dna methylation of adme genes - u-pgxupgx.eu/wp-content/uploads/2017/04/fisel...dna methylation,...

16
DNA Methylation of ADME Genes P Fisel 1,2 , E Schaeffeler 1,2 and M Schwab 1,3,4 The epigenetic regulation of expression of genes involved in the absorption, distribution, metabolism, and excretion (ADME) of drugs contributes to interindividual variability in drug response. Epigenetic mechanisms include DNA methylation, histone modifications, and miRNAs. This review systematically outlines the influence of DNA methylation on ADME gene expression and highlights the consequences for interindividual variability in drug response or drug-induced toxicity and the implications for personalized medicine. ADME GENES AND INTERINDIVIDUAL VARIABILITY OF DRUG RESPONSE Approximately 300 genes contribute to the ADME of xenobiotics, including the majority of drugs (http://pharmaadme.org/). Interin- dividual variability in expression and function of ADME genes are known to affect the individual response of patients to drugs with respect to therapeutic efficacy as well as adverse drug reactions (ADR). The heterogeneity in expression and function of ADME genes among individuals is determined by the interaction of genetic and nongenetic factors, as well as epigenetic modifications. 1 The influence of genetic variation in ADME genes and of nongenetic host factors, e.g., sex, age, disease state, or drug exposure, on drug response is the concept of personalized medicine. Meanwhile, major efforts are spent on implementing the pharmacogenomic (PGx) knowledge into clinical practice (http://www.fda.gov/drugs/ scienceresearch/researchareas/pharmacogenetics/ucm083378.htm; https://www.pharmgkb.org/page/cpic). PGx information is also part of labels of drugs used in various medical disciplines. Of note, about 70% of PGx information for drugs approved by the European Medicines Agency (EMA) between 1995 and 2014 is related to ADME processes. 2 In contrast, the significance of epige- netic regulation of gene expression in the context of drug response and ADME is just evolving and currently not part of drug labels. The term “epigenetics” in general refers to heritable changes of gene expression without underlying changes in the encoding DNA sequence. 3 Epigenetic modifications regulating gene expres- sion include DNA methylation, histone modification, and non- coding RNAs (miRNAs). 4 The interaction of these mechanisms forms a complex network and determines developmental or dif- ferentiation stage-specific expression patterns. In addition, the interplay of epigenetic modifications contributes to the regulation of tissue- and cell type-specific gene expression. Epigenetic modi- fications are dynamic and may change over time or in response to stimuli. 3–5 They are influenced by patient characteristics (e.g., gender, ethnicity), environment, lifestyle, and disease state or concomitant medication and contribute to individual pheno- types. Pharmacoepigenetics, in particular, address the epigenetic basis of interindividual variability of ADME genes, thereby con- tributing to the elucidation of unknown reasons for interindivid- ual variability in drug response. Furthermore, epigenetic regulation of ADME genes can affect endogenous metabolism and deregulation can lead to the pathogenesis of various diseases. Recently, over 60 ADME genes have been considered to be influ- enced by histone modifications, DNA methylation, or miR- NAs 6,7 and evidence for an epigenetic component for the regulation of ADME gene expression has been rising ever since. Best evidence does exist for findings related to in vitro experi- ments using cell lines treated with the DNA methyltransferase (DNMT) inhibitor 5-aza-2 0 -deoxycytidine or the histone deace- tylase (HDAC) inhibitor trichostatin A. Moreover, these findings are supported and/or supplemented by data from cancer tissues, where epigenetic deregulation of gene expression is a common phenomenon. 8 However, the clinical relevance of epigenetic regu- lation by DNA methylation on ADME gene expression and implications for drug safety and efficacy is so far limited. This review provides a summary and update of the currently available knowledge of the impact of DNA methylation on ADME genes, with particular focus on consequences for drug response. DNA METHYLATION One of the three major epigenetic mechanisms for the regulation of gene expression is DNA methylation. DNA methylation is a stable, covalent addition of a methyl (CH 3 ) group, occurring at 5 0 Cs of cytosines primarily within CpG dinucleotides (5-mC). The modification is catalyzed either by de novo DNA 1 Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; 2 University of Tubingen, Tubingen, Germany; 3 Department of Clinical Pharmacology, University Hospital Tubingen, Tubingen, Germany; 4 Department of Pharmacy and Biochemistry, University of Tubingen, Tubingen, Germany. Correspondence: M Schwab ([email protected]) Received 4 December 2015; accepted 20 January 2016; advance online publication 27 January 2016. doi:10.1002/cpt.343 512 VOLUME 99 NUMBER 5 | MAY 2016 | www.wileyonlinelibrary/cpt REVIEWS

Upload: others

Post on 30-Jul-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

DNA Methylation of ADME GenesP Fisel1,2, E Schaeffeler1,2 and M Schwab1,3,4

The epigenetic regulation of expression of genes involved in the absorption, distribution, metabolism, and excretion (ADME)of drugs contributes to interindividual variability in drug response. Epigenetic mechanisms include DNA methylation,histone modifications, and miRNAs. This review systematically outlines the influence of DNA methylation on ADME geneexpression and highlights the consequences for interindividual variability in drug response or drug-induced toxicity and theimplications for personalized medicine.

ADME GENES AND INTERINDIVIDUAL VARIABILITYOF DRUG RESPONSEApproximately 300 genes contribute to the ADME of xenobiotics,including the majority of drugs (http://pharmaadme.org/). Interin-dividual variability in expression and function of ADME genes areknown to affect the individual response of patients to drugs withrespect to therapeutic efficacy as well as adverse drug reactions(ADR). The heterogeneity in expression and function of ADMEgenes among individuals is determined by the interaction of geneticand nongenetic factors, as well as epigenetic modifications.1 Theinfluence of genetic variation in ADME genes and of nongenetichost factors, e.g., sex, age, disease state, or drug exposure, on drugresponse is the concept of personalized medicine. Meanwhile,major efforts are spent on implementing the pharmacogenomic(PGx) knowledge into clinical practice (http://www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/ucm083378.htm;https://www.pharmgkb.org/page/cpic). PGx information is alsopart of labels of drugs used in various medical disciplines. Ofnote, about 70% of PGx information for drugs approved by theEuropean Medicines Agency (EMA) between 1995 and 2014 isrelated to ADME processes.2 In contrast, the significance of epige-netic regulation of gene expression in the context of drug responseand ADME is just evolving and currently not part of drug labels.The term “epigenetics” in general refers to heritable changes of

gene expression without underlying changes in the encodingDNA sequence.3 Epigenetic modifications regulating gene expres-sion include DNA methylation, histone modification, and non-coding RNAs (miRNAs).4 The interaction of these mechanismsforms a complex network and determines developmental or dif-ferentiation stage-specific expression patterns. In addition, theinterplay of epigenetic modifications contributes to the regulationof tissue- and cell type-specific gene expression. Epigenetic modi-

fications are dynamic and may change over time or in responseto stimuli.3–5 They are influenced by patient characteristics(e.g., gender, ethnicity), environment, lifestyle, and disease stateor concomitant medication and contribute to individual pheno-types. Pharmacoepigenetics, in particular, address the epigeneticbasis of interindividual variability of ADME genes, thereby con-tributing to the elucidation of unknown reasons for interindivid-ual variability in drug response. Furthermore, epigeneticregulation of ADME genes can affect endogenous metabolismand deregulation can lead to the pathogenesis of various diseases.Recently, over 60 ADME genes have been considered to be influ-enced by histone modifications, DNA methylation, or miR-NAs6,7 and evidence for an epigenetic component for theregulation of ADME gene expression has been rising ever since.Best evidence does exist for findings related to in vitro experi-ments using cell lines treated with the DNA methyltransferase(DNMT) inhibitor 5-aza-20-deoxycytidine or the histone deace-tylase (HDAC) inhibitor trichostatin A. Moreover, these findingsare supported and/or supplemented by data from cancer tissues,where epigenetic deregulation of gene expression is a commonphenomenon.8 However, the clinical relevance of epigenetic regu-lation by DNA methylation on ADME gene expression andimplications for drug safety and efficacy is so far limited. Thisreview provides a summary and update of the currently availableknowledge of the impact of DNA methylation on ADME genes,with particular focus on consequences for drug response.

DNA METHYLATIONOne of the three major epigenetic mechanisms for the regulationof gene expression is DNA methylation. DNA methylation is astable, covalent addition of a methyl (CH3) group, occurring at50Cs of cytosines primarily within CpG dinucleotides (5-mC).The modification is catalyzed either by de novo DNA

1Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; 2University of T€ubingen, T€ubingen, Germany; 3Department of ClinicalPharmacology, University Hospital T€ubingen, T€ubingen, Germany; 4Department of Pharmacy and Biochemistry, University of T€ubingen, T€ubingen, Germany.Correspondence: M Schwab ([email protected])

Received 4 December 2015; accepted 20 January 2016; advance online publication 27 January 2016. doi:10.1002/cpt.343

512 VOLUME 99 NUMBER 5 | MAY 2016 | www.wileyonlinelibrary/cpt

REVIEWS

Page 2: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

methyltransferases (DNMT3a and DNMT3b) during develop-ment or by maintenance DNA methyltransferases (DNMT1)during DNA replication. In general, DNA methylation in pro-moter regions of genes suppresses their expression by directlyinhibiting the binding of transcription factors or indirectly byrecruiting methyl-CpG-binding proteins, which in turn modulatechromatin structure (Figure 1).4 DNA methylation marks areheritable and can be transmitted transgenerationally.9 Despiteheritability and stability, DNA methylation represents a dynamicprocess which changes throughout the course of a lifetime and inresponse to environmental stimuli and therefore contributes tophenotypic variation in humans. This phenomenon is best corro-borated by studies examining genetically identical monozygotictwins, which show similar epigenetic marks during younger agescompared to older age, when remarkable epigenetic differencescan be observed.10,11

