scholar]
  • 36.Schoedel KA, Hoffmann EB, Rao Y, Sellers EM, Tyndale RF. Ethnic variation in CYP2A6 and association of genetically slow nicotine metabolism and smoking in adult Caucasians. Pharmacogenetics. 2004;14:615–626. doi: 10.1097/00008571-200409000-00006. [DOI] [PubMed] [Google Scholar]
  • 37.Lerman C, Jepson C, Wileyto EP, Patterson F, Schnoll R, Mroziewicz M, Benowitz N, Tyndale RF. Genetic variation in nicotine metabolism predicts the efficacy of extended-duration transdermal nicotine therapy. Clin Pharmacol Ther. 2010;87:553–557. doi: 10.1038/clpt.2010.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wassenaar CA, Dong Q, Wei Q, Amos CI, Spitz MR, Tyndale RF. Relationship between CYP2A6 and CHRNA5-CHRNA3-CHRNB4 variation and smoking behaviors and lung cancer risk. J Natl Cancer Inst. 2011;103:1342–1346. doi: 10.1093/jnci/djr237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Fukami T, Nakajima M, Higashi E, Yamanaka H, Sakai H, McLeod HL, Yokoi T. Characterization of novel CYP2A6 polymorphic alleles (CYP2A6*18 and CYP2A6*19) that affect enzymatic activity. Drug Metab Dispos. 2005;33:1202–1210. doi: 10.1124/dmd.105.004994. [DOI] [PubMed] [Google Scholar]
  • 40.Ho MK, Mwenifumbo JC, Zhao B, Gillam EM, Tyndale RF. A novel CYP2A6 allele, CYP2A6*23, impairs enzyme function in vitro and in vivo and decreases smoking in a population of Black-African descent. Pharmacogenet Genomics. 2008;18:67–75. doi: 10.1097/FPC.0b013e3282f3606e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Desta Z, Kreutz Y, Nguyen AT, Li L, Skaar T, Kamdem LK, Henry NL, Hayes DF, Storniolo AM, Stearns V, et al. Plasma letrozole concentrations in postmenopausal women with breast cancer are associated with CYP2A6 genetic variants, body mass index, and age. Clin Pharmacol Ther. 2011;90:693–700. doi: 10.1038/clpt.2011.174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pitarque M, von Richter O, Oke B, Berkkan H, Oscarson M, Ingelman-Sundberg M. Identification of a single nucleotide polymorphism in the TATA box of the CYP2A6 gene: impairment of its promoter activity. Biochem Biophys Res Commun. 2001;284:455–460. doi: 10.1006/bbrc.2001.4990. [DOI] [PubMed] [Google Scholar]
  • 43.Wang J, Pitarque M, Ingelman-Sundberg M. 3'-UTR polymorphism in the human CYP2A6 gene affects mRNA stability and enzyme expression. Biochem Biophys Res Commun. 2006;340:491–497. doi: 10.1016/j.bbrc.2005.12.035. [DOI] [PubMed] [Google Scholar]
  • 44.Yoshida R, Nakajima M, Nishimura K, Tokudome S, Kwon JT, Yokoi T. Effects of polymorphism in promoter region of human CYP2A6 gene (CYP2A6*9) on expression level of messenger ribonucleic acid and enzymatic activity in vivo and in vitro. Clin Pharmacol Ther. 2003;74:69–76. doi: 10.1016/S0009-9236(03)00090-0. [DOI] [PubMed] [Google Scholar]
  • 45.Xu C, Rao YS, Xu B, Hoffmann E, Jones J, Sellers EM, Tyndale RF. An in vivo pilot study characterizing the new CYP2A6*7, *8, and *10 alleles. Biochem Biophys Res Commun. 2002;290:318–324. doi: 10.1006/bbrc.2001.6209. [DOI] [PubMed] [Google Scholar]
  • 46.Malaiyandi V, Lerman C, Benowitz NL, Jepson C, Patterson F, Tyndale RF. Impact of CYP2A6 genotype on pretreatment smoking behaviour and nicotine levels from and usage of nicotine replacement therapy. Mol Psychiatry. 2006;11:400–409. doi: 10.1038/sj.mp.4001794. [DOI] [PubMed] [Google Scholar]
  • 47.Audrain-McGovern J, Al Koudsi N, Rodriguez D, Wileyto EP, Shields PG, Tyndale RF. The role of CYP2A6 in the emergence of nicotine dependence in adolescents. Pediatrics. 2007;119:e264–e274. doi: 10.1542/peds.2006-1583. [DOI] [PubMed] [Google Scholar]
  • 48.Minematsu N, Nakamura H, Furuuchi M, Nakajima T, Takahashi S, Tateno H, Ishizaka A. Limitation of cigarette consumption by CYP2A6*4, *7 and *9 polymorphisms. Eur Respir J. 2006;27:289–292. doi: 10.1183/09031936.06.00056305. [DOI] [PubMed] [Google Scholar]
  • 49.Nakajima M, Fukami T, Yamanaka H, Higashi E, Sakai H, Yoshida R, Kwon JT, McLeod HL, Yokoi T. Comprehensive evaluation of variability in nicotine metabolism and CYP2A6 polymorphic alleles in four ethnic populations. Clin Pharmacol Ther. 2006;80:282–297. doi: 10.1016/j.clpt.2006.05.012. [DOI] [PubMed] [Google Scholar]
  • 50.Peamkrasatam S, Sriwatanakul K, Kiyotani K, Fujieda M, Yamazaki H, Kamataki T, Yoovathaworn K. In vivo evaluation of coumarin and nicotine as probe drugs to predict the metabolic capacity of CYP2A6 due to genetic polymorphism in Thais. Drug Metab Pharmacokinet. 2006;21:475–484. doi: 10.2133/dmpk.21.475. [DOI] [PubMed] [Google Scholar]
  • 51.Gyamfi MA, Fujieda M, Kiyotani K, Yamazaki H, Kamataki T. High prevalence of cytochrome P450 2A6*1A alleles in a black African population of Ghana. Eur J Clin Pharmacol. 2005;60:855–857. doi: 10.1007/s00228-004-0854-9. [DOI] [PubMed] [Google Scholar]
