ArticlePDF Available

Pharmacogenetics of cytochrome P450 2B6 (CYP2B6): advances on polymorphisms, mechanisms, and clinical relevance

Frontiers
Frontiers in Genetics
Authors:

Abstract and Figures

Cytochrome P450 2B6 (CYP2B6) belongs to the minor drug metabolizing P450s in human liver. Expression is highly variable both between individuals and within individuals, owing to non-genetic factors, genetic polymorphisms, inducibility, and irreversible inhibition by many compounds. Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. CYP2B6 is one of the most polymorphic CYP genes in humans and variants have been shown to affect transcriptional regulation, splicing, mRNA and protein expression, and catalytic activity. Some variants appear to affect several functional levels simultaneously, thus, combined in haplotypes, leading to complex interactions between substrate-dependent and -independent mechanisms. The most common functionally deficient allele is CYP2B6*6 [Q172H, K262R], which occurs at frequencies of 15 to over 60% in different populations. The allele leads to lower expression in liver due to erroneous splicing. Recent investigations suggest that the amino acid changes contribute complex substrate-dependent effects at the activity level, although data from recombinant systems used by different researchers are not well in agreement with each other. Another important variant, CYP2B6*18 [I328T], occurs predominantly in Africans (4–12%) and does not express functional protein. A large number of uncharacterized variants are currently emerging from different ethnicities in the course of the 1000 Genomes Project. The CYP2B6 polymorphism is clinically relevant for HIV-infected patients treated with the reverse transcriptase inhibitor efavirenz, but it is increasingly being recognized for other drug substrates. This review summarizes recent advances on the functional and clinical significance of CYP2B6 and its genetic polymorphism, with particular emphasis on the comparison of kinetic data obtained with different substrates for variants expressed in different recombinant expression systems.
This content is subject to copyright.
“fgene-04-00024” 2013/3/12 14:58 page1—#1
REVIEW ARTICLE
published: 05 March 2013
doi: 10.3389/fgene.2013.00024
Pharmacogenetics of cytochrome P450 2B6 (CYP2B6):
advances on polymorphisms, mechanisms, and clinical
relevance
Ulrich M. Zanger1,2* and Kathrin Klein1,2
1Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
2The University of Tuebingen,Tuebingen, Germany
Edited by:
José A. Agúndez, University of
Extremadura, Spain
Reviewed by:
Chin Eap, University of Lausanne
Medical School, Switzerland
Rajeev K. Mehlotra, Case Western
Reserve University, USA
*Correspondence:
Ulrich M. Zanger, Dr. Margarete
Fischer Bosch Institute of Clinical
Pharmacology, Auerbachstrasse 112,
70376 Stuttgart, Germany.
e-mail: uli.zanger@ikp-stuttgart.de
Cytochrome P450 2B6 (CYP2B6) belongs to the minor drug metabolizing P450s in human
liver. Expression is highly variable both between individuals and within individuals, owing
to non-genetic factors, genetic polymorphisms, inducibility, and irreversible inhibition by
many compounds. Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion,
cyclophosphamide, efavirenz, ketamine, and methadone. CYP2B6 is one of the most
polymorphic CYP genes in humans and variants have been shown to affect transcriptional
regulation, splicing, mRNA and protein expression, and catalytic activity. Some variants
appear to affect several functional levels simultaneously, thus, combined in haplotypes,
leading to complex interactions between substrate-dependent and -independent mech-
anisms. The most common functionally deficient allele is CYP2B6*6 [Q172H, K262R],
which occurs at frequencies of 15 to over 60% in different populations. The allele leads
to lower expression in liver due to erroneous splicing. Recent investigations suggest that
the amino acid changes contribute complex substrate-dependent effects at the activity
level, although data from recombinant systems used by different researchers are not
well in agreement with each other. Another important variant, CYP2B6*18 [I328T], occurs
predominantly in Africans (4–12%) and does not express functional protein. A large number
of uncharacterized variants are currently emerging from different ethnicities in the course of
the 1000 Genomes Project.The CYP2B6 polymorphism is clinically relevant for HIV-infected
patients treated with the reverse transcriptase inhibitor efavirenz, but it is increasingly
being recognized for other drug substrates.This review summarizes recent advances on the
functional and clinical significance of CYP2B6 and its genetic polymorphism, with particular
emphasis on the comparison of kinetic data obtained with different substrates for variants
expressed in different recombinant expression systems.
Keywords: bupropion, cyclophosphamide, cytochrome P450, drug metabolism, drug–drug interaction, efavirenz,
pharmacogenetics, pharmacogenomics
INTRODUCTION
The cytochrome P450 (CYP) enzyme CYP2B6 is one of about a
dozen human CYPs that are primarily involved in the biotrans-
formation of drugs and other xenobiotics. The CYP2B6 gene and
its closely related pseudogene, CYP2B7, are located in a tandem
head-to-tail arrangement within a large CYP2 gene cluster on the
long arm of chromosome 19 (Hoffman et al., 2001;Figure 1). The
orthologous genes in dog, mouse, and rat are termed CYP2B11,
Cyp2b10, and CYP2B1, respectively, but in contrast to other mam-
malian species, CYP2B6 is the only functional isozyme of its
subfamily in humans (Nelson et al., 2004). Owing to the existence
of extensive genetic polymorphism as well as strong inhibitors and
inducers, its activity is highly variable in the population. For some
clinically used drugs including the antiretroviral agents efavirenz
and nevirapine, CYP2B6 single nucleotide polymorphisms have
been shown to be useful predictors of pharmacokinetics and drug
response (reviewed in Zanger et al., 2007;Telenti and Zanger,2008;
Rakhmanina and van den Anker,2010 ). However,recent data indi-
cate that pharmacogenetic mechanisms are complex, appear on
several levels of gene expression from the initial mRNA transcript
to splice variants (pre-mRNA splicing and mRNA expression) to
altered proteins, and affect function in various ways including
substrate-dependent and substrate-independent effects. Several
previous reviews are available that cover the biochemical pharma-
cology, molecular genetics, and pharmacogenetics of this enzyme
at various degrees of detail (Ekins and Wrighton, 1999;Turpeinen
et al., 2006;Hodgson and Rose, 2007;Zanger et al., 2007;Wang
and Tompkins, 2008;Mo et al., 2009;Turpeinen and Zanger, 2012).
The purpose of this review is to summarize recent advances in areas
that have an impact on variable expression of CYP2B6 and the
mechanisms and impact of CYP2B6 polymorphism, as observed
by various in vitro approaches as well as in in vivo studies, and to
discuss their functional and clinical implications.
VARIABILITY OF EXPRESSION AND TRANSCRIPTIONAL
REGULATION
Cytochrome P450 2B6 is primarily expressed in the liver where its
contribution to the total microsomal P450 pool has been estimated
www.frontiersin.org March 2013 |Volume 4 |Article 24 |1
“fgene-04-00024” 2013/3/12 14:58 page2—#2
Zanger and Klein Pharmacogenetics of CYP2B6
FIGURE 1 |Structure of genomic regions on chromosome 19.
Schematic representation of the CYP2ABFGST-cluster on chromosome
19 (A) and of the CYP2B6 gene (B). Boxes in the (A) represent genes,
with direction of transcription indicated by arrows.The (B) represents
CYP2B6 exons and introns and shows the most important SNPs as
triangles.
to be within a range of about 1–10%, with a large inter-individual
variability at protein level of roughly 100-fold (see Zanger et al.,
2007 for review and references therein). Although some earlier
studies reported expression in only a fraction of human liv-
ers, newer studies with better antibodies found CYP2B6 to be
present in all investigated human adult liver samples (Hofmann
et al., 2008) while up to one-third of pediatric samples contained
no detectable protein (Croom et al., 2009). In the latter study,
ontogenic differences were studied in liver microsomes from 217
pediatric liver donors. Hepatic median CYP2B6 protein levels were
about twofold higher in the period between birth and 30 days post-
natal compared to fetal samples, and protein levels varied already
over 25-fold in both of these age groups (Croom et al., 2009). Mat-
uration effects may further depend on genotype, as suggested in
a study on HIV-infected children treated with efavirenz (Sueyoshi
et al., 1999;Wang et al., 2003;Faucette et al., 2004,2007).
One of the most important factors contributing to intra- as
well as inter-individual variability is enzyme induction, i.e., de
novo protein synthesis following exposure to certain chemicals.
Regulation of CYP2B gene expression represents the archetypal
example of enzyme induction (Remmer et al., 1973). Human
CYP2B6 is strongly inducible by several drugs including classi-
cal” inducers such as rifampicin, phenytoin, and phenobarbital
involving a so-called phenobarbital-responsive enhancer module
(PBREM) at 1.7 kb of the CYP2B6 gene promoter, and a dis-
tal xenobiotics-responsive enhancer module (XREM, 8.5 kb),
to which pregnane X receptor (PXR, NR1I2) and/or constitutive
androstane receptor (CAR,NR1I3) bind to mediate increased tran-
scription (Sueyoshi et al., 1999;Wang et al., 2003;Faucette et al.,
2004,2007). Since other CYPs are regulated by overlapping sets
of nuclear receptors, CYP2B6 is often co-induced with CYP2C
enzymes and CYP3A4. CYP2B6 inducers identified to date include
cyclophosphamide (Gervot et al., 1999), hyperforin (Goodwin
et al., 2001), artemisinin antimalarials (Simonsson et al., 2003;
Burk et al., 2005), carbamazepine (Oscarson et al., 2006;Desta
et al., 2007), metamizole (Saussele et al., 2007;Qin et al., 2012),
ritonavir (Kharasch et al., 2008), the insect repellent N,N-diethyl-
m-toluamide (DEET; Das et al., 2008), statins (Feidt et al., 2010),
efavirenz (Ngaimisi et al., 2010;Habtewold et al., 2011). Interest-
ingly, in the latter study, gender influenced the inducibility of
efavirenz 8-hydroxylation, which was higher in women than in the
men (Ngaimisi et al., 2010). In addition to therapeutic drugs, pes-
ticides were found to be powerful inducers of CYP2B6 and other
CYPs through interaction with both PXR and CAR (Das et al.,
2008). Induction of CYP2B6 and other cytochromes P450 and its
clinical consequences has been reviewed by others (Pelkonen et al.,
2008;Mo et al., 2009).
Sex differences in liver expression have been observed in a
number of studies. Females liver donors had higher amounts of
CYP2B6 mRNA (3.9-fold), protein (1.7-fold), and enzyme activity
(1.6-fold) compared to male subjects in a study of 80 ethnically
mixed samples (Lamba et al., 2003). In a study with 235 Cau-
casian liver donors, female samples had 1.6-fold higher expression
level of CYP2B6 mRNA, however, this difference did not trans-
late into higher protein and activity levels and no sex difference
was found when only liver donors without presurgical drug expo-
sure were considered (Hofmann et al., 2008). Discrepant effects
of sex on pharmacokinetics of CYP2B6 substrates, which may be
due to other confounders such as age or smoking status, were
also found in vivo. Higher bupropion hydroxylation rates were
found in adolescent females compared to males (Stewart et al.,
2001) but not in adults (Hsyu et al., 1997). For efavirenz, sev-
eral studies reported elevated plasma concentrations in female
compared to male patients, which is in contrast to the above-
mentioned in vitro findings and may be explained by other factors
such as differences in body fat content and distribution (Burger
et al., 2006;Nyakutira et al., 2008;Mukonzo et al., 2009). The influ-
ence of age on CYP2B6 expression may also depend on sex, as only
males showed a significant increase of liver CYP2B6 at higher age
(Yang et al., 2010).
Frontiers in Genetics |Pharmacogenetics and Pharmacogenomics March 2013 |Volume 4 |Article 24 |2
“fgene-04-00024” 2013/3/12 14:58 page3—#3
Zanger and Klein Pharmacogenetics of CYP2B6
Besides liver, CYP2B6 is also consistently expressed in different
parts of respiratory and gastrointestinal tracts, including lung and
nasal mucosa, and also in skin and the kidneys (Choudhary et al.,
2003;Dutheil et al., 2008;Thelen and Dressman, 2009;Leclerc
et al., 2010). The significance of CYP2B6 in these extrahepatic tis-
sues is currently unknown, but it should be remembered that the
enzyme is probably the most important one for many environ-
mental toxins such as pesticides, and its presence in tissues with
barrier function may thus contribute substantially to protection
against these chemicals. In addition, the presence of CYP2B6 in
brain has been demonstrated in human and primate brain tissue
samples and smoking, alcohol consumption, and genetic poly-
morphism have been suggested to contribute to its variability in
this organ (Miksys et al., 2003). In general, CYP levels in extra-
hepatic tissues are far below those of liver, but the localization
to specific regions in the brain may contribute to the activation
or inactivation of centrally acting drugs and to neurological side
effects of certain medications or abused drugs, e.g., “ecstasy” [1-
methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), see below].
This may also explain why efficacy for some centrally acting drugs
is not well correlated to their plasma levels. The potential role of
brain-expressed CYPs including CYP2B6 in the biotransformation
of centrally acting drugs has been reviewed by others (Meyer et al.,
2007;Ferguson and Tyndale, 2011).
THE CHEMICAL INTERACTION PROFILE OF CYP2B6
Recent studies have revealed crystal structures of the CYP2B6
wild-type and K262R variant in complex with various inhibitors
at providing first views into its active site and its plasticity to
adopt different conformations when binding different ligands
(Gay et al., 2010;Shah et al., 2011;Wilderman and Halpert,
2012). Substrates of CYP2B6 are usually fairly lipophilic, neu-
tral or weakly basic non-planar molecules with one or two
hydrogen bond acceptors (Lewis et al., 1999,2004). The CYP2B6
substrate selectivity comprises many diverse chemicals, includ-
ing not only clinically used drugs but also many environmental
chemicals such as pesticides (Turpeinen et al., 2006;Hodgson
and Rose, 2007;Turpeinen and Zanger, 2012). Therapeutically
important drugs metabolized primarily by CYP2B6 include the
prodrug cyclophosphamide, which is converted to the direct
precursor of the cytotoxic metabolites, phosphoramide mus-
tard and acrolein, by 4-hydroxylation (Huang et al., 2000;Roy
et al., 2005), the non-nucleoside reverse transcriptase inhibitor
(NNRTI), efavirenz, which is 8-hydroxylated to become phar-
macologically inactive (Ward et al., 2003;Desta et al., 2007), the
atypical antidepressant and smoking cessation agent bupropion,
which is converted to pharmacologically active hydroxybupro-
pion (Faucette et al., 2000;Hesse et al., 2000;Turpeinen et al.,
2005b), the anesthetics propofol (Court et al., 2001;Oda et al.,
2001) and ketamine (Desta et al., 2012), the analgesic pethidine
(meperidine; Ramírez et al., 2004); the μ-opioid receptor ago-
nist, methadone (Totah et al., 2008), the antimalarial artemisinin
(Svensson and Ashton, 1999;Asimus and Ashton, 2009), among
numerous additional metabolic pathways of other drugs, to which
CYP2B6 contributes in part, such as the antiretroviral, nevirapine
(Erickson et al., 1999), and many others (Turpeinen and Zanger,
2012). Metabolic pathways suitable as probe for CYP2B6 activity
include S-mephenytoin N-demethylation (Ko et al., 1998), bupro-
pion hydroxylation (Faucette et al., 2000;Fuhr et al., 2007) and
efavirenz, based on in vitro investigations (Ward et al., 2003;Desta
et al., 2007).