Aberrant DNA methylation is associated with the pathogenesisand treatment response of cancer and various other diseases, e.g.,psychiatric or autoimmune diseases,8 and also affects expression

of ADME genes. With respect to drug response, the dynamicnature of DNA methylation represents a major challenge. Thus,age dependency of DNA methylation and subsequent alterationof ADME gene expression need to be taken into consideration,especially in subpopulations such as children and the elderly. Atthe beginning of life and even in preterm infants there is convinc-ing evidence that expression and function of drug metabolizingenzymes (DME) are significantly reduced compared to adults.For instance, CYP3A4 protein levels increase very gradually dur-ing the first year of life, and even 5–15-year-old children showsignificantly lower CYP3A4 levels compared with adults.12

Related to drug transporters, a similar picture can beassumed.13,14 Independent from other factors, there is first evi-dence that differences in DNA methylation patterns may con-tribute to the ontogeny of the expression of DME inchildhood.15 A higher risk for ADRs is well established in olderpatients, which may not only be related to, e.g., different bodycomposition and impaired organ function due to underlying dis-eases as well as polypharmacy, but also to the reduced capability

Figure 1 Epigenetic regulation of ADME gene expression by DNA methylation and DNA hydroxymethylation. Unmethylated DNA in gene promoters allowsbinding of transcription factors (TF) and coactivators (CoA) leading to active gene transcription. DNA methylation, i.e., the covalent addition of a CH3-groupto 50Cs of cytosines within CpG dinucleotides, of gene promoters is catalyzed by DNA methyltransferases (DNMT) and leads to suppression of geneexpression. 5-methylcytosine (5-mC) underlies oxidative DNA demethylation catalyzed by members of the ten-eleven translocation (TET) enzyme family,resulting in hydroxymethylated DNA (5-hmC). Further intermediates of the DNA demethylation process include 5-formylcytosine (5-fC) and5-carboxylcytosine (5-caC). DNA methylation as well as DNA hydroxymethylation are dynamic and can be influenced by environmental or host factors (e.g.,age, sex, disease state).

REVIEWS

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 99 NUMBER 5 | MAY 2016 513

Page 3: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

to metabolize and/or transport drugs.16 Major research activitiesare warranted to fully comprehend the impact of DNA methyla-tion on ADME gene expression and function related to these dif-ferent subgroups.In addition to interindividual variability of DNA methylation,

marked intraindividual differences, particularly in DNA methy-lation patterns between various normal tissues and cell types,are observed. These differences may be even more pronouncedbetween tumor and normal tissues.5

DNA hydroxymethylation is a further recently identified cova-lent modification of DNA with an impact on gene expression.Instead of a methyl group, cytosines are substituted with ahydroxymethyl (CH2-OH) group at the 50C (5-hmC). DNAhydroxymethylation is suggested to be an intermediate in the oxi-dative DNA demethylation pathway mediated by TET enzymes,17

and is proposed to interact with transcriptional and chromatinmodifiers as well. The effect of DNA hydroxymethylation onexpression of the respective gene is not yet fully understood and ispresumably context-dependent. DNA hydroxymethylation in genebodies, i.e., in the gene region downstream of the first exon andupstream of the 30UTR (untranslated region), has been shown tobe associated with active transcription. However, 5-hmC in prox-imity to transcription start sites can either have a repressive or anactivating effect.17 The abundance of 5-hmC is tissue-specific, withhigh levels occurring, e.g., in brain, liver, and kidney.18 In addition,DNA hydroxymethylation is dependent on developmental stage(fetus vs. adults)19 and varies among individuals of different age,20

thereby contributing significantly to interindividual variability ofgene expression. An important aspect to be considered when look-ing at previous DNA methylation studies is that the majority ofthese are based on bisulfite treatment of genomic DNA, which hasbeen accepted as the gold standard to examine DNA methylationprofiles. However, methods based on bisulfite treatment cannotdistinguish between methylated and hydroxymethylated DNA.Meanwhile, methods have been developed, e.g., TET-assistedbisulfite sequencing (TAB-seq) or oxidative bisulfite sequencing(OxBS-seq),21 which are able to differentiate between 5-mC and5-hmC, thereby allowing examination of the individual influenceof DNA methylation or hydroxymethylation on gene expression.The specific influence of DNA hydroxymethylation on the regula-tion of ADME genes in adults has been addressed in a pilot studyby Ivanov et al., investigating DNA methylation as well as hydroxy-methylation in a set of 191 ADME genes using targeted TAB-seqin 20 human liver samples, demonstrating a correlation of theabundance of 5-mC and 5-hmC with interindividual variability inADME gene expression (Abstract: 13th European ISSX Meeting2015; Ivanov, M. et al). However, large-scale studies that evaluatethe role of hydroxymethylation for ADME genes have not yetbeen reported. Such studies will be essential to better delineate theimpact of hydroxymethylation in normal as well as tumor tissuesin terms of drug safety and effectiveness.Finally, two further intermediates, 5-formylcytosine (5-fC) and

5-carboxylcytosine (5-caC), are involved in the oxidative DNAdemethylation process. Both 5-fC and 5-caC are consideredunstable intermediates and their potential functional influenceremains to be elucidated.17

REGULATION OF ADME GENE EXPRESSIONBY DNA METHYLATIONAs genetic variation in ADME genes cannot fully account forinterindividual variability of drug response, current research aimsto identify the epigenetic component of ADME gene regulation.A comprehensive summary of currently available knowledgeregarding regulation of ADME gene expression by DNA methyl-ation is given in Tables 1–3, distinguishing between drug-metabolizing enzymes, drug transporters, and modifiers likenuclear receptors. The role of DNA methylation for selectedgenes is highlighted below.

Phase I and phase II enzymesThe hepatic expression of one of the major drug-metabolizingphase I enzymes, CYP3A4, is extremely variable among individu-als (>100-fold),22 but the contribution of genetic variants to thisinterindividual variability is rather low. Habano et al.23 showedthat DNA methylation of a CpG island in the 50UTR about25 kb distal of the CYP3A4 transcription start site does notaffect gene expression. However, as demonstrated by Kacevskaet al.,24 DNA methylation at CpG sites near or within bindingsites for various transcription factors (e.g., C/EBP or HNF4a) ina 12 kb upstream promoter region has an influence on the expres-sion of CYP3A4. DNA methylation at several CpG sites wasshown to be highly variable and correlated with CYP3A4 expres-sion in adult liver. The lack of CYP3A4 expression in fetal livercould be explained by DNA hypermethylation within the investi-gated region. In contrast, regions with important transcriptionfactor binding sites were hypomethylated in adult liver whereCYP3A4 is expressed. The expression of CYP1A1 has also beendescribed to be influenced by DNA methylation. Hypermethyl-ation of a CYP1A1 enhancer region has been shown to be associ-ated with dioxin responsiveness in prostate cancer cell lines.25

Furthermore, CYP1A1 methylation levels are influenced bysmoking. A dose-dependent reduction of the CYP1A1 promoterDNA methylation from nonsmoker to heavy smokers wasobserved, suggesting a role of DNA methylation for the induc-tion of CYP1A1 in human lung due to tobacco smoking.26

Tekpli et al.27 report that CYP1A1 enhancer methylation wasassociated both with mRNA expression and smoking-inducedDNA adducts in normal human lung. Interestingly, only recentlyHe et al.28 reported an association of CYP1A1 hypermethylationwith a higher susceptibility to antituberculosis drug-induced liverinjury (ADLI) in a cohort of 127 tuberculosis patients developingADLI and 127 tuberculosis patients without liver injury. Theinfluence of DNA methylation on the expression of CYP1B1 hasbeen shown in various cancer tissues. Overexpression of CYP1B1has been related to decreased promoter and enhancer DNAmethylation at ARNT and Sp1 transcription factor binding sitesin prostate cancer.29 Widschwendter et al.30 found that CYP1B1hypermethylation levels are associated with better survival intamoxifen-treated breast cancer patients. Although CYP1B1expression levels were not determined in that study, assumingthat patients with higher methylation levels expressed lessCYP1B1, these patients would be expected to retain higher levelsof the active drug, consistent with the observed improved survival

REVIEWS

514 VOLUME 99 NUMBER 5 | MAY 2016 | www.wileyonlinelibrary/cpt

Page 4: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

Table 1 Epigenetic regulation of phase I and II enzymes by DNA methylation (for references see Supplementary Material)Gene name DNA methylation Reference

ALDH1A2 promoter hypermethylated in human prostate cancer cell lines; expression induced after5-aza-20-deoxycytidine; reduced expression and hypermethylation in human primaryprostate tissue; association with recurrence-free survival

Kim et al., 2005

evidence for association of DNA methylation status at CpG sites near ALDH1A2 genewith loss of control over drinking

Harlaar et al., 2014

ALDH1A3 promoter hypermethylation associated with favorable prognosis in G-CIMP-primaryglioblastoma

Zhang et al., 2013

methylation status is associated with reduced ALDH1A3 gene expression and diseaserecurrence and progression in nonmuscle invasive bladder cancer

Kim et al., 2013

epigenetic silencing by DNA methylation suggested as prognostic biomarker inglioblastoma

Ma et al., 2015

suppressed ALDH1A3 expression due to increased DNA methylation in cervical cancer Lee et al., 2015

CYP1A1 smoking induces hypomethylation of CYP1A1 promoter in lung tissue Anttila et al., 2003

hypermethylation of CYP1A1 in prostate cancer cell line associated with reducedinduction of CYP1A1 mRNA expression by TCDD; hypermethylation also in humanprostate tumors

Okino, 2006

CYP1A1 promoter methylation is not associated with mRNA expression in colorectalcancer cell lines

Habano et al., 2009

in utero exposure to tobacco is associated with promoter hypomethylation andincreased mRNA levels in the placenta

Suter et al., 2010

methylation is not associated with tamoxifen resistance in breast cancer Kim et al., 2010

hypermethylation of CYP1A1 associated with head and neck cancer Sharma et al., 2010

CYP1A1 enhancer methylation inversely linked to mRNA expression and the level ofhydrophilic DNA adducts in normal lung tissue; smoking induces hypomethylation;enhancer hypermethylation and reduced mRNA levels in lung cancer

Tekpli et al., 2012

higher CYP1A1 promoter methylation in tuberculosis patients with anti-tuberculosisdrug-induced liver injury compared to tuberculosis patients without liver injury

He et al., 2015

CYP1A2 variation in methylation status at specific CpG site in the 50flanking region of theCYP1A2 gene associated with interindividual differences in CYP1A2 expression inhuman liver