  • 52.Nurfadhlina M, Foong K, Teh LK, Tan SC, Mohd Zaki S, Ismail R. CYP2A6 polymorphisms in Malays, Chinese and Indians. Xenobiotica. 2006;36:684–692. doi: 10.1080/00498250600715932. [DOI] [PubMed] [Google Scholar]
  • 53.Huang S, Cook DG, Hinks LJ, Chen XH, Ye S, Gilg JA, Jarvis MJ, Whincup PH, Day IN. CYP2A6, MAOA, DBH, DRD4, and 5HT2A genotypes, smoking behaviour and cotinine levels in 1518 UK adolescents. Pharmacogenet Genomics. 2005;15:839–850. doi: 10.1097/01213011-200512000-00002. [DOI] [PubMed] [Google Scholar]
  • 54.Liu T, David SP, Tyndale RF, Wang H, Zhou Q, Ding P, He YH, Yu XQ, Chen W, Crump C, et al. Associations of CYP2A6 genotype with smoking behaviors in southern China. Addiction. 2011;106:985–994. doi: 10.1111/j.1360-0443.2010.03353.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wang H, Bian T, Liu D, Jin T, Chen Y, Lin A, Chen C. Association analysis of CYP2A6 genotypes and haplotypes with 5-fluorouracil formation from tegafur in human liver microsomes. Pharmacogenomics. 2011;12:481–492. doi: 10.2217/pgs.10.202. [DOI] [PubMed] [Google Scholar]
  • 56.Fukami T, Nakajima M, Yoshida R, Tsuchiya Y, Fujiki Y, Katoh M, McLeod HL, Yokoi T. A novel polymorphism of human CYP2A6 gene CYP2A6*17 has an amino acid substitution (V365M) that decreases enzymatic activity in vitro and in vivo. Clin Pharmacol Ther. 2004;76:519–527. doi: 10.1016/j.clpt.2004.08.014. [DOI] [PubMed] [Google Scholar]
  • 57.Moon YJ, Zhang S, Brazeau DA, Morris ME. Effects of the flavonoid biochanin A on gene expression in primary human hepatocytes and human intestinal cells. Mol Nutr Food Res. 2007;51:317–323. doi: 10.1002/mnfr.200600156. [DOI] [PubMed] [Google Scholar]
  • 58.Higashi E, Fukami T, Itoh M, Kyo S, Inoue M, Yokoi T, Nakajima M. Human CYP2A6 is induced by estrogen via estrogen receptor. Drug Metab Dispos. 2007;35:1935–1941. doi: 10.1124/dmd.107.016568. [DOI] [PubMed] [Google Scholar]
  • 59.Benowitz NL, Lessov-Schlaggar CN, Swan GE, Jacob P., 3rd Female sex and oral contraceptive use accelerate nicotine metabolism. Clin Pharmacol Ther. 2006;79:480–488. doi: 10.1016/j.clpt.2006.01.008. [DOI] [PubMed] [Google Scholar]
  • 60.Onica T, Nichols K, Larin M, Ng L, Maslen A, Dvorak Z, Pascussi JM, Vilarem MJ, Maurel P, Kirby GM. Dexamethasone-mediated up-regulation of human CYP2A6 involves the glucocorticoid receptor and increased binding of hepatic nuclear factor 4 alpha to the proximal promoter. Mol Pharmacol. 2008;73:451–460. doi: 10.1124/mol.107.039354. [DOI] [PubMed] [Google Scholar]
  • 61.Draper AJ, Madan A, Parkinson A. Inhibition of coumarin 7-hydroxylase activity in human liver microsomes. Arch Biochem Biophys. 1997;341:47–61. doi: 10.1006/abbi.1997.9964. [DOI] [PubMed] [Google Scholar]
  • 62.Chen Y, Kang Z, Yan J, Yang GP, Tan ZR, Zhou G, Ouyang DS. Liu wei di huang wan, a well-known traditional Chinese medicine, induces CYP1A2 while suppressing CYP2A6 and N-acetyltransferase 2 activities in man. J Ethnopharmacol. 2010;132:213–218. doi: 10.1016/j.jep.2010.08.011. [DOI] [PubMed] [Google Scholar]
  • 63.Hukkanen J, Jacob P, 3rd, Benowitz NL. Metabolism and disposition kinetics of nicotine. Pharmacol Rev. 2005;57:79–115. doi: 10.1124/pr.57.1.3. [DOI] [PubMed] [Google Scholar]
  • 64.McDonagh EM, Whirl-Carrillo M, Garten Y, Altman RB, Klein TE. From pharmacogenomic knowledge acquisition to clinical applications: the PharmGKB as a clinical pharmacogenomic biomarker resource. Biomark Med. 2011;5:795–806. doi: 10.2217/bmm.11.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Benowitz NL, Hukkanen J, Jacob P., 3rd Nicotine chemistry, metabolism, kinetics and biomarkers. Handb Exp Pharmacol. 2009:29–60. doi: 10.1007/978-3-540-69248-5_2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Yamazaki H, Inoue K, Hashimoto M, Shimada T. Roles of CYP2A6 and CYP2B6 in nicotine C-oxidation by human liver microsomes. Arch Toxicol. 1999;73:65–70. doi: 10.1007/s002040050588. [DOI] [PubMed] [Google Scholar]
  • 67.Nakajima M, Yamamoto T, Nunoya K, Yokoi T, Nagashima K, Inoue K, Funae Y, Shimada N, Kamataki T, Kuroiwa Y. Role of human cytochrome P4502A6 in C-oxidation of nicotine. Drug Metab Dispos. 1996;24:1212–1217. [PubMed] [Google Scholar]
  • 68.Brown KM, von Weymarn LB, Murphy SE. Identification of N-(hydroxymethyl) norcotinine as a major product of cytochrome P450 2A6, but not cytochrome P450 2A13-catalyzed cotinine metabolism. Chem Res Toxicol. 2005;18:1792–1798. doi: 10.1021/tx0501381. [DOI] [PubMed] [Google Scholar]
  • 69.Nakajima M, Yamamoto T, Nunoya K, Yokoi T, Nagashima K, Inoue K, Funae Y, Shimada N, Kamataki T, Kuroiwa Y. Characterization of CYP2A6 involved in 3'-hydroxylation of cotinine in human liver microsomes. J Pharmacol Exp Ther. 1996;277:1010–1015. [PubMed] [Google Scholar]