Endogenous substances metabolized by the enzyme include
arachidonic acid, lauric acid, 17beta-estradiol, estrone, ethiny-
lestradiol, and testosterone 16α- and 16β-hydroxylation (Ekins
et al., 1998).
Cytochrome P450 2B6 furthermore participates in the
biotransformation of the abused drug “ecstasy” (N-methyl-
3,4-methylenedioxymethamphetamine, MDMA), which is N-
demethylated leading to potentially neurotoxic metabolites (Kreth
et al., 2000). It also plays a minor role in nicotine metabolism
(Yamazaki et al., 1999;Yamanaka et al., 2005). CYP2B6 has fur ther-
more been found to be of importance in the metabolism of pesti-
cides and other environmental chemicals and pollutants (Hodgson
and Rose, 2007). In particular the bioactivating oxidation of the
organophosphorus insecticides chlorpyrifos (Crane et al., 2012)
and methyl parathion (Ellison et al., 2012a) to their more toxic
oxon metabolites is mainly catalyzed by CYP2B6, a public health
concern due to their worldwide use and documented human
exposures (Ellison et al., 2012b). Further environmental sub-
strates are the insecticide and endocrine disruptor methoxychlor,
the extensively used insect repellent N,N-diethyl-m-toluamide
(Das et al., 2008), profenofos and other pesticides (Abass and
Pelkonen, 2012), as well as the tobacco-specific nitrosamine,
4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK; Smith
et al., 2003), aflatoxin B1 (Code et al., 1997), and others (Hodgson
and Rose, 2007;Abass and Pelkonen, 2012).
Several structurally unrelated drugs have been shown to inhibit
CYP2B6 and many of them do that in a mechanism-based, irre-
versible manner (Turpeinen et al., 2006;Turpeinen and Zanger,
2012). The thienopyridine derivatives clopidogrel and ticlopi-
dine are prodrugs that selectively inhibit platelet aggregation and
have been in clinical use for the prevention of atherothrombotic
events for several years. Both of them are potent mechanism-
based inhibitors of CYP2B6 (Richter et al., 2004;Zhang et al.,
2011a). The established anticancer agent, thioTEPA (N,N,N-
triethylenethiophosphoramide) was also found to be a highly
selective and mechanism-based CYP2B6 inhibitor (Rae et al.,
2002;Harleton et al., 2004;Richter et al., 2005). A compar-
ison of several selective inhibitors revealed that 2-phenyl-2-
(1-piperidinyl)propane is probably the most selective CYP2B6
inhibitor in vitro (Walsky and Obach, 2007). Recent in vitro obser-
vations identified the progesterone receptor antagonist, mifepris-
tone (RU486; Lin et al., 2009); the anti-Parkinsonian agent selegi-
line (the R-enantiomer of deprenyl; Sridar et al., 2012), methadone
(Amunugama et al., 2012), and tamoxifen (Sridar et al., 2012)
as potent mechanism-based inhibitors. In vivo drug–drug inter-
actions have been reported, for example, between thioTEPA
and cyclophosphamide (Huitema et al., 2000), clopidogrel and
bupropion (Turpeinen et al., 2005a), voriconazole and efavirenz
(Liu et al., 2008;Jeong et al., 2009), clopidogrel and efavirenz
(Jiang et al., 2012), and between ticlopidine and ketamine (Pel-
toniemi et al., 2011). Furthermore, certain non-pharmaceutical
compounds like particular benzylpyridine derivatives have been
characterized as very potent inhibitors of CYP2B6 (Korhonen
www.frontiersin.org March 2013 |Volume 4 |Article 24 |3
“fgene-04-00024” 2013/3/12 14:58 page4—#4
Zanger and Klein Pharmacogenetics of CYP2B6
et al., 2007) and have been utilized for structural modeling
experiments (Gay et al., 2010).
PHARMACOGENETICS OF CYP2B6
The CYPalleles website1currently lists 37 distinct star-alleles, i.e.,
gene haplotypes with a distinct variant amino acid sequence or
with demonstrated functional effect (last accessed: February 21st,
2013). More than 30 amino acid-changing single-nucleotide poly-
morphisms (SNPs) occur in different combinations and together
with additional non-coding variants and many more SNPs not
yet assigned to particular haplotypes. The worldwide variations
in SNP frequencies have been reviewed recently (Li et al., 2012).
Tab l e 1 lists the most important variants in terms of frequency
and functional impact and summarizes updated structural, func-
tional, and frequency information for different ethnicities. In
addition to the CYPallele website, further valuable information
about CYP2B6 SNPs and pharmacogenetics are available on the
websites of The Pharmacogenomics Knowledgebase2, the NCBI por-
tal for short genetic variations, dbSNP3, the 1000 Genomes Catalog of
Human Genetic Variation4, as well as the NHLBI exome sequencing
project5.
1http://www.cypalleles.ki.se/cyp2b6.htm
2http://www.pharmgkb.org/
3http://www.ncbi.nlm.nih.gov/projects/SNP/
4http://www.1000genomes.org/
5http://EVS.gs.washington.edu
CYP2B6*6 AND EFAVIRENZ: IN VIVO,EX VIVO,IN VITRO
The most common variant allele in all populations studied to
date harbors two amino acid changes, Q172H and K262R, and
is termed CYP2B6*6. This haplotype occurs in about 15 to over
60% of individuals, depending on ethnicity (Ta bl e 1 ). Although
additional variants occur in the promoter and in introns, their
functional impact appears to be of limited relevance and will not
be further discussed here (Lamba et al., 2003;Hesse et al., 2004;
Hofmann et al., 2008).
Since the discovery that CYP2B6 is the major enzyme for
efavirenz 8-hydroxylation (Ward et al., 2003), pharmacogenetic
studies have linked the Q172H variant to elevated plasma concen-
trations of efavirenz, indicating decreased enzyme function in vivo.
This finding has been reproduced manifold in different ethnicities
throughout the world (summarized by Telenti and Zanger, 2008;
Rakhmanina and van den Anker, 2010). Three CYP2B6 polymor-
phisms, 15631G>T, 21011T>C, and an intron 3 SNP rs4803419,
were also shown to be associated with efavirenz pharmacokinetics
at genome wide significance (Holzinger et al., 2012).
The potent first-generation NNRTI of HIV-1 is recommended
as initial therapy with two NRTIs in highly active antiretrovi-
ral therapy (HAART) regimes, but patients with subtherapeutic
plasma concentrations can develop resistance and treatment fail-
ure, whereas those with too high plasma levels are at increased
risk of central nervous system (CNS) side effects, which can
lead to treatment discontinuation in a fraction of patients (King
and Aberg, 2008). Q172H variant was furthermore associated
Table 1 |Summary data on selected genetic polymorphisms of CYP2B6.
CYP allele
designationa
Key mutation(s)b
rs number
Location, protein effect Allele frequenciescFunctional effect
CYP2B6*4 g.18053(c.516) A>G
rs2279343
K262R (isolated) 0.00 AA, Af
0.04 Ca
0.05–0.12 As
Expression, moderate substrate-
dependent effects
CYP2B6*5 g.25505(c.1459) C>T
rs3211371
R487C 0.01–0.04AA, Af
0.09–0.12 Ca
0.05–0.12 Hs
0.01–0.04 As
Expression, in part compensated
by specific activity
CYP2B6*6 g.15631(c.516) G>T
rs3745274 and
g.18053(c.785)A>G
rs2279343
Q172HK262R 0.33–0.5 AA, Af
0.10–0.21 As
0.14–0.27 Ca
0.62 PNG
Expression; activity with
efavirenz in vivo; some other
substrates show activity
CYP2B6*18 g.21011(c.983)T>C
rs28399499
I328T 0.04–0.08 AA
0.05–0.12, Af
0.01 HS
0.00 As, Ca, PNG
Expression and activity
CYP2B6*22 g.-82T>C
rs34223104
promoter (TATA-box) 0.00–0.025 AA, Af, As
0.024 Ca, Hs
Expression and activity
Inducibility in vitro
aAccording to CYPallele nomenclature homepage http://www.cypalleles.ki.se.
bGenomic (g.) and cDNA (c.) positions are given in bp.
cSelected frequencies of individual ethnicities (AA, African American; Af African; As Asian; Ca Caucasian; Hs, Hispanic; PNG, Papua New Guineans) compiled from
dbSNP http://www.ncbi.nlm.nih.gov/SNP and from the literature cited in the text.
Frontiers in Genetics |Pharmacogenetics and Pharmacogenomics March 2013 |Volume 4 |Article 24 |4
“fgene-04-00024” 2013/3/12 14:58 page5—#5
Zanger and Klein Pharmacogenetics of CYP2B6
with increased neurotoxicity and other CNS side effects (Haas
et al., 2004;King and Aberg, 2008;Lubomirov et al., 2010;Rib-
audo et al., 2010;Maimbo et al., 2011) with HAART-induced
liver injury (Yimer et al., 2011), and with efavirenz treatment
discontinuation and the associated risk of developing drug
resistance (Ribaudo et al., 2006;Lubomirov et al., 2011;Wye n
et al., 2011). Importantly, compound heterozygotes of 516T and
another low activity allele (e.g., *11,*18,*27,*28) also pre-
dict high efavirenz plasma levels (Rotger et al., 2007;Ribaudo
et al., 2010). In prospective, genotype-based dose adjustment
studies the therapeutic dose of efavirenz could be successfully
reduced and CNS-related side effects decreased (Gatanaga et al.,
2007;Gatanaga and Oka, 2009). Using pharmacokinetic modeling
and simulation it was suggested that a priori dose reduction in
homozygous CYP2B6*6 patients would maintain drug exposure
within the therapeutic range in this group of patients (Nyakutira
et al., 2008).
The in vitro data that have accumulated over the years on the
CYP2B6*6 allele draw a more complex picture with functional
consequences on various levels including pre-mRNA splicing, pro-
tein expression, as well as substrate-dependent changes in enzyme
activity and different sensitivity toward irreversible inhibition.
While early studies using recombinantly expressed enzyme vari-
ants found higher 7-ethoxycoumarin O-deethylase activity for
the Q172H variant (Ariyoshi et al., 2001;Jinno et al., 2003), in
genotyped human livers (ex vivo), the *6 allele has been associ-
ated with approximately 50–75% decreased protein levels (Lang
et al., 2001;Desta et al., 2007;Hofmann et al., 2008). An expla-
nation for decreased protein expression was provided based on
the observation that the c.516G>T SNP coding for Q172H in
exon 4 (rs3745274, Tab le 1 ) was correlated to increased amounts
of a hepatic splice variant that lacked exons 4–6, and concur-
rently to decreased amounts of the normal functional transcript.
Recombinant expression of minigene constructs in mammalian
cells proved that the c.516G>T variant was causally involved in
erroneous splicing and lower expression of functional mRNA and
protein (Hofmann et al., 2008). It has been hypothesized that bind-
ing of splice factors to an exonic splicing enhancer(s) located in
exon 4 could be affected by the variant (Zanger and Hofmann,
2008;Sadee et al., 2011). Although reduced expression in liver
appears to satisfactorily explain increased efavirenz plasma con-
centrations in individuals with *6/*6 genotype (Desta et al., 2007),
recent in vitro data of expressed variants seem to indicate that
the amino acid substitutions contribute to changes in catalytic
activity of the enzyme. Structurally this is not easy to compre-
hend, because the Q172H and Lys262Arg amino acid changes
occur in regions of the protein that are not directly located at the
active site or that have been identified as substrate recognition sites
(Figure 2).
Concerning efavirenz and also other substrates, the avail-
able in vitro data are however, not well in agreement with each
other. Tab l e 2 summarizes kinetic parameters for bupropion and
efavirenz for CYP2B6 enzyme variants obtained from different
recombinant expression systems. Using Escherichia coli expression
system, Zhang et al. (2011b) purified six N-terminally truncated
expressed variants to homogeneity and reconstituted them with
NADPH:cytochrome 450 reductase (POR) at a molar ratio of 1:2
FIGURE 2 |CYP2B6 structural model. Selected variant residues are
marked in green (Q172H, K262R, I328T, R487C); substrate recognition sites
(SRS; Nguyen etal., 20 08) and active site residues (ASR; Niu et al., 2011)
are highlighted in blue; the prosthetic heme molecule is shown in red. PDB
file 3IBD was visualized using VMD visual molecular dynamics viewer
http://www.ks.uiuc.edu/Research/vmd/ (Humphrey etal., 1996). The protein
sequence used for determining the crystal structure contains R at position
262 instead of the reference residue K. (A) view along x-axis (light red); (B)
view along y-axis (light green); (C) view along z-axis (light blue)
www.frontiersin.org March 2013 |Volume 4 |Article 24 |5
“fgene-04-00024” 2013/3/12 14:58 page6—#6
Zanger and Klein Pharmacogenetics of CYP2B6
Table 2 |Kinetic properties of recombinantly expressed CYP2B6 protein variants with bupropion and efavirenz.