Hammons et al., 2001

DNA methylation in the CYP1A2 promoter correlates inversely with hepatic CYP1A2mRNA levels; 5-aza-20 -deoxycytidine treatment induces expression in hepatoma cell line

Ghotbi et al., 2009

methylation of GC box in the CYP1A2 promoter contributes to tissue specific regulationof mRNA expression

Miyajima et al., 2009

inverse correlation of mRNA expression and DNA methylation level in normal adult liver Habano et al., 2015

CYP1B1 CYP1B1 promoter methylation is associated with better survival in patients receivingtamoxifen therapy and with inferior survival in patients without tamoxifen therapy

Widschwendter et al., 2004

CYP1B1 overexpression in prostate cancer is regulated by hypomethylation in thepromoter

Tokizane et al., 2005

5-aza-20-deoxycytidine treatment results in increased CYP1B1 mRNA expression incolorectal cancer cell lines, increased CYP1B1 methylation found in a minor fraction ofcolorectal cancer tissues

Habano et al., 2009

CYP1B1 inducibility by dioxin is decreased in HepG2 cells due to promoter methylation Beedanagari et al., 2010

correlation of CYP1B1 methylation with expression, evidence for association of CYP1B1methylation with outcome and disease-free survival in a cohort of adolescent and youngadult patients with acute lymphoblastic leukemia

DiNardo et al., 2013

CYP2A13 CYP2A13 methylation contributes to regulation of gene expression in human lung cancercells

Ling et al., 2007

hypermethylation of CYP2A13 associated with head and neck cancer Sharma et al., 2010

Table 1 Continued on next page

REVIEWS

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 99 NUMBER 5 | MAY 2016 515

Page 5: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

Table 1 Continued

Gene name DNA methylation Reference

CYP2C19 inverse correlation of mRNA expression and DNA methylation level and high variability inmethylation status in normal adult liver

Habano et al., 2015

CYP2D6 methylation is not associated with tamoxifen resistance in breast cancer Kim et al., 2010

inverse correlation of mRNA expression and DNA methylation level in normal adult liver Habano et al., 2015

methylation status of CYP2D6 gene is associated with age and sex and is dependent ongenotype, poor metabolizer phenotype frequently linked to higher methylation level, lowmethylation level was associated with heavy smoking

Tiili et al., 2015

CYP2E1 CYP2E1 is not expressed in fetal liver due to methylation in the 50region, but in adultliver without methylation in this region

Jones et al., 1992

demethylation of 50region of CYP2E1 gene along with increased mRNA in fetal liverduring neonatal period

Vieira et al., 1996

no CYP2E1 expression in human lung and kidney due to methylation in the 50flankingregion of CYP2E1 gene

Vieira et al., 1998

CYP2E1 hypomethylation and mRNA overexpression in brains of patients Parkinson’sdisease

Kaut et al., 2012

prenatal exposure to serotonin reuptake inhibitor antidepressants and maternaldepressed mood leads to aberrant CYP2E1 DNA methylation, which is suggested to beassociated with birth weight

Gurnot et al., 2015

CYP2R1 higher methylation levels in the gene promoter in vitamin D nonresponders than invitamin D responders

Zhou et al., 2014

CYP2W1 CpG island in the first exon/intron junction methylated in cell lines without CYP2W1expression; CYP2W1 expression restored after 5-aza-20 -deoxycytidine treatment

Kalgren et al., 2006

CYP2W1 expression is regulated by DNA methylation of CpG island in the first exon/intron junction in cell lines as well as in colon tumors and normal colon tissue

Gomez et al., 2007

CYP2W1 expression in fetal but not in adult colon samples, associated with highermethylation of 6 CpG sites in the region of the first exon/intron junction in adult colonsamples

Choong et al., 2015

CYP3A4 DNA methylation levels among individuals are variable in 50region of the CYP3A4 regionin key regulatory binding sites; hypermethylation contributes to lacking expression infetal liver; methylation status at single CpG sites correlates with CYP3A4 expression

Kacevska et al., 2012

no significant association between interindividual variability in CYP3A4 expression andDNA methylation observed

Habano et al., 2015

CYP3A7 5-aza-20-deoxycytidine treatment contributes to upregulation of CYP3A7 mRNA expres-sion in HepG2 cells

Dannenberg and Edenberg,2006

CYP7B1 methylation of CYP7B1 promoter region has an influence on gene expression inhigh-grade prostatic intraepithelial neoplasia and adenocarcinomas

Olsson et al., 2007

CYP24A1 CYP24A1 promoter methylated in human full term placenta, but not in other somatictissues

Novakovic et al., 2009

inverse correlation of CYP24A1 expression and promoter DNA methylation in prostatecancer cell lines; 5-aza-20-deoxycytidine treatment induces expression, in vitromethylation reduces promoter activity; DNA promoter methylation contributes to reducedCYP24A1 expression in human prostate cancer

Luo et al., 2010

hypermethylation of 50region of CYP241 gene in calcitriol sensitive tumor-derivedendothelial cells, treatment with 5-aza-20 -deoxycytidine induces resistance

Johnson et al., 2010

higher methylation levels in the CYP24A1 gene in vitamin D nonresponders than invitamin D responders

Zhou et al., 2014

CYP26C1 DNA methylation of CYP26C1 is associated with poor prognosis in neuroblastoma Abe et al., 2008

CYP27B1 DNA methylation of the CYP27B1 promoter contributes to suppression of geneexpression by the vitamin D receptor

Kim et al., 2007

Table 1 Continued on next page

REVIEWS

516 VOLUME 99 NUMBER 5 | MAY 2016 | www.wileyonlinelibrary/cpt

Page 6: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

Table 1 Continued

Gene name DNA methylation Reference

CYP39A1 CYP39A1 is hypermethylated in ovarian cancer Huang et al., 2009

DHRS4L2 DHRS4L2 expression is affected by DNA methylation in human neuroblastoma cells Zhang et al., 2009

DPYD DPYD promoter methylation contributes to the regulation of DPYD expression andinfluences sensitivity to 5-fluorouracil in cancer cell lines

Noguchi et al., 2004

association of aberrant DPYD promoter methylation and unexplained DPYD enzymedeficiency in patients with 5-fluorouracil toxicity

Ezzeldin et al., 2005

no association of DPYD promoter methylation and mRNA expression in colorectalcancer, suggesting major importance of CpG site location and tissue specificity

Yu et al., 2006

DPYD promoter methylation is not associated with 5-fluorouracil severe toxicity inpatients with gastrointestinal cancer

Savva-Bordalo, 2010

GPx3 increased DNA methylation in the first exon of GPx3 leads to reduced GPx3 expressionin prostate cancer

Yu et al., 2007

DNA hypermethylation close to the transcription start site correlates inversely withreduced mRNA expression levels in Barrett’s adenocarcinoma

Peng et al., 2009

GPx3 expression downregulated due to increased promoter methylation in gastriccancer

Zhang et al., 2010

CpG island in the GPx3 promoter hypermethylated accompanied by decreased GPx3mRNA expression in esophageal squamous cell carcinoma

He et al., 2011

GPx3 promoter methylation is associated with resistance to cisplatin treatment inpatients with head and neck cancer

Chen et al., 2011

GPx3 promoter hypermethylation correlates with reduced expression and is associatedwith lymph node metastasis in gastric cancer

Peng et al., 2012

downregulation of GPx3 expression by DNA methylation involved in progression ofinflammatory breast cancer

Mohamed et al., 2014

promoter DNA methylation is associated with reduced GPx3 expression, GPx3expression is associated with lymph node metastasis and progression in cervical cancer

Zhang et al., 2014

GPx3 hypermethylation is associated with poor survival in non-M3 acute myeloidleukemia

Zhou et al., 2015

inverse correlation of GPx3 expression and methylation in chronic myeloid leukemia;re-expression of GPx3 after 5-aza-20-deoxycytidine in CML cell line

Yao et al., 2015

GPx3 expression silenced due to promoter hypermethylation in clear cell renal cellcarcinoma

Liu et al., 2015

GPx7 DNA hypermethylation close to the transcription start site correlates inversely withreduced mRNA expression levels in Barrett’s adenocarcinoma

Peng et al., 2009

DNA methylation in a specific region of the GPx7 promoter CpG island correlatesinversely with expression in oesophageal adenocarcinoma

Peng et al., 2014

GSTA4 inverse correlation of mRNA expression and DNA methylation level and high variability inmethylation status in normal adult liver

Habano et al., 2015

GSTO2 DNA methylation contributes to a larger part to the variation of GSTO2 gene expressionthan SNP genotypes

Bonder et al., 2014

GSTP1 hypermethylation of GSTP1 promoter suggested as biomarker for various cancerentities, including prostate and breast cancer

for review see Schnecken-burger et al., 2014

haplotype structure has an influence on the degree of GSTP1 promoter methylation inbreast cancer

Ronneberg et al., 2008

hypermethylation of GSTP1 promoter negatively correlates with expression and isassociated with improved survival in doxorubicin treated breast cancer patients

Dejeux et al., 2010

methylation of GSTP1 proposed as predictive marker for therapy with DNMT inhibitors inprostate cancer

Chiam et al., 2011

Table 1 Continued on next page

REVIEWS

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 99 NUMBER 5 | MAY 2016 517

Page 7: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

rate. In a cohort of adolescent and young adult patients withacute lymphoblastic leukemia, hypermethylation and reducedCYP1B1 expression in peripheral blood cells was associated withworse overall survival.31 In 1992, Jones et al.32 found DNA meth-ylation in the 50 flanking region of the CYP2E1 gene to beresponsible for the lack of transcription in fetal liver, in contrastto adult liver, where CYP2E1 is expressed and not methylated.Vieira et al.33 subsequently showed that demethylation of the 50

region during the neonatal period resulted in increased CYP2E1

mRNA expression. Interestingly, CYP2E1 overexpression due toDNA hypomethylation has been shown in patients with Parkin-son’s disease.34 CYP2W1 is inherently expressed only in fetalcolon, but not in normal adult tissue. Re-expression of CYP2W1has been observed in colorectal cancer and has therefore beensuggested as a prognostic marker for malignancy in colon can-cer.35 In addition, the cancer-specific expression can be exploitedfor the treatment of colon cancer with prodrugs bioactivated byCYP2W1 (e.g., duocarmycin) to selectively kill cancer cells.36