  • 70.Dempsey D, Tutka P, Jacob P, 3rd, Allen F, Schoedel K, Tyndale RF, Benowitz NL. Nicotine metabolite ratio as an index of cytochrome P450 2A6 metabolic activity. Clin Pharmacol Ther. 2004;76:64–72. doi: 10.1016/j.clpt.2004.02.011. [DOI] [PubMed] [Google Scholar]
  • 71.Nakajima M, Kwon JT, Tanaka N, Zenta T, Yamamoto Y, Yamamoto H, Yamazaki H, Yamamoto T, Kuroiwa Y, Yokoi T. Relationship between interindividual differences in nicotine metabolism and CYP2A6 genetic polymorphism in humans. Clin Pharmacol Ther. 2001;69:72–78. doi: 10.1067/mcp.2001.112688. [DOI] [PubMed] [Google Scholar]
  • 72.Bloom J, Hinrichs AL, Wang JC, von Weymarn LB, Kharasch ED, Bierut LJ, Goate A, Murphy SE. The contribution of common CYP2A6 alleles to variation in nicotine metabolism among European-Americans. Pharmacogenet Genomics. 2011;21:403–416. doi: 10.1097/FPC.0b013e328346e8c0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jarvik ME, Madsen DC, Olmstead RE, Iwamoto-Schaap PN, Elins JL, Benowitz NL. Nicotine blood levels and subjective craving for cigarettes. Pharmacol Biochem Behav. 2000;66:553–558. doi: 10.1016/s0091-3057(00)00261-6. [DOI] [PubMed] [Google Scholar]
  • 74.O'Loughlin J, Paradis G, Kim W, DiFranza J, Meshefedjian G, McMillan-Davey E, Wong S, Hanley J, Tyndale RF. Genetically decreased CYP2A6 and the risk of tobacco dependence: a prospective study of novice smokers. Tob Control. 2004;13:422–428. doi: 10.1136/tc.2003.007070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mwenifumbo JC, Tyndale RF. Molecular genetics of nicotine metabolism. Handb Exp Pharmacol. 2009:235–259. doi: 10.1007/978-3-540-69248-5_9. [DOI] [PubMed] [Google Scholar]
  • 76.Iwahashi K, Aoki J. A review of smoking behavior and smokers evidence (chemical modification, inducing nicotine metabolism, and individual variations by genotype: dopaminergic function and personality traits) Drug Chem Toxicol. 2009;32:301–306. doi: 10.1080/01480540903203976. [DOI] [PubMed] [Google Scholar]
  • 77.Quaak M, van Schayck CP, Knaapen AM, van Schooten FJ. Genetic variation as a predictor of smoking cessation success. A promising preventive and intervention tool for chronic respiratory diseases? Eur Respir J. 2009;33:468–480. doi: 10.1183/09031936.00056908. [DOI] [PubMed] [Google Scholar]
  • 78.Strasser AA, Benowitz NL, Pinto AG, Tang KZ, Hecht SS, Carmella SG, Tyndale RF, Lerman CE. Nicotine metabolite ratio predicts smoking topography and carcinogen biomarker level. Cancer Epidemiol Biomarkers Prev. 2011;20:234–238. doi: 10.1158/1055-9965.EPI-10-0674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lerman C, Tyndale R, Patterson F, Wileyto EP, Shields PG, Pinto A, Benowitz N. Nicotine metabolite ratio predicts efficacy of transdermal nicotine for smoking cessation. Clin Pharmacol Ther. 2006;79:600–608. doi: 10.1016/j.clpt.2006.02.006. [DOI] [PubMed] [Google Scholar]
  • 80.Malaiyandi V, Sellers EM, Tyndale RF. Implications of CYP2A6 genetic variation for smoking behaviors and nicotine dependence. Clin Pharmacol Ther. 2005;77:145–158. doi: 10.1016/j.clpt.2004.10.011. [DOI] [PubMed] [Google Scholar]
  • 81.Iwahashi K, Waga C, Takimoto T. Whole deletion of CYP2A6 gene (CYP2A6AST;4C) and smoking behavior. Neuropsychobiology. 2004;49:101–104. doi: 10.1159/000076418. [DOI] [PubMed] [Google Scholar]
  • 82.Gambier N, Batt AM, Marie B, Pfister M, Siest G, Visvikis-Siest S. Association of CYP2A6*1B genetic variant with the amount of smoking in French adults from the Stanislas cohort. Pharmacogenomics J. 2005;5:271–275. doi: 10.1038/sj.tpj.6500314. [DOI] [PubMed] [Google Scholar]
  • 83.Siedlinski M, Cho MH, Bakke P, Gulsvik A, Lomas DA, Anderson W, Kong X, Rennard SI, Beaty TH, Hokanson JE, et al. Genome-wide association study of smoking behaviours in patients with COPD. Thorax. 2011 doi: 10.1136/thoraxjnl-2011-200154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Haberl M, Anwald B, Klein K, Weil R, Fuss C, Gepdiremen A, Zanger UM, Meyer UA, Wojnowski L. Three haplotypes associated with CYP2A6 phenotypes in Caucasians. Pharmacogenet Genomics. 2005;15:609–624. doi: 10.1097/01.fpc.0000171517.22258.f1. [DOI] [PubMed] [Google Scholar]
  • 85.Al Koudsi N, Tyndale RF. Hepatic CYP2B6 is altered by genetic, physiologic, and environmental factors but plays little role in nicotine metabolism. Xenobiotica. 2010;40:381–392. doi: 10.3109/00498251003713958. [DOI] [PubMed] [Google Scholar]
  • 86.Kimura M, Yamazaki H, Fujieda M, Kiyotani K, Honda G, Saruwatari J, Nakagawa K, Ishizaki T, Kamataki T. Cyp2a6 is a principal enzyme involved in hydroxylation of 1,7-dimethylxanthine, a main caffeine metabolite, in humans. Drug Metab Dispos. 2005;33:1361–1366. doi: 10.1124/dmd.105.004796. [DOI] [PubMed] [Google Scholar]