Variant System Bupropion hydroxylation Efavirenz 8-hydroxylation Reference
Km(μM) Vmax (%) CLint (%) Km(μM) Vmax (%) CLint (%)
2B6.1 COS-1 87 100 100 2.07 100 100 Radloff etal. (2013)
E. coli 95 100 100 7.3 100 100 Zhang etal. (2011b)
Sf9 7.7 100 100 Ariyoshi etal. (2011)
Sf9 64 100 100 3.2 100 100 Xu et al. (2012)
2B6.6 COS-1 72 81 98 1.21 107 183 Radloff et al. (2013)
E. coli 380 175 43 198 563 20 Zhang et al. (2011b)
Sf9 12.4 81 50 Ariyoshi etal. (2011)
Sf9 63 139 143 8.8 133 49 Xu et al. (2012)
2B6.4 E. coli 162 60 35 5.5 73 96 Zhang et al. (2011b)
Sf9 9.16 169 142 Ariyoshi etal. (2011)
2B6.5 COS-1 65 44 59 1.15 46 83 Radloff etal. (2013)
E. coli 134 66 47 53 100 5 138 Zhang et al. (2011b)
Only studies which determined kinetic parameters (Km,V
max,orK
cat) were included.
and measured efavirenz and bupropion kinetics. Using Sf9 insect
cell cotransfection, CYP2B6.1, 2B6.4 and 2B6.6 were expressed in
the presence of 10-fold excess of POR, i.e., under saturating condi-
tions, to measure efavirenz kinetics (Ariyoshi et al., 2011). Another
study determined both bupropion and efavirenz kinetics in protein
preparations also derived from insect cells in the presence or
absence of cytochrome b5 (CYB5) but at somewhat more vari-
able ratios in regard to POR (Xu et al., 2012). Radloff et al. (2013)
used the COS-1 expression system, where P450 monooxygenase
activity is supported by endogenously expressed POR, to deter-
mine bupropion and efavirenz kinetics for several novel CYP2B6
variants in comparison to the known variants 2B6.1, 2B6.5, and
2B6.6.
The compilation of data in Ta b l e 2 shows that differences
between the variants were masked by differences between the
expression systems. For example, efavirenz Kmwas moderately
decreased (58%) for COS-1 cell-expressed 2B6.6 compared to
2B6.1 but moderately larger for both insect cell-expressed proteins.
The E. coli-expressed variant showed, however, 27-fold increased
Km. While the COS-1 proteins had almost identical Vmax ,oneof
the insect cellproteins had decreased Vmax (81%), while the other
had increased activity (133%). Again, the E. coli-expressed variant
showed the biggest difference of almost sixfold higher activity for
the variant. Similar discrepancies, albeit less dramatic, were found
with bupropion as substrate (Ta b l e 2).
This data-comparison illustrates the problems that still exist
with recombinant P450 expression systems, and particularly for
CYP2B6, which appears to be an enzyme that sensibly reacts
with activity changes to expression conditions. It is difficult to
pin down the reasons for these differences exactly. Reconstitution
of recombinant or even purified P450 with POR and CYB5 is a
non-trivial problem especially if different protein variants shall
be compared for quantitative kinetic parameters. Reconstitution
under saturating conditions with respect to electron donators, e.g.,
at a POR:P450 ratio of 10, is a straightforward practical way,
but in hepatocytes, POR is stoichiometrically underrepresented
(ratio about 1:10) and may be limiting for monooxygenase activity
(Gomes et al., 2009). Enzyme variants may interact differently with
the electron donors and catalytic differences could thus depend on
reconstitution conditions. In addition, N-terminal modifications
required to achieve high expression in E. coli may interact with
the DNA-polymorphisms to be analyzed. In the COS-cell system,
on the other hand, the POR:P450 ratio can neither be controlled
nor quantified because expression of P450 is too low for spectral
quantitation.
Taken together, the data from expression systems indicated that
catalytic differences may exist between CYP2B6.6 and CYP2B6.1.
However, except for the E. coli study, the differences were rather
modest and at present it cannot be concluded with certainty
whether the CYP2B6.6 variant is catalytically more or less active
compared to the wild-type, atleast for bupropion and efavirenz.
Taken all evidence together, the decrease in hepatic expression due
to erroneous splicing caused by the c.516G>T SNP (Desta et al.,
2007;Hofmann et al., 2008) most plausibly explains most of the
phenotypic in vivo activity differences observed with efavirenz and
bupropion.
CYP2B6*6 SNP-related functional differences were also
observed with inhibitors. In contrast to the wild-type enzyme
the recombinantly expressed K262R variant was not inactivated
by efavirenz, but both enzymes were irreversibly inhibited by 8-
hydroxyefavirenz (Bumpus et al., 2006;Bumpus and Hollenberg,
2008). Lower susceptibility to inhibition of the K262R variant and
the CYP2B6.6 double variant compared to CYP2B6.1 was also
found with respect to sertraline and clopidogrel, as well as sev-
eral other potent drug inhibitors of CYP2B6 (Talakad et al., 2009).
These data indicate a role of genetic polymorphisms in drug–drug
interaction sensitivity of CYP2B6, a finding that warrants further
investigation in vivo.
Frontiers in Genetics |Pharmacogenetics and Pharmacogenomics March 2013 |Volume 4 |Article 24 |6
“fgene-04-00024” 2013/3/12 14:58 page7—#7
Zanger and Klein Pharmacogenetics of CYP2B6
OTHER CYP2B6 VARIANTS AND OTHER SUBSTRATES
IN VITRO STUDIES
The two amino acid changes that together constitute the *6 allele
also occur in isolation, although at much lower frequencies (Klein
et al., 2005;Zanger et al., 2007;Tab l e 1 ). In most pharmacoge-
netic studies they are not being determined and functional data
is therefore rare, especially concerning *9 (Klein et al., 2005).
Data from recombinant systems as well as liver data suggest
that the K262R variant possesses catalytic activities similar to
the wild-type enzyme, although with different substrates mod-
erately increased or decreased activity was observed (Tables 2 and
3;Desta et al., 2007;Hofmann et al., 2008). The allele with amino
acid change [R487C] in exon 9 (*5 variant; Tab le 1 ) expresses
very low levels of protein which does not translate into simi-
larly reduced activities as measured with bupropion as well as
efavirenz (Lang et al., 2001;Desta et al., 2007;Hofmann et al.,
2008). The variant was shown to have higher specific activity
compared to wild-type (Radloff et al., 2013). In human liver, this
leads to partial compensation of low expression, finally resulting
in a phenotype with moderately decreased activity with bupropion
and efavirenz in vivo. This explains why CYP2B6*5 was not asso-
ciated with efavirenz pharmacokinetics in HIV patients (Burger
et al., 2006).
The second most important functionally deficient allele is
CYP2B6*18 (c.983C>T [I328T]), which occurs predominantly
in African subjects with allele frequencies of 4–11% (Mehlo-
tra et al., 2007;Li et al., 2012). The I328T variant expressed
no detectable protein or activity toward bupropion, 7-ethoxy-4-
trifluoromethylcoumarin (7-EFC), selegiline and artemether in
COS-1 cells whereas a partially defective protein was expressed
in insect cells (Klein et al., 2005;Watanabe et al., 2010;Honda
et al., 2011). This demonstrates another example for expression
system-dependent differences. Most likely the 2B6.18 variant is
temperature-sensitive and thus able to be expressed at the lower
temperature (27C) of insect cell culture but not at 37C. Inter-
estingly, the I328T+Q172H double variant expressed partially
functional protein in HEK293 cells and in yeast (Wang et al.,
2006), indicating that Q172H can stabilize the I328T variant.
The *18 allele is thus phenotypically a null allele, at least in vitro
with some substrates. This is supported by many in vivo studies
(see below).
At least 12 additional null or low-activity alleles have been
described and analyzed with various substrates (Lang et al., 2004;
Klein et al., 2005;Rotger et al., 2007;Watanabe et al., 2010;Honda
et al., 2011). Although they are rather rare in all investigated pop-
ulations they may have profound effects on drug metabolism if
present in compound heterozygous genotypes, e.g., in combina-
tion with *6 or *18 (Rotger et al., 2007). The CYP2B6*22 allele
is a gain-of-function variant associated with increased transcrip-
tion in vitro (Zukunft et al., 2005) and with increased activity in
vivo (Rotger et al., 2007). It was shown that a -82T>C exchange
alters the TATA-box into a functional CCAAT/enhancer-binding
protein binding site that causes increased transcription from an
alternative downstream initiation site (Zukunft et al., 2005). Inter-
estingly, the -82T>C polymorphism also confers synergistically
enhanced CYP2B6 inducibility by the PXR ligand rifampicin in
human primary hepatocytes (Li et al., 2010).
New variants are discoveredpreferentially in previously unchar-
acterized ethnic groups. Restrepo et al. (2011) described two novel
combinations of known amino acid variants in a Colombian
population. Structural variants including a novel CYP2B6/2B7P1
duplicated fusion allele (CYP2B6*30) were found when individ-
uals from various ethnicities were screened for copy number
variations (Martis et al., 2012). Furthermore, three novel and
five previously uncharacterized amino acid variants in different
combinations (CYP2B6*33 to *37) were identified by resequenc-
ing the CYP2B6 gene in a Rwandese cohort of HIV-1-infected
patients (Radloff et al., 2013). The variants were then function-
ally studied by COS-1 cell expression and by in silico prediction
tools. At least four of the variants were shown to result in com-
plete or almost complete loss of function with bupropion and
efavirenz as substrates. The detailed comparison of in vitro func-
tionality of the variants with in silico prediction tools including a
thorough substrate docking simulation analysis points at the chal-
lenge to deal with the hundreds of new variants that exist in all
populations as currently uncovered by next generation sequenc-
ing approaches and large scale population projects (see links
above).
Table 3 |Properties of recombinantly expressed CYP2B6 protein variants with other clinical substrates.
Artemether1
COS-7 cells
(Honda et al., 2011)
Selegiline2
COS-7 cells
(Watanabe et al., 2010)
Chlorpyrifos3
COS-1 cells
(Crane et al., 2012)
Cyclophosphamide4
Sf9 cells
(Ariyoshi et al., 2011)
Cyclophosphamide4
E. coli
(Raccor et al., 2012)
Variant Km(μM) Vmax(%) Km(μM) Vmax (%) Km(μM) Vmax(%) Km(mM) Vmax (%) Km(mM) Vmax (%)
2B6.1 3.1 100 48.2 100 1.84 100 2.68 100 3.6 100
2B6.6 6.72 416 56.6 169 1.97 254 1.62 99 4.0 155.2
2B6.4 2.73 196 45.8 147 1.09 1094 2.75 74 3.5 67.1
2B6.5 6.87 55 70.1 85 0.80 441 5.1 72.4
1O-Demethylation.
2N-Demethylation (mean values were calculated for several expressions of CYP2B6.1).
3Desulfation.
44-Hydroxylation.
www.frontiersin.org March 2013 |Volume 4 |Article 24 |7
“fgene-04-00024” 2013/3/12 14:58 page8—#8
Zanger and Klein Pharmacogenetics of CYP2B6
CLINICAL STUDIES WITH DIFFERENT DRUGS
The widely used anticancer and immunosuppressant pro-
drug cyclophosphamide depends on bioactivation to 4-
hydroxycyclophosphamide for cytotoxic activity. Bioactivation is
highly variable in cancer patients and has been attributed mainly
to CYP2B6 in vitro and in vivo with contributions from CYP2C19
and CYP3A4 (Chang et al., 1993;Raccor et al., 2012). The case
of cyclophosphamide 4-hydroxylation deserves particular atten-
tion, as it exemplifies substrate-dependent effects of CYP2B6
pharmacogenetics. Cyclophosphamide 4-hydroxylation was ini-
tially reported to be enhanced in livers genotyped CYP2B6*6/*6
(Xie et al., 2003), which was confirmed in several later in vivo
studies (Xie et al., 2006;Nakajima et al., 2007;Torimoto and
Kohgo, 2008). However, other in vivo studies analyzing phar-
macokinetics or clinical outcome also presented contradictory or
negative results (Singh et al., 2007;Ekhart et al., 2008;Melanson
et al., 2010;Yao et al., 2010;Raccor et al., 2012). In vitro, insect
cell-expressed recombinant CYP2B6.4 [K262R] had lower activ-
ity for cyclophosphamide 4-hydroxylation (Ariyoshi et al., 2011;
Raccor et al., 2012). The CYP2B6.4 and CYP2B6.6 variants thus
display mirror-inverted catalytic activities toward efavirenz and
cyclophosphamide, in that the former variant is the catalytically
more active one with efavirenz, whereas the opposite is true for the
latter variant (Tab l e 3). A direct comparison of catalytic properties
of the two variants with the reference enzyme expressed in insect
cells supports this inverse behavior of the two variants toward these
two substrates (Ariyoshi et al., 2011). Interestingly, several studies
associated other variants including CYP2B6*4, *5, *8, and *9 with
lower 4-OH cyclophosphamide formation in vivo or with worse
outcome (Takada et al., 2004;Bray et al., 2010;Helsby et al., 2010;
Joy et al., 2012). Taken together, the data concerning cyclophos-
phamide from both in vivo and in vitro indicate that CYP2B6
polymorphism plays a role, although the studies are so far not yet
conclusive. This may be explained by different study size design
as well as lack of consistency in allele definition and genotype
information among studies (Helsby and Tingle, 2011).
In addition to efavirenz, CYP2B6 genotype also affects plasma
levels of the antiretroviral drug nevirapine (Penzak et al., 2007;
Mahungu et al., 2009). The impact of the CYP2B6 516G>T poly-
morphism on nevirapine exposure was confirmed and quantified
in a pharmacometric analysis of nevirapine plasma concentrations
from 271 patients genotyped for 198 SNPs in 45 ADME (absorp-
tion, distribution, metabolism, and excretion) genes and covari-
ates (Lehr et al., 2011). Moreover, nevirapine-related cutaneous
adverse events, which are most likely major histocompatibil-
ity complex (MHC) class I-mediated, were significantly influ-
enced by CYP2B6 polymorphism while hepatic side effects,
most likely MHC class II-mediated, were unaffected by CYP2B6
(Yuan et al., 2011).
CYP2B6 allele variants were also investigated in the context
of the synthetic μ-opioid receptor agonist, methadone, which
is metabolized by CYPs 3A4/5, 2B6, and 2D6, and used as a
maintenance treatment for opioid addiction. In *6/*6 carriers
(S)-methadone plasma levels were increased leading to potentially
higher risk of severe cardiac arrhythmias and methadone asso-
ciated deaths (Crettol et al., 2005;Eap et al., 2007;Bunten et al.,
2011). Methadone dose requirement for effective treatment of
opioid addiction was shown to be significantly reduced in carriers
of this genotype (Levran et al., 2011).
