simultaneous screening of activities of five cytochrome...

10
1521-009X/43/7/11371146$25.00 http://dx.doi.org/10.1124/dmd.114.063016 DRUG METABOLISM AND DISPOSITION Drug Metab Dispos 43:11371146, July 2015 Copyright ª 2015 by The American Society for Pharmacology and Experimental Therapeutics Simultaneous Screening of Activities of Five Cytochrome P450 and Four Uridine 59-Diphospho-glucuronosyltransferase Enzymes in Human Liver Microsomes Using Cocktail Incubation and Liquid ChromatographyTandem Mass Spectrometry s Boram Lee, Hyeon-Kyeong Ji, Taeho Lee, and Kwang-Hyeon Liu College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea Received January 4, 2015; accepted April 22, 2015 ABSTRACT Cytochrome P450 (P450) and uridine 59-diphospho-glucuronosyltransferase (UGT) are major metabolizing enzymes in the biotransformation of most drugs. Altered P450 and UGT activities are a potential cause of adverse drug-drug interaction. A method for the simultaneous evaluation of the activities of five P450s (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A) and four UGTs (UGT1A1, UGT1A4, UGT1A9, and UGT2B7) was developed using in vitro cocktail incubation and tandem mass spectrometry. The nine probe substrates used in this assay were phenacetin (CYP1A2), diclofenac (CYP2C9), S-mephenytoin (CYP2C19), dextromethorphan (CYP2D6), midazolam (CYP3A), 7-ethyl-10-hydroxy-camptothecin (SN-38) (UGT1A1), trifluoperazine (UGT1A4), mycophenolic acid (UGT1A9), and naloxone (UGT2B7). This new method involves incubation of two cocktail doses and single cassette analysis. The two cocktail doses and the concentration of each probe substrate in vitro were determined to minimize mutual drug interactions among substrates. Cocktail A comprised phenac- etin, diclofenac, S-mephenytoin, dextromethorphan, and midazolam, whereas cocktail B comprised SN-38, trifluoperazine, mycophenolic acid, and naloxone. In the incubation study of these cocktails, the reaction mixtures were pooled and simultaneously analyzed using liquid chromatographytandem mass spectrometry. The method was validated by comparing inhibition data obtained from the incubation of each probe substrate alone with data from the cocktail method. The IC 50 values obtained in both cocktail and individual incubations were in agreement with values previously reported in the literature. This cocktail method offers a rapid and robust way to simultaneously evaluate phase I and II enzyme inhibition profiles of many new chemical entities. This new method will also be useful in the drug discovery process and for advancing the mechanistic understanding of drug interactions. Introduction Drug-drug interaction (DDI) can cause intense clinical complications, either by increasing the toxicity or by weakening the therapeutic efficacy of drugs. DDIs are one of the major reasons for drug withdrawal from the market (Huang et al., 2008). Therefore, early detection of DDIs is a critical factor of drug discovery that has led to the development of new screening methods for determining drug interactions. The present guidelines on pharmacokinetics and DDIs from the U.S. Food and Drug Administration and European Medicines Agency noted that phase I and phase II metabolizing enzymes are of clinical relevance. Cytochrome P450 (P450) and uridine 59 -diphospho-glucuronosyltransferase (UGT) enzymes are the representative phase I and II enzymes, respectively, which play important roles in the metabolism of most drugs. In practice, P450s are the enzymes involved in the bio- transformation of about 75% of all drugs metabolized by phase I enzymes (Guengerich, 2008). UGT enzymes are involved in the biotransformation of about 2030% of all drugs (Meech et al., 2012; Stingl et al., 2014). Several in vitro screening methods for the simultaneous evaluation of potential P450-mediated DDIs have been developed, which include a mixture of P450 probe substrates in a cocktail incubation. The resultant P450-mediated metabolites are determined by liquid chromatographytandem mass spectrometry (LC-MS/MS) (Kim et al., 2005; Na et al., 2011; Spaggiari et al., 2014). Recently, three screening methods for UGT enzyme activities using cocktail incubation and tandem mass spectrometry have also been reported (Gagez et al., 2012; Joo et al., 2014; Seo et al., 2014). A screening method for the simultaneous evaluation of P450 and UGT enzyme activities would accelerate the evaluation of the DDI potential of new chemical entities during drug development. To date, however, no such method has been developed. In this study, we report a new screening method that allows the simultaneous evaluation of the activities of five human hepatic P450 and four UGT enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A, UGT1A1, UGT1A4, UGT1A9, and UGT2B7). Their established probe substrates are used for the screening of potential inhibitory interactions This study was supported by grants from the Korea Health Technology R&D Project, Ministry of Health & Welfare, Republic of Korea [Grant No. A111345], and the National Research Foundation of Korea, Ministry of Science, ICT and Future Planning [NRF-2014M3A9D9069714] and Ministry of Education [NRF-2013R1A1A2008442], Republic of Korea. dx.doi.org/10.1124/dmd.114.063016. s This article has supplemental material available at dmd.aspetjournals.org. ABBREVIATIONS: DDI, drug-drug interaction; HLM, human liver microsome; IS, internal standard; LC-MS/MS, liquid chromatographytandem mass spectrometry; P450, cytochrome P450; QC, quality control; SN-38, 7-ethyl-10-hydroxy-camptothecin; SRM, selected reaction monitoring; UDPGA, uridine 59-diphospho-glucuronic acid; UGT, UDP-glucuronosyltransferase. 1137 http://dmd.aspetjournals.org/content/suppl/2015/04/22/dmd.114.063016.DC1 Supplemental material to this article can be found at: at ASPET Journals on November 20, 2020 dmd.aspetjournals.org Downloaded from

Upload: others

Post on 15-Aug-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

1521-009X/43/7/1137–1146$25.00 http://dx.doi.org/10.1124/dmd.114.063016DRUG METABOLISM AND DISPOSITION Drug Metab Dispos 43:1137–1146, July 2015Copyright ª 2015 by The American Society for Pharmacology and Experimental Therapeutics

Simultaneous Screening of Activities of Five Cytochrome P450 andFour Uridine 59-Diphospho-glucuronosyltransferase Enzymes inHuman Liver Microsomes Using Cocktail Incubation and Liquid

Chromatography–Tandem Mass Spectrometry s

Boram Lee, Hyeon-Kyeong Ji, Taeho Lee, and Kwang-Hyeon Liu

College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea

Received January 4, 2015; accepted April 22, 2015

ABSTRACT

CytochromeP450 (P450) and uridine 59-diphospho-glucuronosyltransferase(UGT) are major metabolizing enzymes in the biotransformation ofmost drugs. Altered P450 and UGT activities are a potential cause ofadverse drug-drug interaction. A method for the simultaneousevaluation of the activities of five P450s (CYP1A2, CYP2C9, CYP2C19,CYP2D6, and CYP3A) and four UGTs (UGT1A1, UGT1A4, UGT1A9, andUGT2B7) was developed using in vitro cocktail incubation and tandemmass spectrometry. The nine probe substrates used in this assaywere phenacetin (CYP1A2), diclofenac (CYP2C9), S-mephenytoin(CYP2C19), dextromethorphan (CYP2D6), midazolam (CYP3A),7-ethyl-10-hydroxy-camptothecin (SN-38) (UGT1A1), trifluoperazine(UGT1A4), mycophenolic acid (UGT1A9), and naloxone (UGT2B7). Thisnew method involves incubation of two cocktail doses and singlecassette analysis. The two cocktail doses and the concentration ofeach probe substrate in vitro were determined to minimize mutual

drug interactions among substrates. Cocktail A comprised phenac-etin, diclofenac, S-mephenytoin, dextromethorphan, and midazolam,whereas cocktail B comprised SN-38, trifluoperazine, mycophenolicacid, and naloxone. In the incubation study of these cocktails, thereaction mixtures were pooled and simultaneously analyzed usingliquid chromatography–tandem mass spectrometry. The method wasvalidated by comparing inhibition data obtained from the incubation ofeach probe substrate alone with data from the cocktail method. TheIC50 values obtained in both cocktail and individual incubations werein agreement with values previously reported in the literature. Thiscocktail method offers a rapid and robust way to simultaneouslyevaluate phase I and II enzyme inhibition profiles of many newchemical entities. This new method will also be useful in the drugdiscovery process and for advancing the mechanistic understandingof drug interactions.