  • 87.Thorn CF, Aklillu E, McDonagh EM, Klein TE, Altman RB. PharmGKB summary: caffeine pathway. Pharmacogenet Genomics. 2012 doi: 10.1097/FPC.0b013e3283505d5e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Djordjevic N, Carrillo JA, Gervasini G, Jankovic S, Aklillu E. In vivo evaluation of CYP2A6 and xanthine oxidase enzyme activities in the Serbian population. Eur J Clin Pharmacol. 2010;66:571–578. doi: 10.1007/s00228-010-0785-6. [DOI] [PubMed] [Google Scholar]
  • 89.Poland RE, Pechnick RN, Cloak CC, Wan YJ, Nuccio I, Lin KM. Effect of cigarette smoking on coumarin metabolism in humans. Nicotine Tob Res. 2000;2:351–354. doi: 10.1080/713688151. [DOI] [PubMed] [Google Scholar]
  • 90.Rodriguez-Antona C, Gomez A, Karlgren M, Sim SC, Ingelman-Sundberg M. Molecular genetics and epigenetics of the cytochrome P450 gene family and its relevance for cancer risk and treatment. Hum Genet. 2010;127:1–17. doi: 10.1007/s00439-009-0748-0. [DOI] [PubMed] [Google Scholar]
  • 91.Thorn CF, Marsh S, Carrillo MW, McLeod HL, Klein TE, Altman RB. PharmGKB summary: fluoropyrimidine pathways. Pharmacogenet Genomics. 2011;21:237–242. doi: 10.1097/FPC.0b013e32833c6107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ikeda K, Yoshisue K, Matsushima E, Nagayama S, Kobayashi K, Tyson CA, Chiba K, Kawaguchi Y. Bioactivation of tegafur to 5-fluorouracil is catalyzed by cytochrome P-450 2A6 in human liver microsomes in vitro. Clin Cancer Res. 2000;6:4409–4415. [PubMed] [Google Scholar]
  • 93.Komatsu T, Yamazaki H, Shimada N, Nakajima M, Yokoi T. Roles of cytochromes P450 1A2, 2A6, and 2C8 in 5-fluorouracil formation from tegafur, an anticancer prodrug, in human liver microsomes. Drug Metab Dispos. 2000;28:1457–1463. [PubMed] [Google Scholar]
  • 94.Kajita J, Fuse E, Kuwabara T, Kobayashi H. The contribution of cytochrome P450 to the metabolism of tegafur in human liver. Drug Metab Pharmacokinet. 2003;18:303–309. doi: 10.2133/dmpk.18.303. [DOI] [PubMed] [Google Scholar]
  • 95.Daigo S, Takahashi Y, Fujieda M, Ariyoshi N, Yamazaki H, Koizumi W, Tanabe S, Saigenji K, Nagayama S, Ikeda K, et al. A novel mutant allele of the CYP2A6 gene (CYP2A6*11) found in a cancer patient who showed poor metabolic phenotype towards tegafur. Pharmacogenetics. 2002;12:299–306. doi: 10.1097/00008571-200206000-00005. [DOI] [PubMed] [Google Scholar]
  • 96.Shirasaka T, Shimamato Y, Ohshimo H, Yamaguchi M, Kato T, Yonekura K, Fukushima M. Development of a novel form of an oral 5-fluorouracil derivative (S-1) directed to the potentiation of the tumor selective cytotoxicity of 5-fluorouracil by two biochemical modulators. Anticancer Drugs. 1996;7:548–557. doi: 10.1097/00001813-199607000-00010. [DOI] [PubMed] [Google Scholar]
  • 97.Hirose T, Fujita K, Nishimura K, Ishida H, Yamashita K, Sunakawa Y, Mizuno K, Miwa K, Nagashima F, Tanigawara Y, et al. Pharmacokinetics of S-1 and CYP2A6 genotype in Japanese patients with advanced cancer. Oncol Rep. 2010;24:529–536. doi: 10.3892/or_00000889. [DOI] [PubMed] [Google Scholar]
  • 98.Kim KP, Jang G, Hong YS, Lim HS, Bae KS, Kim HS, Lee SS, Shin JG, Lee JL, Ryu MH, et al. Phase II study of S-1 combined with oxaliplatin as therapy for patients with metastatic biliary tract cancer: influence of the CYP2A6 polymorphism on pharmacokinetics and clinical activity. Br J Cancer. 2011;104:605–612. doi: 10.1038/bjc.2011.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kaida Y, Inui N, Suda T, Nakamura H, Watanabe H, Chida K. The CYP2A6*4 allele is determinant of S-1 pharmacokinetics in Japanese patients with non-small-cell lung cancer. Clin Pharmacol Ther. 2008;83:589–594. doi: 10.1038/sj.clpt.6100484. [DOI] [PubMed] [Google Scholar]
  • 100.Fujita K, Yamamoto W, Endo S, Endo H, Nagashima F, Ichikawa W, Tanaka R, Miya T, Araki K, Kodama K, et al. CYP2A6 and the plasma level of 5-chloro-2,4-dihydroxypyridine are determinants of the pharmacokinetic variability of tegafur and 5-fluorouracil, respectively, in Japanese patients with cancer given S-1. Cancer Sci. 2008;99:1049–1054. doi: 10.1111/j.1349-7006.2008.00773.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Park SR, Kong SY, Nam BH, Choi IJ, Kim CG, Lee JY, Cho SJ, Kim YW, Ryu KW, Lee JH, et al. CYP2A6 and ERCC1 polymorphisms correlate with efficacy of S-1 plus cisplatin in metastatic gastric cancer patients. Br J Cancer. 2011;104:1126–1134. doi: 10.1038/bjc.2011.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kong SY, Lim HS, Nam BH, Kook MC, Kim YW, Ryu KW, Lee JH, Choi IJ, Lee JS, Park YI, et al. Association of CYP2A6 polymorphisms with S-1 plus docetaxel therapy outcomes in metastatic gastric cancer. Pharmacogenomics. 2009;10:1147–1155. doi: 10.2217/pgs.09.48. [DOI] [PubMed] [Google Scholar]
  • 103.Ishii Y, Suzuki S, Takahashi Y, Takayama T, Asai S. Can the 2-(13)C-uracil breath test be used to predict the effect of the antitumor drug S-1? Cancer Chemother Pharmacol. 2010;66:333–343. doi: 10.1007/s00280-009-1168-5. [DOI] [PubMed] [Google Scholar]