Table 1 Continued

Gene name DNA methylation Reference

higher GSTP1 promoter methylation in tuberculosis patients with anti-tuberculosisdrug-induced liver injury compared to tuberculosis patients without liver injury

He et al., 2015

GSTM1 hypermethylation of GSTM1 associated with head and neck cancer Sharma et al., 2010

epigenetic repression due to promoter methylation in retina pigment epithelium (RPE)/choroid of patients with age-related macular degeneration

Hunter et al., 2012

DNA methylation contributes to a larger part to the variation of GSTM1 gene expressionthan SNP genotypes

Bonder et al., 2014

GSTM2 DNA hypermethylation close to the transcription start site correlates inversely withreduced GSTM2 mRNA expression levels in Barrett’s adenocarcinoma

Peng et al., 2009

GSTM5 DNA hypermethylation close to the transcription start site correlates inversely withreduced GSTM5 mRNA expression levels in Barrett’s adenocarcinoma

Peng et al., 2009

inverse correlation of mRNA expression and DNA methylation level and high variability inmethylation status in normal adult liver

Habano et al., 2015

GSTT1 DNA methylation at a specific CpG site in the GSTT1 promoter/enhancer region isassociated with mRNA expression with high significance in human monocytes

Liu et al., 2013

DNA methylation contributes to a larger part to the variation of GSTT1 gene expressionthan SNP genotypes

Bonder et al., 2014

inverse correlation of mRNA expression and DNA methylation level in normal adult liver Habano et al., 2015

NAT1 NAT1 promoter shows reduced methylation levels in breast cancer tissue, associatedwith NAT1 mRNA expression

Kim et al., 2008

higher DNA methylation level in the NAT1 gene is related to tamoxifen-resistant breastcancer

Kim et al., 2010

PON1 DNA methylation contributes to a larger part to the variation of PON1 gene expressionthan SNP genotypes

Bonder et al., 2014

SULT1A1 SULT1A1 is silenced and methylated in breast cancer tissue Kwon et al., 2006

methylation is not associated with tamoxifen resistance Kim et al., 2010

inverse correlation of mRNA expression and DNA methylation level in normal adult liver Habano et al., 2015

UGT1A1 expression variable in primary colon tumors, expression inversely correlated to DNAmethylation with potential influence on irinotecan sensitivity

Gangon et al., 2006

UGT1A1 expression is repressed due to promoter methylation in colon cancer cell lines B�elanger et al., 2010

UGT1A1 promoter methylation contributes to tissue-specific expression in liver andkidney

Oda et al., 2013

DNA methylation contributes to a larger part to the variation of UGT1A1 gene expressionthan SNP genotypes

Bonder et al., 2014

UGT1A10 expressed in intestine, but not in liver; promoter hypomethylated in LS180 cell line andintestine, but hypermethylated in HuH7 and hepatocytes; 5-aza-20-deoxycitidine treat-ment increased UGT1A10 expression only in HuH7 cells

Oda et al., 2014

UGT2B15 DNA methylation is associated with metabolic traits in human blood Petersen et al., 2014

REVIEWS

518 VOLUME 99 NUMBER 5 | MAY 2016 | www.wileyonlinelibrary/cpt

Page 8: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

Table 2 Epigenetic regulation of drug transporters by DNA methylation (for references see Supplementary Material)Gene name DNA methylation Reference

ABCA1 higher DNA methylation in ABCA1 gene in individuals exposed to prenatal famine Tobi et al., 2009

larger interindividual variability of DNA methylation in the ABCA1 gene in old compared toyoung monozygotic twin pairs

Talens et al., 2012

ABCA1 promoter methylation negatively correlates with HDL cholesterol levels and isassociated with coronary artery disease in patients with familial hypercholesterolemia

Guay et al., 2012

ABCA1 hypermethylated in prostate cancer associate with reduced expression and prostatetumor aggressiveness with potential influence for the response to treatment with LXRagonists

Lee et al., 2013

ABCA1 methylation and expression at maternal side of placenta is associated with maternalmetabolic profile and at the fetal side with cord blood lipid profile with consequences for thesusceptibility of newborn to obesity, dyslipidemia and cardiovascular diseases

Houde et al., 2013

higher ABCA1 promoter DNA methylation associated with age, occurrence of coronary arterydisease and low-density cholesterol levels; therapy with acetylsalicylic acid reduces ABCA1promoter methylation

Guay et al., 2014

reduced ABCA1 expression and increased promoter methylation in ovarian cancer cell lines;higher methylation and lower expression in ovarian cancer samples associated with inferiorsurvival

Chou et al., 2015

ABCB1 hypomethylation of ABCB1 is associated with resistance to treatment in various cancer celllines

for review see Baker andEl-Osta, 2004

hypomethylation of the ABCB1 promoter is associated with ABCB1 expression and isassociated with treatment resistance in a T-cell leukemia cell line; association of DNAmethylation with expression also seen in chronic lymphocytic leukemia samples

Kantharidis et al., 1997

less ABCB1 promoter methylation and increased gene expression in multi-drug resistantbreast cancer cells

David et al., 2004

hypomethylation of CpG island in the promoter associated with response to doxorubicin Dejeux et al., 2010

acquisition of drug resistance is accompanied by temporal loss of ABCB1 promotermethylation and increased gene expression in breast cancer cells

Reed et al., 2010

ABCB4 DNA hypermethylation of the ABCB4 promoter silences gene expression in human cancer celllines and in human lung and breast cancer

Kiehl et al., 2014

ABCC6 ABCC6 gene methylation included in a set of DNA methylation markers to detect bladdercancer

Yu et al., 2007

ABCG1 DNA methylation is associated with metabolic traits in human blood Petersen et al., 2014

DNA methylation at CpG site within the ABCG1 gene is associated with fasting insulin levelsand HOMA-IR

Hidalgo et al., 2014

ABCG2 DNA methylation of the ABCG2 gene contributes to reduced expression in renal cell carcinomacell lines

To et al., 2006

topotecan treated patients show higher ABCG2 expression, regulated in parts by promoterdemethylation

Turner et al., 2006

ABCG2 expression correlates inversely with DNA methylation of the ABCG2 gene in small celland non-small cell lung cancer cells, ABCG2 overexpression and hypomethylation in a SN-38treatment resistant lung cancer cell line

Nakano et al., 2008

ABCG2 promoter is demethylated, accompanied by increased gene expression in T-ALL andovarian carcinoma cell lines in response to treatment with antitumor drugs

Bram et al., 2009

SLC5A8 SLC5A8 expression is silenced by DNA methylation in human colon aberrant crypt foci andcancers

Li et al., 2003

DNA methylation in the SLC5A8 promoter silences gene expression in lung cancer Park et al., 2013

SLC6A4 promoter DNA hypermethylation is associated with depression and a better response rate ofdepressed patients treated with escitalopram

Domschke et al., 2014

SLC16A3 DNA methylation of the SLC16A3 promoter region correlates with expression and hasprognostic potential in patients with clear cell renal cell carcinoma

Fisel et al., 2013

Table 2 Continued on next page

REVIEWS

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 99 NUMBER 5 | MAY 2016 519

Page 9: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

Interestingly, Gomez et al.37 showed that DNA methylation inthe CpG-rich first exon/intron junction was involved in the regu-lation of gene expression in colon tumor tissue. Only recently,developmental stage-specific CYP2W1 expression in human fetalcolon was also found to be governed by methylation in this generegion.38 Glutathione peroxidase 3 (GPx3) functions to protectcells from oxidative damage by catalyzing the reduction of reac-tive oxygen species (ROS). GPx3 promoter hypermethylation is amajor contributor to suppressed GPx3 expression in multiplehuman cancer tissues, such as breast, prostate, renal, cervical, andgastric cancer or acute myeloid leukemia (see Table 1). Accord-ing to a study by Chen et al.,39 GPx3 hypermethylation is tightlyrelated with cisplatin resistance and poor survival in patients withhead and neck cancer.The enzyme UGT1A1, involved in phase II drug metabolism,

catalyzes the inactivation of SN-38, the active metabolite ofirinotecan, a drug used to treat metastatic colon cancer.UGT1A1 is variably expressed in colon cancer. In a study byGagnon et al.,40 most colon cancer tissues showed only lowUGT1A1 expression; however, some exhibited high expressionlevels. They could prove that in colon cancer cell lines UGT1A1expression was tightly associated with promoter DNA methyla-tion levels. From these results, the authors hypothesize that aber-

rant UGT1A1 expression, influenced by altered DNAmethylation, contributes to differential sensitivity to irinotecantreatment. In a study by Oda et al.,41 methylation of theUGT1A1 promoter was identified to contribute to the lack ofUGT1A1 expression in human kidney. The N-acetyltransferase1 (NAT1), a phase II enzyme, is involved in the detoxification(N-acetylation) or bioactivation (O-acetylation) of arylamine car-cinogens. NAT1 has been shown to be overexpressed in ER1

breast cancer and to be associated with tamoxifen responsive-ness.42 According to Kim et al.,43 reduced promoter DNA meth-ylation of NAT1 leads to the increased expression in breastcancer tissue.44 In a subsequent study, they compared tamoxifen-resistant and nonresistant breast cancer patients and suggestedthat higher NAT1 methylation was associated with nonresponse.In addition to candidate gene studies, recently a genome-wide

analysis has been performed on evaluating the biological signifi-cance of DNA methylation of DME genes on their mRNAexpression in adult human liver tissues by combining these resultswith corresponding data from the Gene Expression Omnibus(GEO) database of the National Center for Biotechnology Infor-mation (NCBI). Of note, 37 DME genes revealed highly variableDNA methylation, of which DNA methylation was inverselycorrelated with mRNA expression for seven DME genes

Table 2 Continued

Gene name DNA methylation Reference

SLC19A1 CpG island in the SLC19A1 promoter is hypermethylated and not expressed in amethotrexate resistant breast cancer cell line, promoter is unmethylated in a SLC19A1expressing and methotrexate sensitive breast cancer cell line