Introduction

Drug-drug interaction (DDI) can cause intense clinical complications,either by increasing the toxicity or by weakening the therapeutic efficacyof drugs. DDIs are one of the major reasons for drug withdrawal fromthe market (Huang et al., 2008). Therefore, early detection of DDIs isa critical factor of drug discovery that has led to the development of newscreening methods for determining drug interactions. The presentguidelines on pharmacokinetics and DDIs from the U.S. Food and DrugAdministration and European Medicines Agency noted that phase I andphase II metabolizing enzymes are of clinical relevance.Cytochrome P450 (P450) and uridine 59-diphospho-glucuronosyltransferase

(UGT) enzymes are the representative phase I and II enzymes,respectively, which play important roles in the metabolism of most

drugs. In practice, P450s are the enzymes involved in the bio-transformation of about 75% of all drugs metabolized by phase Ienzymes (Guengerich, 2008). UGT enzymes are involved in thebiotransformation of about 20–30% of all drugs (Meech et al., 2012;Stingl et al., 2014). Several in vitro screening methods for thesimultaneous evaluation of potential P450-mediated DDIs have beendeveloped, which include a mixture of P450 probe substrates ina cocktail incubation. The resultant P450-mediated metabolites aredetermined by liquid chromatography–tandem mass spectrometry(LC-MS/MS) (Kim et al., 2005; Na et al., 2011; Spaggiari et al.,2014). Recently, three screening methods for UGT enzyme activitiesusing cocktail incubation and tandem mass spectrometry have alsobeen reported (Gagez et al., 2012; Joo et al., 2014; Seo et al., 2014).A screening method for the simultaneous evaluation of P450 and UGTenzyme activities would accelerate the evaluation of the DDI potentialof new chemical entities during drug development. To date, however,no such method has been developed.In this study, we report a new screening method that allows the

simultaneous evaluation of the activities of five human hepatic P450 andfour UGT enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A,UGT1A1, UGT1A4, UGT1A9, and UGT2B7). Their established probesubstrates are used for the screening of potential inhibitory interactions

This study was supported by grants from the Korea Health Technology R&DProject, Ministry of Health & Welfare, Republic of Korea [Grant No. A111345], and theNational Research Foundation of Korea, Ministry of Science, ICT and Future Planning[NRF-2014M3A9D9069714] and Ministry of Education [NRF-2013R1A1A2008442],Republic of Korea.

dx.doi.org/10.1124/dmd.114.063016.s This article has supplemental material available at dmd.aspetjournals.org.

ABBREVIATIONS: DDI, drug-drug interaction; HLM, human liver microsome; IS, internal standard; LC-MS/MS, liquid chromatography–tandemmass spectrometry; P450, cytochrome P450; QC, quality control; SN-38, 7-ethyl-10-hydroxy-camptothecin; SRM, selected reaction monitoring;UDPGA, uridine 59-diphospho-glucuronic acid; UGT, UDP-glucuronosyltransferase.

1137

http://dmd.aspetjournals.org/content/suppl/2015/04/22/dmd.114.063016.DC1Supplemental material to this article can be found at:

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from

Page 2: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

of test compounds. We explored the optimal incubation conditions toavoid potential interactions between the cocktail compounds. Further-more, we developed an analytical method for the simultaneousdetermination of five P450-specific substrate metabolites and fourUGT-specific glucuronide metabolites using LC-MS/MS. To validateour newly developed method, the IC50 values of known P450 and UGT

inhibitors from the new screening method were directly compared withthe IC50 values from the incubation of individual substrates.

Materials and Methods

Chemicals and Reagents. Acetaminophen, alamethicin, a-naphthoflavone,NADP+, dextromethorphan, hecogenin, glucose-6-phosphate, glucose-6-phosphate

TABLE 1

SRM conditions for the major metabolites of the nine substrates used in all assays

Enzyme Substrate Conc. Metabolite Transition Polarity Collision Energy

mM m/z eV

CYP1A2 Phenacetin 100 Acetaminophen 152 . 110 ESI+ 25CYP2C9 Diclofenac 10 4-Hydroxydiclofenac 312 . 231 ESI+ 23CYP2C19 S-mephenytoin 100 49-Hydroxymephenytion 235 . 150 ESI+ 27CYP2D6 Dextromethorphan 5 Dextrorphan 258 . 157 ESI+ 50CYP3A Midazolam 5 19-Hydroxymidazolam 342 . 203 ESI+ 25UGT1A1 SN-38 0.5 SN-38-glucuronide 569 . 393.4 ESI+ 30UGT1A4 Trifluoperazine 0.5 Trifluoperazine N-glucuronide 584.5 . 408.5 ESI+ 30UGT1A9 Mycophenolic acid 0.2 Mycophenolic acid-glucuronide 495 . 319 ESI2 25UGT2B7 Naloxone 1 Naloxone 3-glucuronide 504 . 310 ESI+ 30IS Terfenadine — 472 . 436 ESI+ 25

Conc., concentration; ESI2, electrospray ionization (negative mode); ESI+, electrospray ionization (positive mode).

Fig. 1. Structures of P450 and UGT isoform–specific probe substrates, their metabolites, and IS.

1138 Lee et al.

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from

Page 3: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

dehydrogenase, ketoconazole, magnolol, mefenamic acid, miconazole, mycophe-nolic acid, naloxone, naloxone 3-glucuronide, niflumic acid, phenacetin, quini-dine, D-saccharic acid 1,4-lactone monohydrate, sulfaphenazole, trifluoperazine,Trizma base, troleandomycin, and UDP-glucuronic acid (UDPGA) were pur-chased from Sigma-Aldrich (St. Louis, MO). 7-Ethyl-10-hydroxy-camptothecin(SN-38) was provided by Santa Cruz Biotechnology (Dallas, TX), andatazanavir, S-benzylnirvanol, bilirubin, dextrorphan, diclofenac, furafyl-line, 4-hydroxydiclofenac, 49-hydroxymephenytoin, 19-hydroxymidazolam,ketoconazole, S-mephenytoin, midazolam, mycophenolic acid-b-D-glucuronide,SN-38 glucuronide, and omeprazole were obtained from Toronto Re-search Chemicals (Toronto, ON, Canada). Solvents were LC-MS grade(Fisher Scientific Co., Pittsburgh, PA). Pooled human liver microsomes(HLMs, catalog no. H2630, mixed gender) were purchased from XenoTech(Lenexa, KS).