CONCLUSION
The polymorphism of the CYP2B6 gene has initially been studied
by reverse genetics approach, i.e., starting from the identification
of genetic variants in DNA and liver samples, followed by in vitro
characterization of genotyped livers and expressed variant pro-
teins. Clinical studies have then contributed to identify the variants
that are important in vivo, and in vitro studies are again needed to
identify and mechanistically explain causal variants. Nevertheless,
CYP2B6 pharmacogenetics has yet to be fully explored, especially
with respect to combined effects of the involved variants on both
expression and catalytic properties, the latter of which additionally
depend on the substrate. While the relevance for HIV-1 therapy
with efavirenz is well established and translational approaches have
already been clinically tested, an increasing number of studies
suggest clinical relevance for additional drug substrates.
ACKNOWLEDGMENTS
Work in the authors’ laboratory was supported by the Ger-
man Federal Ministry of Education and Research (Virtual Liver
Network grant 0315755), the 7FP EU Initial Training Network
program‚ FightingDrugFailure’ (GA-2009–238132), and by the
Robert-Bosch Foundation, Stuttgart, Germany.
REFERENCES
Abass, K., and Pelkonen, O. (2012).
The inhibition of major human
hepatic cytochrome P450 enzymes
by 18 pesticides: comparison of
the N-in-one and single substrate
approaches. Toxicol. In Vitro doi:
10.1016/j.tiv.2012.05.003 [Epub
ahead of print].
Amunugama, H., Zhang, H., and Hol-
lenberg, P. F. (2012). Mechanism-
based inactivation of cytochrome
P450 2B6 by methadone through
destruction of prosthetic heme. Drug
Metab. Dispos. 40, 1765–1770.
Ariyoshi, N., Miyazaki, M., Toide,
K., Sawamura Yi, and Kamataki,
T. (2001). A single nucleotide
polymorphism of CYP2b6 found in
Japanese enhances catalytic activity
by autoactivation. Biochem. Biophys.
Res. Commun. 281, 1256–1260.
Ariyoshi, N., Ohara, M., Kaneko, M.,
Afuso, S., Kumamoto, T., Naka-
mura, H., et al. (2011). Q172H
replacement overcomes effects on
the metabolism of cyclophosphamide
and efavirenz caused by CYP2B6 vari-
ant with Arg262. Drug Metab. Dispos.
39, 2045–2048.
Asimus, S., and Ashton, M. (2009).
Artemisinin–apossible CYP2B6
probe substrate? Biopharm. Drug Dis-
pos. 30, 265–275.
Bray, J., Sludden, J., Griffin, M. J.,
Cole, M., Verrill, M., Jamieson, D.,
et al. (2010). Influence of pharma-
cogenetics on response and toxic-
ity in breast cancer patients treated
with doxorubicin and cyclophos-
phamide. Br. J. Cancer 102, 1003–
1009.
Bumpus, N. N., and Hollenberg, P. F.
(2008). Investigation of the mech-
anisms underlying the differential
effects of the K262R mutation of
P450 2B6 on catalytic activity. Mol.
Pharmacol. 74, 990–999.
Bumpus, N. N., Kent, U. M., and Hol-
lenberg, P. F. (2006). Metabolism of
efavirenz and 8-hydroxyefavirenz by
P450 2B6 leads to inactivation by two
distinct mechanisms. J. Pharmacol.
Exp. Ther. 318, 345–351.
Bunten, H., Liang, W.-J., Pounder, D.,
Seneviratne, C., and Osselton, M. D.
(2011). CYP2B6 and OPRM1 gene
variations predict methadone-related
deaths. Addict. Biol. 16, 142–144.
Burger, D., van der Heiden, I., la Porte,
C., van der Ende, M., Groeneveld, P.,
Richter, C., etal. (2006). Interpatient
variability in the pharmacokinetics
of the HIV non-nucleoside reverse
transcriptase inhibitor efavirenz: the
effect of gender, race, and CYP2B6
polymorphism. Br. J. Clin. Pharma-
col. 61, 148–154.
Burk, O., Arnold, K. A., Nussler,
A. K., Schaeffeler, E., Efimova, E.,
Avery, B. A., etal. (2005). Anti-
malarial artemisinin drugs induce
Frontiers in Genetics |Pharmacogenetics and Pharmacogenomics March 2013 |Volume 4 |Article 24 |8
“fgene-04-00024” 2013/3/12 14:58 page9—#9
Zanger and Klein Pharmacogenetics of CYP2B6
cytochrome P450 and MDR1 expres-
sion by activation of xenosensors
pregnane X receptor and constitutive
androstane receptor. Mol. Pharmacol.
67, 1954–1965.
Chang, T. K., Weber, G. F., Crespi, C. L.,
and Waxman, D. J. (1993). Differen-
tial activation of cyclophosphamide
and ifosphamide by cytochromes P-
450 2B and 3A in human liver micro-
somes. Cancer Res. 53, 5629–5637.
Choudhary, D., Jansson, I., Schenkman,
J. B., Sarfarazi, M., and Stoilov, I.
(2003). Comparative expression pro-
filing of 40 mouse cytochrome P450
genes in embryonic and adult tissues.
Arch. Biochem. Biophys. 414, 91–100.
Code, E. L., Crespi, C. L., Penman, B.
W., Gonzalez, F. J., Chang, T. K.,
and Waxman, D. J. (1997). Human
cytochrome P4502B6: interindivid-
ual hepatic expression, substrate
specificity, and role in procarcinogen
activation. Drug Metab. Dispos. 25,
985–993.
Court, M. H., Duan, S. X., Hesse, L.
M., Venkatakrishnan, K., and Green-
blatt, D. J. (2001). Cytochrome P-450
2B6 is responsible for interindivid-
ual variability of propofol hydroxy-
lation by human liver microsomes.
Anesthesiology 94, 110–119.
Crane, A. L., Klein, K., and Olson, J. R.
(2012). Bioactivation of chlorpyrifos
by CYP2B6 variants. Xenobiotica 42,
1255–1262.
Crettol, S., Déglon, J.-J., Besson, J.,
Croquette-Krokkar, M., Gothuey,
I., Hämmig, R., et al. (2005).
Methadone enantiomer plasma lev-
els, CYP2B6, CYP2C19, and CYP2C9
genotypes, and response to treat-
ment. Clin. Pharmacol. Ther. 78,
593–604.
Croom, E. L., Stevens, J. C., Hines,
R. N., Wallace, A. D., and Hodg-
son, E. (2009). Human hepatic
CYP2B6 developmental expression:
the impact of age and genotype.
Biochem. Pharmacol. 78, 184–190.
Das, P. C., Cao, Y., Rose, R. L., Cher-
rington, N., and Hodgson, E. (2008).
Enzyme induction and cytotoxicity
in human hepatocytes by chlorpyri-
fos and N,N-diethyl-m-toluamide
(DEET). Drug Metab. Drug Interact.
23, 237–260.
Desta, Z., Moaddel, R., Ogburn, E. T.,
Xu, C., Ramamoorthy, A., Venkata,
S. L. V., etal. (2012). Stereose-
lective and regiospecific hydroxyla-
tion of ketamine and norketamine.
Xenobiotica 42, 1076–1087
Desta, Z., Saussele, T.,Ward, B., Bliever-
nicht, J., Li, L., Klein, K., et al. (2007).
Impact of CYP2B6 polymorphism
on hepatic efavirenz metabolism in
vitro. Pharmacogenomics 8, 547–558.
Dutheil, F., Beaune, P., and Loriot,
M.-A. (2008). Xenobiotic metaboliz-
ing enzymes in the central nervous
system: contribution of cytochrome
P450 enzymes in normal and patho-
logical human brain. Biochimie 90,
426–436.
Eap, C. B., Crettol, S., Rougier, J.-
S., Schläpfer, J., Sintra Grilo, L.,
Déglon, J.-J., etal. (2007). Stereos-
elective block of hERG channel by
(S)-methadone and QT interval pro-
longation in CYP2B6 slow metab-
olizers. Clin. Pharmacol. Ther. 81,
719–728.
Ekhart, C., Doodeman, V. D., Roden-
huis, S., Smits, P. H. M., Beijnen, J.
H., and Huitema, A. D. R. (2008).
Influence of polymorphisms of drug
metabolizing enzymes (CYP2B6,
CYP2C9, CYP2C19, CYP3A4,
CYP3A5, GSTA1, GSTP1, ALDH1A1
and ALDH3A1) on the pharma-
cokinetics of cyclophosphamide
and 4-hydroxycyclophosphamide.
Pharmacogenet. Genomics 18,
515–523.
Ekins, S., Vandenbranden, M., Ring, B.
J., Gillespie, J. S., Yang, T. J., Gelboin,
H. V., et al. (1998). Further character-
ization of the expression in liver and
catalytic activity of CYP2B6. J. Phar-
macol. Exp. Ther. 286, 1253–1259.
Ekins, S., and Wrighton, S. A. (1999).
The role of CYP2B6 in human xeno-
biotic metabolism. Drug Metab. Rev.
31, 719–754.
Ellison, C. A., Tian, Y., Knaak, J. B.,
Kostyniak, P. J., and Olson, J. R.
(2012a). Human hepatic cytochrome
p450-specific metabolism of the
organophosphorus pesticides methyl
parathion and diazinon. Drug Metab.
Dispos. 40, 1–5.
Ellison, C. A., Abou El-Ella, S. S., Taw-
fik, M., Lein, P. J., and Olson, J.
R. (2012b). Allele and genotype fre-
quencies of CYP2B6 and CYP2C19
polymorphisms in Egyptian agricul-
tural workers. J. Toxicol. Environ.
Health A. 75, 232–241.
Erickson, D. A., Mather, G., Trager, W.
F.,Levy, R. H., and Keirns, J. J. (1999).
Characterization of the in vitro bio-
transformation of the HIV-1 reverse
transcriptase inhibitor nevirapine by
human hepatic cytochromes P-450.
Drug Metab. Dispos. 27, 1488–1495.
Faucette, S. R., Hawke, R. L., Lecluyse,
E. L., Shord, S. S., Yan, B., Laethem,
R. M., et al. (2000). Validation of
bupropion hydroxylation as a selec-
tive marker of human cytochrome
P450 2B6 catalytic activity. Drug
Metab. Dispos. 28, 1222–1230.
Faucette, S. R., Wang, H., Hamilton, G.
A., Jolley, S. L., Gilbert, D., Lind-
ley, C., etal. (2004). Regulation of
CYP2B6 in primary human hepato-
cytes by prototypical inducers. Drug
Metab. Dispos. 32, 348–358.
Faucette, S. R., Zhang, T.-C., Moore,
R., Sueyoshi, T., Omiecinski, C.
J., LeCluyse, E. L., etal. (2007).
Relative activation of human preg-
nane X receptor versus constitutive
androstane receptor defines distinct
classes of CYP2B6 and CYP3A4
inducers. J. Pharmacol. Exp. Ther.
320, 72–80.
Feidt, D. M., Klein, K., Hofmann,
U., Riedmaier, S., Knobeloch, D.,
Thasler, W. E., et al. (2010). Profil-
ing induction of cytochrome p450
enzyme activity by statins using a new
liquid chromatography-tandem mass
spectrometry cocktail assay in human
hepatocytes. Drug Metab. Dispos. 38,
1589–1597.
Ferguson, C. S., and Tyndale, R. F.
(2011). Cytochrome P450 enzymes
in the brain: emerging evidence of
biological significance. Trends Phar-
macol. Sci. 32, 708–714.
Fuhr, U., Jetter, A., and Kirchheiner,
J. (2007). Appropriate phenotyp-
ing procedures for drug metabolizing
enzymes and transporters in humans
and their simultaneous use in the
“cocktail”approach. Clin. Pharmacol.
Ther. 81, 270–283.
Gatanaga, H., Hayashida, T., Tsuchiya,
K., Yoshino, M., Kuwahara, T.,
Tsukada, H., etal. (2007). Suc-
cessful efavirenz dose reduction in
HIV type 1-infected individuals with
cytochrome P450 2B6 *6 and *26.
Clin. Infect. Dis. 45, 1230–1237.
Gatanaga, H., and Oka, S. (2009). Suc-
cessful genotype-tailored treatment
with small-dose efavirenz. AIDS 23,
433–434.
Gay, S. C., Shah, M. B., Talakad, J. C.,
Maekawa, K., Roberts, A. G., Wil-
derman, P. R., etal. (2010). Crystal
structure of a cytochrome P450 2B6
genetic variant in complex with the
inhibitor 4-(4-chlorophenyl) imida-
zole at 2.0-A resolution. Mol. Phar-
macol. 77, 529–538.
Gervot, L., Rochat, B., Gautier, J. C.,
Bohnenstengel, F., Kroemer, H., de
Berardinis, V., et al. (1999). Human
CYP2B6: expression, inducibility and
catalytic activities. Pharmacogenetics
9, 295–306.
Gomes, A. M., Winter, S., Klein,
K., Turpeinen, M., Schaeffeler,
E., Schwab, M., etal. (2009).
Pharmacogenomics of human liver
cytochrome P450 oxidoreductase:
multifactorial analysis and impact on
microsomal drug oxidation. Pharma-
cogenomics 10, 579–599.
Goodwin, B., Moore, L. B., Stoltz, C.
M., McKee, D. D., and Kliewer, S.
A. (2001). Regulation of the human
CYP2B6 gene by the nuclear preg-
nane X receptor. Mol. Pharmacol. 60,
427–431.
Haas, D. W., Ribaudo, H. J., Kim, R. B.,
Tierney, C., Wilkinson, G. R., Gulick,
R. M., etal. (2004). Pharmacogenet-
ics of efavirenz and central nervous
system side effects: an Adult AIDS
Clinical Trials Group study. AIDS 18,
2391–2400.
Habtewold, A., Amogne, W., Makon-
nen, E., Yimer, G., Riedel, K.-D.,
Ueda, N., et al. (2011). Long-term
effect of efavirenz autoinduction on
plasma/peripheral blood mononu-
clear cell drug exposure and CD4
count is influenced by UGT2B7
and CYP2B6 genotypes among HIV
patients. J. Antimicrob. Chemother.
66, 2350–2361.
Harleton, E., Webster, M., Bum-
pus, N. N., Kent, U. M., Rae,
J. M., and Hollenberg, P. F.
(2004). Metabolism of N,N,N -
triethylenethiophosphoramide by
CYP2B1 and CYP2B6 results in the
inactivation of both isoforms by two
distinct mechanisms. J. Pharmacol.
Exp. Ther. 310, 1011–1019.
Helsby, N., and Tingle, M. (2011).