Worm et al., 2001

rate of complete remission in response to methotrexate therapy is associated with SLC19A1promoter methylation

Ferreri et al., 2004

reverse correlation of SLC19A1 promoter methylation with expression in a panel of malignantcell lines

Yang et al., 2008

reduced expression and hypermethylation in a methotrexate resistant lymphoblast cell lineand in treatment na€ıve diffuse large B-cell lymphomas

Kastrup et al., 2008

SLC22A1 SLC22A1 expression is reduced due to hypermethylation of the promoter region inhepatocellular carcinoma with potential consequences for cisplatin-based chemotherapy

Schaeffeler et al., 2011

long-term cisplatin exposure leads to increased SLC22A1 promoter methylation conferringcisplatin resistance in human esophageal cancer cell lines

Lin et al., 2013

SLC22A2 SLC22A2 promoter DNA methylation regulates kidney-specific expression Aoki et al., 2008

SLC22A3 DNA methylation of the SLC22A3 promoter region regulates gene expression in prostate andcolon cancer cell lines; low expression accompanied by high promoter methylation levels inprostate cancer

Chen et al., 2013

SLC22A8 DNA methylation contributes to the regulation of SLC22A8 gene expression in Caco2 andHEK293 cells

Kikuchi et al., 2006

SLC26A4 methylation of the SLC26A4 gene is suggested to be an early event in the tumorigenesis ofthyroid cancer

Xing et al., 2003

SLCO1B3 5-aza-20 -deoxycytidine treatment increased SLCO1B3 mRNA expression in HepG2 and Caco-2cells

Ichihara et al., 2010

SLC22A18 increased promoter methylation contributes to downregulation of SLC22A18 expression ingliomas; expression is increased upon 5-aza-20 -deoxycytidine treatment in glioma cell lines

Chu et al., 2011

SLC22A18 protein expression is a better predictive marker than promoter DNA methylation inglioblastoma cells receiving temozolomide therapy

Chu et al., 2013

REVIEWS

520 VOLUME 99 NUMBER 5 | MAY 2016 | www.wileyonlinelibrary/cpt

Page 10: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

(CYP1A2, CYP2C19, CYP2D6, GSTA4, GSTM5, GSTT1, andSULT1A1). Moreover, the authors provide evidence that DNAmethylation status around the first exon is responsible for tissue-specific (e.g., hepatic vs. intestinal tissue) and age-dependentexpression of UGT1A isoforms by guiding correct alternativesplicing of the UGT1A gene.45

Very recently, the contribution of genetic variation and epige-netic factors to the regulation of gene expression includingADME genes in human liver was investigated by a genome-wideapproach. Interestingly, a larger effect of methylation level at sin-gle CpG sites on hepatic expression was found for five ADMEgenes (GSTT1, GSTM1, UGT1A1, GST01, and PON1) whoseexpression levels are altered additionally by gene-specific variants.This study provides first evidence that a combination of geneticand epigenetic regulation by DNA methylation may better pre-dict interindividual variability of gene expression than either ofthe two alone.15

Interestingly, Petersen et al.46 recently suggested that DNAmethylation affects human metabolism. They performed anepigenome-wide association study (EWAS) between DNA meth-ylation and metabolic traits in human blood and found signifi-cant associations also for CpG loci associated with the ADMEgenes UGT2B15 and ABCG1. However, in general the effects ofDNA methylation on the metabolic traits were not as strong asthe effects observed for genotypes. Direct monitoring of DNAmethylation-based biomarkers in body fluids, such as plasma orserum, as currently suggested for cancer or disease biomarkers,would presumably allow better prediction and could facilitateclinical implementation.47

TransportersEpigenetic regulation of transporter gene expression by DNAmethylation has been shown for members of the ABC as well asthe SLC transporter family (Table 2). It is well known that

Table 3 Epigenetic regulation of nuclear receptors by DNA methylation (for references see Supplementary Material)Gene name DNA methylation Reference

HNF1A 5-aza-20-deoxycytidine treatment restores HNF1A expression in colon cancer cell lines B�elanger et al., 2010

HNF1A gene methylation silences its expression and is associated with the composition ofglycans on plasma glycoproteins

Zoldo�s et al., 2012

PPARG hypomethylation of PPARG promoter is associated with gastric cancer Hong et al., 2010

PPARG promoter hypermethylation leads to reduced expression and is associated withadvanced tumors and poor outcome in patients with colorectal cancer

Pancione et al., 2010

PPARG promoter DNA methylation leads to reduced mRNA expression in patients with chronichepatitis B and is associated with liver inflammation and fibrosis

Zhao et al., 2013

PXR hypermethylation of PXR and reduced gene expression in aggressive neuroblastomas Misawa 2005

low expression of PXR in colon cancer cells by promoter methylation in the PXR gene, linked toreduced transactivation of the CYP3A4 gene, hypomethylation of the PXR promoteraccompanied by increased gene expression in colorectal cancer tissue

Habano et al., 2011

RARB RARB expression silenced by DNA methylation in breast cancer Widschwendteret al., 2000

aberrant RARB methylation found in head and neck cancer Youssef et al., 2004

RXRA higher methylation level at specific CpG site in the RXRA gene in umbilical cord tissue isassociated with maternal pregnancy diet and childhood adiposity

Godfrey et al., 2011

RXRG downregulation of RXRG gene expression by DNA methylation may play different roles inNSCLCs depending on smoking status of patients

Lee 2010

SOD2 SOD2 expression is repressed by DNA hypermethylation in the SOD2 gene promoter in breastcancer cell lines, expression is restored by 5-aza-20-deoxycytidine treatment

Hitchler et al., 2006

SOD2 expression silenced by SOD2 gene DNA methylation in pancreatic cancer cell lines Hurt et al., 2007

downregulation of SOD2 gene expression in human cartilage with osteoarthritis accompaniedby increased SOD2 promoter methylation

Scott et al., 2010

SOD3 hypermethylation of the SOD3 promoter contributes to loss of SOD3 expression in lung cancer Teoh-Fitzgeraldet al., 2012

decreased SOD3 expression due to promoter hypermethylation in mammaryadenocarcinomas

Teoh-Fitzgeraldet al., 2014

SOD3 gene methylation associated with adiposity Agha et al., 2015

VDR VDR expression is silenced in due to VDR promoter methylation leading to calcitriolinsensitivity in breast cancer

Marik et al., 2010

DNA hypermethylation in the VDR promoter in reduced VDR expression in HIV infected T-cells Chandel et al., 2013

REVIEWS

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 99 NUMBER 5 | MAY 2016 521

Page 11: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

increased expression of the ABCB1 gene, encoding for the multi-drug resistance protein MDR1 (P-glycoprotein), may lead toacquired cancer treatment resistance. Besides other mechanisms(e.g., transcriptional induction by transcription factors such asHIF1a48), DNA methylation in the ABCB1 gene promotercontributes to MDR1 overexpression in vitro (cancer cell lines).In vivo the expression of MDR1 in cancer tissues of patients diag-nosed with acute myeloid leukemia, acute lymphocytic leukemia,chronic lymphocytic leukemia, colorectal cancer, and bladdercancer was linked to promoter DNA methylation.49 In addition,hypomethylation of the ABCB1 promoter was associated withpoor drug response and, together with hypomethylation of theGSTP1 promoter, related to inferior survival in vivo in breastcancer patients treated with doxorubicin.50 For the breast cancerresistance protein (BCRP), mediating efflux of chemotherapeuticdrugs, such as methotrexate, doxorubicin, or SN-38 (the activemetabolite of irinotecan), drug resistance is linked to the DNAmethylation status in the ABCG2 promoter, which results inoverexpression of this transporter protein. As shown by Bramet al.,51 leukemic and ovarian cancer cell lines upregulated

ABCG2 expression upon exposure to chemotherapeutic drugsaccompanied by demethylation of the ABCG2 promoter, result-ing in a significant increase in drug resistance compared tountreated cells.Aberrant drug uptake mediated by members of the SLC trans-

porter family can also mediate drug resistance or toxicity. TheSLC19A1 gene encodes for a carrier mediating the uptake ofreduced folate and antifolate drugs such as methotrexate. Hyper-methylation of the SLC19A1 promoter is associated withreduced expression in breast cancer cell lines52 and has been asso-ciated with a lower response rate to methotrexate treatment inpatients with CNS lymphomas.53 Furthermore, members of theSLC22A gene family have been shown to be regulated by DNAmethylation. The organic cation transporters (OCT) 1, 2, and 3,encoded by the genes SLC22A1, SLC22A2, and SLC22A3respectively, mediate the uptake of several clinically relevant drugsincluding cancer agents (e.g., cisplatin or oxaliplatin).54 OCT1 isprimarily expressed in the liver,55 but not in kidney, where the50UTR of the SLC22A1 gene is highly methylated compared toliver tissue (Figure 2).56,57 In hepatocellular carcinoma, OCT1

Figure 2 Expression and DNA methylation of OCT1/SLC22A1, OCT2/SLC22A2 and OCT3/SLC22A3 in liver and kidney. Immunofluorescent staining ofOCT1, OCT2, and OCT3 and DNA methylation profiles of the regulatory 50UTR regions of SLC22A1, SLC22A2, and SLC22A3, investigated by MALDI-TOFmass spectrometry, in human liver (n 5 100) and kidney (n 5 18) tissue, respectively (see refs. 55, 57, 75, unpublished data). Median methylation lev-els at investigated CpG sites are represented by diamonds; 25%/75%-quantiles are depicted as shaded areas. OCT1 is expressed in human liver, but notin human kidney. Accordingly, DNA methylation in the 50UTR of the SLC22A1 gene is low in liver (blue) and high in kidney tissues (yellow). In contrast,OCT2 is a kidney-specific transporter, showing low methylation levels in the 50 UTR of the SLC22A2 gene in kidney (yellow), but high methylation levels inliver (blue), where OCT2 is not expressed. OCT3 is expressed in liver as well as in kidney. The 50UTR region of the SLC22A3 gene does not show differen-ces in methylation profiles between the tissues.