Microsomal Incubations. All microsomal incubations were done underlinear incubation time and protein concentration conditions for theformation of metabolites. Incubations contained either each substratecocktail set (A set: phenacetin, diclofenac, S-mephenytoin, dextromethor-phan, and midazolam; B set: SN-38, trifluoperazine, mycophenolic acid,and naloxone) or an individual substrate. The final concentration of

methanol in the cocktail incubation conditions was 1.0% (v/v) (Easterbrooket al., 2001; Uchaipichat et al., 2004). The incubation mixtures (finalvolume: 100 ml) for the cocktail A set contained 0.25 mg/ml microsomalprotein, 0.1 M potassium phosphate buffer (pH 7.4), and various P450enzyme–specific substrates or a substrate cocktail A set. After a 5-minutepreincubation at 37�C, the reactions were initiated by adding a NADPH-generating system containing 3.3 mM glucose-6-phosphate, 1.3 mMb-NADP+, 3.3 mM MgCl2, and 500 unit/ml glucose-6-phosphatedehydrogenase, and further incubated for 15 minutes at 37�C in athermoshaker (Kim et al., 2005). The incubation mixtures (final volume:100 ml) for the cocktail B set consisted of 0.25 mg/ml microsomal protein,25 mg/ml alamethicin, 0.1 M Tris-HCl (pH 7.4), 10 mM MgCl2, andvarious UGT enzyme–specific substrates or a substrate cocktail B set (Jooet al., 2014). After preincubation on ice for 15 minutes, the reactions wereinitiated by the addition of 5 mM UDPGA and incubated for 1 hour at 37�C. Allreactions were terminated by the addition of 50 ml of cold acetonitrilecontaining 5 ng/ml terfenadine [internal standard (IS)] to the mixtures, and werecentrifuged at 10,000g for 5 minutes at 4�C. The supernatants of the individualincubation samples and pooled cocktail reaction samples (A set/B set, 1/1) wereanalyzed by LC-MS/MS.

Fig. 2. Representative selected reaction monitoring chromatograms of P450- and UGT-mediated metabolites of probe substrate and internal standard: acetaminophen (A),4-hydroxydiclofenac (B), 49-hydroxymephenytoin (C), dextrorphan (D), 19-hydroxymidazolam (E), SN-38 glucuronide (F), trifluoperazine N-glucuronide (G), mycophenolicacid glucuronide (H), naloxone 3-glucuronide (I), and terfenadine (internal standard) (J).

Simultaneous Evaluation of P450 and UGT Enzyme Activities 1139

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from

Page 4: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

LC-MS/MS Analysis. The samples were analyzed using a Thermo Vantagetriple quadrupole mass spectrometer coupled with a high-performance liquidchromatography system (Thermo Fisher Scientific, San Jose, CA). Analytesseparation was performed on a Phenomenex (Torrance, CA) Kinetex XB-C18high-performance liquid chromatography column (2.6 mm, 100 Å, 100� 2.10 mm).The mobile phase consisted of 0.1% formic acid in acetonitrile (A) and 0.1%formic acid in water (B), which formed the following gradient: 0 minute (0% A),0–1 minute (40% A), 1–5 minutes (50% A), 5–5.1 minutes (0% A), and 5.1–8 min-utes (0% A) (Joo et al., 2014). Quantitation was performed by selected reac-tion monitoring (SRM) of the [M+H]+ (or [M–H]–) ion and the related production for each metabolite, using an IS to establish peak area ratios. The SRMtransitions and collision energies determined for each metabolite and IS arelisted in Table 1.

Method Validation. Four inter- and intraday validations were performedto validate the LC-MS/MS method for the simultaneous quantification of thefive P450 and four UGT probe metabolites in microsomal incubates.Calibration standards were prepared at six different concentrations from 20 to5000 nM (acetaminophen, 4-hydroxydiclofenac, 49-hydroxymephenytoin,trifluoperazine N-glucuronide, and mycophenolic acid glucuronide) or 2 to500 nM (dextrorphan, 19-hydroxymidazolam, SN-38 glucuronide, andnaloxone 3-glucuronide) in a blank microsomal mixture. Quality control(QC) samples were prepared separately at two different concentrations (5 and20 nM for dextrorphan, 19-hydroxymidazolam, SN-38 glucuronide, andnaloxone 3-glucuronide; 50 and 200 nM for other metabolites) for assays.Inter- and intraday precision and accuracy were determined by analyzingreplicates of the QC samples (n = 4).

Comparison of the Cocktail and Individual Substrates for InhibitionScreening. Known inhibitors of specific P450 and UGT enzymes (a-naphthoflavonefor CYP1A2, sulfaphenazole for CYP2C9, S-benzylnirvanol for CYP2C19,quinidine for CYP2D6, ketoconazole for CYP3A, atazanavir for UGT1A1,hecogenin for UGT1A4, niflumic acid for UGT1A9, and mefenamic acid forUGT2B7) were incubated with each substrate cocktail set and with theindividual substrates alone, and the results were compared. The incubationswere performed (as described earlier) with various inhibitor concentrations, andall incubations were performed in triplicate. Furafylline, paroxetine, andtroleandomycin were preincubated for 10 minutes at 37�C with HLMs and aNADPH-generating system (as described earlier). The reactions were initiatedby the addition of the individual or cocktail substrate. With the exception of theaddition of P450 or UGT isoform–specific inhibitors, all other incubationconditions were as described earlier.

Data Analysis. The P450 or UGT isoform–mediated activities in the presenceof inhibitors were expressed as percentages of the corresponding control values.The percentage of inhibition was calculated by the ratio of the amounts ofmetabolites formed, in the presence and absence of the specific inhibitor. Tocalculate the enzyme inhibition IC50 values, the relevant data were fitted to aninhibitory effect model [i.e., v = Emax � (12 [I]/(IC50 + [I]))] using WinNonlin(Pharsight, Mountain View, CA).

Results

Substrate Selection and Optimization of Microsomal Incuba-tions. The structures of the P450 and UGT probe substrates, theirmetabolites, and the internal standard used in the assays are shown inFig. 1. In this study, we selected a P450 isoform–specific probesubstrate based on the preferred and accepted P450 substrates forassessing activity in vitro (Tucker et al., 2001; Na et al., 2011). TheUGT isoform–specific substrates were selected on the basis of theirspecificity and previously published data (Hanioka et al., 2001; DiMarco et al., 2005; Picard et al., 2005; Uchaipichat et al., 2006; Jooet al., 2014). The probe substrates for the P450 and UGT isoforms wereas follows: phenacetin, CYP1A2; diclofenac, CYP2C9; S-mephenytoin,CYP2C19; dextromethorphan, CYP2D6; midazolam, CYP3A; SN-38,UGT1A1; trifluoperazine, UGT1A4; mycophenolic acid, UGT1A9; andnaloxone, UGT2B7. The optimum microsomal incubation times were15 minutes and 1 hour for phase I (cocktail A set) and phase II (cocktail Bset), respectively, with 0.25 mg/ml microsomal protein. The concentrationof each probe substrate was optimized to avoid interactions betweenthem: 100 mM for phenacetin, 10 mM for diclofenac, 100 mM forS-mephenytoin, 5 mM for dextromethorphan, 5 mM for midazolam,0.5 mM for SN-38, 0.5 mM for trifluoperazine, 0.2 mM for mycophenolicacid, and 1 mM for naloxone (Table 1). These concentrations were lowerthan their respective Km values.Development of a Simultaneous Analytical Method Using