The importance of correct assign-
ment of CYP2B6 genetic variants
with respect to cyclophosphamide
metabolizer status. Am. J. Hematol.
86, 383–384; author reply 384.
Helsby, N. A., Hui, C.-Y., Goldthorpe,
M. A., Coller,J. K., Soh, M. C., Gow, P.
J., et al. (2010). The combined impact
of CYP2C19 and CYP2B6 pharmaco-
genetics on cyclophosphamide bioac-
tivation. Br. J. Clin. Pharmacol. 70,
844–853.
Hesse, L. M., He, P., Krishnaswamy,
S., Hao, Q., Hogan, K., von Moltke,
L. L., etal. (2004). Pharmacogenetic
determinants of interindividual vari-
ability in bupropion hydroxylation
by cytochrome P450 2B6 in human
liver microsomes. Pharmacogenetics
14, 225–238.
Hesse, L. M., Venkatakrishnan, K.,
Court, M. H., von Moltke, L. L.,
Duan, S. X., Shader, R. I., et al.
(2000). CYP2B6 mediates the in
vitro hydroxylation of bupropion:
potential drug interactions with other
antidepressants. Drug Metab. Dispos.
28, 1176–1183.
Hodgson, E., and Rose, R. L. (2007). The
importance of cytochrome P450 2B6
in the human metabolism of environ-
mental chemicals. Pharmacol. Ther.
113, 420–428.
Hoffman, S. M., Nelson, D. R., and
Keeney, D. S. (2001). Organization,
structure and evolution of the CYP2
gene cluster on human chromosome
www.frontiersin.org March 2013 |Volume 4 |Article 24 |9
“fgene-04-00024” 2013/3/12 14:58 page 10 #10
Zanger and Klein Pharmacogenetics of CYP2B6
19. Pharmacogenetics 11, 687–
698.
Hofmann, M. H., Blievernicht, J. K.,
Klein, K., Saussele, T., Schaeffeler, E.,
Schwab, M., etal. (2008). Aberrant
splicing caused by single nucleotide
polymorphism c.516G>T [Q172H],
a marker of CYP2B6*6, is responsible
for decreased expression and activity
of CYP2B6 in liver. J. Pharmacol. Exp.
Ther. 325, 284–292.
Holzinger, E. R., Grady, B., Ritchie,
M. D., Ribaudo, H. J., Acosta, E.
P., Morse, G. D., et al. (2012).
Genome-wide association study of
plasma efavirenz pharmacokinetics
in AIDS Clinical Trials Group proto-
cols implicates several CYP2B6 vari-
ants. Pharmacogenet. Genomics 22,
858–867.
Honda, M., Muroi, Y., Tamaki, Y.,
Saigusa, D., Suzuki, N., Tomioka,
Y., et al. (2011). Functional char-
acterization of CYP2B6 allelic vari-
ants in demethylation of antimalarial
artemether. Drug Metab. Dispos. 39,
1860–1865.
Hsyu, P. H., Singh, A., Giargiari, T. D.,
Dunn, J. A., Ascher, J. A., and John-
ston, J. A. (1997). Pharmacokinetics
of bupropion and its metabolites in
cigarette smokers versusnonsmokers.
J. Clin. Pharmacol. 37, 737–743.
Huang, Z., Roy, P., and Waxman,
D. J. (2000). Role of human liver
microsomal CYP3A4 and CYP2B6
in catalyzing N-dechloroethylation
of cyclophosphamide and ifosfamide.
Biochem. Pharmacol. 59, 961–972.
Huitema, A. D., Kerbusch, T., Tibben,
M. M., Rodenhuis, S., and Bei-
jnen, J. H. (2000). Reduction
of cyclophosphamide bioactivation
by thioTEPA: critical sequence-
dependency in high-dose chemother-
apy regimens. Cancer Chemother.
Pharmacol. 46, 119–127.
Humphrey, W., Dalke,A., and Schulten,
K. (1996). VMD: visual molecular
dynamics. J. Mol. Graph. 14, 33–38,
27–28.
Jeong, S., Nguyen, P. D., and Desta,
Z. (2009). Comprehensive in vitro
analysis of voriconazole inhibition
of eight cytochrome P450 (CYP)
enzymes: major effect on CYPs 2B6,
2C9, 2C19, and 3A. Antimicrob.
Agents Chemother. 53, 541–551.
Jiang, F., Desta, Z., Shon, J.-H.,
Yeo, C.-W., Kim, H.-S., Liu, K.-
H., et al. (2012). Effects of clopi-
dogrel and itraconazole on the
disposition of efavirenz and its
hydroxyl-metabolites: exploration of
a novel CYP2B6 phenotyping index.
Br. J. Clin. Pharmacol. 75, 244–253.
Jinno, H., Tanaka-Kagawa, T., Ohno, A.,
Makino,Y., Matsushima, E., Hanioka,
N., et al. (2003). Functional charac-
terization of cytochrome P450 2B6
allelic variants. Drug Metab. Dispos.
31, 398–403.
Joy, M. S., La, M., Wang, J., Bridges,
A. S., Hu, Y., Hogan, S. L., et al.
(2012). Cyclophosphamide and
4-hydroxycyclophosphamide phar-
macokinetics in patients with
glomerulonephritis secondary to
lupus and small vessel vasculitis. Br.
J. Clin. Pharmacol. 74, 445–455.
Kharasch, E. D., Mitchell, D.,
Coles, R., and Blanco, R. (2008).
Rapid clinical induction of hep-
atic cytochrome P4502B6 activity
by ritonavir. Antimicrob. Agents
Chemother. 52, 1663–1669.
King, J., and Aberg, J. A. (2008). Clini-
cal impact of patient population dif-
ferences and genomic variation in
efavirenz therapy. AIDS 22, 1709–
1717.
Klein, K., Lang, T.,Saussele, T., Barbosa-
Sicard, E., Schunck, W.-H., Eichel-
baum, M., et al. (2005). Genetic
variability of CYP2B6 in populations
of African and Asian origin: allele fre-
quencies, novel functional variants,
and possible implications for anti-
HIV therapy with efavirenz. Pharma-
cogenet. Genomics 15, 861–873.
Ko, J. W., Desta, Z., and Flockhart, D.
A. (1998). Human N-demethylation
of (S)-mephenytoin by cytochrome
P450s 2C9 and 2B6. Drug Metab.
Dispos. 26, 775–778.
Korhonen, L. E., Turpeinen, M., Rah-
nasto, M., Wittekindt, C., Poso,
A., Pelkonen, O., et al. (2007).
New potent and selective cytochrome
P450 2B6 (CYP2B6) inhibitors based
on three-dimensional quantitative
structure-activity relationship (3D-
QSAR) analysis. Br. J. Pharmacol.
150, 932–942.
Kreth, K., Kovar, K., Schwab, M., and
Zanger, U. M. (2000). Identifica-
tion of the human cytochromes P450
involved in the oxidative metabolism
of “Ecstasy”-related designer drugs.
Biochem. Pharmacol. 59, 1563–
1571.
Lamba, V., Lamba, J., Yasuda, K., Strom,
S., Davila, J., Hancock, M. L., et al.
(2003). Hepatic CYP2B6 expression:
gender and ethnic differences and
relationship to CYP2B6 genotype and
CAR (constitutive androstane recep-
tor) expression. J. Pharmacol. Exp.
Ther. 307, 906–922.
Lang, T., Klein, K., Fischer, J., Nüssler,
A. K., Neuhaus, P., Hofmann, U.,
et al. (2001). Extensive genetic poly-
morphism in the human CYP2B6
gene with impact on expression and
function in human liver. Pharmaco-
genetics 11, 399–415.
Lang, T., Klein, K., Richter, T., Zibat,
A., Kerb, R., Eichelbaum, M., etal.
(2004). Multiple novel nonsynony-
mous CYP2B6 gene polymorphisms
in Caucasians: demonstration of phe-
notypic null alleles. J. Pharmacol.
Exp. Ther. 311, 34–43.
Leclerc, J., Tournel, G., Courcot-
Ngoubo Ngangue, E., Pottier, N.,
Lafitte, J.-J., Jaillard, S., et al.
(2010). Profiling gene expression
of whole cytochrome P450 super-
family in human bronchial and
peripheral lung tissues: differential
expression in non-small cell lung can-
cers. Biochimie 92, 292–306.
Lehr, T., Yuan, J., Hall, D., Zimdahl-
Gelling, H., Schaefer, H. G., Staab, A.,
et al. (2011). Integration of absorp-
tion, distribution, metabolism, and
elimination genotyping data into a
population pharmacokinetic analy-
sis of nevirapine. Pharmacogenet.
Genomics 21, 721–730.
Levran, O., Peles, E., Hamon, S., Ran-
desi, M., Adelson, M., and Kreek,
M. J. (2011). CYP2B6 SNPs are
associated with methadone dose
required for effective treatment of
opioid addiction. Addict. Biol. doi:
10.1111/j.1369-1600.2011.00349.x.
[Epub ahead of print].
Lewis, D. F., Lake, B. G., Dickins, M.,
Eddershaw, P. J., Tarbit, M. H., and
Goldfarb, P. S. (1999). Molecular
modelling of CYP2B6, the human
CYP2B isoform, by homology with
the substrate-bound CYP102 crystal
structure: evaluation of CYP2B6 sub-
strate characteristics, the cytochrome
b5 binding site and comparisons with
CYP2B1 and CYP2B4. Xenobiotica
29, 361–393.
Lewis, D. F. V., Lake, B. G., and
Dickins, M. (2004). Substrates of
human cytochromes P450 from fam-
ilies CYP1 and CYP2: analysis of
enzyme selectivity and metabolism.
Drug Metab. Drug Interact. 20,
111–142.
Li, H., Ferguson, S. S., and Wang,
H. (2010). Synergistically enhanced
CYP2B6 inducibility between a
polymorphic mutation in CYP2B6
promoter and pregnane X recep-
tor activation. Mol. Pharmacol. 78,
704–713.
Li, J., Menard, V., Benish, R. L., Jurevic,
R. J., Guillemette, C., Stoneking,
M., et al. (2012). Worldwide vari-
ation in human drug-metabolism
enzyme genes CYP2B6 and UGT2B7:
implications for HIV/AIDS treat-
ment. Pharmacogenomics 13,
555–570.
Lin, H. L., Zhang, H., and Hol-
lenberg, P. F. (2009). Metabolic
activation of mifepristone (RU486)
by mammalian P450s and the
mechanism-based inactivation of
human CYP2B6. J. Pharmacol. Exp.
Ther. 329, 26–37.
Liu, P., Foster, G., LaBadie, R. R.,
Gutierrez, M. J., and Sharma, A.
(2008). Pharmacokinetic interaction
between voriconazole and efavirenz
at steady state in healthy male sub-
jects. J. Clin. Pharmacol. 48, 73–84.
Lubomirov, R., Colombo, S., di Iulio,
J., Ledergerber, B., Martinez, R.,
Cavassini, M., et al. (2011). Asso-
ciation of pharmacogenetic markers
with premature discontinuation of
first-line anti-HIV therapy: an obser-
vational cohort study. J. Infect. Dis.
203, 246–257.
Lubomirov, R., di Iulio, J., Fayet, A.,
Colombo, S., Martinez, R., Marzolini,
C., et al. (2010). ADME phar macoge-
netics: investigation of the pharma-
cokinetics of the antiretroviral agent
lopinavir coformulated with riton-
avir. Pharmacogenet. Genomics 20,
217–230.
Mahungu, T., Smith, C., Turner, F.,
Egan, D., Youle, M., Johnson, M.,
et al. (2009). Cytochrome P450 2B6
516G–>T is associated with plasma
concentrations of nevirapine at both
200 mg twice daily and 400 mg once
daily in an ethnically diverse popula-
tion. HIV Med. 10, 310–317.
Maimbo, M., Kiyotani, K., Mushi-
roda, T., Masimirembwa, C., and
Nakamura, Y. (2011). CYP2B6 geno-
typeisastrongpredictorofsystemic
exposure to efavirenz in HIV-infected
Zimbabweans. Eur. J. Clin. Pharma-
col. 68, 267–271.
Martis, S., Mei, H., Vijzelaar, R., Edel-
mann, L., Desnick, R. J., and Scott, S.
A. (2012). Multi-ethnic cytochrome-
P450 copy number profiling: novel
pharmacogenetic alleles and mecha-
nism of copy number variation for-
mation. Pharmacogenomics J. doi:
10.1038/tpj.2012.48 [Epub ahead of
print].
Mehlotra, R. K., Bockarie, M. J., and
Zimmerman, P. A. (2007). CYP2B6
983T>C polymorphism is preva-
lent in West Africa but absent in
Papua New Guinea: implications for
HIV/AIDS treatment. Br. J. Clin.
Pharmacol. 64, 391–395.
Melanson, S. E. F., Stevenson, K.,
Kim, H., Antin, J. H., Court, M.
H., Ho, V. T., et al. (2010). Allelic
variations in CYP2B6 and CYP2C19
and survival of patients receiving
cyclophosphamide prior to myeloab-
lative hematopoietic stem cell trans-
plantation. Am. J. Hematol. 85,
967–971.
Meyer, R. P., Gehlhaus, M., Knoth, R.,
and Volk, B. (2007). Expression and
Frontiers in Genetics |Pharmacogenetics and Pharmacogenomics March 2013 |Volume 4 |Article 24 |10
“fgene-04-00024” 2013/3/12 14:58 page 11 #11
Zanger and Klein Pharmacogenetics of CYP2B6
function of cytochrome p450 in brain
drug metabolism. Curr. Drug Metab.
8, 297–306.
Miksys, S., Lerman, C., Shields, P.
G., Mash, D. C., and Tyndale, R.
F. (2003). Smoking, alcoholism and
genetic polymorphisms alter CYP2B6
levels in human brain. Neuropharma-
cology 45, 122–132.
Mo, S.-L., Liu, Y.-H., Duan, W., Wei,
M. Q., Kanwar, J. R., and Zhou, S.-F.
(2009). Substrate specificity, regula-
tion, and polymorphism of human
cytochrome P450 2B6. Curr. Drug
Metab. 10, 730–753.
Mukonzo, J. K., Röshammar, D.,
Waako, P., Andersson, M., Fuka-
sawa, T., Milani, L., et al. (2009). A
novel polymorphism in ABCB1 gene,
CYP2B6*6 and sex predict single-
dose efavirenz population pharma-
cokinetics in Ugandans. Br. J. Clin.
Pharmacol. 68, 690–699.
Nakajima, M., Komagata, S., Fujiki, Y.,
Kanada, Y., Ebi, H., Itoh, K., et al.
(2007). Genetic polymorphisms of
CYP2B6 affect the pharmacokinet-
ics/pharmacodynamics of cyclophos-
phamide in Japanese cancer patients.
Pharmacogenet. Genomics 17,
431–445.
Nelson, D. R., Zeldin, D. C., Hoffman,
S. M. G., Maltais, L. J., Wain, H. M.,
and Nebert, D. W. (2004). Compari-
son of cytochrome P450 (CYP) genes
from the mouse and human genomes,
including nomenclature recommen-
dations for genes, pseudogenes and
alternative-splice variants. Pharma-
cogenetics 14, 1–18.
Ngaimisi, E., Mugusi, S., Minzi, O. M.,
Sasi, P., Riedel, K.-D., Suda, A., et al.
(2010). Long-term efavirenz autoin-
duction and its effect on plasma
exposure in HIV patients. Clin. Phar-
macol. Ther. 88, 676–684.
Nguyen, T.-A., Tychopoulos, M., Bichat,
F., Zimmermann, C., Flinois, J.-P.,
Diry, M., et al. (2008). Improvement
of cyclophosphamide activation by
CYP2B6 mutants: from in silico to
ex vivo. Mol. Pharmacol. 73, 1122–
1133.
Niu, R.-J., Zheng, Q.-C., Zhang, J.-L.,
and Zhang, H.-X. (2011). Analy-
sis of clinically relevant substrates of
CYP2B6 enzyme by computational
methods. J. Mol. Model. 17, 2839–
2846.
Nyakutira, C., Röshammar, D.,
Chigutsa, E., Chonzi, P., Ashton,
M., Nhachi, C., et al. (2008). High
prevalence of the CYP2B6 516G–
>T(*6) variant and effect on the
population pharmacokinetics of
efavirenz in HIV/AIDS outpatients
in Zimbabwe. Eur. J. Clin. Pharmacol.
64, 357–365.
Oda, Y., Hamaoka, N., Hiroi, T.,
Imaoka, S., Hase, I., Tanaka, K.,
et al. (2001). Involvement of human
liver cytochrome P4502B6 in the
metabolism of propofol. Br. J. Clin.
Pharmacol. 51, 281–285.
Oscarson, M., Zanger, U. M., Rifki,
O. F., Klein, K., Eichelbaum, M.,
and Meyer, U. A. (2006). Transcrip-
tional profiling of genes induced in
the livers of patients treated with car-
bamazepine. Clin. Pharmacol. Ther.
80, 440–456.
Pelkonen, O., Turpeinen, M., Hakkola,
J., Honkakoski, P., Hukkanen, J.,
and Raunio, H. (2008). Inhibition
and induction of human cytochrome
P450 enzymes: current status. Arch.
Tox i c o l . 82, 667–715.
Peltoniemi, M. A., Saari, T. I., Hagel-
berg, N. M., Reponen, P., Turpeinen,
M., Laine, K., et al. (2011). Expo-
sure to oral S-ketamine is unaffected
by itraconazole but greatly increased
by ticlopidine. Clin. Pharmacol. Ther.
90, 296–302.
Penzak, S. R., Kabuye, G., Mugyenyi, P.,
Mbamanya, F., Natarajan, V., Alfaro,
R. M., etal. (2007). Cytochrome
P450 2B6 (CYP2B6) G516T influ-
ences nevirapine plasma concentra-
tions in HIV-infected patients in
Uganda. HIV Med. 8, 86–91.
Qin, W.-J., Zhang, W., Liu, Z.-Q., Chen,
X.-P., Tan, Z.-R., Hu, D.-L., et al.
(2012). Rapid clinical induction of
bupropion hydroxylationby metami-
zole in healthy Chinese men. Br. J.
Clin. Pharmacol. 74, 999–1004.
Raccor,B. S., Claessens, A. J., Dinh, J. C.,
Park, J. R., Hawkins, D. S., Thomas,
S. S., et al. (2012). Potential contri-
bution of cytochrome P450 2B6 to
hepatic 4-hydroxycyclophosphamide
formation in vitro and in vivo. Drug
Metab. Dispos. 40, 54–63.
Radloff, R., Gras, A., Zanger, U.
M., Masquelier, C., Arumugam, K.,
Karasi, J. C., et al. (2013). Novel
CYP2B6 enzyme variants in a Rwan-
dese population: functional charac-
terization and assessment of in silico
prediction tools. Hum. Mutat. doi:
10.1002/humu.22295 [Epub ahead of
print].
Rae, J. M., Soukhova, N. V., Flockhart,
D. A., and Desta, Z. (2002). Tri-
ethylenethiophosphoramide is a spe-
cific inhibitor of cytochrome P450
2B6: implications for cyclophos-
phamide metabolism. Drug Metab.
Dispos. 30, 525–530.
Rakhmanina, N. Y., and van den Anker,
J. N. (2010). Efavirenz in the therapy
of HIV infection. Expert Opin. Drug
Metab. Toxicol. 6, 95–103.
Ramírez, J., Innocenti, F., Schuetz, E.
G., Flockhart, D. A., Relling, M. V.,
Santucci, R., et al. (2004). CYP2B6,
CYP3A4, and CYP2C19 are responsi-
ble for the in vitro N-demethylation
of meperidine in human liver micro-
somes. Drug Metab. Dispos. 32,
930–936.
Remmer, H., Schoene, B., and Fleis-
chmann, R. A. (1973). Induction of
the unspecific microsomal hydroxy-
lase in the human liver. Drug Metab.
Dispos. 1, 224–230.
Restrepo, J. G., Martínez, C., García-
Agúndez, G., Gaviria, E., Laguna, J.
J., García-Martín, E., et al. (2011).
Cytochrome P450 CYP2B6 geno-
types and haplotypesin a Colombian
population: identification of novel
variant CYP2B6 alleles. Pharmaco-
genet. Genomics 21, 773–778.
Ribaudo, H. J., Haas, D. W., Tierney, C.,
Kim, R. B., Wilkinson, G. R., Gulick,
R. M., etal. (2006). Pharmacogenet-
ics of plasma efavirenz exposure after
treatment discontinuation: an Adult
AIDS Clinical Trials Group Study.
Clin. Infect. Dis. 42, 401–407.
Ribaudo, H. J., Liu, H., Schwab,
M., Schaeffeler, E., Eichelbaum,
M., Motsinger-Reif, A. A., et al.
(2010). Effect of CYP2B6, ABCB1,
and CYP3A5 polymorphisms on
efavirenz pharmacokinetics and
treatment response: an AIDS Clinical
Trials Group study. J. Infect. Dis. 202,
717–722.
Richter, T., Mürdter, T. E., Heinkele,
G., Pleiss, J., Tatzel, S., Schwab,
M., et al. (2004). Potent mechanism-
based inhibition of human CYP2B6
by clopidogrel and ticlopidine. J.
Pharmacol. Exp. Ther. 308, 189–197.
Richter,T., Schwab,M., Eichelbaum, M.,
and Zanger, U. M. (2005). Inhibition
of human CYP2B6 by N,N,N-
triethylenethiophosphoramide
is irreversible and mechanism-
based. Biochem. Pharmacol. 69,
517–524.
Rotger, M., Tegude, H., Colombo, S.,
Cavassini, M., Furrer, H., Décosterd,
L., et al. (2007). Predictive value of
known and novel alleles of CYP2B6
for efavirenz plasma concentrations
in HIV-infected individuals. Clin.
Pharmacol. Ther. 81, 557–566.
Roy, J.-N., Lajoie, J., Zijenah, L. S.,
Barama, A., Poirier, C., Ward, B. J.,
et al. (2005). CYP3A5 genetic poly-
morphisms in different ethnic pop-
ulations. Drug Metab. Dispos. 33,
884–887.
Sadee, W., Wang, D., Papp, A. C.,
Pinsonneault, J. K., Smith, R. M.,
Moyer, R. A., et al. (2011). Pharma-
cogenomics of the RNA world: struc-
tural RNA polymorphisms in drug
therapy. Clin. Pharmacol. Ther. 89,
355–365.
Saussele, T., Burk, O., Blievernicht, J.
K., Klein, K., Nussler, A., Nussler,
N., et al. (2007). Selective induction
of human hepatic cytochromes P450
2B6 and 3A4 by metamizole. Clin.
Pharmacol. Ther. 82, 265–274.
Shah, M. B., Pascual, J., Zhang, Q.,
Stout, C. D.,and Halpert, J. R. (2011).
Structures of cytochrome P450 2B6
bound to 4-benzylpyridine and 4-
(4-nitrobenzyl) pyridine: insight into
inhibitor binding and rearrangement
of active site side chains. Mol. Phar-
macol. 80, 1047–1055.
Simonsson, U. S. H., Jansson, B., Hai,
T. N., Huong, D. X., Tybring, G.,
and Ashton, M. (2003). Artemisinin
autoinduction is caused by involve-
ment of cytochrome P450 2B6 but
not 2C9. Clin. Pharmacol. Ther. 74,
32–43.
Singh, G., Saxena, N., Aggarwal, A.,
and Misra, R. (2007). Cytochrome
P450 polymorphism as a predictor of
ovarian toxicity to pulse cyclophos-
phamide in systemic lupus erythe-
matosus. J. Rheumatol. 34, 731–733.
Smith, G. B. J., Bend, J. R., Bedard,
L. L., Reid, K. R., Petsikas, D., and
Massey, T. E. (2003). Biotransfor-
mation of 4-(methylnitrosamino)-
1-(3-pyridyl)-1-butanone (NNK) in
peripheral human lung microsomes.
Drug Metab. Dispos. 31, 1134–
1141.
Sridar, C., Kenaan, C., and Hol-
lenberg, P. F. (2012). Inhibition
of bupropion metabolism by selegi-
line: mechanism-based inactivation
of human CYP2B6 and character-
ization of glutathione and peptide
adducts. Drug Metab. Dispos. 40,
2256–2266.
Stewart, J. J., Berkel, H. J., Parish, R.
C., Simar, M. R., Syed, A., Boc-
chini, J. A., etal. (2001). Single-dose
pharmacokinetics of bupropion in
adolescents: effects of smoking status
and gender. J. Clin. Pharmacol. 41,
770–778.
Sueyoshi, T., Kawamoto, T., Zelko,
I., Honkakoski, P., and Negishi, M.
(1999). The repressed nuclear recep-
tor CAR responds to phenobarbital in
activating the human CYP2B6 gene.
J. Biol. Chem. 274, 6043–6046.
Svensson, U. S., and Ashton, M.
(1999). Identification of the human
cytochrome P450 enzymes involved
in the in vitro metabolism of
artemisinin. Br. J. Clin. Pharmacol.
48, 528–535.
Takada, K., Arefayene, M., Desta, Z.,
Yarboro, C. H., Boumpas, D. T.,
Balow,J. E., et al. (2004). Cytochrome
P450 pharmacogenetics as a predic-
tor of toxicity and clinical response
to pulse cyclophosphamide in lupus
www.frontiersin.org March 2013 |Volume 4 |Article 24 |11
“fgene-04-00024” 2013/3/12 14:58 page 12 #12
Zanger and Klein Pharmacogenetics of CYP2B6
nephritis. Arthritis Rheum. 50, 2202–
2210.
Talakad, J. C., Kumar, S., and Halpert,
J. R. (2009). Decreased susceptibility
of the cytochrome P450 2B6 variant
K262R to inhibition by several clin-
ically important drugs. Drug Metab.
Dispos. 37, 644–650.
Telenti, A., and Zanger, U. M. (2008).
Pharmacogenetics of anti-HIV drugs.
Annu. Rev. Pharmacol. Toxicol. 48,
227–256.
Thelen, K., and Dressman, J. B.
(2009). Cytochrome P450-mediated
metabolism in the human gut wall. J.
Pharm. Pharmacol. 61, 541–558.
Torimoto, Y., and Kohgo, Y. (2008).
[Cyclophosphamide and CYP2B6].
Gan To Kagaku Ryoho 35, 1090–1093.
Totah, R. A., Sheffels, P., Roberts,
T., Whittington, D., Thummel, K.,
and Kharasch, E. D. (2008). Role
of CYP2B6 in stereoselective human
methadone metabolism. Anesthesiol-
ogy 108, 363–374.
Turpeinen, M., Raunio, H., and Pelko-
nen, O. (2006). The functional role of
CYP2B6 in human drug metabolism:
substrates and inhibitors in vitro, in
vivo and in silico. Curr. Drug Metab.
7, 705–714.
Turpeinen, M., Tolonen, A., Uusitalo, J.,
Jalonen, J., Pelkonen, O., and Laine,
K. (2005a). Effect of clopidogrel and
ticlopidine on cytochrome P450 2B6
activity as measured by bupropion
hydroxylation. Clin. Pharmacol. Ther.
77, 553–559.
Turpeinen, M., Uusitalo, J., Jouko,
U., Jalonen, J., Jorma, J., Pelko-
nen, O., etal. (2005b). Multiple
P450 substrates in a single run: rapid
and comprehensive in vitro interac-
tion assay. Eur. J. Pharm. Sci. 24,
123–132.
Turpeinen, M., and Zanger, U. M.
(2012). Cytochrome P450 2B6: func-
tion, genetics, and clinical relevance.
Drug Metabol. Drug Interact. 27,
185–197.
Walsky, R. L., and Obach, R. S. (2007).
A comparison of 2-phenyl-2-(1-
piperidinyl)propane (ppp), 1,1,1-
phosphinothioylidynetrisaziridine
(thioTEPA), clopidogrel, and ticlo-
pidine as selective inactivators of
human cytochrome P450 2B6. Drug
Metab. Dispos. 35, 2053–2059.
Wang, H., Faucette, S., Sueyoshi,
T., Moore, R., Ferguson, S.,
Negishi, M., et al. (2003). A novel
distal enhancer module regulated
by pregnane X receptor/constitutive
androstane receptor is essential for
the maximal induction of CYP2B6
gene expression. J. Biol. Chem. 278,
14146–14152.
Wang, J., Sönnerborg, A., Rane, A.,
Josephson, F., Lundgren, S., Ståhle, L.,
et al. (2006). Identification of a novel
specific CYP2B6 allele in Africans
causing impaired metabolism of the
HIV drug efavirenz. Pharmacogenet.
Genomics 16, 191–198.
Wang, H., and Tompkins, L. M.
(2008). CYP2B6: new insights into
a historically overlooked cytochrome
P450 isozyme. Curr. Drug Metab. 9,
598–610.
Ward, B. A., Gorski, J. C., Jones, D.
R., Hall, S. D., Flockhart, D. A., and
Desta, Z. (2003). 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. Pharma-
col. Exp. Ther. 306, 287–300.
Watanabe, T., Sakuyama, K., Sasaki,
T., Ishii, Y., Ishikawa, M., Hirasawa,
N., et al. (2010). Functional char-
acterization of 26 CYP2B6 allelic
variants (CYP2B6.2-CYP2B6.28,
except CYP2B6.22). Pharmacogenet.
Genomics 20, 459–462.
Wilderman, P. R., and Halpert, J. R.
(2012). Plasticity of CYP2B enzymes:
structural and solution biophysical
methods. Curr. Drug Metab. 13,
167–176.
Wyen, C., Hendra, H., Siccardi,
M., Platten, M., Jaeger, H., Har-
rer, T., et al. (2011). Cytochrome
P450 2B6 (CYP2B6) and constitutive
androstane receptor (CAR) polymor-
phisms are associated with early dis-
continuation of efavirenz-containing
regimens. J. Antimicrob. Chemother.
66, 2092–2098.
Xie, H., Griskevicius, L., Ståhle, L.,
Hassan, Z., Yasar, U., Rane, A.,
et al. (2006). Pharmacogenetics of
cyclophosphamide in patients with
hematological malignancies. Eur. J.
Pharm. Sci. 27, 54–61.
Xie, H.-J., Yasar, U., Lundgren, S.,
Griskevicius, L., Terelius, Y., Hassan,
M., et al. (2003). Role of polymor-
phic human CYP2B6 in cyclophos-
phamide bioactivation. Pharmacoge-
nomics J. 3, 53–61.
Xu, C., Ogburn, E. T., Guo, Y., and
Desta, Z. (2012). Effects of the
CYP2B6*6 allele on catalytic prop-
erties and inhibition of CYP2B6 in
vitro: implication for the mechanism
of reduced efavirenz metabolism and
other CYP2B6 substrates in vivo.
Drug Metab. Dispos. 40, 717–725.
Yamanaka, H., Nakajima, M., Fukami,
T., Sakai, H., Nakamura, A., Katoh,
M., et al. (2005). CYP2A6 AND
CYP2B6 are involved in nornicotine
formation from nicotine in humans:
interindividual differences in these
contributions. Drug Metab. Dispos.
33, 1811–1818.
Yamazaki, H., Inoue, K., Hashimoto,
M., and Shimada, T. (1999). Roles
of CYP2A6 and CYP2B6 in nicotine
C-oxidation by human liver micro-
somes. Arch. Toxicol. 73, 65–70.
Yang, X., Zhang, B., Molony, C., Chudin,
E., Hao, K., Zhu, J., etal. (2010). Sys-
tematic genetic and genomic analysis
of cytochrome P450 enzyme activi-
ties in human liver. Genome Res. 20,
1020–1036.
Yao, S., Barlow,W.E., Albain, K. S., Choi,
J.-Y., Zhao, H., Livingston, R. B.,
et al. (2010). Gene polymorphisms in
cyclophosphamide metabolism path-
way, treatment-related toxicity, and
disease-free survival in SWOG 8897
clinical trial for breast cancer. Clin.
Cancer Res. 16, 6169–6176.
Yimer, G., Amogne, W., Habtewold, A.,
Makonnen, E., Ueda, N., Suda, A.,
et al. (2011). High plasma efavirenz
level and CYP2B6*6 are associ-
ated with efavirenz-based HAART-
induced liver injury in the treatment
of naïve HIV patients from Ethiopia:
a prospective cohort study. Pharma-
cogenomics J. 12, 499–506.
Yuan, J., Guo, S., Hall, D., Cam-
mett, A. M., Jayadev, S., Distel,
M., et al. (2011). Toxicogenomics
of nevirapine-associated cutaneous
and hepatic adverse events among
populations of African, Asian, and
European descent. AIDS 25, 1271–
1280.
Zanger, U. M., and Hofmann, M.
H. (2008). Polymorphic cytochromes
P450 CYP2B6 and CYP2D6: recent
advances on single nucleotide poly-
morphisms affecting splicing. Acta
Chim. Slov. 55, 38.
Zanger, U. M., Klein, K., Saussele, T.,
Blievernicht, J., Hofmann, M. H.,
and Schwab, M. (2007). Polymorphic
CYP2B6: molecular mechanisms and
emerging clinical significance. Phar-
macogenomics 8, 743–759.
Zhang, H., Amunugama, H., Ney,
S., Cooper, N., and Hollenberg, P.
F. (2011a). Mechanism-based inac-
tivation of human cytochrome P450
2B6 by clopidogrel: involvement of
both covalent modification of cys-
teinyl residue 475 and loss of heme.
Mol. Pharmacol. 80, 839–847.
Zhang, H., Sridar, C., Kenaan, C.,
Amunugama, H., Ballou, D. P., and
Hollenberg, P. F. (2011b). Polymor-
phic variants of cytochrome P450
2B6 (CYP2B6.4-CYP2B6.9) exhibit
altered rates of metabolism for
bupropion and efavirenz: a charge-
reversal mutation in the K139E vari-
ant (CYP2B6.8) impairs formation
of a functional cytochrome p450-
reductase complex. J. Pharmacol.
Exp. Ther. 338, 803–809.
Zukunft, J., Lang, T., Richter,T., Hirsch-
Ernst, K. I., Nussler, A. K., Klein, K.,
et al. (2005). A natural CYP2B6 TATA
box polymorphism (-82T–>C) lead-
ing to enhanced transcription and
relocation of the transcriptional start
site. Mol. Pharmacol. 67, 1772–
1782.
Conflict of Interest Statement: Ulrich
M. Zanger is a named coinventor of
a patent on the detection of specific
CYP2B6 polymorphisms for diagnos-
tic purposes. Kathrin Klein declares no
conflict of interest.
Received: 19 October 2012; accepted:
14 February 2013; published online: 05
March 2013.
Citation: Zanger UM and Klein K (2013)
Pharmacogenetics of cytochrome P450
2B6 (CYP2B6): advances on polymor-
phisms, mechanisms, and clinical rele-
vance. Front. Genet. 4:24. doi: 10.3389/
fgene.2013.00024
This article was submitted to Frontiers
in Pharmacogenetics and Pharmacoge-
nomics, a specialty of Frontiers in Genet-
ics.
Copyright © 2013 Zanger and Klein.
This is an open-access article distributed
under the terms of the Creative Commons
Attribution License, which permits use,
distribution and reproduction in other
forums, provided the original authors and
source are credited and subject to any
copyright notices concerning any third-
party graphics etc.
Frontiers in Genetics |Pharmacogenetics and Pharmacogenomics March 2013 |Volume 4 |Article 24 |12
... CYP2B6 accounts for 2-5% of total CYP content in liver and is thought to metabolize about 8% of clinical drugs, including bupropion, EFV, nevirapine, meperidine, methadone, ketamine, propofol, mephobarbital, and cyclophosphamide [15][16][17]. Owing to genetic variants, inducibility or inhibition by many compounds, CYP2B6 activity is highly variable between individuals. Some variants of CYP2B6 affect transcriptional regulation, splicing, protein expression and thus catalytic activity. ...
... Due to differences among expression systems and substrates used in vitro enzymatic activity analysis may produce inconsistent results [17,31,32]. For example, the CYP2B6*6 allele is associated with enhanced cyclophosphamide metabolism, but appears to confer decreased EFV metabolism [17,33]. ...
... Due to differences among expression systems and substrates used in vitro enzymatic activity analysis may produce inconsistent results [17,31,32]. For example, the CYP2B6*6 allele is associated with enhanced cyclophosphamide metabolism, but appears to confer decreased EFV metabolism [17,33]. (S)-methadone plasma levels in CYP2B6*6/*6 carriers were increased and resulted in methadone associated deaths or higher risk of severe cardiac arrhythmias [34,35]. ...
Article
Full-text available
Amino acid variants in protein may result in deleterious effects on enzymatic activity. In this study we investigate the DNA variants on activity of CYP2B6 gene in a Chinese Han population for potential use in precision medicine. All exons in CYP2B6 gene from 1483 Chinese Han adults (Zhejiang province) were sequenced using Sanger sequencing. The effects of nonsynonymous variants on recombinant protein catalytic activity were investigated in vitro with Sf12 system. The haplotype of novel nonsynonymous variants with other single nucleotide variants in the same allele was determined using Nanopore sequencing. Of 38 alleles listed on the Pharmacogene Variation Consortium, we detected 7 previously reported alleles and 18 novel variants, of which 11 nonsynonymous variants showed lower catalytic activity (0.00–0.60) on bupropion compared to CYP2B6*1. Further, these 11 novel star-alleles (CYP2B6*39–49) were assigned by the Pharmacogene Variation Consortium, which may be valuable for pharmacogenetic research and personalized medicine.
... Te gene for CYP2B6 is located in the middle of chromosome 19 [63] and is mainly expressed in the liver; however, it has been detected in extrahepatic tissues such as the intestines, kidneys, brain, lungs, and skin [64][65][66][67]. CYP2B6 has been reported to be involved in the metabolism of many other drugs including ifosfamide, diazepam, and efavirenz as well as in the synthesis of endogenous compounds such as cholesterol and steroids [65,[68][69][70]. ...
Article
Full-text available
Cyclophosphamide (Cy) is a prodrug that is mainly bioactivated by cytochrome P450 (CYP) 2B6 enzyme. Several other enzymes are also involved in its bioactivation and affect its kinetics. Previous studies have shown the effect of the enzymes’ genetic polymorphisms on Cy kinetics and its clinical outcome. These results were controversial primarily because of the involvement of several interacting enzymes in the Cy metabolic pathway, which can also be affected by several clinical factors as well as other drug interactions. In this review article, we present the effect of CYP2B6 polymorphisms on Cy kinetics since it is the main bioactivating enzyme, as well as discussing all previously reported enzymes and clinical factors that can alter Cy efficacy. Additionally, we present explanations for key Cy side effects related to the nature and site of its bioactivation. Finally, we discuss the role of busulphan in conditioning regimens in the Cy metabolic pathway as a clinical example of drug-drug interactions involving several enzymes. By the end of this article, our aim is to have provided a comprehensive summary of Cy pharmacogenomics and the effect on its kinetics. The utility of these findings in the development of new strategies for Cy personalized patient dose adjustment will aid in the future optimization of patient specific Cy dosages and ultimately in improving clinical outcomes. In conclusion, CYP2B6 and several other enzyme polymorphisms can alter Cy kinetics and consequently the clinical outcomes. However, the precise quantification of Cy kinetics in any individual patient is complex as it is clearly under multifactorial genetic control. Additionally, other clinical factors such as the patient’s age, diagnosis, concomitant medications, and clinical status should also be considered.
... Te gene for CYP2B6 is located in the middle of chromosome 19 [63] and is mainly expressed in the liver; however, it has been detected in extrahepatic tissues such as the intestines, kidneys, brain, lungs, and skin [64][65][66][67]. CYP2B6 has been reported to be involved in the metabolism of many other drugs including ifosfamide, diazepam, and efavirenz as well as in the synthesis of endogenous compounds such as cholesterol and steroids [65,[68][69][70]. ...
... Senescence pathways may vary according to organs and cell types so there is a need to identify cell-specific molecu- enzymes that are primarily involved in the biotransformation of drugs and xenobiotics in the liver (Zanger & Klein, 2013). Cyp2b10 is only present in rodents and the orthologous gene is Cyp2b6 ...
Article
Full-text available
The aging heart is characterized by a number of structural changes leading to ventricular stiffness, impaired resistance to stress and increased risk of developing heart failure (HF). Genetic or pharmacological removal of senescent cells has recently demonstrated the possibility to relieve some cardiac aging features such as hypertrophy and fibrosis. However, the contribution of the different cell types in cardiac aging remains fragmentary due to a lack of cell‐specific markers. Cardiomyocytes undergo post‐mitotic senescence in response to telomere damage, characterized by persistent DNA damage response and expression of the classical senescence markers p21 and p16, which are shared by many other cell types. In the present study, we used transcriptomic approaches to discover new markers specific for cardiomyocyte senescence. We identified Prominin2 (Prom2), encoding a transmembrane glycoprotein, as the most upregulated gene in cardiomyocytes of aged mice compared to young mice. We showed that Prom2 was upregulated by a p53‐dependent pathway in stress‐induced premature senescence. Prom2 expression correlated with cardiomyocyte hypertrophy in the hearts of aged mice and was increased in atrial samples of patients with HF with preserved ejection fraction. Consistently, Prom2 overexpression was sufficient to drive senescence, hypertrophy and resistance to cytotoxic stress while Prom2 shRNA silencing inhibited these features in doxorubicin‐treated cardiac cells. In conclusion, we identified Prom2 as a new player of cardiac aging, linking cardiomyocyte hypertrophy to senescence. These results could provide a better understanding and targeting of cell‐type specific senescence in age‐associated cardiac diseases.
... Cyclophosphamide is an alkylating anticancer prodrug, which requires enzymatic activation in the liver. Carriers of CYP2B6*9 are considered to have worse pharmacotherapeutic outcomes from treatment with cyclophosphamide [16]. Determination of the CYP2B6 genotype of these patients could improve their pharmacotherapeutic effects and anticancer therapy outcomes. ...
Article
Full-text available
The cytochrome P450 superfamily consists of hemeproteins involved in the detoxication of different xenobiotics, including drugs. The CYP2 gene family is responsible for the metabolism of 80% of the drugs in clinical use. There are considerable interindividual and interethnic variabilities in the rate of drug metabolism as a result of genetic polymorphisms. The goal of our study was to determine the frequency of 10 genetic polymorphisms in CYP2 family genes to give light on the pharmacogenetic defects of the main CYPs, involved in drug metabolism, in Bulgarian individuals. We detected high allele frequency for CYP2D6*10 (0.27), CYP2D6*4 (0.22), and CYP2B6*9 (0.24), followed by CYP2C19*2 (0.14), CYP2C9*3 (0.11) and CYP2C9*2 (0.09). The genotype frequencies were also determined for all investigated variants. In total 47.2% of the analyzed individuals carried CYP2D6 genetic polymorphisms – 5.6% carried a single variant and 41.6% were found to have two or more such variants. Homozygotes for CYP2D6 variants were established among 14% of Bulgarian individuals. Determination of the prevailing pharmacogenetic polymorphisms of the CYPs, most responsible for drug metabolism, will lead to a lower risk of drug toxicity, increased drug efficacy, and drug dose optimization.
Article
Objectives Understanding the influence of fetal and maternal genetics on prenatal drug exposure could potentially improve benefit-risk evaluation. In this study, we investigated the impact of two functional polymorphisms in CYP2B6 on prenatal exposure to efavirenz. Methods Dried blood spot (DBS) samples were collected from HIV-positive pregnant women ( n = 112) and their newborns ( n = 107) at delivery. They were genotyped for single nucleotide polymorphisms in CYP2B6. Efavirenz was quantified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Results Significant correlations were observed in efavirenz concentration between maternal and newborn ( r = 0.46, R ² = 0.21, P < 0.001), and maternal and cord ( r = 0.83, R ² = 0.68, P < 0.001) samples. Median (interquartile range) newborn plasma-to-maternal plasma and cord-to-maternal plasma ratios were 0.85 (0.03–3.49) and 0.78 (0.23–1.96), respectively. Newborn efavirenz concentration in DBS varied significantly based on composite maternal CYP2B6 genotype: fast ( CYP2B6 516GG and 983TT, n = 26), 747 ng/ml (602–1060); intermediate ( CYP2B6 516GT or 983TC n = 50), 1177 ng/ml (898–1765); and slow ( CYP2B6 516GT and 983TC or 516TT or 983CC, n = 14), 3094 ng/ml (2126–3812). Composite newborn CYP2B6 genotype was, however, not significantly associated with prenatal exposure. Efavirenz concentration in newborn stratified as fast ( n = 25), intermediate ( n = 36), and slow metabolizers ( n = 19) from prenatal exposure was 999.7 (774–1285), 1240 (709–1984), and 1792 ng/ml (1201–3188), respectively. Conclusion The clinical relevance of the observed influence of maternal genetics on prenatal efavirenz exposure requires further investigation.
Article
The efficacy and tolerability of psychotropic medications can vary significantly among children and adolescents, and some of this variability relates to pharmacogenetic factors. Pharmacogenetics (PGx) in child and adolescent psychiatry can potentially improve treatment outcomes and minimize adverse drug reactions. This article reviews key pharmacokinetic and pharmacodynamic genes and principles of pharmacogenetic testing and discusses the evidence base for clinical decision-making concerning PGx testing. This article reviews current guidelines from the United States Food and Drug Administration (FDA), the Clinical Pharmacogenetics Implementation Consortium (CPIC), and the Dutch Pharmacogenetics Working Group (DPWG) and explores potential future directions. This review discusses key clinical considerations for clinicians prescribing psychotropic medications in children and adolescents, focusing on antidepressants, antipsychotics, stimulants, norepinephrine reuptake inhibitors, and alpha-2 agonists. Finally, this review synthesizes the practical use of pharmacogenetic testing and clinical decision support systems.
Article
Full-text available
Cytochromes P450 (CYPs) are a group of monooxygenases that can be found in almost all kinds of organisms. For CYPs to receive electrons from co-substrate NADPH, the activity of NADPH-Cytochrome-P450-oxidoreductase (CPR) is required as well. In humans, CYPs are an integral part of liver-based phase-1 biotransformation, which is essential for the metabolization of multiple xenobiotics and drugs. Consequently, CYPs are important players during drug development and therefore these enzymes are implemented in diverse screening applications. For these applications it is usually advantageous to use mono CYP microsomes containing only the CYP of interest. The generation of mono-CYP containing mammalian cells and vesicles is difficult since endogenous CYPs are present in many cell types that contain the necessary co-factors. By obtaining translationally active lysates from a modified CHO-CPR cell line, it is now possible to generate mono CYPs in a cell-free protein synthesis process in a straightforward manner. As a proof of principle, the synthesis of active human CYPs from three different CYP450 gene families (CYP1A2, CYP2B6 and CYP3A4), which are of outstanding interest in industry and academia was demonstrated. Luciferase based activity assays confirm the activity of the produced CYPs and enable the individual adaptation of the synthesis process for efficient cell-free enzyme production. Furthermore, they allow for substrate and inhibitor screenings not only for wild-type CYPs but also for mutants and further CYP isoforms and variants. As an example, the turnover of selected CYP substrates by cell-free synthesized CYPs was demonstrated via an indirect luciferase assay-based screening setup.
Chapter
Mental health disorders are prevalent, complex, and difficult to treat illnesses. Psychopharmacotherapy is the cornerstone of their treatment. Drug selection is still heuristic, due to the lack of reliable biological biomarkers to predict treatment response. Genetic variants of pharmacokinetics and pharmacodynamic genes contribute to a quarter of total drug response variability across the population. The promise of pharmacogenomics in psychiatry is a better prediction of psychotropic drug response. This chapter will focus on genetic variants with demonstrated clinical impact related to antidepressants, antipsychotics, mood stabilizers, and other psychotropics.
Article
Full-text available
Recent work on functional polymorphisms in drug metabolizing cytochromes P450 CYP2B6 and CYP2D6 emphasizes the role of single nucleotide polymorphisms (SNPs) in non-consensus splicing elements such as exonic and intronic splicing enhancers. In the CYP2D6 gene the intron 6 SNP 2988G > A (allele *41) shifts the balance of spliced transcripts towards a variant that lacks exon 6. In the CYP2B6 gene, the 516G > T SNP, a marker of allele CYP2B6*6, encodes an amino acid change in exon 4 [Q172H] but also leads to increased amounts of a transcript lacking exons 4 to 6. In both cases aberrant splicing results in reduced amounts of functional transcript and reduced amounts of functional protein in the liver. Although expression of functional protein is only partially diminished, reduced activity phenotypes arise in homozygous genotypes or in compound heterozygotes carrying other severely affected alleles. We here describe the elucidation of these genetic polymorphisms and their mechanisms as well as their clinical relevance.
Article
Full-text available
To determine the role of CYP450 copy number variation (CNV) beyond CYP2D6, 11 CYP450 genes were interrogated by multiplex ligation-dependent probe amplification and quantitative PCR in 542 African-American, Asian, Caucasian, Hispanic and Ashkenazi Jewish individuals. The CYP2A6, CYP2B6 and CYP2E1 combined deletion/duplication allele frequencies ranged from 2 to 10% in these populations. High-resolution microarray-based comparative genomic hybridization (aCGH) localized CYP2A6, CYP2B6 and CYP2E1 breakpoints to directly oriented low-copy repeats. Sequencing localized the CYP2B6 breakpoint to a 529-bp intron 4 region with high homology to CYP2B7P1, resulting in the CYP2B6*29 partial deletion allele and the reciprocal, and novel, CYP2B6/2B7P1 duplicated fusion allele (CYP2B6*30). Together, these data identified novel CYP450 CNV alleles (CYP2B6*30 and CYP2E1*1Cx2) and indicate that common CYP450 CNV formation is likely mediated by non-allelic homologous recombination resulting in both full gene and gene-fusion copy number imbalances. Detection of these CNVs should be considered when interrogating these genes for pharmacogenetic drug selection and dosing.The Pharmacogenomics Journal advance online publication, 20 November 2012; doi:10.1038/tpj.2012.48.
Article
Full-text available
Selegiline, the R-enantiomer of deprenyl, is used in the treatment of Parkinson's disease. Bupropion, an antidepressant, often used to treat patients in conjunction with selegiline, is metabolized primarily by CYP2B6. The effect of selegiline on the enzymatic activity of human cytochrome CYP2B6 in a reconstituted system and its effect on the metabolism of bupropion were examined. Selegiline was found to be a mechanism-based inactivator of the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation (7EFC) activity of CYP2B6 as well as bupropion metabolism. The inactivations were time-, concentration-, and NADPH-dependent and were characterized by K(I) values of 0.14 μM and 0.6 μM, k(inact) values of 0.022 min(-1) and 0.029 min(-1) and t(1/2) values of 31.5 and 24 min, respectively. In standard inhibition assays, selegiline increased the K(m) of CYP2B6 for bupropion from 10 to 92 μM and decreased the k(cat) by about 50%. The reduced carbon-monoxide difference spectrum revealed over a 50% loss in the P450 spectrum in the inactivated sample with no loss in heme and there was about 70% loss in enzyme activity. Trapping of the reactive metabolite using glutathione (GSH) led to the identification of a GSH-selegiline conjugate with a m/z 528 that could be explained by hydroxylation of selegiline followed by the addition of glutathione to the propargyl moiety after oxygenation to form the ketene intermediate. LC-MS/MS analysis of the labeled protein following digestion with trypsin revealed the peptide (64)DVFTVHLGPR(73) as the peptide modified by the reactive metabolite of selegiline and the site of adduct formation is Asp64.
Article
Cytochrome P450 CYP2B6 is a highly polymorphic enzyme that metabolizes numerous drugs, pesticides, and environmental toxins. Sequence analysis of a Rwandese population identified eight functionally uncharacterized non-synonymous variants c.329G>T (p.G110V), c.341T>C (p.I114T), c.444G>T (p.E148D), c.548T>G (p.V183G), c.637T>C (p.F213L), c.758G>A (p.R253H), c.835G>C (p.A279P), c.1459C>A (p.R487S) and five novel alleles termed CYP2B6*33 to CYP2B6*37 were assigned. Recombinant expression in COS-1 cells and functional characterization using the antidepressant bupropion and the antiretroviral efavirenz as substrates demonstrated complete or almost complete loss-of-function for variants p.G110V, p.I114T, p.V183G, and p.F213L, while p.E148D, p.R253H, p.A279P, and p.R487S variants were functional. The data were used to assess the predictive power of eight online available functional prediction programs for amino acid changes. While none of the programs correctly predicted the functionality of all variants, substrate docking simulation analyses indicated similar conformational changes by all four deleterious mutations within the enzyme's active site, thus explaining lack of enzymatic function for both substrates. Because low-activity alleles of CYP2B6 are associated with impaired efavirenz metabolism and adverse drug response, these results are of potential utility for personalized treatment strategies in HIV/AIDS therapy.
Article
Abstract Cytochrome P450 (CYP) 2B6 belongs to the set of important hepatic drug-metabolizing CYPs. It makes up roughly 3%-6% of total hepatic CYP content and metabolizes several pharmaceuticals including bupropion, efavirenz, cyclophosphamide, pethidine, ketamine and propofol. The enzyme is susceptible to drug-drug interactions by enzyme induction and inhibition. In addition to drugs, CYP2B6 is able to both detoxify and bioactivate a number of procarcinogens and environmental agents including pesticides and herbicides. There is an extensive interindividual variability in the expression of CYP2B6, which is in part explained by extensive genetic polymorphism. CYP2B6 is one of the most polymorphic CYP genes in humans with over 100 described SNPs, numerous complex haplotypes and distinct ethnic and racial frequencies. This review summarizes the basic properties of CYP2B6 and the main characteristics of clinical relevance.
Article
Background and objective: Recent in vitro studies have suggested an important role of cytochrome P450 (CYP) 2B6 and CYP2C19 in methadone metabolism. We aimed to determine the influence of CYP2B6, CYP2C9, and CYP2C19 genetic polymorphism on methadone pharmacokinetics and on the response to treatment. Methods: We included 209 patients in methadone maintenance treatment on the basis of their response to treatment and their daily methadone dose. Patients were genotyped for CYP2B6, CYP2C9, and CYP2C19. Steady-state trough and peak (R)-, (S)-, and (R,S)-plasma levels and peak-to-trough plasma level ratios were measured. Results: CYP2B6 genotype influences (S)-methadone and, to a lesser extent, (R)-methadone plasma levels, with the median trough (S)-methadone plasma levels being 105, 122, and 209 ng . kg/mL . mg for the noncarriers of allele *6, heterozygous carriers, and homozygous carriers (*6/*6), respectively (P = .0004). CYP2C9 and CYP2C19 genotypes do not influence methadone plasma levels. Lower peak and trough plasma levels of methadone and higher peak-to-trough ratios were measured in patients considered as nonresponders [median (R,S)-methadone trough plasma levels of 183 and 249 ng . kg/mL . mg (P = .0004) and median peak-to-trough ratios of 1.82 and 1.58 for high-dose nonresponders and high-dose responders, respectively (P = .0003)]. Conclusion: Although CYP2B6 influences (S)-methadone plasma levels, given that only (R)-methadone contributes to the opioid effect of this drug, a major influence of CYP2B6 genotype on response to treatment is unlikely and has not been shown in this study. Lower plasma levels of methadone in nonresponders, suggesting a higher clearance, and higher peak-to-trough ratios, suggesting a shorter elimination half-life, are in agreement with the usual clinical measures taken for such patients, which are to increase methadone dosages and to split the daily dose into several intakes.
Article
Objectives: Prior candidate gene studies have associated CYP2B6 516G→T [rs3745274] and 983T→C [rs28399499] with increased plasma efavirenz exposure. We sought to identify novel variants associated with efavirenz pharmacokinetics. Materials and methods: Antiretroviral therapy-naive AIDS Clinical Trials Group studies A5202, A5095, and ACTG 384 included plasma sampling for efavirenz pharmacokinetics. Log-transformed trough efavirenz concentrations (Cmin) were previously estimated by population pharmacokinetic modeling. Stored DNA was genotyped with Illumina HumanHap 650Y or 1MDuo platforms, complemented by additional targeted genotyping of CYP2B6 and CYP2A6 with MassARRAY iPLEX Gold. Associations were identified by linear regression, which included principal component vectors to adjust for genetic ancestry. Results: Among 856 individuals, CYP2B6 516G→T was associated with efavirenz estimated Cmin (P=8.5×10). After adjusting for CYP2B6 516G→T, CYP2B6 983T→C was associated (P=9.9×10). After adjusting for both CYP2B6 516G→T and 983T→C, a CYP2B6 variant (rs4803419) in intron 3 was associated (P=4.4×10). After adjusting for all the three variants, non-CYP2B6 polymorphisms were associated at P-value less than 5×10. In a separate cohort of 240 individuals, only the three CYP2B6 polymorphisms replicated. These three polymorphisms explained 34% of interindividual variability in efavirenz estimated Cmin. The extensive metabolizer phenotype was best defined by the absence of all three polymorphisms. Conclusion: Three CYP2B6 polymorphisms were independently associated with efavirenz estimated Cmin at genome-wide significance, and explained one-third of interindividual variability. These data will inform continued efforts to translate pharmacogenomic knowledge into optimal efavirenz utilization.