  • 104.Smith IE, Dowsett M. Aromatase inhibitors in breast cancer. N Engl J Med. 2003;348:2431–2442. doi: 10.1056/NEJMra023246. [DOI] [PubMed] [Google Scholar]
  • 105.Tanii H, Shitara Y, Horie T. Population pharmacokinetic analysis of letrozole in Japanese postmenopausal women. Eur J Clin Pharmacol. 2011 doi: 10.1007/s00228-011-1042-3. Epub ahead of press. [DOI] [PubMed] [Google Scholar]
  • 106.Murai K, Yamazaki H, Nakagawa K, Kawai R, Kamataki T. Deactivation of anti-cancer drug letrozole to a carbinol metabolite by polymorphic cytochrome P450 2A6 in human liver microsomes. Xenobiotica. 2009;39:795–802. doi: 10.3109/00498250903171395. [DOI] [PubMed] [Google Scholar]
  • 107.Jeong S, Woo MM, Flockhart DA, Desta Z. Inhibition of drug metabolizing cytochrome P450s by the aromatase inhibitor drug letrozole and its major oxidative metabolite 4,4'-methanol-bisbenzonitrile in vitro. Cancer Chemother Pharmacol. 2009;64:867–875. doi: 10.1007/s00280-009-0935-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.letrozole (Femara) tablet drug label, Novartis Pharmaceuticals Corporation on the DailyMed website: http://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?id=49536
  • 109.Kerb R, Fux R, Morike K, Kremsner PG, Gil JP, Gleiter CH, Schwab M. Pharmacogenetics of antimalarial drugs: effect on metabolism and transport. Lancet Infect Dis. 2009;9:760–774. doi: 10.1016/S1473-3099(09)70320-2. [DOI] [PubMed] [Google Scholar]
  • 110.Grimberg BT, Mehlotra RK. Expanding the Antimalarial Drug Arsenal-Now, But How? Pharmaceuticals (Basel) 2011;4:681–712. doi: 10.3390/ph4050681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Rakhmanina NY, van den Anker JN. Efavirenz in the therapy of HIV infection. Expert Opin Drug Metab Toxicol. 2010;6:95–103. doi: 10.1517/17425250903483207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Stahle L, Moberg L, Svensson JO, Sonnerborg A. Efavirenz plasma concentrations in HIV-infected patients: inter- and intraindividual variability and clinical effects. Ther Drug Monit. 2004;26:267–270. doi: 10.1097/00007691-200406000-00008. [DOI] [PubMed] [Google Scholar]
  • 113.Marzolini C, Telenti A, Decosterd LA, Greub G, Biollaz J, Buclin T. Efavirenz plasma levels can predict treatment failure and central nervous system side effects in HIV-1-infected patients. AIDS. 2001;15:71–75. doi: 10.1097/00002030-200101050-00011. [DOI] [PubMed] [Google Scholar]
  • 114.Csajka C, Marzolini C, Fattinger K, Decosterd LA, Fellay J, Telenti A, Biollaz J, Buclin T. Population pharmacokinetics and effects of efavirenz in patients with human immunodeficiency virus infection. Clin Pharmacol Ther. 2003;73:20–30. doi: 10.1067/mcp.2003.22. [DOI] [PubMed] [Google Scholar]
  • 115.Leger P, Dillingham R, Beauharnais CA, Kashuba AD, Rezk NL, Fitzgerald DW, Pape JW, Haas DW. CYP2B6 variants and plasma efavirenz concentrations during antiretroviral therapy in Port-au-Prince, Haiti. J Infect Dis. 2009;200:955–964. doi: 10.1086/605126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Ward BA, Gorski JC, Jones DR, Hall SD, Flockhart DA, Desta Z. The cytochrome P450 2B6 (CYP2B6) is the main catalyst of efavirenz primary and secondary metabolism: implication for HIV/AIDS therapy and utility of efavirenz as a substrate marker of CYP2B6 catalytic activity. J Pharmacol Exp Ther. 2003;306:287–300. doi: 10.1124/jpet.103.049601. [DOI] [PubMed] [Google Scholar]
  • 117.Desta Z, Saussele T, Ward B, Blievernicht J, Li L, Klein K, Flockhart DA, Zanger UM. Impact of CYP2B6 polymorphism on hepatic efavirenz metabolism in vitro. Pharmacogenomics. 2007;8:547–558. doi: 10.2217/14622416.8.6.547. [DOI] [PubMed] [Google Scholar]
  • 118.di Iulio J, Fayet A, Arab-Alameddine M, Rotger M, Lubomirov R, Cavassini M, Furrer H, Gunthard HF, Colombo S, Csajka C, et al. In vivo analysis of efavirenz metabolism in individuals with impaired CYP2A6 function. Pharmacogenet Genomics. 2009;19:300–309. doi: 10.1097/FPC.0b013e328328d577. [DOI] [PubMed] [Google Scholar]
  • 119.Elens L, Vandercam B, Yombi JC, Lison D, Wallemacq P, Haufroid V. Influence of host genetic factors on efavirenz plasma and intracellular pharmacokinetics in HIV-1-infected patients. Pharmacogenomics. 2010;11:1223–1234. doi: 10.2217/pgs.10.94. [DOI] [PubMed] [Google Scholar]
  • 120.Rotger M, Colombo S, Furrer H, Bleiber G, Buclin T, Lee BL, Keiser O, Biollaz J, Decosterd L, Telenti A. Influence of CYP2B6 polymorphism on plasma and intracellular concentrations and toxicity of efavirenz and nevirapine in HIV-infected patients. Pharmacogenet Genomics. 2005;15:1–5. doi: 10.1097/01213011-200501000-00001. [DOI] [PubMed] [Google Scholar]