LC-MS/MS. We developed a method for simultaneously analyzingmetabolites of five P450 and four UGT probe substrates usingLC-MS/MS. Among the nine metabolites, only mycophenolic acidglucuronide was detected in the negative mode because of its poorionization property in the positive mode. The remaining eightmetabolites and the IS were monitored in the positive mode. TheSRM transitions and optimal MS/MS collision energy are described inTable 1. The representative chromatograms for the five P450 and fourUGT probe metabolites and the IS in microsomal incubation mixturesare presented in Fig. 2. As shown in Fig. 2, all of the metabolites wereeluted in less than 6.5 minutes and separated into their individual SRMchannels. The retention times for acetaminophen, 4-hydroxydiclofenac,49-hydroxymephenytoin, dextrorphan, 19-hydroxymidazolam, SN-38glucuronide, trifluoperazine N-glucuronide, mycophenolic acid glucu-ronide, naloxone 3-glucuronide, and terfenadine were approximately3.3, 6.2, 3.9, 3.4, 3.9, 3.6, 4.8, 4.0, 3.0, and 5.8 minutes, respectively.For acetaminophen, three peaks were observed in the microsomalincubation. Similar results have also been observed in other reportedstudies (Joo and Liu, 2013; Pillai et al., 2013; Song et al., 2013). The

Fig. 3. Comparison of the results from the cocktailincubation with the individual incubations for each offive P450 (A) and four UGT (B) enzyme activities.Incubations were performed with the individual sub-strate (j) and each substrate cocktail set (u) in pooledHLMs. The activities are the means of triplicateincubations.

1140 Lee et al.

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from

Page 5: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

peak with a retention time of 3.3 minutes was identified as acet-aminophen by comparing it with the retention time of an authenticstandard.

Cocktail Dose Selection. The inhibition potential of each P450 orUGT substrate was evaluated by comparing the reaction of eachmetabolite in the single-substrate incubations to the response of thesame metabolite formed in incubations with the two-substratecocktail set. The change in each P450 and UGT enzyme activitylevel was less than 15% in each cocktail set, compared with theindividual incubation (Fig. 3). The relative standard deviationsranged from 2.4 to 10.3% (n = 3) for the results of the cocktailincubation procedure.

Method Validation. The inter- and intraday precision and accuracyof the method were determined by analyzing four QC replicates. Themethod accuracy was expressed as the percentage of the metaboliteconcentration measured in each sample relative to the known amount ofmetabolites added (Joo et al., 2014). Calibration curves produced goodcorrelation coefficients for nine metabolites in the mixture (r . 0.999).The interday and intraday accuracy and precision data for the five P450and four UGT probe metabolites in human liver microsomal incubatesare summarized in Tables 2 and 3. Overall, the inter- and intradayaccuracy was 88.2–110.9% with a precision of less than 15.3%.

Validation of the Screening Method Using Selective Inhibitors.The newly developed screening method, used as a tool for determiningP450 and UGT inhibition, was validated using known selectiveinhibitors of the isoforms (a-naphthoflavone, CYP1A2; sulfaphenazole,CYP2C9; S-benzylnirvanol, CYP2C19; quinidine, CYP2D6; ketocona-zole, CYP3A; atazanavir, UGT1A1; hecogenin, UGT1A4; niflumicacid, UGT1A9; and mefenamic acid, UGT2B7) with their correspond-ing substrates. The IC50 value of each P450 and UGT isoform–selectiveinhibitor was estimated in both individual and cocktail incubations(Table 4). The inhibition curves (Fig. 4) show there was no substantialdifference between the individual inhibitor profiles for the two differentincubation methods (single versus cocktail).

Discussion

Pharmacodynamic interactions between drugs alter the response toone or both drugs while also affecting their plasma concentrations(Singh, 2006). Therefore, the risk of metabolism-based DDIs is alwaysa potential problem to consider during the drug development process.For this reason, in vitro drug interaction studies using HLMs are usedfor the early estimation and prediction of the in vivo drug interactionpotential of new candidate drugs (Baranczewski et al., 2006). SeveralLC-MS/MS–based P450 (Dierks et al., 2001; Kim et al., 2005; Smith

TABLE 2

Intraday (n = 4) validation data for the simultaneous determination of five P450- and four UGT-specific probemetabolites in human liver microsomal incubations using the LC-MS/MS method

Intraday accuracy and precision were determined at two different concentration levels (n = 4).

Metabolites50 nM 200 nM

Conc. Founda Accuracy RSD Conc. Founda Accuracy RSD

nM % % nM % %

Acetaminophen 49.3 98.6 8.1 202.1 101.0 5.54-Hydroxydiclofenac 45.8 91.7 6.4 182.0 91.0 11.749-Hydroxymephenytoin 51.5 103.0 13.4 202.9 101.5 14.1Dextrorphanb 4.41 88.2 15.3 20.0 99.9 3.919-Hydroxymidazolamb 4.85 97.1 7.2 19.2 95.8 12.1SN-38 glucuronideb 5.30 106.0 10.8 19.1 95.7 6.4Trifluoperazine N-glucuronide 45.4 90.8 1.9 179.9 89.9 8.5Mycophenolic acid glucuronide 47.2 94.4 6.3 182.5 91.2 6.8Naloxone 3-glucuronideb 5.55 110.9 3.8 19.4 94.6 3.7

Conc., concentration; RSD, relative standard deviation.aEach value represents the mean of four replicates.bDextrorphan, 19-hydroxymidazolam, SN-38 glucuronide, and naloxone 3-glucuronide concentrations were 5 and 20 nM.

TABLE 3

Interday (n = 4) validation data for the simultaneous determination of five P450- and four UGT-specific probemetabolites in human liver microsomal incubations using the LC-MS/MS method

Interday accuracy and precision were determined at two different concentration levels (n = 4).

Metabolites50 nM 200 nM

Conc. Founda Accuracy RSD Conc. Founda Accuracy RSD

nM % % nM % %

Acetaminophen 52.0 104.1 4.9 202.0 101.0 3.04-Hydroxydiclofenac 50.1 100.1 9.6 198.5 99.2 8.449-Hydroxymephenytoin 51.3 102.7 3.4 203.8 101.9 2.3Dextrorphanb 4.61 92.1 8.5 20.6 103.0 7.119-Hydroxymidazolamb 5.35 107.0 5.7 20.2 100.7 8.7SN-38 glucuronideb 4.81 96.2 9.9 19.9 99.3 5.4Trifluoperazine N-glucuronide 50.7 101.5 3.3 201.4 100.7 5.3Mycophenolic acid glucuronide 46.0 92.9 8.0 188.8 94.4 4.2Naloxone 3-glucuronideb 5.46 109.2 4.9 20.2 101.0 3.2

Conc., concentration; RSD, relative standard deviation.aEach value represents the mean of four replicates.bDextrorphan, 19-hydroxymidazolam, SN-38 glucuronide, and naloxone 3-glucuronide concentrations were 5 and 20 nM.