REVIEWS

522 VOLUME 99 NUMBER 5 | MAY 2016 | www.wileyonlinelibrary/cpt

Page 12: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

expression is downregulated due to DNA methylation, indicatingimpaired uptake and subsequent lower intracellular concentra-tion of cancer agents, resulting in potential nonresponse.57 Linet al.58 showed that long-term treatment with cisplatin leads tohypermethylation of the SLC22A1 gene and in consequence tocisplatin resistance in human esophageal cancer cells. The expres-sion of OCT2 is kidney-specific. According to this tissue-specificexpression, DNA methylation levels in the SLC22A2 promoterregion is low in kidney tissue as compared to human livermissing OCT2 expression due to hypermethylation of SLC22A2(Figure 2).56,57 OCT3 is expressed in liver as well as in kidneyand does not show different DNA methylation patterns inthese organs (Figure 2). However, aberrant DNA methylation ofthe SLC22A3 promoter has been identified as a regulatory factorin prostate cancer, corroborating the strong tissue-specific natureof epigenetic regulation.59 The monocarboxylate transporter4 (MCT4), encoded by the gene SLC16A3, is an important lac-tate transporter responsible for the extrusion of lactate in highlyglycolytic cells, such as tumor cells. Independent from unspecificMCT4 inhibitors (e.g., statins), specific MCT4 inhibitors aspromising cancer agents are currently under investigation. Interest-ingly, DNA methylation at a specific CpG site in the 50 regulatoryregion of the SLC16A3 promoter has been shown to correlatewith expression of this lactate transporter and is strongly associatedwith survival of patients with clear cell renal cell carcinoma.60,61

Nuclear receptorsNuclear receptors are a family of ligand binding transcriptionfactors acting to induce expression of genes involved in variousphysiological processes in response to endogenous or exogenousstimuli, including the expression of ADME genes in response toxenobiotics. Nuclear receptors not only contribute to epigeneticregulation of gene expression by recruiting coactivators and core-pressors mediating epigenetic modifications in target genes, butare also regulated epigenetically themselves. It can be assumedthat aberrant expression of nuclear receptors due to changes inDNA methylation profiles results in aberrant activation ofresponsive ADME genes, and hence drug response. A majorxenobiotic sensor and regulator of several ADME genes, includ-ing CYP3A4, UGT1A1 or ABCB1, is the pregnane X receptor(PXR/NR1I2). A comparison of PXR low- and PXR high-expression colon cancer cell lines showed that decreased expres-sion of PXR is linked to promoter methylation in the NR1I2gene and reduced transactivation of the CYP3A4 gene. In colo-rectal cancer tissues DNA methylation alters PXR expression aswell, in that PXR expression is upregulated due to NR1I2 pro-moter hypomethylation.23 Misawa et al.62 found a link betweenNR1I2 DNA methylation in a CpG island in close proximity toexon3 and reduced NR1I2 gene expression in advanced neuro-blastoma tumors, suggesting PXR to target genes with growthinhibitory functions. The effect of lower PXR levels due tohypermethylation on target gene expression like CYP3A4, how-ever, was not analyzed in that study. Regarding the peroxisomeproliferator-activated receptor g (PPARg), hypermethylation incolorectal cancer tissue is accompanied by reduced gene expres-sion, advanced tumor stage, and poor patient survival.63 Since

PPARg agonists are supposed to potentiate the efficacy for anumber of chemotherapeutics, these authors hypothesize that arational combination of chemotherapeutics with a PPARg ago-nist and an epigenetic drug reactivating PPARg expression inpatients with silenced PPARg would sensitize tumors to anti-cancer treatment. The vitamin D receptor (VDR/NR1I1) playsan important role in mediating the response to calcitriol, whichis the active metabolite of vitamin D. Calcitriol has been ascribedto mediate antiproliferative effects on tumor cells; however, can-cer tissue and cell lines often show insensitivity to calcitriol treat-ment.64 The expression of the VDR receptor is influenced byDNA methylation in colon and endometrial cancers. Interest-ingly, Marik et al.64 show that combined treatment of breast can-cer cell lines with the demethylating agent 50aza-20-deoxycytidineand calcitriol enhanced growth inhibitory effects accompanied byinduction of VDR mRNA expression. Indeed, the NR1I1 pro-moter was hypermethylated in breast cancer cell lines as well asprimary breast cancer tissue, resulting in reduced VDR expressionand presumably to insensitivity to calcitriol treatment.

CONSEQUENCES FOR DRUG THERAPYWhereas the contribution of DNA methylation on interindivid-ual variability of expression of a number of ADME genes relatedto drug metabolism and transport has repeatedly been shown, theclinical relevance of DNA methylation to explain drug nonres-ponse and drug-induced toxicity needs further intensive evalua-tion. DNA methylation of specific ADME genes (e.g., GSTP1,GPx3, or SLC16A3) is currently proposed as biomarker for thediagnosis and prognosis of various cancers and has been associ-ated with acquired drug resistance (Tables 1–3). A major draw-back of findings from in vitro studies with cultivated cancer celllines generally used to investigate associations between ADMEgene expression and DNA methylation is that extrapolation tothe in vivo situation is limited, since expression and DNA meth-ylation profiles do not necessarily resemble the profiles found invivo. It is well known that gene expression and DNA methylationprofiles of a number of genes involved in drug metabolism andtransport are considerably different between cell lines and pri-mary tissues.65 Much less information is available with respect tothe impact of DNA methylation in ADME genes and conse-quences for the treatment of diseases other than cancer. Oneexplanation might be that investigations on DNA methylationare restricted methodologically by the availability of tissue-specific DNA, which is readily available from tumor biopsies orresection material, but rarely from nontumor tissue. Nevertheless,regarding psychiatric disorders, increased promoter DNA methyl-ation of the serotonin transporter SLC6A4 in peripheral bloodleukocytes has recently been associated with an increased risk fordepression.66 Interestingly, decreased promoter methylation levelsin blood of depressed patients were associated with a lowerresponse rate to antidepressant treatment using escitalopram.67

In addition, DNA methylation of a CpG site in the ABCG1gene was associated with fasting insulin levels and homeostasismodel assessment of insulin resistance (HOMA-IR) in an EWASwith metabolically healthy volunteers. This indicates that DNAmethylation at this CpG site might represent a marker for risk of

REVIEWS

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 99 NUMBER 5 | MAY 2016 523

Page 13: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

metabolic disease or insulin resistance, with potential consequen-ces for drug therapy.68 However, a major limitation of these stud-ies so far is the rather small sample size and limited phenotypiccharacterization of study populations.The continuous development of high-throughput technologies

to systematically analyze DNA methylation profiles on a genome-wide level, i.e., EWAS, and their use in large-scale projects, suchas the International Human Epigenome Consortium or the NIHRoadmap Epigenomics Mapping Consortium, will enable a morecomprehensive understanding of the epigenetic component ofADME gene regulation, and will provide additional informationfor personalized drug treatment in the future. Moreover, theadvancement of technologies to reliably and efficiently quantify5-mC specifically in selected ADME genes, such as the methodrecently developed by Ivanov et al.,69 which uses next generationsequencing of bisulfite-converted DNA specifically enriched forcoding and regulatory regions of 174 ADME genes, will also facil-itate high-throughput analysis of DNA methylation with respectto drug response.Most important, the function and relevance of DNA hydroxy-

methylation needs to be further evaluated to elucidate its rolewithin the epigenetic machinery. Thus, the consideration ofnovel high-throughput technologies for quantification of notonly 5-mC but also 5-hmc and further intermediates (5-fC and5-caC) opens a new avenue in the understanding of epigeneticcontrol of ADME genes.The dynamic nature of DNA methylation, being influenced,

e.g., by age, sex, environmental, and lifestyle factors, represents amajor challenge for the integration of the knowledge into clinicalpractice. In contrast to genetic variations, DNA methylation, asepigenetic modification, is dynamic and reversible. Use of DNAmethylation profiles as contributors to interindividual variabilityof drug response would require repeated investigation of bio-marker gene DNA methylation profiles depending on patientage, treated target organ, or during long-term treatment. More-over, not only interindividual variability of DNA methylation ofADME genes, but also of drug target genes may account foraltered response to drug treatment. For instance, hypermethyl-ation of the promoter of the epidermal growth factor receptor(EGFR) gene, for example, is associated with resistance to theEGFR-tyrosine kinase inhibitor gefitinib in nonsmall-cell lungcancer cells.70 The concept of systems pharmacology1 and theintegration of epigenomic information not only on ADME genesbut also other pharmacogenes will provide promising progress tobetter understand drug resistance and nonresponse.On the other hand, the plasticity of DNA methylation pro-

vides an attractive opportunity for therapeutic intervention withso-called epidrugs. DNMT inhibitors, such as the nucleoside ana-logs 5-azacytidine and 5-aza-20-deoxycytidine, are approved by theUS Food and Drug Administration (FDA) for the treatment ofmyelodysplastic syndrome and for elderly patients with acutemyeloid leukemia. Just as shown for tumor suppressor genes,ADME-related gene expression could be increased by drugs inhib-iting DNA methylation. However, treatment with DNMT inhib-itors is unspecific and causes genome-wide DNA demethylation,with potentially serious side effects (Figure 3). Furthermore, both

5-azacytidine and 5-aza-20-deoxycytidine rely on transporter-mediated uptake into the cells to be incorporated into theDNA, via, e.g., the human concentrative nucleoside transporter 1(hCNT1).71 Advancements in technologies to epigenetically editselected target DNA sequences holds much promise to overcomethe problem of global off-target effects. Different targetedapproaches to epigenetically modify expression of specific geneshave been established as powerful tools in biological research. Therationale of these approaches is to fuse a catalytically active effec-tor domain, e.g., DNA methylating (DNMTs) or demethylating(TETs) enzymes, to a DNA-binding domain selectively targetingthe effector to the target gene sequence. The currently most com-monly applied tools differ in the way of mediating targetsequence recognition, which can be achieved via protein/DNA-based interaction (zinc finger proteins or transcription activatorlike effectors [TALEs]) or RNA/DNA-based interaction (clus-tered regularly interspaced short palindromic repeats/CRISPR-associated 9 system [CRISPR/Cas9 system]) (Figure 3). Zincfinger-DNA binding domains coupled to nucleases have already

Figure 3 Schematic overview of methods to interfere with aberrant DNAmethylation. (a) Treatment with either 5-azacytidine or 5-aza-20 -deoxyazacytidine inhibits DNMT, eventually leading to global DNA hypome-thylation. (b) Targeted epigenetic editing at ADME gene promoters can beachieved by use of zinc finger proteins, TALEs, or the CRISPR/Cas9 sys-tem, specifically targeting DNA sequences of genes of interest, coupled toeffector proteins, i.e., DNMTs or TETs.

REVIEWS

524 VOLUME 99 NUMBER 5 | MAY 2016 | www.wileyonlinelibrary/cpt

Page 14: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

been shown to be useful for therapeutic approaches and are cur-rently in clinical trials for the treatment of HIV. TALE nucleasesand the CRISPR/Cas9 system have proven their usefulness aswell for clinical applications in vivo in animal models.72 Refine-ment of these tools with respect to their use for targeted modifi-cation of DNA methylation will not only push forward this areaof research, but will also further their application for therapeuticpurposes including the alteration of responsiveness to drugtreatment.Although this review focuses specifically on the influence

of DNA methylation on ADME genes, the contribution of his-tone modifications and miRNAs to interindividual variability ofADME gene expression needs to be considered carefully. Bothhistone modifications and miRNAs significantly affect ADMEgene expression, with a putative role for variability of drugresponse.73 There is evidence that all three epigenetic mecha-nisms interact with each other by a highly complex network ofgene regulation. For instance, DNA methylation can eitherdirectly or indirectly recruit histone-modifying enzymes such ashistone deacetylases. Conversely, DNMTs may act depending oncertain precedent histone modifications.4 Moreover, the expres-sion of miRNAs may be altered by DNA methylation or histonemodifications.74 Therefore, improved understanding not only ofDNA methylation, but the whole network of epigenetic regula-tion in combination with genetic and nongenetic host factors isneeded to elucidate the interindividual variability of ADMEgenes and the consequences for drug safety and efficacy.Extending the current knowledge with additional studies in the

field of pharmacoepigenomics in general, and pharmacogeneDNA methylation in particular, will reveal how ADME DNAmethylation-based biomarkers can be implemented into clinicalpractice in the future in order to better predict drug response andavoid ADR. Advancement of technologies to feasibly and accu-rately monitor these biomarkers is inevitably necessary to facilitateclinical application. Currently, analysis of DNA methylation inbody fluids, e.g., cell-free circulating DNA in blood, is emergingand represents a promising opportunity to detect disease or to pre-dict response to therapeutic intervention, especially for cancer dis-eases.47 This noninvasive approach might be similarly applicablefor other diseases with disease- or response-specific DNA methyla-tion patterns, where availability of tissue biopsies could represent amajor limitation for diagnosis or prediction of treatment response.Since the regulation of ADME gene expression is a highly com-

plex network of various factors, the integration of high-throughput“omic” technologies, including genomics, epigenomics, transcrip-tomics, proteomics, and metabolomics,1 will help to identify novelpotential biomarkers for drug efficacy or safety and combinationof the generated data will help to determine the clinically usefulones. The most promising candidates have to be validated in pro-spective trials to prove their specificity and sensitivity for clinicalapplication. This concept will strengthen the idea of precisionmedicine and the implementation into clinical practice.

Additional Supporting Information may be found in the online version ofthis article.

ACKNOWLEDGMENTSThe first two authors contributed equally to this review. This work wassupported by the Robert Bosch Foundation (Stuttgart, Germany), theFederal Ministry for Education and Research (BMBF, Berlin, Germany,FKZ 031L0037 and 01EK1509A), the Horizon 2020-PHC-2015 grantU-PGx 668353, the German Cancer Consortium (DKTK) and GermanCancer Research Center (DKFZ, Heidelberg, Germany), and ICEPHAGraduate School Tuebingen-Stuttgart. The authors thank Bernd Borstelfor drafting the figures and Prof. Anne T. Nies for providing figurematerial.

CONFLICT OF INTERESTThe authors have no conflicts of interest to disclose.

VC 2016 American Society for Clinical Pharmacology and Therapeutics

1. Meyer, U.A., Zanger, U.M. & Schwab, M. Omics and drug response.Annu. Rev. Pharmacol. Toxicol. 53, 475–502 (2013).

2. Ehmann, F. et al. Pharmacogenomic information in drug labels:European Medicines Agency perspective. PharmacogenomicsJ. 15(3), 201–210 (2015).

3. Holliday, R. Epigenetics: a historical overview. Epigenetics 1(2),76–80 (2006).

4. Goldberg, A.D., Allis, C.D. & Bernstein, E. Epigenetics: a landscapetakes shape. Cell 128(4), 635–638 (2007).

5. Jones, P.A. & Baylin, S.B. The epigenomics of cancer. Cell 128(4),683–692 (2007).

6. Kacevska, M., Ivanov, M. & Ingelman-Sundberg, M. Perspectiveson epigenetics and its relevance to adverse drug reactions. Clin.Pharmacol. Ther. 89(6), 902–907 (2011).

7. Kacevska, M., Ivanov, M. & Ingelman-Sundberg, M. Epigenetic-dependent regulation of drug transport and metabolism: an update.Pharmacogenomics 13(12), 1373–1385 (2012).

8. Feinberg, A.P. Phenotypic plasticity and the epigenetics of humandisease. Nature 447(7143), 433–440 (2007).

9. Szyf, M. Nongenetic inheritance and transgenerational epigenetics.Trends Mol. Med. 21(2), 134–144 (2015).

10. Fraga, M.F. et al. Epigenetic differences arise during the lifetimeof monozygotic twins. Proc. Natl. Acad. Sci. U. S. A. 102(30),10604–10609 (2005).

11. Talens, R.P. et al. Epigenetic variation during the adult lifespan:cross-sectional and longitudinal data on monozygotic twin pairs.Aging Cell. 11(4), 694–703 (2012).

12. Stevens, J.C. et al. Developmental expression of the major humanhepatic CYP3A enzymes. J. Pharmacol. Exp. Ther. 307(2), 573–582(2003).

13. Brouwer, K.L. et al. Human ontogeny of drug transporters: review andrecommendations of the Pediatric Transporter Working Group. Clin.Pharmacol. Ther. 98(3), 266–287 (2015).

14. Mooij, M.G. et al. Development of human membrane transporters:drug disposition and pharmacogenetics. Clin. Pharmacokinet. (2015);e-pub ahead of print.

15. Bonder, M.J. et al. Genetic and epigenetic regulation of geneexpression in fetal and adult human livers. BMC Genomics 15, 860(2014).

16. Seripa, D., Panza, F., Daragjati, J., Paroni, G. & Pilotto, A. Measuringpharmacogenetics in special groups: geriatrics. Expert Opin. DrugMetab. Toxicol. 11(7), 1073–1088 (2015).

17. Branco, M.R., Ficz, G. & Reik, W. Uncovering the role of 5-hydroxymethylcytosine in the epigenome. Nat. Rev. Genet. 13(1),7–13 (2012).

18. Li, W. & Liu, M. Distribution of 5-hydroxymethylcytosine in differenthuman tissues. J. Nucleic Acids 2011, 870726 (2011).

19. Ivanov, M. et al. Ontogeny, distribution and potential roles of5-hydroxymethylcytosine in human liver function. Genome Biol. 14(8),R83 (2013).

20. Xiong, J. et al. DNA hydroxymethylation age of human blooddetermined by capillary hydrophilic-interaction liquid chromatography/mass spectrometry. Clin. Epigenetics 7(1), 72 (2015).

REVIEWS

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 99 NUMBER 5 | MAY 2016 525

Page 15: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

21. Song, C.X., Yi, C. & He, C. Mapping recently identified nucleotidevariants in the genome and transcriptome. Nat. Biotechnol. 30(11),1107–1116 (2012).

22. Zanger, U.M. & Schwab, M. Cytochrome P450 enzymes in drugmetabolism: regulation of gene expression, enzyme activities, andimpact of genetic variation. Pharmacol. Ther. 138(1), 103–141(2013).

23. Habano, W. et al. Involvement of promoter methylation in theregulation of Pregnane X receptor in colon cancer cells BMC Cancer11, 81 (2011).

24. Kacevska, M. et al. DNA methylation dynamics in the hepatic CYP3A4gene promoter. Biochimie. 94(11), 2338–2344 (2012).

25. Okino, S.T. et al. Epigenetic inactivation of the dioxin-responsivecytochrome P4501A1 gene in human prostate cancer. Cancer Res.66(15), 7420–7428 (2006).

26. Anttila, S. et al. Methylation of cytochrome P4501A1 promoter inthe lung is associated with tobacco smoking. Cancer Res. 63(24),8623–8628 (2003).

27. Tekpli, X., Zienolddiny, S., Skaug, V., Stangeland, L., Haugen, A. &Mollerup, S. DNA methylation of the CYP1A1 enhancer is associatedwith smoking-induced genetic alterations in human lung. Int. J. Cancer131(7), 1509–1516 (2012).

28. He, L. et al. Involvement of cytochrome P450 1A1 and glutathioneS-transferase P1 polymorphisms and promoter hypermethylation inthe progression of anti-tuberculosis drug-induced liver injury: a case-control study. PLoS One 10(3), e0119481 (2015).

29. Tokizane, T. et al. Cytochrome P450 1B1 is overexpressed andregulated by hypomethylation in prostate cancer. Clin. Cancer Res.11(16), 5793–5801 (2005).

30. Widschwendter, M. et al. Association of breast cancer DNAmethylation profiles with hormone receptor status and response totamoxifen. Cancer Res. 64(11), 3807–3813 (2004).

31. DiNardo, C.D. et al. Impact of aberrant DNA methylation patternsincluding CYP1B1 methylation in adolescents and young adults withacute lymphocytic leukemia. Am. J. Hematol. 88(9), 784–789 (2013).

32. Jones, S.M., Boobis, A.R., Moore, G.E. & Stanier, P.M. Expression ofCYP2E1 during human fetal development: methylation of the CYP2E1gene in human fetal and adult liver samples. Biochem. Pharmacol.43(8), 1876–1879 (1992).

33. Vieira, I., Sonnier, M. & Cresteil, T. Developmental expression ofCYP2E1 in the human liver. Hypermethylation control of geneexpression during the neonatal period. Eur. J. Biochem. 238(2),476–483 (1996).

34. Kaut, O., Schmitt, I. & Wullner, U. Genome-scale methylation analysisof Parkinson’s disease patients’ brains reveals DNA hypomethylationand increased mRNA expression of cytochrome P450 2E1.Neurogenetics 13(1), 87–91 (2012).

35. Stenstedt, K., Hallstrom, M., Johansson, I., Ingelman-Sundberg, M.,Ragnhammar, P. & Edler, D. The expression of CYP2W1: a prognosticmarker in colon cancer. Anticancer Res. 32(9), 3869–3874 (2012).

36. Travica, S. et al. Colon cancer-specific cytochrome P450 2W1converts duocarmycin analogues into potent tumor cytotoxins. Clin.Cancer Res. 19(11), 2952–2961 (2013).

37. Gomez, A., Karlgren, M., Edler, D., Bernal, M.L., Mkrtchian, S. &Ingelman-Sundberg, M. Expression of CYP2W1 in colon tumors:regulation by gene methylation. Pharmacogenomics 8(10),1315–1325 (2007).

38. Choong, E. et al. Developmental regulation and induction ofcytochrome P450 2W1, an enzyme expressed in colon tumors. PLoSOne 10(4), e0122820 (2015).

39. Chen, B., Rao, X., House, M.G., Nephew, K.P., Cullen, K.J. & Guo, Z.GPx3 promoter hypermethylation is a frequent event in human cancerand is associated with tumorigenesis and chemotherapy response.Cancer Lett. 309(1), 37–45 (2011).

40. Gagnon, J.F., Bernard, O., Villeneuve, L., Tetu, B. & Guillemette, C.Irinotecan inactivation is modulated by epigenetic silencing ofUGT1A1 in colon cancer. Clin. Cancer Res. 12(6), 1850–1858(2006).

41. Oda, S., Fukami, T., Yokoi, T. & Nakajima, M. Epigenetic regulation isa crucial factor in the repression of UGT1A1 expression in the humankidney. Drug Metab. Dispos. 41(10), 1738–1743 (2013).

42. Bieche, I., Girault, I., Urbain, E., Tozlu, S. & Lidereau, R. Relationshipbetween intratumoral expression of genes coding for xenobiotic-metabolizing enzymes and benefit from adjuvant tamoxifen in

estrogen receptor alpha-positive postmenopausal breast carcinoma.Breast Cancer Res. 6(3), R252–R263 (2004).

43. Kim, S.J. et al. Methylation patterns of genes coding for drug-metabolizing enzymes in tamoxifen-resistant breast cancer tissues.J. Mol. Med. (Berl). 88(11), 1123–1131 (2010).

44. Kim, S.J. et al. Promoter hypomethylation of the N-acetyltransferase 1gene in breast cancer. Oncol. Rep. 19(3), 663–668 (2008).

45. Habano, W., Kawamura, K., Iizuka, N., Terashima, J., Sugai, T. &Ozawa, S. Analysis of DNA methylation landscape reveals the roles ofDNA methylation in the regulation of drug metabolizing enzymes. Clin.Epigenetics 7, 105 (2015).

46. Petersen, A.K. et al. Epigenetics meets metabolomics: anepigenome-wide association study with blood serum metabolic traits.Hum. Mol. Genet. 23(2), 534–545 (2014).

47. How, K.A., Nielsen, H.M. & Tost, J. DNA methylation basedbiomarkers: practical considerations and applications. Biochimie.94(11), 2314–2337 (2012).

48. Comerford, K.M., Wallace, T.J., Karhausen, J., Louis, N.A., Montalto,M.C. & Colgan, S.P. Hypoxia-inducible factor-1-dependent regulationof the multidrug resistance (MDR1) gene. Cancer Res. 62(12),3387–3394 (2002).

49. Baker, E.K. & El-Osta, A. MDR1, chemotherapy and chromatinremodeling. Cancer Biol. Ther. 3(9), 819–824 (2004).

50. Dejeux, E. et al. DNA methylation profiling in doxorubicin treatedprimary locally advanced breast tumours identifies novel genesassociated with survival and treatment response. Mol. Cancer 9, 68(2010).

51. Bram, E.E., Stark, M., Raz, S. & Assaraf, Y.G. Chemotherapeuticdrug-induced ABCG2 promoter demethylation as a novel mechanismof acquired multidrug resistance. Neoplasia. 11(12), 1359–1370(2009).

52. Worm, J., Kirkin, A.F., Dzhandzhugazyan, K.N. & Guldberg, P.Methylation-dependent silencing of the reduced folate carrier gene ininherently methotrexate-resistant human breast cancer cells. J. Biol.Chem. 276(43), 39990–40000 (2001).

53. Ferreri, A.J. et al. Aberrant methylation in the promoter region of thereduced folate carrier gene is a potential mechanism of resistance tomethotrexate in primary central nervous system lymphomas. Br. J.Haematol. 126(5), 657–664 (2004).

54. Nies, A.T., Koepsell, H., Damme, K. & Schwab, M. Organic cationtransporters (OCTs, MATEs), in vitro and in vivo evidence for theimportance in drug therapy. Handb. Exp. Pharmacol. (201), 105–167(2011).

55. Nies, A.T. et al. Expression of organic cation transportersOCT1 (SLC22A1) and OCT3 (SLC22A3) is affected by geneticfactors and cholestasis in human liver. Hepatology 50(4),1227–1240 (2009).

56. Aoki, M. et al. Kidney-specific expression of human organic cationtransporter 2 (OCT2/SLC22A2) is regulated by DNA methylation. Am.J. Physiol. Renal Physiol. 295(1), F165–F170 (2008).

57. Schaeffeler, E. et al. DNA methylation is associated withdownregulation of the organic cation transporter OCT1 (SLC22A1) inhuman hepatocellular carcinoma. Genome Med. 3(12), 82 (2011).

58. Lin, R. et al. Long-term cisplatin exposure promotes methylation ofthe OCT1 gene in human esophageal cancer cells. Dig. Dis. Sci.58(3), 694–698 (2013).

59. Chen, L. et al. Genetic and epigenetic regulation of the organic cationtransporter 3, SLC22A3. Pharmacogenomics J. 13(2), 110–120(2013).

60. Fisel, P. et al. DNA methylation of the SLC16A3 promoter regulatesexpression of the human lactate transporter MCT4 in renal cancerwith consequences for clinical outcome. Clin. Cancer Res. 19(18),5170–5181 (2013).

61. Fisel, P. et al. MCT4 surpasses the prognostic relevance of theancillary protein CD147 in clear cell renal cell carcinoma. Oncotarget.6(31), 30615–30627 (2015).

62. Misawa, A. et al. Methylation-associated silencing of the nuclearreceptor 1I2 gene in advanced-type neuroblastomas, identified bybacterial artificial chromosome array-based methylated CpG islandamplification. Cancer Res. 65(22), 10233–10242 (2005).

63. Pancione, M. et al. Epigenetic silencing of peroxisome proliferator-activated receptor gamma is a biomarker for colorectal cancerprogression and adverse patients’ outcome. PLoS One 5(12),e14229 (2010).

REVIEWS

526 VOLUME 99 NUMBER 5 | MAY 2016 | www.wileyonlinelibrary/cpt

Page 16: DNA Methylation of ADME Genes - U-PGxupgx.eu/wp-content/uploads/2017/04/Fisel...DNA methylation, i.e., the covalent addition of aCH 3-group to 50Cs of cytosines within CpG dinucleotides,

64. Marik, R. et al. DNA methylation-related vitamin D receptorinsensitivity in breast cancer. Cancer Biol. Ther. 10(1), 44–53(2010).

65. Varley, K.E. et al. Dynamic DNA methylation across diversehuman cell lines and tissues. Genome Res. 23(3), 555–567(2013).

66. Zhao, J., Goldberg, J., Bremner, J.D. & Vaccarino, V. Associationbetween promoter methylation of serotonin transporter gene anddepressive symptoms: a monozygotic twin study. Psychosom. Med.75(6), 523–529 (2013).

67. Domschke, K. et al. Serotonin transporter gene hypomethylationpredicts impaired antidepressant treatment response. Int. J.Neuropsychopharmacol. 17(8), 1167–1176 (2014).

68. Hidalgo, B. et al. Epigenome-wide association study of fastingmeasures of glucose, insulin, and HOMA-IR in the Genetics of LipidLowering Drugs and Diet Network study. Diabetes 63(2), 801–807(2014).

69. Ivanov, M., Kals, M., Kacevska, M., Metspalu, A., Ingelman-Sundberg, M. & Milani, L. In-solution hybrid capture of bisulfite-

converted DNA for targeted bisulfite sequencing of 174 ADME genes.Nucleic Acids Res. 41(6), e72 (2013).

70. Li, X.Y. et al. Blockade of DNA methylation enhances the therapeuticeffect of gefitinib in non-small cell lung cancer cells. Oncol. Rep.29(5), 1975–1982 (2013).

71. Rius, M. et al. Human concentrative nucleoside transporter1-mediated uptake of 5-azacytidine enhances DNA demethylation.Mol. Cancer Ther. 8(1), 225–231 (2009).

72. Cox, D.B., Platt, R.J. & Zhang, F. Therapeutic genome editing:prospects and challenges. Nat. Med. 21(2), 121–131 (2015).

73. Ivanov, M. & Barragan, I., Ingelman-Sundberg, M. Epigeneticmechanisms of importance for drug treatment. Trends Pharmacol.Sci. 35(8), 384–396 (2014).

74. Malumbres, M. miRNAs and cancer: an epigenetics view.Mol. Aspects Med. 34(4), 863–874 (2013).

75. Fisel, P., Renner, O., Nies, A.T., Schwab, M. & Schaeffeler, E. Solutecarrier transporter and drug-related nephrotoxicity: the impact ofproximal tubule cell models for preclinical research. Expert Opin. DrugMetab. Toxicol. 10(3), 395–408 (2014).

REVIEWS

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 99 NUMBER 5 | MAY 2016 527