  • 121.Arab-Alameddine M, Di Iulio J, Buclin T, Rotger M, Lubomirov R, Cavassini M, Fayet A, Decosterd LA, Eap CB, Biollaz J, et al. Pharmacogenetics-based population pharmacokinetic analysis of efavirenz in HIV-1-infected individuals. Clin Pharmacol Ther. 2009;85:485–494. doi: 10.1038/clpt.2008.271. [DOI] [PubMed] [Google Scholar]
  • 122.Kwara A, Lartey M, Sagoe KW, Rzek NL, Court MH. CYP2B6 (c.516G-->T) and CYP2A6 (*9B and/or *17) polymorphisms are independent predictors of efavirenz plasma concentrations in HIV-infected patients. Br J Clin Pharmacol. 2009;67:427–436. doi: 10.1111/j.1365-2125.2009.03368.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kwara A, Lartey M, Sagoe KW, Kenu E, Court MH. CYP2B6, CYP2A6 and UGT2B7 genetic polymorphisms are predictors of efavirenz mid-dose concentration in HIV-infected patients. AIDS. 2009;23:2101–2106. doi: 10.1097/QAD.0b013e3283319908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Pan-Zhou XR, Cretton-Scott E, Zhou XJ, Yang MX, Lasker JM, Sommadossi JP. Role of human liver P450s and cytochrome b5 in the reductive metabolism of 3'-azido-3'-deoxythymidine (AZT) to 3'-amino-3'-deoxythymidine. Biochem Pharmacol. 1998;55:757–766. doi: 10.1016/s0006-2952(97)00538-8. [DOI] [PubMed] [Google Scholar]
  • 125.Medhi B, Patyar S, Rao RS, Byrav DSP, Prakash A. Pharmacokinetic and toxicological profile of artemisinin compounds: an update. Pharmacology. 2009;84:323–332. doi: 10.1159/000252658. [DOI] [PubMed] [Google Scholar]
  • 126.Svensson US, Ashton M. Identification of the human cytochrome P450 enzymes involved in the in vitro metabolism of artemisinin. Br J Clin Pharmacol. 1999;48:528–535. doi: 10.1046/j.1365-2125.1999.00044.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Li XQ, Bjorkman A, Andersson TB, Gustafsson LL, Masimirembwa CM. Identification of human cytochrome P(450)s that metabolise anti-parasitic drugs and predictions of in vivo drug hepatic clearance from in vitro data. Eur J Clin Pharmacol. 2003;59:429–442. doi: 10.1007/s00228-003-0636-9. [DOI] [PubMed] [Google Scholar]
  • 128.Asimus S, Hai TN, Van Huong N, Ashton M. Artemisinin and CYP2A6 activity in healthy subjects. Eur J Clin Pharmacol. 2008;64:283–292. doi: 10.1007/s00228-007-0406-1. [DOI] [PubMed] [Google Scholar]
  • 129.Veiga MI, Asimus S, Ferreira PE, Martins JP, Cavaco I, Ribeiro V, Hai TN, Petzold MG, Bjorkman A, Ashton M, Gil JP. Pharmacogenomics of CYP2A6, CYP2B6, CYP2C19, CYP2D6, CYP3A4, CYP3A5 and MDR1 in Vietnam. Eur J Clin Pharmacol. 2009;65:355–363. doi: 10.1007/s00228-008-0573-8. [DOI] [PubMed] [Google Scholar]
  • 130.Elsherbiny DA, Asimus SA, Karlsson MO, Ashton M, Simonsson US. A model based assessment of the CYP2B6 and CYP2C19 inductive properties by artemisinin antimalarials: implications for combination regimens. J Pharmacokinet Pharmacodyn. 2008;35:203–217. doi: 10.1007/s10928-008-9084-6. [DOI] [PubMed] [Google Scholar]
  • 131.Simonsson US, Jansson B, Hai TN, Huong DX, Tybring G, Ashton M. Artemisinin autoinduction is caused by involvement of cytochrome P450 2B6 but not 2C9. Clin Pharmacol Ther. 2003;74:32–43. doi: 10.1016/S0009-9236(03)00092-4. [DOI] [PubMed] [Google Scholar]
  • 132.Chateauvieux S, Morceau F, Dicato M, Diederich M. Molecular and therapeutic potential and toxicity of valproic acid. J Biomed Biotechnol. 2010;2010 doi: 10.1155/2010/479364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Kiang TK, Ho PC, Anari MR, Tong V, Abbott FS, Chang TK. Contribution of CYP2C9, CYP2A6, and CYP2B6 to valproic acid metabolism in hepatic microsomes from individuals with the CYP2C9*1/*1 genotype. Toxicol Sci. 2006;94:261–271. doi: 10.1093/toxsci/kfl096. [DOI] [PubMed] [Google Scholar]
  • 134.Rettie AE, Rettenmeier AW, Howald WN, Baillie TA. Cytochrome P-450--catalyzed formation of delta 4-VPA, a toxic metabolite of valproic acid. Science. 1987;235:890–893. doi: 10.1126/science.3101178. [DOI] [PubMed] [Google Scholar]
  • 135.Kesterson JW, Granneman GR, Machinist JM. The hepatotoxicity of valproic acid and its metabolites in rats. I. Toxicologic, biochemical and histopathologic studies. Hepatology. 1984;4:1143–1152. doi: 10.1002/hep.1840040609. [DOI] [PubMed] [Google Scholar]
  • 136.Wen X, Wang JS, Kivisto KT, Neuvonen PJ, Backman JT. In vitro evaluation of valproic acid as an inhibitor of human cytochrome P450 isoforms: preferential inhibition of cytochrome P450 2C9 (CYP2C9) Br J Clin Pharmacol. 2001;52:547–553. doi: 10.1046/j.0306-5251.2001.01474.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Tan L, Yu JT, Sun YP, Ou JR, Song JH, Yu Y. The influence of cytochrome oxidase CYP2A6, CYP2B6, and CYP2C9 polymorphisms on the plasma concentrations of valproic acid in epileptic patients. Clin Neurol Neurosurg. 2010;112:320–323. doi: 10.1016/j.clineuro.2010.01.002. [DOI] [PubMed] [Google Scholar]