Simultaneous Evaluation of P450 and UGT Enzyme Activities 1141

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from

Page 6: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

et al., 2007; Zambon et al., 2010; Na et al., 2011; Pillai et al., 2013) orUGT (Gagez et al., 2012; Joo et al., 2014; Seo et al., 2014) inhibitionmethods for the evaluation of in vitro drug interaction potential havebeen reported. To date, however, a simultaneous evaluation methodfor both P450 and UGT enzyme activities has not been developed. Theaim of this study was to develop and validate a cocktail assay tosimultaneously evaluate the activity of hepatic P450 and UGTisoforms in HLMs using LC-MS/MS.P450 and UGT are representative phase I and II enzymes responsible

for the biotransformation of a wide variety of drugs (Evans andRelling, 1999). Among the numerous P450 and UGT enzymesidentified to date, five P450 (CYP1A2, CYP2C9, CYP2C19,CYP2D6, and CYP3A) and four UGT (UGT1A1, UGT1A4,UGT1A9, and UGT2B7) enzymes have been shown to play animportant role in the metabolism of marketed drugs. P450 enzymesbiotransform about 75% of all drugs, and the aforementioned fiveP450 human isoforms biotransform about 95% of marketed drugs

(Baj-Rossi et al., 2011). In the case of UGT enzymes, UGT2B7 wassuggested to be responsible for the hepatic glucuronidation of 40% ofdrugs, whereas UGT1A1, 1A4, and 1A9 contribute to an additional47% (Williams et al., 2004; Burchell et al., 2005). Based on these data,we attempted to develop a method for the simultaneous evaluation ofthe activities of five P450 (P450s 1A2, 2C9, 2C19, 2D6, and 3A) andfour UGT (UGTs 1A1, 1A4, 1A9, and 2B7) enzymes, which areprimarily responsible for drug metabolism.The optimum incubation conditions for the assessment of enzyme

activities are different between the P450 and UGT enzymes. Themicrosomal incubation of P450 enzymes was conducted in phosphatebuffer, whereas UGT enzymes were incubated in Tris-HCl buffer.UGT enzyme activity was assessed in the presence of alamethicin,a membrane permeabilizing agent (Fisher et al., 2000), and saccharolactone,a b-glucuronidase inhibitor (Oleson and Court, 2008), using a longerincubation time (1 hour). In addition, UDPGA also has inhibitorypotential against CYP2C9 and CYP2C19 activity to some degree (Yan

Fig. 4. Inhibition curves determined using individual substrates and the substrate cocktails. Each P450- and UGT-selective inhibitor was incubated in a separate experimentwith cocktail substrates (d) or an individual substrate set (s). The activity is expressed as the percentage of the remaining activity compared with a control sample containingno inhibitor. Inhibition of phenacetin O-deethylation by a-naphthoflavone (A), diclofenac 4-hydroxylation by sulfaphenazole (B), S-mephenytoin 49-hydroxylation byS-benzylnirvanol (C), dextromethorphan O-demethylation by quinidine (D), midazolam 19-hydroxylation by ketoconazole (E), SN-38 glucuronidation by atazanavir (F),trifluoperazine N-glucuronidation by hecogenin (G), mycophenolic acid glucuronidation by niflumic acid (H), and naloxone 3-glucuronidation by mefenamic acid (I).

1142 Lee et al.

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from

Page 7: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

and Caldwell, 2003). Therefore, we used two separate microsomalincubations with P450 and UGT probe substrates. The substrates weredivided into two cocktail groups (cocktail A for five P450 probesubstrates and cocktail B for four UGT probe substrates). These twococktail mixtures were pooled after incubation and analyzed togetherusing LC-MS/MS to reduce the assay time.The selection of specific probe substrates for each P450 and UGT

enzyme is important because multiple enzymes are involved in themetabolism of a single drug. Therefore, in this study, we chosespecific probe substrates for the nine major P450 and UGT enzymes inthe human liver based on previous reports (Table 1). Phenacetin,dextromethorphan, and midazolam are the most frequently used probesubstrates for in vitro activity assessment of CYP1A2, CYP2D6, andCYP3A enzymes, respectively, using the cocktail approach, and arealso the preferred probes for regulatory authorities (Kim et al., 2005;Otten et al., 2011; Pillai et al., 2013). S-mephenytoin (Dierks et al.,2001; Kim et al., 2005; Smith et al., 2007; Zambon et al., 2010; Ottenet al., 2011; Pillai et al., 2013) and omeprazole (Testino and Patonay,

2003; He et al., 2007; Tolonen et al., 2007; Song et al., 2013) are usedas CYP2C19 substrates in cocktail incubation studies. In ourpreliminary study, omeprazole (20 mM) inhibited CYP3A-mediatedmidazolam hydroxylase activity (40% of control) and CYP1A2-mediated phenacetin O-deethylation activity (.20% of control)(Supplemental Fig. 1). Diclofenac (Dierks et al., 2001; Smith et al.,2007; Pillai et al., 2013) and tolbutamide (Bu et al., 2001; Testino andPatonay, 2003; Kim et al., 2005; He et al., 2007; Tolonen et al., 2007;Otten et al., 2011; Joo and Liu, 2013) are generally used as CYP2C9probe substrates. In this study, diclofenac was selected as the CYP2C9probe substrate because it showed higher detection intensity thantolbutamide in the proposed LC-MS/MS method. Based on ourpreliminary findings, phenacetin, diclofenac, S-mephenytoin, dextro-methorphan, and midazolam were selected as the CYP1A2, CYP2C9,CYP2C19, CYP2D6, and CYP3A probe substrates, respectively(cocktail A set). For the cocktail B set (UGT enzymes), SN-38,trifluoperazine, mycophenolic acid, and naloxone were selected asprobe substrates for UGT1A1, UGT1A4, UGT1A9, and UGT2B7,

Fig. 5. Inhibitory effects of a-naphthoflavone (A), sulfaphenazole (B), S-benzylnirvanol (C), quinidine (D), ketoconazole (E), atazanavir (F), hecogenin (G), niflumic acid(H), and mefenamic acid (I) on phenacetin O-deethylation (d), diclofenac 4-hydroxylation (s), S-mephenytoin 49-hydroxylation (.), dextromethorphan O-demethylation(n), midazolam 19-hydroxylation (j), SN-38 glucuronidation (u), trifluoperazine N-glucuronidation (♦), mycophenolic acid glucuronidation ()), and naloxone3-glucuronidation (m) on incubation with pooled HLMs. Data are the means of triplicate experiments.

Simultaneous Evaluation of P450 and UGT Enzyme Activities 1143

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from

Page 8: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

respectively, based on previously reported data (Uchaipichat et al.,2004; Di Marco et al., 2005; Picard et al., 2005) and our recentlypublished data (Joo et al., 2014).Following the selection of the P450 and UGT probe substrates for

the cocktail incubation study, the potential metabolic interactionsamong the five P450 metabolites and four UGT metabolites of theselected probe substrates were evaluated. The simultaneous incubationof five or four substrates may lead to interactions. As shown in Fig. 3,P450 and UGT enzyme activities in the cocktail incubations were inaccordance with the results from individual incubations, suggestingthat drug interactions among probe substrates in the present cocktailset were negligible.P450 and UGT isoform–selective inhibitors represent the most

powerful tools available for metabolizing enzyme phenotyping. Thewell known P450 and UGT inhibitors include a-naphthoflavone forCYP1A2 (von Moltke et al., 1996), sulfaphenazole for CYP2C9(Khojasteh et al., 2011), S-benzylnirvanol for CYP2C19 (Suzuki et al.,2002), quinidine for CYP2D6 (Khojasteh et al., 2011), ketoconazolefor CYP3A (Khojasteh et al., 2011), hecogenin for UGT1A4(Uchaipichat et al., 2006), and niflumic acid for UGT1A9 (Minerset al., 2011). Atazanavir (Zhang et al., 2005) and mefenamic acid(Mano et al., 2007) are known inhibitors of UGT1A1 and UGT2B7,respectively. Our results demonstrated that a-naphthoflavone, sulfa-phenazole, S-benzylnirvanol, quinidine, hecogenin, and niflumic acidstrongly and selectively inhibited CYP1A2, CYP2C9, CYP2C19,

CYP2D6, UGT1A4, and UGT1A9, respectively (Fig. 5; Table 5). Liuet al. (2011) also reported that selective P450 inhibitors such asa-naphthoflavone, sulfaphenazole, and quinidine have negligibleinhibitory potential against UGT isoforms. Ketoconazole alsoselectively inhibits CYP3A with an IC50 value of 0.03 mM, whichis at least 100-fold more selective compared with its effects against allother P450s and UGTs tested (Fig. 5E) (Ren et al., 2013). Theinhibitory potential of ketoconazole on UGT1A1-mediated SN-38glucuronidation (IC50 = 4.8 mM) concurred with previous findings inwhich ketoconazole had inhibitory potential on UGT1A1-catalyzedb-estradiol glucuronidation (IC50 = 4.1 mM) (Liu et al., 2011).Hecogenin and niflumic acid are UGT1A4 and UGT1A9 selectiveinhibitors, respectively, and they have a negligible inhibitory effect onthe P450 isoforms tested (IC50 . 20 mM; Table 5). Atazanavirstrongly inhibited UGT1A1 activity with an IC50 value of 0.4 mM;however, it also inhibited CYP3A activity (IC50 = 1.8 mM; Fig. 5F).Mefenamic acid inhibited UGT2B7 activity (IC50 = 5.0 mM), but alsoweakly inhibited CYP1A2 and CYP2C9 activity with IC50 values of8.3 and 12 mM, respectively (Fig. 5I).We also evaluated the IC50 values of the characterized inhibitors

for each P450 and UGT enzyme using both the individual and thecocktail substrates (Fig. 4). The IC50 value of each cocktail set usingthis approach was comparable to those of the individual substratesand was in agreement with those previously reported (Table 4). Thisconfirms that the IC50 values of P450 and UGT inhibitors can be

TABLE 4

Comparison of IC50 values estimated using the individual substrate and the substrate cocktails of well known P450- and UGT-selective inhibitors

Inhibitors of P450 and UGT enzymes were incubated with each substrate cocktail set and with individual substrate alone. IC50 values were calculated using a nonlinear least-squares regressionanalysis.

Enzyme Substrates InhibitorsIC50 (mM 6 S.D.)a

ReferencesIndividual Substrate Cocktail Substrate Literature

CYP1A2 Phenacetin a-Naphthoflavone 0.014 6 0.003 0.013 6 0.004 0.01–0.27 (Gao et al., 2007; Zhang et al., 2008; Perloff et al., 2009)CYP2C9 Diclofenac Sulfaphenazole 0.70 6 0.08 0.68 6 0.08 0.3–1.5 (Dierks et al., 2001; Kim et al., 2005)CYP2C19 S-mephenytoin S-benzylnirvanol 1.02 6 0.13 1.20 6 0.06 0.4–0.41 (Walsky and Obach, 2003; Walsky and Obach, 2004)CYP2D6 Dextromethorphan Quinidine 0.11 6 0.01 0.13 6 0.01 0.02–0.68 (Dierks et al., 2001; Kim et al., 2005)CYP3A Midazolam Ketoconazole 0.038 6 0.001 0.032 6 0.0004 0.09–0.24 (Dierks et al., 2001; Kim et al., 2005)UGT1A1 SN-38 Atazanavir 0.37 6 0.11 0.38 6 0.13 0.4–2.5 (Zhang et al., 2005)UGT1A4 Trifluoperazine Hecogenin 0.89 6 0.21 0.86 6 0.11 0.64–1.5 (Uchaipichat et al., 2006)UGT1A9 Mycophenolic acid Niflumic acid 0.78 6 0.10 0.83 6 0.18 0.1–8 (Vietri et al., 2000; Mano et al., 2006; Miners et al., 2011)UGT2B7 Naloxone Mefenamic acid 4.18 6 1.01 5.04 6 1.34 0.3–370 (Mano et al., 2007; Knights et al., 2009)

aValues represent the mean (6S.D.) of experiments performed in triplicate.

TABLE 5

Effects of specific enzyme inhibitors on cytochrome P450 and uridine 59-diphospho-glucuronosyltransferase metabolicactivities in pooled human liver microsomes

Enzyme Inhibitors

IC50a

P450s UGTs

1A2 2C9 2C19 2D6 3A 1A1 1A4 1A9 2B7

mM mM mM mM mM mM mM mM mM

CYP1A2 a-Naphthoflavone 0.01 — — — — — — — —

CYP2C9 Sulfaphenazole — 0.7 — — — — — — —

CYP2C19 S-benzylnirvanol — — 1.2 — — — — — —

CYP2D6 Quinidine — — — 0.2 — — — — —

CYP3A Ketoconazole — — — — 0.03 4.8 — — —

UGT1A1 Atazanavir 15 20 10 13 1.8 0.4 15 — —

UGT1A4 Hecogenin — — — — — — 0.9 — —

UGT1A9 Niflumic acid 15 — — — — 20 — 0.8 —

UGT2B7 Mefenamic acid 8.3 12 — — — — — — 5.0

—, IC50 . 50 mΜ.aValues represent the mean of experiments performed in triplicate.

1144 Lee et al.

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from

Page 9: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

accurately determined using the cocktail assay instead of individualsubstrate incubations, which would save considerable time in thescreening process for new chemical entities. One exception observedwas in the inhibition of CYP2C19 by S-benzylnirvanol, whichshowed a slight difference compared with previously publishedvalues. The published IC50 value of 0.41 mM for S-benzylnirvanolagainst CYP2C19-mediated S-mephenytoin hydroxylation activity(Walsky and Obach, 2003) was lower than our present data (1.2 mM;Table 4). Such a difference is not unusual and could be due to the useof different microsomal incubation conditions (Dierks et al., 2001).Nonetheless, our results demonstrate that the IC50 values ofinhibitors against the nine enzymes can be determined accuratelyby cocktail incubation and cassette analysis, thereby reducing thetime required.In conclusion, we developed a LC-MS/MS method for the si-

multaneous determination of five P450 and four UGT enzyme ac-tivities. Nine substrates were divided into two cocktail sets forincubation and pooled for LC-MS/MS analysis in a single run. Thisprocess allowed us to simultaneously evaluate the activity of five P450and four UGT enzyme activities, using well known P450 and UGTisoform–specific inhibitors. In addition, this cocktail method involvingmultiple substrates provided inhibition profiles similar to thoseobtained from single-substrate incubations. Therefore, these resultssuggest that this newly developed assay can be a useful tool for robustand rapid screening, which accelerates the prediction of the P450 andUGT inhibitory potential of new chemical entities.

Authorship ContributionsParticipated in research design: B. Lee, T. Lee, Liu.Conducted experiments: B. Lee, Ji, Liu.Contributed new reagents or analytic tools: B. Lee, Liu.Performed data analysis: B. Lee, T. Lee, Liu.Wrote or contributed to the writing of the manuscript: B. Lee, T. Lee, Liu.

References

Baj-Rossi C, De Micheli G, and Carrara S (2011) Based Nano-Bio-Sensors for PersonalizedMedicine, p 450, INTECH Open Access Publisher, Rijeka, Croatia.

Baranczewski P, Sta�nczak A, Sundberg K, Svensson R, Wallin A, Jansson J, Garberg P,and Postlind H (2006) Introduction to in vitro estimation of metabolic stability and druginteractions of new chemical entities in drug discovery and development. Pharmacol Rep 58:453–472.

Bu HZ, Magis L, Knuth K, and Teitelbaum P (2001) High-throughput cytochrome P450 (CYP)inhibition screening via a cassette probe-dosing strategy. VI. Simultaneous evaluation of in-hibition potential of drugs on human hepatic isozymes CYP2A6, 3A4, 2C9, 2D6 and 2E1.Rapid Commun Mass Spectrom 15:741–748.

Burchell B, Lockley DJ, Staines A, Uesawa Y, and Coughtrie MW (2005) Substrate specificity ofhuman hepatic udp-glucuronosyltransferases. Methods Enzymol 400:46–57.

Dierks EA, Stams KR, Lim HK, Cornelius G, Zhang H, and Ball SE (2001) A method for thesimultaneous evaluation of the activities of seven major human drug-metabolizing cytochromeP450s using an in vitro cocktail of probe substrates and fast gradient liquid chromatographytandem mass spectrometry. Drug Metab Dispos 29:23–29.

Di Marco A, D’Antoni M, Attaccalite S, Carotenuto P, and Laufer R (2005) Determination ofdrug glucuronidation and UDP-glucuronosyltransferase selectivity using a 96-well radiometricassay. Drug Metab Dispos 33:812–819.

Easterbrook J, Lu C, Sakai Y, and Li AP (2001) Effects of organic solvents on the activities ofcytochrome P450 isoforms, UDP-dependent glucuronyl transferase, and phenol sulfotransfer-ase in human hepatocytes. Drug Metab Dispos 29:141–144.

Evans WE and Relling MV (1999) Pharmacogenomics: translating functional genomics intorational therapeutics. Science 286:487–491.

Fisher MB, Campanale K, Ackermann BL, VandenBranden M, and Wrighton SA (2000) In vitroglucuronidation using human liver microsomes and the pore-forming peptide alamethicin.Drug Metab Dispos 28:560–566.

Gagez AL, Rouguieg-Malki K, Sauvage FL, Marquet P, and Picard N (2012) Simultaneousevaluation of six human glucuronidation activities in liver microsomes using liquidchromatography-tandem mass spectrometry. Anal Biochem 427:52–59.

Gao ZW, Shi XJ, Yu C, Li SJ, and Zhong MK (2007) [Simultaneous determination of theinhibitory potency of compounds on the activity of five cytochrome P-450 enzymes usinga cocktail probe substrates method]. Yao Xue Xue Bao 42:589–594.

Guengerich FP (2008) Cytochrome p450 and chemical toxicology. Chem Res Toxicol 21:70–83.Hanioka N, Ozawa S, Jinno H, Ando M, Saito Y, and Sawada J (2001) Human liver UDP-glucuronosyltransferase isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin.Xenobiotica 31:687–699.

He F, Bi HC, Xie ZY, Zuo Z, Li JK, Li X, Zhao LZ, Chen X, and Huang M (2007) Rapiddetermination of six metabolites from multiple cytochrome P450 probe substrates in human

liver microsome by liquid chromatography/mass spectrometry: application to high-throughputinhibition screening of terpenoids. Rapid Commun Mass Spectrom 21:635–643.

Huang SM, Strong JM, Zhang L, Reynolds KS, Nallani S, Temple R, Abraham S, Habet SA,Baweja RK, and Burckart GJ, et al. (2008) New era in drug interaction evaluation: US Foodand Drug Administration update on CYP enzymes, transporters, and the guidance process.J Clin Pharmacol 48:662–670.

Joo J, Lee B, Lee T, and Liu K-H (2014) Screening of six UGT enzyme activities in human livermicrosomes using liquid chromatography/triple quadrupole mass spectrometry. Rapid CommunMass Spectrom 28:2405–2414.

Joo JM and Liu KH (2013) Inhibitory effect of honokiol and magnolol on cytochrome P450enzyme activities in human liver microsomes. Mass Spectrom Lett 4:34–37.

Khojasteh SC, Prabhu S, Kenny JR, Halladay JS, and Lu AY (2011) Chemical inhibitors ofcytochrome P450 isoforms in human liver microsomes: a re-evaluation of P450 isoform se-lectivity. Eur J Drug Metab Pharmacokinet 36:1–16.

Kim MJ, Kim H, Cha IJ, Park JS, Shon JH, Liu KH, and Shin JG (2005) High-throughputscreening of inhibitory potential of nine cytochrome P450 enzymes in vitro using liquidchromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom 19:2651–2658.

Knights KM, Winner LK, Elliot DJ, Bowalgaha K, and Miners JO (2009) Aldosteroneglucuronidation by human liver and kidney microsomes and recombinant UDP-glucuronosyl-transferases: inhibition by NSAIDs. Br J Clin Pharmacol 68:402–412.

Liu Y, She M, Wu Z, and Dai R (2011) The inhibition study of human UDP-glucuronosyltransferases with cytochrome P450 selective substrates and inhibitors. J EnzymeInhib Med Chem 26:386–393.

Mano Y, Usui T, and Kamimura H (2006) In vitro inhibitory effects of non-steroidal anti-inflammatory drugs on 4-methylumbelliferone glucuronidation in recombinant human UDP-glucuronosyltransferase 1A9—potent inhibition by niflumic acid. Biopharm Drug Dispos 27:1–6.

Mano Y, Usui T, and Kamimura H (2007) Inhibitory potential of nonsteroidal anti-inflammatorydrugs on UDP-glucuronosyltransferase 2B7 in human liver microsomes. Eur J Clin Pharmacol63:211–216.

Meech R, Miners JO, Lewis BC, and Mackenzie PI (2012) The glycosidation of xenobiotics andendogenous compounds: versatility and redundancy in the UDP glycosyltransferase super-family. Pharmacol Ther 134:200–218.

Miners JO, Bowalgaha K, Elliot DJ, Baranczewski P, and Knights KM (2011) Characterization ofniflumic acid as a selective inhibitor of human liver microsomal UDP-glucuronosyltransferase1A9: application to the reaction phenotyping of acetaminophen glucuronidation. Drug MetabDispos 39:644–652.

Na DH, Ji HY, Park EJ, Kim MS, Liu KH, and Lee HS (2011) Evaluation of metabolism-mediated herb-drug interactions. Arch Pharm Res 34:1829–1842.

Oleson L and Court MH (2008) Effect of the beta-glucuronidase inhibitor saccharolactone onglucuronidation by human tissue microsomes and recombinant UDP-glucuronosyltransferases.J Pharm Pharmacol 60:1175–1182.

Otten JN, Hingorani GP, Hartley DP, Kragerud SD, and Franklin RB (2011) An in vitro, highthroughput, seven CYP cocktail inhibition assay for the evaluation of new chemical entitiesusing LC-MS/MS. Drug Metab Lett 5:17–24.

Perloff ES, Mason AK, Dehal SS, Blanchard AP, Morgan L, Ho T, Dandeneau A, Crocker RM,Chandler CM, and Boily N, et al. (2009) Validation of cytochrome P450 time-dependentinhibition assays: a two-time point IC50 shift approach facilitates kinact assay design. Xenobiotica39:99–112.

Picard N, Ratanasavanh D, Prémaud A, Le Meur Y, and Marquet P (2005) Identification of theUDP-glucuronosyltransferase isoforms involved in mycophenolic acid phase II metabolism.Drug Metab Dispos 33:139–146.

Pillai VC, Strom SC, Caritis SN, and Venkataramanan R (2013) A sensitive and specific CYPcocktail assay for the simultaneous assessment of human cytochrome P450 activities in primarycultures of human hepatocytes using LC-MS/MS. J Pharm Biomed Anal 74:126–132.

Ren S, Zeng J, Mei Y, Zhang JZ, Yan SF, Fei J, and Chen L (2013) Discovery and character-ization of novel, potent, and selective cytochrome P450 2J2 inhibitors. Drug Metab Dispos 41:60–71.

Seo KA, Kim HJ, Jeong ES, Abdalla N, Choi CS, Kim DH, and Shin JG (2014) In vitro assay ofsix UDP-glucuronosyltransferase isoforms in human liver microsomes, using cocktails of probesubstrates and liquid chromatography-tandem mass spectrometry. Drug Metab Dispos 42:1803–1810.

Singh SS (2006) Preclinical pharmacokinetics: an approach towards safer and efficacious drugs.Curr Drug Metab 7:165–182.

Smith D, Sadagopan N, Zientek M, Reddy A, and Cohen L (2007) Analytical approaches todetermine cytochrome P450 inhibitory potential of new chemical entities in drug discovery.J Chromatogr B Analyt Technol Biomed Life Sci 850:455–463.

Song M, Hong M, Choi HG, Jahng Y, Lee SH, and Lee S (2013) Effects of Mollugin on HepaticCytochrome P450 in Male ICR Mice as Determined by Liquid Chromatography/Tandem MassSpectrometry. Mass Spectrom Lett 3:104–107.

Spaggiari D, Geiser L, Daali Y, and Rudaz S (2014) A cocktail approach for assessing the in vitroactivity of human cytochrome P450s: an overview of current methodologies. J Pharm BiomedAnal 101:221–237.

Stingl JC, Bartels H, Viviani R, Lehmann ML, and Brockmöller J (2014) Relevance of UDP-glucuronosyltransferase polymorphisms for drug dosing: A quantitative systematic review.Pharmacol Ther 141:92–116.

Suzuki H, Kneller MB, Haining RL, Trager WF, and Rettie AE (2002) (+)-N-3-Benzyl-nirvanoland (-)-N-3-benzyl-phenobarbital: new potent and selective in vitro inhibitors of CYP2C19.Drug Metab Dispos 30:235–239.

Testino SA, Jr and Patonay G (2003) High-throughput inhibition screening of major humancytochrome P450 enzymes using an in vitro cocktail and liquid chromatography-tandem massspectrometry. J Pharm Biomed Anal 30:1459–1467.

Tolonen A, Petsalo A, Turpeinen M, Uusitalo J, and Pelkonen O (2007) In vitro interactioncocktail assay for nine major cytochrome P450 enzymes with 13 probe reactions and a singleLC/MSMS run: analytical validation and testing with monoclonal anti-CYP antibodies. J MassSpectrom 42:960–966.

Tucker GT, Houston JB, and Huang SM (2001) Optimizing drug development: strategies toassess drug metabolism/transporter interaction potential—towards a consensus. Br J ClinPharmacol 52:107–117.

Simultaneous Evaluation of P450 and UGT Enzyme Activities 1145

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from

Page 10: Simultaneous Screening of Activities of Five Cytochrome ...dmd.aspetjournals.org/content/dmd/43/7/1137.full.pdf · separation was performed on a Phenomenex (Torrance, CA) Kinetex

Uchaipichat V, Mackenzie PI, Elliot DJ, and Miners JO (2006) Selectivity of substrate (tri-fluoperazine) and inhibitor (amitriptyline, androsterone, canrenoic acid, hecogenin, phenylbutazone,quinidine, quinine, and sulfinpyrazone) “probes” for human udp-glucuronosyltransferases. DrugMetab Dispos 34:449–456.

Uchaipichat V, Mackenzie PI, Guo XH, Gardner-Stephen D, Galetin A, Houston JB, and Miners JO(2004) Human udp-glucuronosyltransferases: isoform selectivity and kinetics of 4-methylumbelliferoneand 1-naphthol glucuronidation, effects of organic solvents, and inhibition by diclofenac and probenecid.Drug Metab Dispos 32:413–423.

Vietri M, Pietrabissa A, Mosca F, and Pacifici GM (2000) Mycophenolic acid glucuronidationand its inhibition by non-steroidal anti-inflammatory drugs in human liver and kidney. Eur JClin Pharmacol 56:659–664.

von Moltke LL, Greenblatt DJ, Duan SX, Schmider J, Kudchadker L, Fogelman SM, Harmatz JS,and Shader RI (1996) Phenacetin O-deethylation by human liver microsomes in vitro: in-hibition by chemical probes, SSRI antidepressants, nefazodone and venlafaxine. Psycho-pharmacology (Berl) 128:398–407.

Walsky RL and Obach RS (2003) Verification of the selectivity of (+)N-3-benzylnirvanol asa CYP2C19 inhibitor. Drug Metab Dispos 31:343.

Walsky RL and Obach RS (2004) Validated assays for human cytochrome P450 activities. DrugMetab Dispos 32:647–660.

Williams JA, Hyland R, Jones BC, Smith DA, Hurst S, Goosen TC, Peterkin V, Koup JR,and Ball SE (2004) Drug-drug interactions for UDP-glucuronosyltransferase substrates:

a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. DrugMetab Dispos 32:1201–1208.

Yan Z and Caldwell GW (2003) Metabolic assessment in liver microsomes by co-activating cyto-chrome P450s and UDP-glycosyltransferases. Eur J Drug Metab Pharmacokinet 28:223–232.

Zambon S, Fontana S, and Kajbaf M (2010) Evaluation of cytochrome P450 inhibition assaysusing human liver microsomes by a cassette analysis /LC-MS/MS. Drug Metab Lett 4:120–128.

Zhang D, Chando TJ, Everett DW, Patten CJ, Dehal SS, and Humphreys WG (2005) In vitroinhibition of UDP glucuronosyltransferases by atazanavir and other HIV protease inhibitorsand the relationship of this property to in vivo bilirubin glucuronidation. Drug Metab Dispos33:1729–1739.

Zhang L, Wei MJ, Zhao CY, and Qi HM (2008) Determination of the inhibitory potential of 6fluoroquinolones on CYP1A2 and CYP2C9 in human liver microsomes. Acta Pharmacol Sin29:1507–1514.

Address correspondence to: Kwang-Hyeon Liu, College of Pharmacy andResearch Institute of Pharmaceutical Sciences, Kyungpook National University,80 Daehak-ro, Buk-gu, Daegu 702-701, Korea. E-mail: [email protected]

1146 Lee et al.

at ASPE

T Journals on N

ovember 20, 2020

dmd.aspetjournals.org

Dow

nloaded from