  • 138.Endo T, Ban M, Hirata K, Yamamoto A, Hara Y, Momose Y. Involvement of CYP2A6 in the formation of a novel metabolite, 3-hydroxypilocarpine, from pilocarpine in human liver microsomes. Drug Metab Dispos. 2007;35:476–483. doi: 10.1124/dmd.106.013425. [DOI] [PubMed] [Google Scholar]
  • 139.Endo T, Nakajima M, Fukami T, Hara Y, Hasunuma T, Yokoi T, Momose Y. Genetic polymorphisms of CYP2A6 affect the in-vivo pharmacokinetics of pilocarpine. Pharmacogenet Genomics. 2008;18:761–772. doi: 10.1097/FPC.0b013e328303c034. [DOI] [PubMed] [Google Scholar]
  • 140.Kasperska-Zajac A, Brzoza Z, Rogala B. Platelet-activating factor (PAF): a review of its role in asthma and clinical efficacy of PAF antagonists in the disease therapy. Recent Pat Inflamm Allergy Drug Discov. 2008;2:72–76. doi: 10.2174/187221308783399306. [DOI] [PubMed] [Google Scholar]
  • 141.Nunoya K, Yokoi Y, Kimura K, Kodama T, Funayama M, Inoue K, Nagashima K, Funae Y, Shimada N, Green C, Kamataki T. (+)-cis-3,5-dimethyl-2-(3-pyridyl) thiazolidin-4-one hydrochloride (SM-12502) as a novel substrate for cytochrome P450 2A6 in human liver microsomes. J Pharmacol Exp Ther. 1996;277:768–774. [PubMed] [Google Scholar]
  • 142.Nunoya KI, Yokoi T, Kimura K, Kainuma T, Satoh K, Kinoshita M, Kamataki T. A new CYP2A6 gene deletion responsible for the in vivo polymorphic metabolism of (+)-cis-3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one hydrochloride in humans. J Pharmacol Exp Ther. 1999;289:437–442. [PubMed] [Google Scholar]
  • 143.Komatsu T, Yamazaki H, Asahi S, Gillam EM, Guengerich FP, Nakajima M, Yokoi T. Formation of a dihydroxy metabolite of phenytoin in human liver microsomes/cytosol: roles of cytochromes P450 2C9, 2C19, and 3A4. Drug Metab Dispos. 2000;28:1361–1368. [PubMed] [Google Scholar]
  • 144.Thorn CF, Leckband SG, Kelsoe J, Steven Leeder J, Muller DJ, Klein TE, Altman RB. PharmGKB summary: carbamazepine pathway. Pharmacogenet Genomics. 2011 doi: 10.1097/FPC.0b013e328348c6f2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Pearce RE, Vakkalagadda GR, Leeder JS. Pathways of carbamazepine bioactivation in vitro I. Characterization of human cytochromes P450 responsible for the formation of 2- and 3-hydroxylated metabolites. Drug Metab Dispos. 2002;30:1170–1179. doi: 10.1124/dmd.30.11.1170. [DOI] [PubMed] [Google Scholar]
  • 146.Torchin CD, McNeilly PJ, Kapetanovic IM, Strong JM, Kupferberg HJ. Stereoselective metabolism of a new anticonvulsant drug candidate, losigamone, by human liver microsomes. Drug Metab Dispos. 1996;24:1002–1008. [PubMed] [Google Scholar]
  • 147.Kimber-Trojnar Z, Borowicz KK, Malek R, Sobieszek G, Piskorska B, Czuczwar SJ. Perspectives of losigamone in epilepsy treatment. Pol J Pharmacol. 2003;55:675–682. [PubMed] [Google Scholar]
  • 148.Restrepo JG, Garcia-Martin E, Martinez C, Agundez JA. Polymorphic drug metabolism in anaesthesia. Curr Drug Metab. 2009;10:236–246. doi: 10.2174/138920009787846305. [DOI] [PubMed] [Google Scholar]
  • 149.Rossini A, de Almeida Simao T, Albano RM, Pinto LF. CYP2A6 polymorphisms and risk for tobacco-related cancers. Pharmacogenomics. 2008;9:1737–1752. doi: 10.2217/14622416.9.11.1737. [DOI] [PubMed] [Google Scholar]
  • 150.Rotunno M, Yu K, Lubin JH, Consonni D, Pesatori AC, Goldstein AM, Goldin LR, Wacholder S, Welch R, Burdette L, et al. Phase I metabolic genes and risk of lung cancer: multiple polymorphisms and mRNA expression. PLoS One. 2009;4:e5652. doi: 10.1371/journal.pone.0005652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Tamaki Y, Arai T, Sugimura H, Sasaki T, Honda M, Muroi Y, Matsubara Y, Kanno S, Ishikawa M, Hirasawa N, Hiratsuka M. Association between Cancer Risk and Drug Metabolizing Enzyme Gene (CYP2A6, CYP2A13, CYP4B1, SULT1A1, GSTM1, and GSTT1) Polymorphisms in Japanese Cases of Lung Cancer. Drug Metab Pharmacokinet. 2011 doi: 10.2133/dmpk.dmpk-11-rg-046. [DOI] [PubMed] [Google Scholar]
  • 152.Fujieda M, Yamazaki H, Saito T, Kiyotani K, Gyamfi MA, Sakurai M, Dosaka-Akita H, Sawamura Y, Yokota J, Kunitoh H, Kamataki T. Evaluation of CYP2A6 genetic polymorphisms as determinants of smoking behavior and tobacco-related lung cancer risk in male Japanese smokers. Carcinogenesis. 2004;25:2451–2458. doi: 10.1093/carcin/bgh258. [DOI] [PubMed] [Google Scholar]
  • 153.Gemignani F, Landi S, Szeszenia-Dabrowska N, Zaridze D, Lissowska J, Rudnai P, Fabianova E, Mates D, Foretova L, Janout V, et al. Development of lung cancer before the age of 50: the role of xenobiotic metabolizing genes. Carcinogenesis. 2007;28:1287–1293. doi: 10.1093/carcin/bgm021. [DOI] [PubMed] [Google Scholar]
  • 154.Han GW, Craven BM. Crystal structure of cholestanyl caprylate and binary phase behavior with cholesteryl caprylate. J Lipid Res. 1991;32:1187–1194. [PubMed] [Google Scholar]
  • 155.Topcu Z, Chiba I, Fujieda M, Shibata T, Ariyoshi N, Yamazaki H, Sevgican F, Muthumala M, Kobayashi H, Kamataki T. CYP2A6 gene deletion reduces oral cancer risk in betel quid chewers in Sri Lanka. Carcinogenesis. 2002;23:595–598. doi: 10.1093/carcin/23.4.595. [DOI] [PubMed] [Google Scholar]
  • 156.Canova C, Hashibe M, Simonato L, Nelis M, Metspalu A, Lagiou P, Trichopoulos D, Ahrens W, Pigeot I, Merletti F, et al. Genetic associations of 115 polymorphisms with cancers of the upper aerodigestive tract across 10 European countries: the ARCAGE project. Cancer Res. 2009;69:2956–2965. doi: 10.1158/0008-5472.CAN-08-2604. [DOI] [PubMed] [Google Scholar]
  • 157.Kadlubar S, Anderson JP, Sweeney C, Gross MD, Lang NP, Kadlubar FF, Anderson KE. Phenotypic CYP2A6 variation and the risk of pancreatic cancer. JOP. 2009;10:263–270. [PMC free article] [PubMed] [Google Scholar]
  • 158.Mwenifumbo JC, Lessov-Schlaggar CN, Zhou Q, Krasnow RE, Swan GE, Benowitz NL, Tyndale RF. Identification of novel CYP2A6*1B variants: the CYP2A6*1B allele is associated with faster in vivo nicotine metabolism. Clin Pharmacol Ther. 2008;83:115–121. doi: 10.1038/sj.clpt.6100246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Fukami T, Nakajima M, Yamanaka H, Fukushima Y, McLeod HL, Yokoi T. A novel duplication type of CYP2A6 gene in African-American population. Drug Metab Dispos. 2007;35:515–520. doi: 10.1124/dmd.106.013557. [DOI] [PubMed] [Google Scholar]
  • 160.Thorgeirsson TE, Gudbjartsson DF, Surakka I, Vink JM, Amin N, Geller F, Sulem P, Rafnar T, Esko T, Walter S, et al. Sequence variants at CHRNB3-CHRNA6 and CYP2A6 affect smoking behavior. Nat Genet. 2010;42:448–453. doi: 10.1038/ng.573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Ho MK, Mwenifumbo JC, Al Koudsi N, Okuyemi KS, Ahluwalia JS, Benowitz NL, Tyndale RF. Association of nicotine metabolite ratio and CYP2A6 genotype with smoking cessation treatment in African-American light smokers. Clin Pharmacol Ther. 2009;85:635–643. doi: 10.1038/clpt.2009.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Kitagawa K, Kunugita N, Katoh T, Yang M, Kawamoto T. The significance of the homozygous CYP2A6 deletion on nicotine metabolism: a new genotyping method of CYP2A6 using a single PCR-RFLP. Biochem Biophys Res Commun. 1999;262:146–151. doi: 10.1006/bbrc.1999.1182. [DOI] [PubMed] [Google Scholar]
  • 163.Yamanaka H, Nakajima M, Nishimura K, Yoshida R, Fukami T, Katoh M, Yokoi T. Metabolic profile of nicotine in subjects whose CYP2A6 gene is deleted. Eur J Pharm Sci. 2004;22:419–425. doi: 10.1016/j.ejps.2004.04.012. [DOI] [PubMed] [Google Scholar]
  • 164.Yoshida R, Nakajima M, Watanabe Y, Kwon JT, Yokoi T. Genetic polymorphisms in human CYP2A6 gene causing impaired nicotine metabolism. Br J Clin Pharmacol. 2002;54:511–517. doi: 10.1046/j.1365-2125.2002.01667.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Ariyoshi N, Miyamoto M, Umetsu Y, Kunitoh H, Dosaka-Akita H, Sawamura Y, Yokota J, Nemoto N, Sato K, Kamataki T. Genetic polymorphism of CYP2A6 gene and tobacco-induced lung cancer risk in male smokers. Cancer Epidemiol Biomarkers Prev. 2002;11:890–894. [PubMed] [Google Scholar]
  • 166.Minematsu N, Nakamura H, Iwata M, Tateno H, Nakajima T, Takahashi S, Fujishima S, Yamaguchi K. Association of CYP2A6 deletion polymorphism with smoking habit and development of pulmonary emphysema. Thorax. 2003;58:623–628. doi: 10.1136/thorax.58.7.623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Ando M, Hamajima N, Ariyoshi N, Kamataki T, Matsuo K, Ohno Y. Association of CYP2A6 gene deletion with cigarette smoking status in Japanese adults. J Epidemiol. 2003;13:176–181. doi: 10.2188/jea.13.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Tan W, Chen GF, Xing DY, Song CY, Kadlubar FF, Lin DX. Frequency of CYP2A6 gene deletion and its relation to risk of lung and esophageal cancer in the Chinese population. Int J Cancer. 2001;95:96–101. doi: 10.1002/1097-0215(20010320)95:2<96::aid-ijc1017>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • 169.Zhang X, Amemo K, Ameno S, Iwahashi K, Kinoshita H, Kubota T, Mostofa J, Ijiri I. Lack of association between smoking and CYP2A6 gene polymorphisms in A Japanese population. Nihon Arukoru Yakubutsu Igakkai Zasshi. 2001;36:486–490. [PubMed] [Google Scholar]
  • 170.Ariyoshi N, Sawamura Y, Kamataki T. A novel single nucleotide polymorphism altering stability and activity of CYP2a6. Biochem Biophys Res Commun. 2001;281:810–814. doi: 10.1006/bbrc.2001.4422. [DOI] [PubMed] [Google Scholar]
  • 171.Al Koudsi N, Ahluwalia JS, Lin SK, Sellers EM, Tyndale RF. A novel CYP2A6 allele (CYP2A6*35) resulting in an amino-acid substitution (Asn438Tyr) is associated with lower CYP2A6 activity in vivo. Pharmacogenomics J. 2009;9:274–282. doi: 10.1038/tpj.2009.11. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES