<<

1521-0081/68/1/168–241$25.00 http://dx.doi.org/10.1124/pr.115.011411 PHARMACOLOGICAL REVIEWS Pharmacol Rev 68:168–241, January 2016 Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics

ASSOCIATE EDITOR: MARKKU KOULU Role of 2C8 in and Interactions

Janne T. Backman, Anne M. Filppula, Mikko Niemi, and Pertti J. Neuvonen Department of Clinical Pharmacology, University of Helsinki (J.T.B., A.M.F., M.N., P.J.N.), and Helsinki University Hospital, Helsinki, Finland (J.T.B., M.N., P.J.N.)

Abstract ...... 169 I. Introduction ...... 169 II. Basic Characteristics of Cytochrome P450 2C8 ...... 170 A. Genomic Organization and Transcriptional Regulation ...... 170 B. Protein Structure ...... 171 C. Expression ...... 172 III. Substrates of Cytochrome P450 2C8...... 173 A. ...... 173 1. Anticancer Agents...... 173 Downloaded from 2. Antidiabetic Agents...... 183 3. Antimalarial Agents...... 183 4. Lipid-lowering Drugs...... 184 5. Other Drugs...... 184 6. Glucuronide Metabolites...... 186

B. Endogenous and Natural Compounds...... 187 by guest on March 4, 2019 IV. Pharmacogenetics...... 187 A. Population Genetics ...... 187 B. Functional Studies ...... 191 C. Effects on in Humans ...... 192 V. In Vitro Inhibition and Induction of Cytochrome P450 2C8 ...... 193 A. Reversible Inhibition ...... 193 1. Drugs That Act as Inhibitors of Cytochrome P450 2C8...... 193 2. Natural Compounds...... 210 B. Metabolism-dependent Inhibition ...... 210 C. Induction ...... 210 VI. Clinical Drug Interactions Mediated via Cytochrome P450 2C8 ...... 212 A. General Aspects...... 212 B. as Prototypical Inhibitor ...... 214 1. In Vitro Versus In Vivo...... 214 2. Gemfibrozil Dose Versus CYP2C8 Inhibition...... 216 3. Onset and Duration of CYP2C8 Inhibition by Gemfibrozil...... 216 4. Quantification of CYP2C8-Mediated Drug Interactions in Humans...... 216 C. Inhibition-Mediated Drug Interactions and Their Clinical Significance ...... 217 1. ...... 217 2. Other Oral Antidiabetic Drugs...... 218 3. Amodiaquine...... 219 4. ...... 219 5. Anticancer Drugs...... 220

This work was supported by grants from the Academy of Finland [Grant decision 278123, 2014], the Helsinki University Central Hospital Research Fund, and the Sigrid Juselius Foundation (Helsinki, Finland). Address correspondence to: Prof. Janne T. Backman, Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, P.O. Box 705, FI-00029 HUS, Finland. E-mail: [email protected] dx.doi.org/10.1124/pr.115.011411.

168 Role of CYP2C8 in Drug Metabolism and Interactions 169

6. Antiviral Drugs...... 221 7. Antiasthmatic Drugs...... 221 8. Other or Inhibitor Drugs...... 221 D. Induction-Mediated Drug Interactions ...... 222 1. Rifampin ()...... 222 VII. Points to Consider When Investigating Cytochrome P450 2C8-Mediated Drug Metabolism and Interactions ...... 222 A. InVitro...... 222 1. General Aspects...... 222 2. Assessment of CYP2C8 Activity In Vitro...... 223 3. In Vitro Methods to Estimate the Contribution of CYP2C8 in the Metabolism of a Drug...... 224 B. In Vivo ...... 224 1. General Aspects...... 224 2. In Vivo Cytochrome P450 2C8 Probe Substrates...... 226 3. In Vivo Cytochrome P450 2C8 Probe Inhibitors...... 226 VIII. Conclusions and Future Prospects...... 227 Acknowledgments...... 228 References ...... 228

Abstract——During the last 10-15 years, cytochrome glucuronide metabolites interact with CYP2C8 as P450 (CYP) 2C8 has emerged as an important drug- substrates or inhibitors, suggesting that an interplay metabolizing . CYP2C8 is highly expressed in between CYP2C8 and glucuronides is common. Lack of human and is known to metabolize more than 100 fully selective and safe probe substrates, inhibitors, drugs. CYP2C8 substrate drugs include amodiaquine, and inducers challenges execution and interpretation , dasabuvir, , imatinib, of drug-drug interaction studies in humans. Apart from loperamide, , , pioglitazone, drug-drug interactions, some CYP2C8 genetic variants repaglinide, and , and the number is are associated with altered CYP2C8 activity and exhibit increasing. Similarly, many drugs have been identified significant interethnic frequency differences. Herein, as CYP2C8 inhibitors or inducers. In vivo, already a small we review the current knowledge on substrates, inhibitors, dose of gemfibrozil, i.e., 10% of its therapeutic dose, is a inducers, and pharmacogenetics of CYP2C8, as well as strong, irreversible inhibitor of CYP2C8. Interestingly, its role in clinically relevant drug interactions. In recent findings indicate that the acyl-b-glucuronides addition, implications for selection of CYP2C8 marker of gemfibrozil and cause metabolism- and perpetrator drugs to investigate CYP2C8-mediated dependent inactivation of CYP2C8, leading to a strong drug metabolism and interactions in preclinical and potential for drug interactions. Also several other clinical studies are discussed.

I. Introduction (HMG-CoA) reductase inhibitor cerivastatin, a CYP2C8 substrate, resulting in rhabdomyolysis cases and fatalities Cytochrome P450 (CYP) 2C8 accounts for approxi- mately 6–7% of the total hepatic CYP content (Rowland brought attention to the importance of CYP2C8 in drug Yeoetal.,2004;Inoueetal.,2006; Rostami-Hodjegan and metabolism (Backman et al., 2002; Staffa et al., 2002; Tucker, 2007; Achour et al., 2014). The importance of Wang et al., 2002; Chang et al., 2004; Huang et al., 2008). CYP2C8 causing variation in drug response via drug-drug The event was the onset of a broadening scientific in- interactions and pharmacogenetic polymorphisms has terestinCYP2C8,promptlyconvincingdrugregulatory been recognized only for the last 10–15 years. In the authorities to acknowledge CYP2C8 as one of the major beginning of the millennium, the pharmacokinetic drug- drug-metabolizing CYP . drug interaction between the fibric acid derivative gemfi- Drugs that were introduced into clinical use before brozil and the 3-hydroxy-3-methylglutaryl-coenzyme A the role of CYP2C8 was recognized may have deficient

ABBREVIATIONS: AUC, area under the plasma concentration-time curve; C/EBPa, CCAAT/enhancer-binding protein a; CAR, constitutive androstane receptor; CITCO, [6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime;

CLint, intrinsic clearance; Cmax, peak concentration; CYP, cytochrome P450; EMA, European Medicines Agency; FDA, Food and Drug Administration; GR, receptor; HLM, human liver microsomes; HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A; HNF, hepatic nuclear factor; I, inhibitor concentration; Ki, reversible inhibition constant; KI, inhibitor concentration supporting half of the maximal rate of enzyme inactivation; kinact, maximal rate of inactivation; Km, Michaelis-Menten constant; MRL-C, 2-[[5,7-dipropyl-3- (trifluoromethyl)-1,2-benzisoxazol-6-yl]oxy]-2-methylpropanoic acid; mRNA, messenger ribonucleic acid; OAT, organic anion transporter; OATP, organic anion-transporting polypeptide; PPAR, peroxisome proliferator activated receptor; PXR, pregnane X receptor; ROR, retinoic acid-related orphan receptors; SIFT, sorting intolerant from tolerant; SNV, single nucleotide variation; t1/2, elimination half-life; tmax,time to peak concentration; UGT, uridine-59-diphosphoglucuronosyltransferase; VDR, D receptor. 170 Backman et al. or incorrect product information regarding their in- roles of CYP2C8 have been thoroughly examined and teraction potential (Neuvonen, 2012). One example is discussed in several previous reviews (Kirchheiner et al., the leukotriene montelukast. 2005; Totah and Rettie, 2005; Garcia-Martin et al., 2006; Early preclinical studies concluded that CYP2C9 and Gil and Gil Berglund, 2007; Agundez et al., 2009; Chen CYP3A4 are the most important enzymes involved in its and Goldstein, 2009; Daily and Aquilante, 2009; Lai et al., metabolism, whereas the role of CYP2C8 was not 2009; Aquilante et al., 2013b; Fleming, 2014; Xiaoping evaluated (Chiba et al., 1997). In interaction studies et al., 2013). Thus, the reader will be directed to these performed more than a decade later, the strong CYP2C8 earlier works for some previously known aspects related inhibitor gemfibrozil increased the plasma exposure to CYP2C8. Herein, our intention is to review and update to montelukast almost fivefold, whereas the strong the current knowledge on substrates, inhibitors, in- CYP3A4 inhibitor itraconazole had no significant effect ducers, and pharmacogenetics of CYP2C8, as well as its on its (Karonen et al., 2010, 2012). role in clinically relevant drug interactions. In addition, The clinical findings were corroborated by in vitro data, implications for selection of CYP2C8 marker and perpe- showing that CYP2C8 is the main enzyme involved in trator drugs to investigate CYP2C8-mediated drug me- the oxidative metabolism of montelukast (Filppula tabolism and interactions in preclinical and clinical et al., 2011; VandenBrink et al., 2011). studies are discussed. The large, trifurcated cavity of CYP2C8 is able to accommodate substrates of different shapes and II. Basic Characteristics of Cytochrome P450 2C8 sizes (Schoch et al., 2008). Today, CYP2C8 is known to participate in the metabolism of more than 100 drugs, A. Genomic Organization and Transcriptional Regulation including amodiaquine, cerivastatin, dasabuvir, enzalu- The CYP2C8 enzyme is encoded by the CYP2C8 gene, tamide, imatinib, loperamide, montelukast, paclitaxel, which is located on the 10q24 in the pioglitazone, repaglinide, and rosiglitazone. The num- gene cluster centromere-2C18-2C19-2C9-2C8-telomere ber of drugs that are identified as CYP2C8 substrates in close proximity of the CYP2C9 gene (Fig. 1; Gray or inhibitors, as well as CYP2C8-mediated drug-drug et al., 1995; Klose et al., 1999). CYP2C8 is the smallest interactions is continuously increasing. The strong of the human CYP2C genes; it spans a 31-kb region and CYP2C8 inhibition by gemfibrozil observed in vivo is contains 9 exons (Klose et al., 1999; Lai et al., 2009). It due to its acyl-b-glucuronide metabolite, which is a shares 74% with CYP2C9 (Daily potent mechanism-based inhibitor of CYP2C8 (Ogilvie and Aquilante, 2009). et al., 2006). Also other glucuronide metabolites of drugs The transcriptional regulation of CYP2C8 is mediated were recently found to interact with CYP2C8 either as via several transcriptional factors and distinct nuclear substrates or inhibitors. For instance, the acyl-b-D- receptors that can activate the respective responsive glucuronide metabolite of clopidogrel is a metabolism- elements within the 59-flanking promoter region of the dependent inhibitor of CYP2C8, causing more than a gene (Ferguson et al., 2005; Johnson and Stout, 2005; fivefold increase in the plasma exposure to repaglinide Kojima et al., 2007; Chen and Goldstein, 2009). Such in healthy subjects (Tornio et al., 2014). In another factors/receptors include the constitutive androstane re- recent in vitro study, CYP2C8 metabolized the glucu- ceptor (CAR), pregnane X receptor (PXR), vitamin D ronide metabolite of to its pharmacolog- receptor (VDR), glucocorticoid receptor (GR), hepatic ically active 3-hydroxydesloratadine metabolite (Kazmi nuclear factor-4a (HNF4a), HNF3g, CCAAT/enhancer- et al., 2015). These and other data suggest that an binding protein a (C/EBPa), and retinoic acid-related interplay between CYP2C8 and glucuronide metabo- orphan receptors (RORs) (Fig. 1; Ferguson et al., 2005; lites may be more a rule than an exception. Chen and Goldstein, 2009; Rana et al., 2010; Aquilante There are several common nonsynonymous varia- et al., 2013b). Although HNF4a,HNF3g, C/EBPa,and tions in the CYP2C8 gene (Daily and Aquilante, 2009; RORs seem to mainly regulate the constitutive expression Aquilante et al., 2013b). For example, the CYP2C8*3 of CYP2C genes in liver, the other receptors are more allele has been associated with decreased metabolism important to the xenobiotic-mediated induction of CYP2C8 of several substrates, e.g., paclitaxel, in vitro (Dai et al., expression, as described in more detail in section V.C. 2001). In contrast, clinical data indicate that the CYP2C8*3 After activation by endo- or xenobiotics, CAR, PXR, allele is often associated with increased metabolism of and VDR form heterodimers with the retinoid X CYP2C8 substrates, such as repaglinide (Niemi et al., receptor, whereas GR forms homodimers (Chen and 2003c). Thus, although complete lack of function variants Goldstein, 2009). These dimers are thereafter recog- in CYP2C8 arerare,thepossiblesubstratedependency nized by specific response elements within the CYP2C8 of the functional consequences of the common CYP2C8 promoter. By using in vitro gel shift assays, responsive variants and their potential clinical significance have elements/motifs within the CYP2C8 promoter regions raised a lot of interest toward CYP2C8 pharmacogenetics. have been identified for CAR, PXR, and GR (Gerbal- The structural properties, regulation of expression, Chaloin et al., 2002; Ferguson et al., 2005; Chen and pharmacogenetics, substrates, inhibitors, and physiologic Goldstein, 2009). Role of CYP2C8 in Drug Metabolism and Interactions 171

Fig. 1. The CYP2C8 gene is located to the CYP2C gene cluster on . C/EBPa, CCAAT/enhancer-binding protein a; CAR, constitutive androstane receptor; GR, glucocorticoid receptor; HNF, hepatic nuclear factor; PXR, pregnane X receptor; ROR, retinoic acid-related orphan receptor; VDR, vitamin D receptor.

After activation, the orphan nuclear receptor HNF4a 2004), which is similar to that of CYP3A4 (1,386 Å3) binds as a homodimer to a DR1 type element and also but larger than those of CYP1A2 (375 Å3), CYP2A6 to the Hep-G2 specific P450 factor-1 motif (Venepally (260 Å3), CYP2C9 (;470 Å3), CYP2D6 (;540 Å3), and et al., 1992), whereas HNF3g binds to DNA as a CYP2E1 (190 Å3) (Williams et al., 2003; Yano et al., monomer (Bort et al., 2004). At least two Hep-G2 2004; Rowland et al., 2006; Sansen et al., 2007; specific P450 factor-1 motifs and several putative Porubsky et al., 2008). Although CYP3A4 has a uni- HNF3g binding sites have been identified within the formly distributed active site cavity, that of CYP2C8 is promoter of CYP2C8 (Bort et al., 2004; Ferguson et al., trifurcated, resembling a T or Y shape (Schoch et al., 2005; Chen and Goldstein, 2009). RORs are constitu- 2008). The bottom branch of the cavity provides access tively active orphan nuclear receptors, which have to the heme, and the two other terminate in solvent and natural ligands, such as all-trans-retinoic acid that substrate access channels that exit the active site cavity can influence their activity. Also RORs seem to be on either side of the helix B-C loop. involved in the constitutive regulation of CYP2C8, In the X-ray crystallography study, the N-terminal and at least two ROR responsive elements have been anchor domains of both CYP2C8 molecules were lo- identified in the gene promoter (Chen et al., 2009). cated on the same side of the dimer, indicating an Transcriptional regulation of CYP2C8 has been re- orientation compatible with membrane binding (Schoch viewed thoroughly by Chen and Goldstein (2009). et al., 2004). The proximal surfaces of each protein were roughly parallel, suggesting that they are accessible for B. Protein Structure interaction with the membrane-bound CYP oxidoreduc- The crystal structure of CYP2C8 was resolved in 2004 tase. Another study demonstrated that the dimeric (Schochetal.,2004).AsingleCYP2C8crystaldif- structure observed in the crystal structure of CYP2C8 fracted to 2.7 Å and had the molecular weight approx- may also be present in membrane-bound native CYP2C8 imated to 54 kDa. Interestingly, CYP2C8 crystallized as (Hu et al., 2010). The signal anchor/linker regions of a symmetric dimer formed by interactions between the native CYP2C8 formed a second dimerization interface, helix F to G regions of the two monomers. Two palmitic and it was suggested that this interaction is required acid molecules were bound in the dimer interface, for the formation of the dimer of the native protein. stabilizing the dimer. Thus, the two fatty acids may Although direct evidence for a functional significance form a peripheral , which may affect the of the dimerization is lacking, such interactions structural dynamics of the active site and influence have been shown to affect activities of other CYPs and reactions catalyzed by CYP2C8. The active site volume membrane proteins in the endoplasmic reticulum (Hu of CYP2C8 was estimated to 1,438 Å3 (Schoch et al., et al., 2010). 172 Backman et al.

According to the X-ray crystallographic data, the where Ser-100 and Ser-103 reside (Fig. 2; Baer et al., active site cavity of CYP2C8 is capable of binding 2009; Tornio et al., 2014). structurally diverse substrates without major changes Although the large active site of CYP2C8 and in its tertiary structure (Schoch et al., 2008). Ligands of diversity of its substrates (section III) may complicate CYP2C8 may bind to the active site differently, filling the use of a general pharmacophore model, analysis of the cavity either partially or completely or occupying it eight CYP2C8 substrates showed that the majority of with two molecules simultaneously. For instance, mon- these compounds contained a terminal anionic or polar telukast, a large anionic molecule with a tripartite group ;13 Å from the oxidation site, and one or two structure, complemented the size and shape of the secondary polar moieties ;4.5 Å and ;8.5 Å from the whole active-site cavity. The linearly shaped troglita- oxidation site (Melet et al., 2004). The pharmacophore zone molecule occupied the upper portion of the cavity, model and previously reported homology models for leaving a significant part of the cavity empty, whereas CYP2C8 have been comprehensively reviewed by Lai two molecules of 9-cis-retinoic acid were simultaneously and colleagues (2009). present in the substrate-binding cavity of CYP2C8 (Schoch et al., 2008). The interactions between CYP2C8 C. Expression and its substrates were predominantly hydrophobic. According to meta-analyses, the mean hepatic CYP2C8 In addition, the distal region of the CYP2C8 active concentration approximates to 22–24 pmol/mg and site cavity contains a number of polar amino acid side 14 pmol/mg in adult Caucasian and Japanese , chains and exposed peptide backbone hydrogen bond respectively (Rowland Yeo et al., 2004; Inoue et al., donors and acceptors (Schoch et al., 2008). Accordingly, 2006; Rostami-Hodjegan and Tucker, 2007; Achour for example, the residues Ser-100, Ser-103, Asn-204, et al., 2014). The interindividual variability of CYP2C8 Asn-217, and Arg-241 form hydrogen bonds involved in protein expression in liver is high, with coefficients of the binding of the CYP2C8 substrates retinoic acid, variation of 68–95%.Theproteinexpressionlevel troglitazone and montelukast. Of note, a pronounced of CYP2C8 seems to be highly correlated with both side chain movement was observed in crystallized its enzyme activity and messenger ribonucleic acid complexes with troglitazone and retinoic acid, where (mRNA) expression level (Ohtsuki et al., 2012). Arg-241 was reoriented to the inside of the cavity, where Hepatic CYP2C8 mRNA and protein are expressed it could provide a strong, charge-stabilized hydrogen early in the prenatal development and reaches adult bond with the substrate. Interestingly, according to levels already in early childhood (Treluyer et al., 1997; computational docking simulations, the glucuronide Blanco et al., 2000; Naraharisetti et al., 2010; Cizkova moieties of gemfibrozil 1-O-b glucuronide and clopidog- et al., 2014; Johansson et al., 2014). CYP2C8 seems to be rel acyl 1-b-D-glucuronide are oriented toward the same the predominant CYP2C isoform in fetal livers (Hak- hydrophilic area in the active site close to helix B9, kola et al., 1994; Nishimura et al., 2003; Johansson

Fig. 2. Stereoimage of three independent docking simulations of the interaction between clopidogrel acyl 1-b-D-glucuronide and the active site of CYP2C8. Clopidogrel acyl-b-D-glucuronide is rendered with gray sticks depicting carbon atoms. The distance between clopidogrel acyl-b-D-glucuronide thiophene ring carbon and heme iron is indicated by a green line. Other atoms of the clopidogrel acyl-b-D-glucuronide molecule are colored red for oxygen, blue for , yellow for sulfur, and green for chlorine. Role of CYP2C8 in Drug Metabolism and Interactions 173 et al., 2014). In a recent study, CYP2C8 mRNA was III. Substrates of Cytochrome P450 2C8 expressed in all fetal tissues studied (adrenal, kidney, CYP2C8 participates in the metabolism of numerous liver, and lung tissue), whereas CYP2C9 mRNA was drugs and some endogenous and natural compounds. It restricted to the liver (Johansson et al., 2014). Another catalyzes a variety of oxidative reactions, in particular study detected CYP2C8, CYP2C9, and CYP2C19 pro- hydroxylations, N-demethylations, and N-deethyla- tein in fetal liver, intestine, and kidney (Cizkova et al., tions (Tables 1–4). Because of its large, sinuous active 2014). One explanation for the role of CYP2C8 in the site, CYP2C8 can accommodate substrates of different fetus during early pregnancy may be a need for CYP2C8 sizes and structures. The molecular weight of drugs to metabolize endogenous compounds such as retinoic significantly metabolized by CYP2C8 (.20%; Table 1) acids and hence protect the fetus from retinoic acid- ranges from 206 to 854 g/mol, with a median of 451 g/mol induced embryotoxicity (Johansson et al., 2014). (Fig. 3). In adults, CYP2C8 mRNA has been detected in Most, if not all, of the drugs significantly metabolized numerous extrahepatic tissues, including the adrenal by CYP2C8 are also substrates of other CYP enzymes gland, arteries, brain, duodenum, heart, kidney, lung, (Table 1), with about 75% being metabolized by CYP3A4 mammary gland, ovary, prostate, retina; testis, and and ;30% by CYP2C9. However, the metabolic prod- uterus, but not in placenta (Zeldin et al., 1995; Mace ucts generated by CYP2C8 and CYP3A4 are often et al., 1998; McFayden et al., 1998; Klose et al., 1999; different. For instance, CYP2C8 metabolizes paclitaxel Thum and Borlak, 2000; Nishimura et al., 2003; to 6a-hydroxypaclitaxel, whereas CYP3A4 exclusively Delozier et al., 2007; Dutheil et al., 2009; Capozzi et al., generates 39-hydroxypaclitaxel (Rahman et al., 1994), 2014). CYP2C8 protein has been detected in heart, suggesting that compounds that are substrates of both hepatocytes, kidney, salivary ducts, small and large CYP2C8 and CYP3A4 bind differently to their active intestine, adrenal cortical cells, and tonsils (Läpple sites. et al., 2003; Enayetallah et al., 2004; Delozier et al., 2007; Cizkova et al., 2014). The expression of CYP2C8 and other CYP enzymes has recently been reviewed by A. Drugs Shahabi et al. (2014). CYP2C8 is involved in the metabolism of more than Analysis of liver samples has recently shown that a 100 clinically used drugs (Table 1). Typical substrate nearly full-length form of CYP2C8 (wild type) and an drugs of CYP2C8 include anticancer, antidiabetic, N-terminal truncated splice variant 3 are expressed in antimalarial, and lipid-lowering agents (Fig. 4). In- mitochondria (Bajpai et al., 2014). Although the wild- terestingly, some glucuronide metabolites of drugs in- type protein was detected only at low levels in mito- teract with CYP2C8. chondria (,25%), variant 3 was primarily targeted to 1. Anticancer Agents. The antimicrotubule agent mitochondria and minimally to the endoplasmic re- paclitaxel with a molecular weight of 853.9 g/mol is ticulum. Interestingly, although molecular modeling one of the largest substrates of CYP2C8. In vitro, showed that both the heme binding pocket and the paclitaxel is primarily metabolized by CYP2C8 to its substrate binding cavity were nearly intact in variant main 6a-hydroxy metabolite and by CYP3A4 to 39- 3, it was unable to catalyze paclitaxel 6-hydroxylation phenyl-hydroxypaclitaxel, and the further metabolism in human hepatocellular liver carcinoma cells. How- of these metabolites results in the formation of 6a,39-p- ever, it did metabolize smaller substrates such as dihydroxypaclitaxel (Cresteil et al., 1994, 2002; Harris and dibenzylfluorescein. Further- et al., 1994; Kumar et al., 1994; Rahman et al., 1994). more, the variant generated higher levels of reactive Paclitaxel 6a-hydroxylation is recommended by drug oxygen species and showed a higher level of mitochon- authorities as a marker reaction for CYP2C8 acti- drial respiratory dysfunction than wild type CYP2C8, vity in vitro (EMA, 2012b; http://www.fda.gov/Drugs/ suggesting that the mitochondrially targeted variant 3 DevelopmentApprovalProcess/DevelopmentResources/ may contribute to oxidative stress in tissues (Bajpai DrugInteractionsLabeling/ucm093664.htm), and it has et al., 2014). been widely used in preclinical studies. In a mass balance In living organisms, CYP enzymes undergo natural study in patients, metabolites accounted for about 40% of degradation that can be described as a first-order process the total systemic drug exposure, and the excreted 6a- (Yang et al., 2008). Therefore, the expression level of hydroxypaclitaxel corresponded to almost one-third of the enzyme is determined by the rate of enzyme the administered dose (Walle et al., 1995), suggesting synthesis and the degradation half-life of the enzyme. that ;30–40% of a paclitaxel dose is converted by The extent and dose and time dependency of enzyme CYP2C8 to 6a-hydroxy paclitaxel. induction and inactivation are thus also dependent on Cabazitaxel, a taxane approved in 2010, is also the degradation half-life. Based on clinical studies metabolized by CYP2C8 to a small extent in vitro with the CYP2C8 inactivator gemfibrozil, the degra- (FDA, 2010a), whereas there is conflicting data re- dation (turnover) half-life of CYP2C8 is approximately garding the role of CYP2C8 in the metabolism of 22 hours (Backman et al., 2009). docetaxel. One in vitro study demonstrated that 174 Backman et al.

TABLE 1 Drugs that are metabolized by CYP2C8, grouped by the importance of CYP2C8 in their elimination (Major, intermediate, minor)

Other CYP Enzymes Substrate Therapeutic Use and/or Metabolic Pathway(s) Involved in Overall References Drug Class Catalyzed by CYP2C8 Metabolisma Major (.70%) Amodiaquine Antimalarial N-deethylation (CYP1A1, CYP1B1) Li et al., 2002 Cerivastatin (acid, Antihyperlipidemic, HMG- 6-hydroxylation (M-23), CYP3A4 Backman et al., 2002 parent) CoA reductase inhibitor demethylation (M-1) Daprodustat Antianemic, prolyl CYP3A4 Johnson et al., 2014 (GSK1278863) hydroxylase inhibitor Dasabuvir (ABT-333) Antiviral, NSB5 inhibitor M1 formation CYP3A4, CYP2D6 FDA, 2014g Enzalutamide Anticancer, Hydroxylation (M6), CYP3A4/5 FDA, 2012k; Gibbons et al., N-demethylation (M2) 2015 Montelukast Antiasthmatic, LTRA 36-hydroxylation (M6), CYP3A4, CYP2C9 Karonen et al., 2010; 25-hydroxylation Filppula et al., 2011 (M3), M4 formation Pioglitazone Antidiabetic, PPAR-g Hydroxylation CYP3A4/5, CYP1A1 Jaakkola et al., 2006c; FDA, 2013a Repaglinide Antidiabetic, meglitinide M2 and M4 formation CYP3A4 Bidstrup et al., 2003; analog Kajosaari et al., 2005a

Intermediate (20–70%) 9cUAB30 Anticancer, retinoid M1-M5 formation CYP2C9, CYP2C19, Gorman et al., 2007 (CYP1A2, CYP2B6) Acotiamide (Z-338) Antidyspeptic, Deisopropylation (M-4) CYP1A1, CYP3A4 Furuta et al., 2004; PMDA, 2014 inhibitor Alitretinoin (9-cis- Antipsoriatic, retinoid 4-hydroxylation CYP2C9, CYP3A4, Marill et al., 2002 retinoic acid) CYP26A1 Amiodarone Antiarrhythmic N-deethylation CYP3A4, (CYP1A2, Ohyama et al., 2000 CYP2C19, CYP2D6) Chloroquine Antimalarial N-deethylation CYP3A4/5, (CYP2D6) Kim et al., 2003; Projean et al., 2003a (2)(+)- Gastroprokinetic, 5-HT4 N-dealkylation, 4- CYP3A4, CYP2B6 Desta et al., 2000, 2001 receptor agonist hydroxylation, 2- hydroyxlation Compound A ERA Hydroxylation Ma et al., 2004 Dabrafenib Anticancer, PKI Hydroxylation CYP3A4/5, (CYP2C9, Lawrence et al., 2014; CYP2C19) Suttle et al., 2015 Fenretinide Anticancer, retinoid 49-hydroxylation, CYP3A4/5 Illingworth et al., 2011 49-oxidation R/S- , SSRI N-demethylation CYP2C9, CYP2D6 Wang et al., 2014b R-Ibuprofen Anti-inflammatory, NSAID 2-hydroxylation, CYP2C9 Hamman et al., 1997; 3-hydroxylation Tornio et al., 2007; Chang et al., 2008 Imatinib Anticancer, PKI N-demethylation CYP3A4/5 Nebot et al., 2010; Filppula et al., 2013a,b Irosustat Anticancer, STS inhibitor M9 and M13 formation CYP2C9, CYP3A4/5, Ventura et al., 2011 (CYP2E1) Isotretinoin (13-cis- Antiacne, retinoid 4-hydroxylation CYP3A4 Marill et al., 2002 retinoic acid) Loperamide Antidiarrheal, N-demethylation CYP3A4, CYP2B6, CYP2D6 Kim et al., 2004; Niemi et al., 2006 N-demethylation CYP1A2, CYP2D6, CYP3A4 Korprasertthaworn et al., 2015 Olodaterol Antiasthmatic, LABA O-demethylation CYP2C9, (CYP3A4) FDA, 2014f Paclitaxel (taxol) Anticancer, taxane 6a-hydroxylation CYP3A4 Cresteil et al., 1994; Rahman et al., 1994; Walle et al., 1995 Paritaprevir (ABT-450) Antiviral, NS3-4A inhibitor CYP3A4 FDA, 2014g Propanoic acid Drug metabolite Hydroxylation (M10 and CYP1A1 Klieber et al., 2014 dronedarone M11) R483 Antidiabetic, PPAR-g M1 and M4 formation CYP2C19, CYP3A4, Bogman et al., 2010 agonist (CYP2C9) Rosiglitazone Antidiabetic, PPAR-g p-hydroxylation, CYP2C9 Baldwin et al., 1999 agonist N-demethylation acid Antihyperlipidemic, HMG- Oxidation (M1-M3) CYP3A4/5 Prueksaritanont et al., 2003 CoA reductase inhibitor Tazarotenic acid Antipsoriatic, drug Sulfoxidation Attar et al., 2003 metabolite (active) Tozasertib (MK 0457, Anticancer, PKI N-demethylation CYP3A4 Ballard et al., 2007 VX6, VX 680) Treprostinil Antihypertensive CYP2C9 FDA, 2009b Troglitazone Antidiabetic, PPAR-g Quinone metabolite CYP3A4 Yamazaki et al., 1999b agonist formation (continued) Role of CYP2C8 in Drug Metabolism and Interactions 175

TABLE 1—Continued

Other CYP Enzymes Substrate Therapeutic Use and/or Metabolic Pathway(s) Involved in Overall References Drug Class Catalyzed by CYP2C8 Metabolisma R/S-Verapamil Antihypertensive, CCB N-dealkylation, N- CYP3A4/5, (CYP2E1) Busse et al., 1995; Tracy demethylation, O- et al., 1999 demethylation Vidupiprant (AMG 853) Antiasthmatic, PGD2 t-butyl hydroxylation CYP2J2, CYP3A Foti et al., 2012 receptor antagonist (M2), cyclopropyl hydroxylation (M3) , GABA receptor N-demethylation, N- CYP3A4, CYP2C9 Becquemont et al., 1999; agonist oxidation Tornio et al., 2006 Unknown or Minor (;,20%) 5-MeO-DIPT (Foxy) Hallucinogenic N-deisopropylation CYP2D6, CYP1A2, Narimatsu et al., 2006 CYP3A4, CYP2C19 7-Epi-10-deacetyl- Paclitaxel derivative Hydroxylation CYP3A4 Zhang et al., 2009a paclitaxel 7-Epi-cephalomannine Paclitaxel derivative M-2 formation CYP3A4 Zhang et al., 2009a 7-Epi-paclitaxel Paclitaxel epimer M-2 formation CYP3A4 Zhang et al., 2009b 10-Aceyldocetaxel Docetaxel derivative 6-hydroxylation CYP3A4 Cresteil et al., 2002 10-Deacetylpaclitaxel Paclitaxel derivative 6-hydroxylation CYP3A4 Cresteil et al., 2002 17a-Ethinylestradiol Contraceptive, hormone 2-hydroxylation Wang et al., 2004 derivative 17b- (estradiol) Hormonal replacement 2-hydroxylation, 4- CYP1A1, CYP1B1 Spink et al., 1992 therapy hydroxylation Aminophenazone N-demethylation CYP2C19, CYP2B6, Niwa et al., 1999, 2000 (aminopyrine) CYP2D6 Antidepressant, TCA N-demethylation CYP3A4/5, CYP2C19 Venkatakrishnan et al., 2001 Anticancer, Hydroxylation CYP3A4, CYP3A5 Kamdem et al., 2010 Inhibitor Apixaban , factor Xa O-demethylation CYP3A4, CYP1A2, FDA, 2012d inhibitor CYP2C9, CYP2C19, CYP2J2 Apremilast Antipsoriatic, PDE4 M5 formation CYP3A4, CYP2A6, FDA, 2014e inhibitor (CYP1A2, CYP2C9, CYP2E1) Artelinic acid Antimalarial 3-hydroxylation CYP3A4/5 Grace et al., 1999 (acid, Antihyperlipidemic, HMG- p-hydroxylation CYP3A4/5 Jacobsen et al., 2000b parent) CoA reductase inhibitor Azilsartan Antihypertensive, ARB Decarboxylation (M-I), CYP2C9, CYP2B6 FDA, 2011c O-dealkylation (M-II) Bedaquiline , ATP synthase N-demethylation CYP3A4, CYP2C19 Liu et al., 2014 inhibitor Brinzolamide Antiglaucoma, carbonic CYP3A4, CYP2A6, EMA, 2014 anhydrase inhibitor CYP2B6, CYP2C9 Brivaracetam Antiepileptic Hydroxylation CYP2C9, CYP3A4 Whomsley et al., 2007; Nicolas et al., 2012 Analgesic, opioid N-dealkylation, M1 CYP3A4 Moody et al., 2002; Picard formation et al., 2005; Chang et al., 2006 CYP3A4 Karlsson et al., 2013 BYZX Antidementia, N-deethylation (M3) CYP3A4 Yu et al., 2013a acetylcholinesterase inhibitor BYZX M2 Drug metabolite N-deethylation (M1) CYP3A4 Yu et al., 2013a Cabazitaxel Anticancer, taxane RPR 112698 formation CYP3A4/5 FDA, 2010a Psychostimulant N-demethylation, CYP3A4, CYP1A2, CYP2C9 Kot and Daniel, 2008 C-8-hydroxylation Capravirine Antiviral, NNRTI Sulfoxidation (C23), CYP3A4, CYP2C9, Bu et al., 2006 N-oxidation (C26), CYP2C19 hydroxylation (C19) Antiepileptic 10,11-epoxidation, CYP3A4 Kerr et al., 1994; Pelkonen 3-hydroxylation et al., 2001 Cephalomannine Paclitaxel derivative 4a-hydroxylation Zhang et al., 2009a Antidementia, 5-HT6 Demethylation CYP3A4 Tse et al., 2014 receptor antagonist Cilostazol Antithrombotic, PDE3 OPC-13217 formation CYP3A4/5, CYP1B1, Hiratsuka et al., 2007 inhibitor CYP2C19 Gastroprokinetic, 5-HT4 CYP3A4 Robert et al., 2007 receptor agonist E-Clomiphene Ovulation inducer, SERM Deethylation, CYP3A4/5, CYP2D6 Mürdter et al., 2012 hydroxylation Antipsychotic N-demethylation, oxidation CYP1A2, Linnet and Olesen, 1997 (CYP3A4) (continued) 176 Backman et al.

TABLE 1—Continued

Other CYP Enzymes Substrate Therapeutic Use and/or Metabolic Pathway(s) Involved in Overall References Drug Class Catalyzed by CYP2C8 Metabolisma CPI-613 Anticancer, CYP3A4, (CYP2C9, Lee et al., 2011 antimitochondrial CYP2C19) metabolism agent Antipsychotic N-demethylation CYP1A2, CYP3A4, CYP2C9 Arbus et al., 2007 Cyclophosphamide Anticancer, alkylating 4-hydroxylation CYP2C9, CYP2A6, Chang et al., 1993; Huang agent CYP2B6, CYP3A4 et al., 2000 Cyclosporine Immunosuppressant, CYP3A4 Karlsson et al., 2013 calcineurin inhibitor Dapsone N-hydroxylation CYP2C9 Winter et al., 2000 Anxiolytic, N-demethylation, 3- CYP2C9, CYP3A4, Sai et al., 2000 hydroxylation CYP2C19 Dibenzylfluorescein Fluorescent CYP probe O-debenzylation CYP3A4, CYP2C19, Miller et al., 2000 CYP2C9, CYP3A5, CYP3A7 Anti-inflammatory, NSAID 49-hydroxylation, 5- CYP2C9, CYP3A4, Mancy et al., 1999 hydroxylation CYP2C18/19 Diltiazem Antihypertensive, CCB N-demethylation CYP3A4, CYP2C9, CYP2D6 Sutton et al., 1997 Docetaxel Anticancer, taxane Baccatin ring hydroxylation CYP3A4 Komoroski et al., 2005 Dovitinib Anticancer, PKI CYP1A1/2, CYP2D6, Kim et al., 2011c CYP3A4 DY-9760e Calmodulin antagonist oxidation (M8), CYP3A4, CYP2C9, Tachibana et al., 2005 N-dealkylation (DY- CYP2C19 9836), O-demethylation (M5), phenyl hydroxylation (M3) Eltrombopag , c-mpl Monooxygenation (J and CYP1A2 FDA, 2008c receptor agonist M6) Antidepressant N-demethylation (M4) CYP3A4 Kamel et al., 2013 Erlotinib Anticancer, PKI CYP3A4/5, CYP1A2, FDA 2004; Ling et al., 2006 CYP1A1, CYP1B1 , solvent Acetaldehyde formation CYP2E1, CYP1A2 Hamitouche et al., 2006 Etodolac Anti-inflammatory, NSAID 6-hydroxylation, 7- CYP2C9 Tougou et al., 2004 hydroxylation Evatanepag (CP-533,536) Prostaglandin EP2 receptor Formation of M3, M4, M20, CYP3A4/5 Prakash et al., 2008 agonist M22-M6 Everolimus Immunosuppressant, PKI Hydroxylation CYP3A4/5 Jacobsen et al., 2001; Picard et al., 2011 Antihyperuricemic, XO Hydroxylation (67M-2) CYP1A2, CYP2C9 Mukoyoshi et al., 2008; inhibitor FDA, 2009c Felodipine Antihypertensive, CCB CYP3A4 Karlsson et al., 2013 Flutamide Anticancer, antiandrogen Flu-1-G2 formation CYP1A2, CYP3A4, CYP2C9 Kang et al., 2008 (acid, parent) Antihyperlipidemic, HMG- 5-hydroxylation CYP2C9, CYP1A1, Fischer et al., 1999 CoA reductase inhibitor CYP2D6, CYP3A4 Gallopamil Antiarrhythmic, CCB Oxidation CYP3A4, CYP2D6 Suzuki et al., 1999 Genistein Anticancer, PKI 39-hydroxylation CYP1A2, CYP2E1 Hu et al., 2003 Gliclazide Antidiabetic, sulfonylurea 6b-hydroxylation, 7b- CYP2C9, CYP2C19 Elliot et al., 2007 hydroxylation Glyburide Antidiabetic, sulfonylurea 4-trans- (M1) and 3-cis- CYP3A4, CYP2C9, CYPC19 Zharikova et al., 2009 (glibenclamide) hydroxycyclohexyl (M2b) glyburide formation Halofantrine Antimalarial N-debutylation CYP3A4/5 Baune et al., 1999 Ibrolipim (NO-1886) Antihyperlipidemic O-deethylation (M2) CYP3A4 Morioka et al., 2002 ID951551 Acotiamide analog Deisopropylation CYP3A4, CYP1A1 Furuta et al., 2004 Ifosfamide Anticancer, alkylating 4-hydroxylation CYP2C9, CYP2A6, Chang et al., 1993; Huang agent CYP2B6, CYP3A4 et al., 2000 IN-1130 Anticancer, PKI M1 and M3 formation CYP3A4, CYP2D6, Kim et al., 2008 CYP2C19 K11777 , cysteine Formation of N-oxide CYP3A4 Jacobsen et al., 2000a protease inhibitor b-hydroxy-homoPhe and N- demethyl metabolites Karenicetin Anticancer CYP3A4, CYP2D6 Smith et al., 2003 L-775,606 Antimigraine, Hydroxylation (M1), N- CYP3A4 Prueksaritanont et al., 2000 dealkylation (M2) Lansoprazole Antiulcerative, PPI 5-hydroxylation CYP2C19, CYP3A4 Pichard et al., 1995 Anticancer, PKI O-dealkylation, N- CYP3A4/5, CYP2C19 FDA, 2007e; Teng et al., dealkylation 2010 Licofelone Anti-inflammatory, NSAID Hydroxylation (M2 and M4) CYP2C9, CYP2C19, Albrecht et al., 2008 CYP2D6, CYP2J2, CYP3A4 Lonafarnib Anticancer, FTI Hydroxylation (M4) CYP3A4/5, CYP1A1 Ghosal et al., 2006 Macitentan Antihypertensive, ERA Depropylation CYP3A4, CYP2C9, Sidharta et al., 2015 CYP2C19 (continued) Role of CYP2C8 in Drug Metabolism and Interactions 177

TABLE 1—Continued

Other CYP Enzymes Substrate Therapeutic Use and/or Metabolic Pathway(s) Involved in Overall References Drug Class Catalyzed by CYP2C8 Metabolisma Mavoglurant Anti-Parkinson, mGLUR5 M7 formation CYP3A4, CYP1A1 Walles et al., 2013 antagonist Methadone Analgesic, opioid N-demethylation CYP2B6, CYP3A4, CYP2D6 Iribarne et al., 1996; Wang and DeVane, 2003; Chang et al., 2011 Mirodenafil Erectogenic, PDE5 N-dealkylation CYP3A4, CYP2D6 Lee et al., 2008 inhibitor Antidepressant, NaSSA N-demethylation CYP2D6, CYP1A2, CYP3A4 Störmer et al., 2000 Analgesic, opioid N-demethylation CYP3A4 Projean et al., 2003b Muraglitazar Antidiabetic, PPARa, and g O-demethylation, O- CYP2C9, CYP2C19, Zhang et al., 2007a agonist dealkylation CYP2D6, CYP3A4 hydroxylation, N-acetyl- imide metabolite formation (2)(+)- Antihypertensive M1-M5 formation CYP2C9, CYP2C19 Zhu et al., 2014 Nalfurafine , opioid Decyclopropylmethylation CYP3A4, CYP2C9, Ando et al., 2012 CYP2C19 Naproxen Anti-inflammatory, NSAID O-demethylation CYP2C9, CYP1A2 Rodrigues et al., 1996, Tracy et al., 1997 Nicotine Psychostimulant 5-hydroxylation CYP2A6, CYP2B6 Yamazaki et al., 1999a Nifedipine Antihypertensive, CCB CYP3A4/5 Karlsson et al., 2013 Nilotinib Anticancer, PKI CYP3A4, CYP1A1/2, FDA, 2007c CYP2J2 R/S-Norverapamil Drug metabolite O-demethylation, N- CYP3A4/5 Tracy et al., 1999 dealkylation Odanacatib Antiosteoporotic, cathepsin Methyl hydroxylation (M8) CYP3A4/5 Kassahun et al., 2014 K Ombitasvir (ABT-267) Antiviral, NS5A inhibitor CYP3A4, CYP3A5 FDA, 2014k Omeprazole Antiulcerative, PPI 5-hydroxylation CYP2C19, CYP3A4 Karam et al., 1996 Pafuramidine maleate Antiparasitic O-demethylation (M1) CYPF4 Wang et al., 2006 (DB289) Anticancer, PKI Mono-oxygenation CYP3A4, CYP1A2 FDA, 2009d Antipsychotic MX 1, 10-11614, CO-UK2, CYP3A4, CYP2D6, Mizuno et al., 2003; and CO-UK3 formation (CYP1A1) Kitamura et al., 2005 Antipsychotic N-dealkylation CYP1A2, CYP3A4, Olesen and Linnet, 2000 CYP2C19, CYP2D6, (CYP2C18) Phenazone (antipyrine) Analgesic N-demethylation, 3- CYP3A4, CYP1A2, CYP2C9 Engel et al., 1996 hydroxylation, 4- hydroxylation Phenprocoumon Antithrombotic, VKA S-49-hydroxylation CYP2C9, CYP3A4 Ufer et al., 2004 Antiepileptic 4-hydroxylation CYP2C9, CYP2C19 Doecke et al., 1990 Piperaquine Antimalarial CYP3A4 Lee et al., 2012c acid Antihyperlipidemic, HMG- CYP2C9 Fujino et al., 2004 CoA reductase inhibitor Anticancer, PKI CYP3A4, CYP2D6, CYP3A5 FDA, 2012e Hormonal replacement CYP2C19, CYP3A4 Waxman et al., 1991 therapy 4-hydroxylation CYP2C9, CYP1A2, CYP2B6 Guitton et al., 1998 Antihypertensive, sGC N-demethylation CYP1A1, CYP3A4, CYP2J2 FDA, 2013b stimulator Anti-Parkinson, Desthienylethyl rotigotine CYP1A2, CYP2C9, CYP3A4 FDA, 2007b agonist formation Antipsychotic M203, EMD148107, EMD CYP3A4, CYP2C9, CYP1A2 Gallemann et al., 2010 329989, and M364d formation Selegiline Anti-Parkinson, MAO-B N-demethylation CYP2B6, CYP2C19 Hidestrand et al., 2001, inhibitor Salonen et al., 2003 Semagacestat Antidementia, g-secretase Benzylic hydroxylation CYP3A4/5 Yi et al., 2010 inhibitor (M3) Antiasthmatic, TXRA 5-methyl hydroxylation, 49- CYP3A4/5, CYP2C9 Kumar et al., 1997 hydroxylation Sildenafil Erectogenic, PDE5 CYP3A4, CYP2C9 Karlsson et al., 2013 inhibitor (TMC435) Antiviral, protease M21 and M2 formation CYP3A4/5, CYP2C19 FDA, 2013h inhibitor Sipoglitazar Antidiabetic, PPARa Hydroxylation (M-II) Nishihara et al., 2012 agonist Sirolimus Immunosuppressant Hydroxylation CYP3A4/5 Jacobsen et al., 2001 Antidiabetic, DPP-4 M2 and M5 formation CYP3A4 Vincent et al., 2007 inhibitor Sulfadiazine Antimicrobial N-hydroxylation CYP2C9 Winter and Unadkat, 2005 (continued) 178 Backman et al.

TABLE 1—Continued

Other CYP Enzymes Substrate Therapeutic Use and/or Metabolic Pathway(s) Involved in Overall References Drug Class Catalyzed by CYP2C8 Metabolisma Sunitinib Anticancer, PKI N-deethylation CYP3A4, CYP2B6, FDA, 2006b CYP2C9/19 T-5 Erectogenic, PDE5 N-oxidation CYP3A5 Li et al., 2014a inhibitor Tacrolimus Immunosuppressant, CYP3A4/5 Karlsson et al., 2013 calcineurin inhibitor Tamoxifen Anticancer, SERM M-I formation CYP3A4/5, CYP2D6 Desta et al., 2004 Tamoxifen N-oxide Drug metabolite Reduction to tamoxifen CYP2A6, CYP1A1, CYP3A4 Parte and Kupfer, 2005 Tegafur Anticancer, prodrug 5-hydroxylation CYP1A2, CYP2A6 Komatsu et al., 2000b, 2001 Sedative, benzodiazepine N-demethylation Yang et al., 1998 Terbinafine N-demethylation, side CYP2C9, CYP1A2, CYP3A4 Vickers et al., 1999 chain oxidation Hormonal replacement CYP3A4/5, CYP2B6 Waxman et al., 1991 therapy Tienilic acid 5-hydroxylation CYP2C9 Lopez Garcia et al., 1993; Bonierbale et al., 1999 Tipifarnib Anticancer, FTI CYP3A4, CYP2C19, Perez-Ruixo et al., 2006 CYP2A6, CYP2D6, CYP2C9 R-Tofisopam Anxiolytic M3 formation CYP3A4, CYP2C9, Cameron et al., 2007 (CYP3A5, CYP2C19) Tolbutamide Antidiabetic, sulfonylurea p-methyl hydroxylation CYP2C9, CYP2C19 Relling et al., 1990; Veronese et al., 1993; Rettie et al., 1994; Komatsu et al., 2000a Torsemide (torasemide) Diuretic Methyl hydroxylation CYP2C9 Miners et al., 2000; Ong et al., 2000 Trabectedin Anticancer N-demethylation CYP3A4, CYP2D6 Vermeir et al., 2009 Tretinoin (all-trans- Antiacne, retinoid 4-hydroxylation, CYP26A1, CYP3A4/5, Leo et al., 1989; Nadin and retinoic acid) 18-hydroxylation, CYP2B6, CYP1A2, Murray, 1999; Marill 5,6-epoxy metabolite CYP26, CYP2C9 et al., 2000; Thatcher formation et al., 2010 Trimethadione Antiepileptic N-demethylation CYP2E1, CYP3A4, CYP2C9 Kurata et al., 1998 (troxidone) Vanoxerine Antiarrhythmic, DRI CYP3A4, CYP2E1 Cherstniakova et al., 2001 R-Warfarin Antithrombotic, VKA 4-hydroxylation, CYP3A4, CYP1A1, Scordo et al., 2002; Kim 7-hydroxylation CYP2C19, CYP1A2 et al., 2012 Vitamin A (retinol) Anti-acne, retinoid Hydroxylation CYP1A1, CYP1A2, Leo et al., 1989 CYP1B1, CYP2D6, CYP3A4 Antidepressant, SMS Sulfoxide (M4a) formation CYP2D6, CYP3A4/5, FDA, 2013j; Chen et al., CYP2C19, CYP2C9, 2014 CYP2A6 Antiviral, NRTI Reduction CYP2C9 Eagling et al., 1994

5-HT, 5-hydroxytryptamine (); 5-MeO-DIPT, 5-methoxy-N,N-; 9cUAB30, (2E,4E,6Z,8E)-8-(39,4’-dihydro-1’(2’H)-naphthalen-1’-ylidene)-3,7-dimethyl- 2,4,6- octatrienoic acid; ARB, angiotensin II receptor blocker; ATP, adenosine triphosphate; BYZX, [(E)-2-(4-((diethylamino)methyl)benzylidene)-5,6-dimethoxy-2,3-dihydroinden-one]; CCB, ; Compound A, [(+)-(5S,6R,7R)-2-isopropylamino-7-[4-methoxy-2-((2R)-3-methoxy-2-methylpropyl)-5-(3,4-methylenedioxyphenyl) cyclopenteno[1,2-b] pyridine 6-carboxylic acid]; DPP, dipeptidyl peptidase; DRI, dopamine reuptake inhibitor; DY-9760, 3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4- imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate; ERA, endothelin receptor antagonist; FTI, farnesyl inhibitor; GABA, g-aminobutyric acid; HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A; IN-1130, 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-1H-imidazol-2-yl)methyl)benzamide; K11777, N-methyl--Phe-homoPhe- vinylsulfone-phenyl; L-775,606, (1-(3-(5-(1,2, 4-triazol-4-yl)-1H-indol-3-yl)propyl)-4-(2-(3-fluorophenyl)ethyl)piperazine); LABA, long-acting b-adrenoceptor agonist; LTRA, leukotriene receptor antagonist; MAO, monoamine oxidase; mGLUR, metabotropic glutamate receptor; c-mpl, myeloproliferative leukemia; NaSSA, noradrenergic and specific antidepressant; NNRTI, non-nucleoside reverse transcriptase inhibitor; NRTI, nucleoside analog reverse transcriptase inhibitor; NS, nonstructural protein; NSAID, nonsteroidal anti-inflammatory drug; PDE, phosphodiesterase; PGD2, prostaglandin D2; PKI, protein kinase inhibitor; PPAR, peroxisome proliferator- activated receptor; PPI, proton pump inhibitor; sGC, soluble guanylate cyclase; SERM, selective estrogen ; SMS, serotonin modulator and stimulator; SSRI, selective serotonin reuptake inhibitor; STS, steroid sulfatase; T-5, methyl 2-(4-aminophenyl)-1-oxo-7-(pyridin-2-ylmethoxy)-4-(3,4,5-trimethox- yphenyl)-1,2-dihydroisoquinoline-3-carboxylate; TCA, ; TXRA, thromboxane A2 receptor antagonist; VKA, vitamin K antagonist; XO, oxidase. aThis information is either based on the references given in the table or on data from the UW Metabolism and Transport Drug Interaction Database (DIDB), Copyright University of Washington 1999-2015 (DIDB Accessed May-September, 2015). docetaxel at high concentrations was metabolized by The androgen receptor antagonist enzalutamide, in- CYP2C8, but no enzyme kinetic parameters were dicated for treatment of castration-resistant prostate determined (Komoroski et al., 2005). An earlier cancer, is mainly metabolized by CYP2C8 and CYP3A4/ study, however, suggested that CYP2C8 is unable to 5 to enzalutamide M6 in vitro (FDA, 2012k). Then, M6 is accommodate docetaxel in its active site because of further metabolized by CYP2C8 to the active metabolite the absence of a side chain in the docetaxel mole- N-demethyl enzalutamide (M2), which accounts for cule (Cresteil et al., 2002). The side chain, which is approximately 50% of the total drug exposure in present in the paclitaxel molecule, is required for plasma. CYP2C8 seems to be the predominant enzyme a correct orientation of it into the active site of involved in enzalutamide pharmacokinetics also in vivo CYP2C8. (section VI.C.5; Gibbons et al., 2015). Role of CYP2C8 in Drug Metabolism and Interactions 179

TABLE 2 Glucuronide metabolites that are metabolized by CYP2C8

Metabolic pathway Other CYP enzymes involved Substrate catalyzed by CYP2C8 in overall metabolism References

a Clopidogrel acyl 1-b-D-glucuronide Tornio et al., 2014 Desloratadine N-glucuronide 3-hydroxylation Kazmi et al., 2015 Diclofenac acyl glucuronide 49-hydroxylation Kumar et al., 2002 Estradiol-17b-glucuronide 2-hydroxylation CYP3A7 Delaforge et al., 2005 Gemfibrozil 1-O-b glucuronide Benzylic oxidation Ogilvie et al., 2006; Baer et al., 2009 Licofelone 1-O-acyl glucuronide (M1) Hydroxylation (M3) Albrecht et al., 2008 Lu AA34893 carbamoyl glucuronidea Kazmi et al., 2010 MRL-C acyl glucuronide Hydroxylation CYP3A4 Kochansky et al., 2005 Sipoglitazar b-1-O-acyl glucuronide O-dealkylation (M-I) Nishihara et al., 2012

MRL-C, 2-[[5,7-dipropyl-3-(trifluoromethyl)-1,2-benzisoxazol-6-yl]oxy]-2-methylpropanoic acid. aThese glucuronides are likely to be substrates of CYP2C8 based on their metabolism-dependent inhibitory effect on CYP2C8.

Several protein kinase inhibitors are metabolized CYP2C8 only catalyzes the formation of the main by CYP2C8 to various degrees in vitro (Table 1). In metabolite, N-demethylimatinib (Table 4; Rochat et al., vitro, the majority (60–70%) of dabrafenib, a selective 2008; Filppula et al., 2013a). The relative roles of BRAF inhibitor, is metabolized by CYP2C8, and a CYP2C8 and CYP3A4 in the in vivo pharmacokinetics smaller part by CYP3A4 (;25%) and CYP2C9 (#10%) of imatinib are complex (section VI.C.5; Filppula et al., (Lawrence et al., 2014). Recombinant CYP2C8 pro- 2013b). In vitro, the multitargeted kinase duced only hydroxydabrafenib, whereas CYP3A4 formed inhibitor ponatinib is mainly metabolized by CYP3A4, both hydroxydabrafenib and carboxydabrafenib. How- followed by CYP2C8, CYP2D6, and CYP3A5. The con- ever, the in vivo importance of CYP2C8 in dabrafenib tributions of CYP3A4 and CYP2C8 to the in vivo pharmacokinetics seems to be quite modest (section VI. elimination of ponatinib are estimated to 34% and 19%, C.5; Suttle et al., 2015). Imatinib, the first tyrosine respectively (FDA, 2012e). Furthermore, CYP2C8 plays kinase inhibitor approved for clinical use, is metabo- a major role in the N-demethylation of the aurora kinase lized by several CYP enzymes in vitro, with CYP2C8 inhibitor tozasertib (Table 4; Ballard et al., 2007). With and CYP3A4 being the most important ones (Nebot the exception of dabrafenib and imatinib, the in vivo et al., 2010; Filppula et al., 2013a). CYP3A4 is involved importance of CYP2C8 in the metabolism of protein in several metabolic pathways of imatinib, whereas kinase inhibitors seems to have been poorly studied.

TABLE 3 Natural and endogenous compounds that are metabolized by CYP2C8

Metabolic Pathway(s) Other CYP Enzymes Involved Substrate Description Catalyzed by CYP2C8 in Overall Metabolisma References 1-Hydroxyl-2,3,5-trimethoxyxanthone Constituent of M1-M4 formation CYP3A4, CYP1A2, Feng et al., 2014 Halenia elliptica CYP2A6, CYP2B6, CYP2C9, CYP2C19 7a- and 7b-Hydroxy-D8-THC Marijuana constituent Oxidation CYP3A4, CYP2C9 Watanabe et al., 2007 8-Prenylnaringenin (flavaprenin) Prenylflavonoid M2 formation CYP2C19 Guo et al., 2006 Arachidonic acid Endogenous CYP2C9, CYP1A2, Daikh et al., 1994; Rifkind compound CYP2E1, CYP2J2 et al., 1995; Zeldin et al., 1995; Barbosa-Sicard et al., 2005 Endogenous CYP2C9/11/23 Barbosa-Sicard et al., 2005 compound Eupatilin Flavone 4-O-demethylation CYP1A2 Lee et al., 2007 R/S-Limonene Terpene CYP2C19, CYP2C9, Miyazawa et al., 2002 CYP3A4 Magnolin Constituent of Shin-i O-demethylation CYP2C9, CYP2C19, Kim et al., 2011a (M1 and M2), CYP3A4 hydroxylation (M4) Mesaconitine M1-M2, M7-M9 CYP3A4, CYP2C9, Ye et al., 2011 formation CYP2D6 Nitidine chloride Alkaloid CYP3A4 Li et al., 2014b Silybin () O-demethylation (CYP3A4) Jancova et al., 2007 Tanshinol ester Combination of the M1-M5 formation (CYP3A4) Liu et al., 2010b natural compounds danshensu and borneol

THC, . aThis information is either based on the references given in the table or on data from the UW Metabolism and Transport Drug Interaction Database (DIDB), Copyright University of Washington 1999-2015 (DIDB Accessed May-September, 2015). 180 Backman et al.

TABLE 4 CYP2C8-mediated reactions in vitro

a Substrate Metabolic Pathway Km Vmax CLint Test System References

mM pmol/min/pmol ml/min/pmol (pmol/min/mg) (ml/min/mg) 5-MeO-DIPT (Foxy) N-deisopropylation 291 1.7 0.0058 rCYP2C8 Narimatsu et al., 2006 7-Epi-10-deacetyl- Hydroxylation 18.0 3.038 0.17 rCYP2C8 Zhang et al., 2009a paclitaxel 7-Epi-cephalomannine M-2 formation 2.6 1.882 0.72 rCYP2C8 Zhang et al., 2009a 7-Epi-paclitaxel M-2 formation 1.4 1.409 1.0 rCYP2C8 Zhang et al., 2009b 8-prenylnaringenin M2 formation 3.72 4.64 1.3 rCYP2C8 Guo et al., 2006 17a-ethinylestradiol 2-hydroxylation 12 0.064 0.0053 rCYP2C8 Wang et al., 2004 Acotiamide (Z-338) Deisopropylation 152 (12.7) (0.084) rCYP2C8 Furuta et al., 2004 318 (347) (1.19) HLM Furuta et al., 2004 Alitretinoin (9-cis-retinoic 4-hydroxylation 7 0.948 0.14 rCYP2C8 Marill et al., 2002 acid) Aminophenazone N-demethylation 5,300 188 0.035 rCYP2C8 Niwa et al., 1999 (aminopyrine) Amiodarone N-deethylation 8.6 2.3 0.27 rCYP2C8 Ohyama et al., 2000 5.22 (12.2) (2.3) rCYP2C8 Soyama et al., 2002 Amitriptyline N-demethylation 0.072 rCYP2C8 Venkatakrishnan et al., 2001 Amodiaquine N-deethylation 0.9-1.2 2.6-3.9 2.1-4.4 rCYP2C8 Li et al., 2002 2.4 (1,462) (610) HLM Li et al., 2002 0.728 11.2 15 rCYP2C8 Walsky and Obach, 2004 1.89 (1,480) (780) HLM Walsky and Obach, 2004 0.81 0.23 0.28 rCYP2C8 Parikh et al., 2007 1 11 11 rCYP2C8 O’Donnell et al., 2007 1.95 6.87b 3.5b rCYP2C8 Baer et al., 2009 1.6 (9,130) (5,700) HLM Perloff et al., 2009 3.0 5.7b 1.9b rcCYP2C8 Kaspera et al., 2011 3.33-5.17 (1,180-2,770) (220-830) HLM Sjogren et al., 2012 3.9-7.3 (791-861) (120-200) HLM Yang et al., 2012 1.9 (2,196) (1,200) HLM Misaka et al., 2013 1.8 (7.3) (4.1) HIM Misaka et al., 2013 58.8 3,234c 55c Hep Kosugi et al., 2014 0.22-42.44 19.91-1,140c 24-95c Hep Li and Schlicht, 2014 Anastrozole Hydroxylation 86.8 0.00005 ,0.001 rCYP2C8 Kamdem et al., 2010 Arachidonic acid Total oxidative 6.0 4.6d 0.77d rCYP2C8 Barbosa-Sicard et al., 2005 metabolism Epoxidation 71 0.078 0.0011 rCYP2C8 Lundblad et al., 2005 Atorvastatin (acid, parent) p-hydroxylation 35.9 0.29 0.0081 rCYP2C8 Jacobsen et al., 2000b Bedaquiline N-demethylation 13.1 rCYP2C8 Liu et al., 2014 Buprenorphine N-dealkylation 12.4 (176.3) (14) rCYP2C8 Picard et al., 2005 Buspirone Total oxidative metabolism 0.073 rCYP2C8 Karlsson et al., 2013 BYZX N-deethylation (M3) 62.1 0.099e 0.0016e rCYP2C8 Yu et al., 2013a BYZX M2 N-deethylation (M1) 143.2 (0.000370) (,0.001) rCYP2C8 Yu et al., 2013a Caffeine 1-N-demethylation 920 0.014 ,0.001 rCYP2C8 Kot and Daniel, 2008 3-N-demethylation 200 0.016 ,0.001 rCYP2C8 Kot and Daniel, 2008 7-N-demethylation 3,560 0.172 ,0.001 rCYP2C8 Kot and Daniel, 2008 C-8-hydroxylation 3,370 0.319 ,0.001 rCYP2C8 Kot and Daniel, 2008 Carbamazepine 10,11-epoxidation 757 0.669 ,0.001 rCYP2C8 Cazali et al., 2003 Cephalomannine 6a-hydroxylation 41.3 5.267 0.13 rCYP2C8 Zhang et al., 2009a Cerivastatin (acid, parent) 6-hydroxylation 23 0.22 0.0096 rcCYP2C8 Kaspera et al., 2010 Demethylation 24 0.57 0.024 rcCYP2C8 Kaspera et al., 2010 Cerlapirdine Demethylation 3.3 3.4 1.0 rCYP2C8 Tse et al., 2014 Chloroquine N-deethylation 430 52.1 0.12 rCYP2C8 Kim et al., 2003 111 8.33 0.075 rCYP2C8 Projean et al., 2003a Cilostazol OPC-13217 formation 33.8 0.30 0.089 rCYP2C8 Hiratsuka et al., 2007 Cisapride 4-hydroxylation ;5.9 0.71 ;0.12 rCYP2C8 Desta et al., 2000 N-dealkylation ;0.91 0.29 ;0.32 rCYP2C8 Desta et al., 2000 2-hydroxylation 5.80 0.0267 0.0046 rCYP2C8 Pearce et al., 2001 4-hydroxylation 3.40 0.289 0.085 rCYP2C8 Pearce et al., 2001 N-dealkylation 2.0 0.109 0.055 rCYP2C8 Pearce et al., 2001 (2)-Cisapride 4-hydroxylation 13.3 0.15 0.011 rCYP2C8 Desta et al., 2001 (+)-Cisapride 4-hydroxylation 12.6 0.24 0.019 rCYP2C8 Desta et al., 2001 Cyclosporine Total oxidative metabolism 0.40 rCYP2C8 Karlsson et al., 2013 Dapsone N-hydroxylation 58-75 0.440 0.0059-0.0076 rCYP2C8 Winter et al., 2000 Dibenzylfluorescein O-debenzylation 1.0 0.4d 0.4d n/a Miller et al., 2000 29.16 0.79 0.027 rCYP2C8 Ghosal et al., 2003 1.9 (1.3) (0.68) THLE Donato et al., 2004 Diclofenac 49-hydroxylation 630 1.2b,d 0.0019b,d rCYP2C8 Mancy et al., 1999 5-hydroxylation 280 7b,d 0.025b,d rCYP2C8 Mancy et al., 1999 DY-9760e Imidazole oxidation (M8) 2.6 0.0732 0.028 rCYP2C8 Tachibana et al., 2005 N-dealkylation (DY-9836) 15.2 0.0231 0.0015 rCYP2C8 Tachibana et al., 2005 O-demethylation (M5) 3.1 0.0128 0.0041 rCYP2C8 Tachibana et al., 2005 Phenyl hydroxylation (M3) 2.5 0.2955 0.12 rCYP2C8 Tachibana et al., 2005 (continued) Role of CYP2C8 in Drug Metabolism and Interactions 181

TABLE 4—Continued

a Substrate Metabolic Pathway Km Vmax CLint Test System References Eicosapentaenoic acid Total oxidative metabolism 5.4 6.2d 1.1d rCYP2C8 Barbosa-Sicard et al., 2005 Estradiol-17b-glucuronide 2-hydroxylation 88 1.86 0.021 rCYP2C8 Delaforge et al., 2005 Ethanol Acetaldehyde formation 8,300 (0.0043) (,0.001) rCYP2C8 Hamitouche et al., 2006 Eupatilin 4-O-demethylation 4.5 0.94 0.21 rCYP2C8 Lee et al., 2007 Felodipine Total oxidative metabolism 1.1 rCYP2C8 Karlsson et al., 2013 Fenretidine 49-hydroxylation 2.2 282f 130f rCYP2C8 Illingworth et al., 2011 49-oxidation 5.0 30f 6.0f rCYP2C8 Illingworth et al., 2011 R-Fluoxetine N-demethylation 153.8 (6.08) (0.040) rcCYP2C8 Wang et al., 2014b S-Fluoxetine N-demethylation 195.0 (6.68) (0.034) rcCYP2C8 Wang et al., 2014b Fluvastatin (acid, parent) 5-hydroxylation 2.8 0.13 0.046 rCYP2C8 Fischer et al., 1999 Gliclazide 6b-hydroxylation 984 0.63 ,0.001 rCYP2C8 Elliot et al., 2007 7b-hydroxylation 346 0.06 ,0.001 rCYP2C8 Elliot et al., 2007 Glyburide (glibenclamide) Total oxidative metabolism 10.2 0.9 0.09 rCYP2C8 Zharikova et al., 2009 7.7 2.5 0.32 rCYP2C8 Zhou et al., 2010 0.08 rCYP2C8 Varma et al., 2014 Halofantrine N-debutylation 156 0.039 ,0.001 rCYP2C8 Baune et al., 1999 Ibrolipim (NO-1886) O-deethylation (M2) 28.4–53.9 0.0334–0.10 0.0012–0.0019 rCYP2C8 Morioka et al., 2002 R-Ibuprofen 2-hydroxylation 3.5–74 rCYP2C8 Hamman et al., 1997 282 9.4 0.033 rCYP2C8 Chang et al., 2008 341.3 4.92e 0.014e rCYP2C8 Yu et al., 2013b S-Ibuprofen 2-hydroxylation 292 5.4 0.018 rCYP2C8 Chang et al., 2008 388.8 3.02e 0.0078e rCYP2C8 Yu et al., 2013b Imatinib N-demethylation 1.4 0.408 0.29 rCYP2C8 Nebot et al., 2010 4.28 4.07 0.95 rCYP2C8 Filppula et al., 2013a 5 0.553 0.1 rCYP2C8 Khan et al., 2015 Isotretinoin (13-cis-retinoic 4-hydroxylation 13.8 134.6g 9.8g rCYP2C8 Rowbotham et al., 2010 acid) L-775,606 Hydroxylation (M1) 42 0.62 0.015 rCYP2C8 Prueksaritanont et al., 2000 N-dealkylation (M2) 64 0.03 ,0.001 rCYP2C8 Prueksaritanont et al., 2000 Loperamide N-demethylation 11.3 0.0052 ,0.001 rCYP2C8 Kim et al., 2004 Magnolin O-demethylation (M1) 17.7 1.9 0.11 rCYP2C8 Kim et al., 2011a O-demethylation (M2) 21.2 0.3021 0.014 rCYP2C8 Kim et al., 2011a Hydroxylation (M4) 29.7 0.7099 0.024 rCYP2C8 Kim et al., 2011a Mavoglurant Total oxidative metabolism 17.1 5.06 0.30 rCYP2C8 Walles et al., 2013 Mirodenafil N-dealkylation (SK3541) 121 0.85 0.0070 rCYP2C8 Lee et al., 2008 Montelukast 36-hydroxylation (M6) 0.050 0.18 3.6 rCYP2C8 Filppula et al., 2011 0.014 ;0.24 ;17 rCYP2C8 VandenBrink et al., 2011 0.065 ;0.09 ;1 HLM VandenBrink et al., 2011 0.31 0.015 0.048 rCYP2C8 Oliveira Cardoso et al., 2015 25-hydroxylation (M3) 0.33 0.002 0.006 rCYP2C8 Oliveira Cardoso et al., 2015 Morphine N-demethylation 4,800 5.41 0.0011 rCYP2C8 Projean et al., 2003b Nifedipine Total oxidative metabolism 0.38 rCYP2C8 Karlsson et al., 2013 Nitidine chloride Total oxidative metabolism 1.17 0.0705 0.060 rCYP2C8 Li et al., 2014b R-Norverapamil D-620 formation 56 5.3 0.095 rCYP2C8 Tracy et al., 1999 PR-22 formation 38 29 0.76 rCYP2C8 Tracy et al., 1999 S-Norverapamil D-620 formation 80 14 0.18 rCYP2C8 Tracy et al., 1999 PR-22 formation 80 12 0.15 rCYP2C8 Tracy et al., 1999 Olanzapine N-demethylation 30 1.370 0.046 rCYP2C8 Korprasertthaworn et al., 2015 Omeprazole 5-hydroxylation 300 3.3 0.011 rCYP2C8 Karam et al., 1996 Paclitaxel 6a-hydroxylation 5.4 30 5.6 rCYP2C8 Rahman et al., 1994 4.0 (870) (220) HLM Rahman et al., 1994 15 0.12 0.0080 HLM Monsarrat et al., 1997 17 HLM Ando et al., 1998 26 HLM Desai et al., 1998 34.8 (1632) (47) HLM Fischer et al., 1998 4.9 1.14 0.23 rCYP2C8 Masimirembwa et al., 1999 6 (234) (39) rCYP2C8 Ong et al., 2000 12.2 (142) (12) HLM Ong et al., 2000 2.85 5.667 2.0 rCYP2C8 Ohyama et al., 2000 2.58–4.55 0.224–0.583 0.070–0.19 HLM Ohyama et al., 2000 6.8 3.0d 0.44d rCYP2C8 Yamazaki et al., 2000 15 0.8 0.053 rCYP2C8 Dai et al., 2001 4.3 (147) (34) rCYP2C8 Fujino et al., 2001 27.4 (359) (13) HLM Fujino et al., 2001 16.2 29.8 1.8 rCYP2C8 Soyama et al., 2001 15 1.950 0.13 HLM Cresteil et al., 2002 9.3 (60.9) (6.8) HLM Václavíková et al., 2003 13.3 (109.1) (8.2) HLM Donato et al., 2004 16.3 (81.0) (5.0) THLE Donato et al., 2004 7.50 (70.2) (9.4) HLM Polasek et al., 2004 8.3 1.718 0.21 rCYP2C8 Zhang et al., 2009b 18.3 (250) (14) HLM Zhang et al., 2009b (continued) 182 Backman et al.

TABLE 4—Continued

a Substrate Metabolic Pathway Km Vmax CLint Test System References 4.17 2.4 0.58 rCYP2C8 Gao et al., 2010 2.33 4.05 1.7 rCYP2C8 Hanioka et al., 2010 3.7 0.29b 0.078b rcCYP2C8 Kaspera et al., 2011 2.58 3.53 1.4 rCYP2C8 Wattanachai et al., 2011 7.08 (137) (19) HLM Wattanachai et al., 2011 8.65 69.83 8.1 rCYP2C8 Yu et al., 2013b 5.2 (222.1) (43) HLM Wang et al., 2014a 5.41–15.9 (52.6–230) (3.3–26) HLM Kudo et al., 2015 Pafuramidine maleate O-demethylation (M1) 2.6 0.12 0.046 rCYP2C8 Wang et al., 2006 (DB289) Perospirone Total oxidative metabolism 1.09 1.93 1.8 rCYP2C8 Kitamura et al., 2005 Perphenazine N-dealkylation 28 1.35 0.048 rCYP2C8 Olesen and Linnet, 2000 Phenazone (antipyrine) N-demethylation 30,400 (156.4) (0.0051) rCYP2C8 Engel el al., 1996 3-hydroxylation 22,000 (43.9) (0.0020) rCYP2C8 Engel el al., 1996 4-hydroxylation 61,000 (140.9) (0.0023) rCYP2C8 Engel el al., 1996 Phenprocoumon S-49-hydroxylation 3.78 0.027 0.0071 rCYP2C8 Ufer et al., 2004 Pioglitazone M-IV formation 10.2 9.2 0.91 rCYP2C8 Tornio et al., 2008b 9.8 (640) (65) HLM Tornio et al., 2008b 29.5 1.702 0.058 rCYP2C8 Muschler et al., 2009 R483 Hydroxylation (M1) 1.4 (1,000) (700) n/a Bogman et al., 2010 Repaglinide Total oxidative metabolism 2.8 4.9 1.8 rCYP2C8 Kajosaari et al., 2005a M1 formation 25 0.08 0.003 rCYP2C8 Säll et al., 2012 M4 formation 5.7 0.35 0.061 rCYP2C8 Säll et al., 2012 M4 formation 9.0 (130) (14) HLM Säll et al., 2012 M4 formation 28 13c 0.46c Hep Säll et al., 2012 M4 formation 13 (18) (1.4) S9 Säll et al., 2012 12.01 15.69e 1.3e rCYP2C8 Yu et al., 2013b Rosiglitazone p-hydroxylation 44 (2,900) (66) rCYP2C8 Baldwin et al., 1999 4.3–7.7 (550–883) (93–130) HLM Baldwin et al., 1999 N-demethylation 10 (2,430) (240) rCYP2C8 Baldwin et al., 1999 p-hydroxylation 4.0 0.42b 0.11b rcCYP2C8 Kaspera et al., 2011 N-demethylation 2.9 0.38b 0.13b rcCYP2C8 Kaspera et al., 2011 Selegiline Demethylation 82 3 0.04 rCYP2C8 Hidestrand et al., 2001 Levomethamphetamine 630 7 0.01 rCYP2C8 Hidestrand et al., 2001 formation Seratrodast 49-hydroxylation 28.2 0.1438 0.0051 rCYP2C8 Kumar et al., 1997 5-methylhydroxylation 32.9 0.4983 0.015 rCYP2C8 Kumar et al., 1997 Sildenafil Total oxidative metabolism 0.055 rCYP2C8 Karlsson et al., 2013 Simvastatin acid M1 formation 88 2,800 32 rCYP2C8 Prueksaritanont et al., 2003 M2 formation 36 850 24 rCYP2C8 Prueksaritanont et al., 2003 M3 formation 16 600 38 rCYP2C8 Prueksaritanont et al., 2003 T-5 N-oxidation 1.6 0.22 0.14 rCYP2C8 Li et al., 2014a Tacrolimus Total oxidative metabolism 0.19 rCYP2C8 Karlsson et al., 2013 Tanshinol borneol ester M3 formation 45.2 4.28h 0.095h rCYP2C8 Liu et al., 2010b Terbinafine Deamination 24.8 0.512 0.021 rCYP2C8 Vickers et al., 1999 N-demethylation 13.6 2.06 0.15 rCYP2C8 Vickers et al., 1999 Side chain oxidation 26.4 0.825 0.031 rCYP2C8 Vickers et al., 1999 Total oxidative metabolism 15.3 4.47 0.29 rCYP2C8 Vickers et al., 1999 R-Tofisopam M3 formation 52 0.43 0.0083 rCYP2C8 Cameron et al., 2007 Tolbutamide Hydroxylation 650.5 rCYP2C8 Veronese et al., 1993 531 0.39 ,0.001 rCYP2C8 Rettie et al., 1994 1,160 (10.2) (0.0088) rCYP2C8 Pang et al., 2012 Torsemide (torasemide) Methyl hydroxylation 184 1.8 0.0098 rCYP2C8 Miners et al., 2000 170 (35) (0.21) rCYP2C8 Ong et al., 2000 Tozasertib (MK 0457, VX6, N-demethylation 64 129 2.0 rCYP2C8 Ballard et al., 2007 VX 680) Tretinoin (all-trans-retinoic 4-hydroxylation 6.1 0.18 0.030 rCYP2C8 Nadin and Murray, 1999 acid) 4-hydroxylation 50 1.211 0.024 rCYP2C8 Marill et al., 2000 18-hydroxylation 17 0.033 0.0019 rCYP2C8 Marill et al., 2000 5,6-epoxy metabolite 130 0.450 0.0035 rCYP2C8 Marill et al., 2000 formation 4-hydroxylation 13.4 4.8 0.36 rCYP2C8 Thatcher et al., 2010 Troglitazone Quinone formation 2.7 4.2d 1.6d rCYP2C8 Yamazaki et al., 1999b Verapamil O-demethylation 48.4 (13) (0.27) rCYP2C8 Busse et al., 1995 Total oxidative metabolism 0.39 rCYP2C8 Karlsson et al., 2013 R-Verapamil D-617 formation 127 8.0 0.063 rCYP2C8 Tracy et al., 1999 Norverapamil formation 127 6.9 0.054 rCYP2C8 Tracy et al., 1999 PR-22 formation 33 2.2 0.067 rCYP2C8 Tracy et al., 1999 S-Verapamil D-617 formation 185 8.0 0.043 rCYP2C8 Tracy et al., 1999 Norverapamil formation 154 15 0.097 rCYP2C8 Tracy et al., 1999 PR-22 formation 141 1.6 0.011 rCYP2C8 Tracy et al., 1999 (continued) Role of CYP2C8 in Drug Metabolism and Interactions 183

TABLE 4—Continued

a Substrate Metabolic Pathway Km Vmax CLint Test System References Vidupiprant (AMG 853) t-butyl hydroxylation (M2) 1.21 0.031 0.026 rCYP2C8 Foti et al., 2012 Cyclopropyl hydroxylation 49.1 0.250 0.0051 rCYP2C8 Foti et al., 2012 (M3) Vitamin A (retinol) 4-hydroxylation 71 1.73 0.024 rcCYP2C8 Leo et al., 1989 Zopiclone N-demethylation 71 2.5 0.035 rCYP2C8 Becquemont et al., 1999 N-oxidation 59 1.0 0.017 rCYP2C8 Becquemont et al., 1999

5-MeO-DIPT, 5-methoxy-N,N-diisopropyltryptamine; BYZX, [(E)-2-(4-((diethylamino)methyl)benzylidene)-5,6-dimethoxy-2,3-dihydroinden-one]; CLint, intrinsic clearance; Hep, hepatocytes; HIM, human intestinal microsomes; HLM, human liver microsomes; Km, Michaelis-Menten constant; n/a, not available; rCYP2C8, recombinant CYP2C8; rcCYP2C8, reconstituted CYP2C8; S9, S9 fraction; THLE, immortalized human liver epithelial cells; Vmax, maximal velocity. a Calculated as Vmax/Km. b kcat reported instead of Vmax,CLint calculated as kcat/Km. c 6 6 Vmax as pmol/min/10 cells, CLint as pmol/min/10 cells/mM. d Vmax as 1/min, CLint as 1/min/mM. e Vmax as arbitrary unit (AU), CLint as AU/mM. f Vmax as unit/min, CLint as unit/min/mM. g Vmax as counts/s/s, CLint as counts/s/s/mM. h Vmax as AUC×min/nmol, CLint as AUC×min/nmol/mM.

In addition, CYP2C8 participates to various degree to Drug Administration (FDA) as a marker reaction for the metabolism of several other anticancer agents, as in vitro CYP2C8 activity (http://www.fda.gov/Drugs/ listed in Table 1. DevelopmentApprovalProcess/DevelopmentResources/ 2. Antidiabetic Agents. The nonsulfonylurea insulin DrugInteractionsLabeling/ucm093664.htm). In vitro, secretagogue repaglinide is metabolized primarily by troglitazone is metabolized by CYP2C8 to its qui- CYP2C8 (Table 4) and CYP3A4, but it also undergoes none metabolite (M3) (Table 4) at a two- to eightfold direct glucuronidation by uridine-59-diphosphoglucuro- higher rate than by CYP2C19, CYP3A4, and CYP2C9 nosyltransferase (UGT) 1A1 (Bidstrup et al., 2003; (Yamazaki et al., 1999b). M3 is a minor metabolite of Kajosaari et al., 2005a,b; Gan et al., 2010). In addition, troglitazone, but it has been suggested to be responsible there is in vitro data suggesting that aldehyde de- for the drug-induced hepatotoxicity associated with hydrogenase is involved in its metabolism (Säll et al., troglitazone use (Yamazaki et al., 1999b; Smith, 2012). Moreover, repaglinide is a substrate of the 2003). Furthermore, the R483 is hepatic uptake transporter organic anion-transporting primarily metabolized by CYP2C8 and CYP2C19 in polypeptide (OATP) 1B1 (Niemi et al., 2005b, 2011). The vitro (Bogman et al., 2010). CYP2C8 catalyzes the formation of the main metabolites of repaglinide, an formation of the weakly active M1 metabolite (Table 4) oxidized dicarboxylic acid (M2) and, in particular, 39- and its further metabolism to M4, which is the main hydroxyl repaglinide (M4), is largely dependent on metabolite of R483 in plasma. In turn, CYP2C19 forms CYP2C8, whereas the less important aromatic amine M2, which shows similar pharmacological activity as metabolite (M1) is primarily formed by CYP3A4 (Bidstrup parent R483 (Bogman et al., 2010). et al., 2003; Kajosaari et al., 2005a,b). Additionally, CYP2C8 is involved, to a minor extent, Pioglitazone, a thiazolidinedione peroxisome prolif- in the metabolism of the sulfonylureas gliclazide, erator activated receptor (PPAR) g agonist, is primarily glyburide, and tolbutamide, the dipeptidyl peptidase 4 metabolized by CYP2C8 in vitro, with smaller contri- inhibitor sitagliptin, and the PPARa agonist sipoglita- butions by CYP3A4 and the extrahepatic CYP1A1 zar (Table 1; Relling et al., 1990; Srivastava et al., 1991; (Jaakkola et al., 2006c; FDA, 2013a). In vitro, CYP2C8 Veronese et al., 1993; Elliot et al., 2007; Vincent et al., forms the pharmacologically active hydroxypioglita- 2007; Zharikova et al., 2009; Nishihara et al., 2012). zone (M-IV) and ketopioglitazone (M-III) (Jaakkola Tolbutamide p-methyl hydroxylation has been used as et al., 2006c; Tornio et al., 2008b), which are the main a marker reaction for CYP2C8 activity in several in metabolites in human serum with concentrations equal vitro studies. However, its Michaelis-Menten constant to or greater than those of the parent drug (Eckland (Km) for CYP2C8 is very high (.530 mM; Table 4), and and Danhof, 2000). In vivo studies support the central it is effectively metabolized by CYP2C9 at lower role of CYP2C8 in pioglitazone metabolism observed concentrations. in vitro (section VI.C.2). 3. Antimalarial Agents. The 4-aminoquinoline de- Rosiglitazone, another PPAR-g agonist, is also a rivative amodiaquine, widely used for treatment of substrate of CYP2C8. In vitro, it undergoes CYP2C8- malaria for more than 60 years, is a substrate of mediated p-hydroxylation and N-demethylation (Table 4), CYP2C8 (Li et al., 2002). It is also metabolized by the followed by sulfate and glucuronic acid conjugation extrahepatic enzymes CYP1A1 and CYP1B1 to a minor (Baldwin et al., 1999; Kaspera et al., 2011; FDA, extent (Li et al., 2002). Amodiaquine N-deethylation is a 2014a). CYP2C9 also participates in its metabolism frequently used in vitro marker reaction for CYP2C8 to a minor extent (Baldwin et al., 1999). Rosiglitazone because of its high affinity (Km typically around 2 mM) p-hydroxylation is recommended by the Food and and high turnover rate (Table 4). N-desethylamodiaquine, 184 Backman et al.

Fig. 3. Molecular descriptors of drugs classified as "major" or "intermediate" CYP2C8 substrates in Table 1. The molecular descriptors were obtained from SciFinder (American Chemical Society). which is the main metabolite of amodiaquine, is assumed gemfibrozil and cerivastatin is discussed in section to be the main entity responsible for the pharmacological VI.C.4. response to amodiaquine (Churchill et al., 1985; Mount The parent simvastatin lactone is either oxidized by et al., 1986). CYP3A4/5 or hydrolyzed to its acid form, which is Chloroquine, also a 4-aminoquinoline, is mainly pharmacologically active (Prueksaritanont et al., 1997, metabolized to its N-desethyl metabolite by CYP2C8 2003). In human liver microsomes (HLM), the metabo- (Table 4) and CYP3A4, with a small contribution by lism of simvastatin acid was catalyzed primarily CYP2D6 in vitro (Kim et al., 2003; Projean et al., 2003a). ($80%) by CYP3A4/5, with a smaller contribution Furthermore, CYP2C8 also seems to play a small role (#20%) by CYP2C8 (Prueksaritanont et al., 2003). in the in vitro metabolism of the antimalarial agents Recombinant CYP2C8 formed all three simvastatin dapsone, halofantrine, and piperaquine (Table 1; Baune acid metabolites (M1-M3) observed in HLM (Table 4; et al., 1999; Winter et al., 2000; Lee et al., 2012c). Prueksaritanont et al., 2003). In vitro, fluvastatin is 4. Lipid-lowering Drugs. CYP2C8 participates to a mainly metabolized by CYP2C9 into three metabolites, small extent in the metabolism of several HMG-CoA but CYP1A1, CYP2C8, CYP2D6, and CYP3A4 form reductase inhibitors (statins), but it has a major role for 5-hydroxyfluvastatin (Fischer et al., 1999). Both atorvas- the biotransformation of cerivastatin. Cerivastatin is tatin (acid) and its lactone are primarily metabolized extensively metabolized in humans (Boberg et al., 1997; by CYP3A4 to their hydroxylated metabolites in vitro, Mück, 2000). Parent cerivastatin (acid) is metabolized but CYP2C8 is involved in the formation of p-hydroxy by CYP2C8 and CYP3A4, whereas cerivastatin lac- atorvastatin acid to a small extent (Jacobsen et al., tone is predominantly metabolized by CYP3A4 (Boberg 2000b). Furthermore, pitavastatin acid is metabolized et al., 1997; Wang et al., 2002; Fujino et al., 2004). The by CYP2C9 and CYP2C8 in vitro, whereas its lactone is formation of the major metabolite of cerivastatin, metabolized by CYP3A4 and CYP2D6 (Fujino et al., 6-hydroxycerivastatin (M-23), is primarily mediated by 2004). CYP2C8, whereas both CYP2C8 and CYP3A4 produce 5. Other Drugs. Early in vitro studies concluded demethylcerivastatin (M1) (Wang et al., 2002; Kaspera that the leukotriene receptor antagonist montelukast et al., 2010). The notorious in vivo interaction between is mainly metabolized by CYP2C9 and CYP3A4 (Chiba Role of CYP2C8 in Drug Metabolism and Interactions 185

Fig. 4. The number of "major" and "intermediate" CYP2C8 substrates by drug class, as listed in Table 1. et al., 1997), whereas the role of CYP2C8 was not 2009b). Incubation of treprostinil with recombinant evaluated. However, in vitro studies performed more CYP2C8 for 15 minutes resulted in a 95% depletion of than a decade later demonstrated that CYP2C8 is the treprostinil concentrations, whereas only 22% was con- key enzyme involved in the oxidative metabolism of sumed by recombinant CYP2C9. CYP2C8 seems to be of montelukast (Filppula et al., 2011; VandenBrink et al., importance in the in vivo pharmacokinetics of treprosti- 2011). CYP2C8 catalyzes the main metabolic pathway nil (FDA, 2009b). of montelukast; formation of the pharmacologically The sedative agent zopiclone is metabolized by active 36-hydroxymontelukast (M6), and its subsequent CYP2C8 and CYP3A4 in vitro (Becquemont et al., metabolism to the secondary metabolite M4, a dicar- 1999). In HLM, CYP2C8 was the main enzyme catalyz- boxylic acid (Table 4). In addition, CYP2C8 forms 25- ing N-demethylation of zopiclone, followed by CYP2C9 hydroxymontelukast (M3) (Filppula et al., 2011). These and CYP3A4. CYP2C8 also participated in the forma- in vitro findings are in agreement with X-ray crystal- tion of N-oxide zopiclone, together with CYP3A4 (major) lography data, demonstrating a ligand-protein binding and CYP2C9 (Becquemont et al., 1999). However, in interaction between montelukast and CYP2C8 (Schoch another in vitro study, montelukast (CYP2C8 inhibitor) et al., 2008). The montelukast molecule was positioned and gemfibrozil (CYP2C8 and CYP2C9 inhibitor) had with its benzyl ring in close proximity to the heme iron no effect on the elimination of clinically relevant con- of CYP2C8. The montelukast metabolites M3, M4, and centrations of zopiclone (Tornio et al., 2006), supporting M6 formed by CYP2C8 in vitro, all result from the in vivo data showing a lack of effect of gemfibrozil on oxidation of the benzyl ring of montelukast. zopiclone concentrations in healthy subjects (section The novel prolyl hydroxylase inhibitor daprodustat VI.C.8). (GSK1278863), an antianemic agent, is primarily me- Based on in vitro studies, CYP2C8 likely plays tabolized by CYP2C8, with a smaller contribution by an intermediate role in the elimination of 9cUAB30, CYP3A4 in vitro (Johnson et al., 2014). It seems to alitretionin, amiodarone, cisapride, fenretinide, fluox- be more sensitive than repaglinide to CYP2C8 inhibi- etine, irosustat, isotretionin, loperamide, olanzapine, tion by gemfibrozil in vivo (section VI.C.8; Johnson olodaterol, propanoic acid, dronedarone, tazarotenic et al., 2014). acid, verapamil, and vidupiprant (AMG 853) (Table 1). The novel nonstructural 5B nonnucleoside polymer- For instance, CYP2C8 catalyzes dealkylation of both ase inhibitor dasabuvir is extensively metabolized by enantiomers of the calcium channel blocker verapamil CYP2C8, with a small contribution by CYP3A (FDA, and its metabolite norverapamil (Busse et al., 1995; 2014g). CYP2C8 also plays an intermediate/small role Tracy et al., 1999). Tazarotenic acid, the active moiety of in the metabolism of the nonstructural protein 3/4 A the antipsoriatic agent tazarotene, is mainly metabo- protease inhibitor paritaprevir and nonstructural pro- lized by CYP2C8 and flavin-containing monooxyge- tein 5A inhibitor ombitasvir (FDA, 2014g; Menon et al., nases in vitro (Attar et al., 2003). When tazarotenic 2015). However, no in vitro metabolism data have yet acid was incubated with 10 individual recombinant been published for these compounds. CYP enzymes, only CYP2C8 markedly catalyzed sul- The prostacyclin analog treprostinil is primarily me- foxidation, which is the main metabolic pathway of tabolized by CYP2C8, followed by CYP2C9 in vitro (FDA, tazarotenic acid. 186 Backman et al.

There is a vast amount of in vitro data suggesting that estradiol-17b-glucuronide, gemfibrozil 1-O-b glucuronide, CYP2C8 may be of relevance in the metabolism of a licofelone 1-O-acyl glucuronide, Lu AA34893 carbamoyl number of other drugs (Table 1). For the majority of these glucuronide, 2-[[5,7-dipropyl-3-(trifluoromethyl)-1,2- compounds, the role of CYP2C8 in their in vivo elimina- benzisoxazol-6-yl]oxy]-2-methylpropanoic acid (MRL-C) tion is likely to be small or negligible. However, for acyl glucuronide, and sipoglitazar b-1-O-acyl glucuronide some drugs, the in vivo contribution of CYP2C8 to their (see Table 2 for references). Thus, CYP2C8 makes yet metabolism cannot be estimated based on available another exception to the old concept that drug metabolism information, and may, in fact, exceed 20%. Alternatively, is divided into sequential phase I and phase II reactions, CYP2C8 may become a determinant in their metabolism i.e., functionalization and conjugation, respectively (Fig. 5). after inhibition of other enzymes important for their Kumar et al. (2002) demonstrated the first example of elimination. For instance, CYP2C8 is involved in the CYP2C8-mediated metabolism of a glucuronide conju- metabolism of seratrodast, a thromboxane A2 receptor gate when they showed that the conversion of diclofenac antagonist in vitro (Kumar et al., 1997). The main acyl glucuronide to its 49-hydroxy derivative is exclusively metabolic pathway of seratrodast, 5-methylhydroxylation, mediated by CYP2C8 in vitro. In 2005, it was reported is primarily catalyzed by CYP3A and CYP2C9, but CYP2C8 that CYP2C8 is also able to directly catalyze the contributes to a small degree. However, CYP2C8 is a 2-hydroxylation of estradiol-17b-glucuronide in vitro major contributor to seratrodast 49-hydroxylation, a minor (Delaforge et al., 2005). Docking of the glucuronide of metabolic route (Kumar et al., 1997). estradiol into the crystal structure of CYP2C8 showed 6. Glucuronide Metabolites. Several glucuronide me- that the active site is large enough to inhabit the tabolites have been reported to undergo metabolism by conjugate. Also the fetal CYP3A isoform CYP3A7 oxidized CYP2C8, including clopidogrel acyl 1-b-D-glucuronide, estradiol-17b-glucuronide, but CYP2C8 was five times desloratadine glucuronide, diclofenac acyl glucuronide, more active than CYP3A7. However, CYP3A4 was not

Fig. 5. Schematic illustration of the interaction between CYP2C8 and its glucuronide substrates. CYP2C8 and the UGT are localized on opposite sites of the endoplasmic membrane (A). The drug is glucuronidated by the UGT (B). Hereafter, the glucuronide crosses the endoplasmic membrane and binds into CYP2C8 (C). Then, the glucuronide is either metabolized by CYP2C8 and released as a metabolite (D, left), e.g., desloratadine glucuronide, and diclofenac acyl glucuronide, or it is metabolized to a reactive agent that inactivates CYP2C8 (D, right), as for clopidogrel acyl 1-b-D-glucuronide and gemfibrozil 1-O-b glucuronide. CYP2C8 has been suggested to exist as a dimer (Hu et al., (2010), Schoch et al., (2004)). ER, endoplasmic reticulum. Role of CYP2C8 in Drug Metabolism and Interactions 187 able to metabolize estradiol-17b-glucuronide (Delaforge acid, 13-cis-retinoic acid, and 9cUAB30 (Marill et al., et al., 2005). Moreover, the acyl glucuronide of the dual 2002; Gorman et al., 2007). PPAR a/b agonist MRL-C was oxidized by CYP2C8 and to Some in vitro data suggest that CYP2C8 may contrib- a minor extent by CYP3A4, but not by CYP2C9 (Kochansky ute to the metabolism of the steroids 17b-estradiol, et al., 2005). Furthermore, the main elimination pathway progesterone, and testosterone (Waxman et al., 1991; of licofelone, a dual inhibitor of cyclooxygenases 1 and 2 Spink et al., 1992, 1994). However, there seems to be no and 5-lipoxygenase, is glucuronidation of its carboxylic evidence for a role of CYP2C8 in the metabolism of acid metabolite, followed by CYP2C8-catalyzed hydroxyl- androgens in vivo. Although CYP2C8 participates in the ation of the acyl glucuronide M1 to form the hydroxylated metabolism of several endogenous compounds, no car- glucuronide M3 (Albrecht et al., 2008). diovascular or other potentially CYP2C8-related adverse For two compounds, the formation of an unconjugated effects were observed in the Helsinki Heart Study, in hydroxyl metabolite involves oxidation and subsequent which over 2000 middle-aged men with primary dyslipi- deconjugation of a glucuronide metabolite. In vitro, the demia ingested the strong CYP2C8 inhibitor gemfibrozil antidiabetic agent sipoglitazar was first glucuronidated 600 mg twice daily for several years (Frick et al., 1987). to sipoglitazar b-1-O-acyl glucuronide (sipoglitazar-G1). Furthermore, some natural compounds have been Sipoglitazar-G1 was then metabolized to the main metab- reported to undergo metabolism by CYP2C8 in vitro olite M-I by O-dealkylation by CYP2C8 and subsequent (Table 3). CYP2C8 and CYP3A4 are the primary deconjugation (Nishihara et al., 2012). A similar finding was enzymes involved in the in vitro metabolism of recently observed for desloratadine (Kazmi et al., 2015). The 1-hydroxyl-2,3,5-trimethoxyxanthone, a constituent of main metabolite of desloratadine, 3-hydroxydesloratadine, the Tibetan medicinal plant Halenia elliptica (Feng which is active, was formed via CYP2C8-mediated oxida- et al., 2014). CYP2C8 is responsible for the main tion of desloratadine glucuronide and a deconjugation event metabolic pathway of silybin, the active component of (Kazmi et al., 2015). Thus, it seems that phase II metabo- silymarin in vitro (Jancova et al., 2007). The CYP2C8 lism occurs before phase I for these compounds (Fig. 5). inhibitor inhibited silybin O-demethylation Also the glucuronide metabolites of clopidogrel, gem- by 80% in HLM, and recombinant CYP2C8 was the fibrozil, and Lu AA34893 are likely to be substrates of major enzyme forming O-demethyl silybin, with a small CYP2C8 (Ogilvie et al., 2006; Baer et al., 2009; Kazmi contribution by CYP3A4. Furthermore, CYP2C8 is the et al., 2010; Tornio et al., 2014). All three compounds are major enzyme responsible for the in vitro metabolism of metabolism-dependent inhibitors of CYP2C8, as dis- tanshinol borneol ester, a combination of the natural cussed in sections V.B and VI.B. compounds danshensu and borneol (Liu et al., 2010a). Recombinant CYP2C8 generated all five tanshinol B. Endogenous and Natural Compounds borneol metabolites (M1-M5) observed in HLM incuba- CYP2C8 metabolizes some endogenous and natu- tions, whereas recombinant CYP3A4 only produced the ral compounds (Table 3). CYP2C8 participates in M4 metabolite. the metabolism of arachidonic acid to biologically active epoxyeicosatrienoic acids (e.g., 11-, 13-, or 15- IV. Pharmacogenetics hydroxyeicosatrienoic acid), involved in the regulation of numerous physiologic processes, e.g., vascular function, Nearly 100 nonsynonymous single nucleotide varia- pressure regulation, pancreatic peptide hormone tions (SNV) and short deletions, as well as essential secretion, and aggregation (Daikh et al., 1994; splice site variants have been found in the CYP2C8 Rifkind et al., 1995; Zeldin et al., 1995). The roles of gene. The variants described in the literature, dbSNP CYP2C8 and other CYP enzymes in inflammation, database, or the 1000 Genomes project database are cardiovascular disease, and cancer were recently listed in Table 5, together with their continental reviewed by Chen and Wang (2015) and Fleming (2014). frequencies and predicted or experimentally deter- CYP2C8 and CYP3A enzymes have generally been mined effects on protein function. The vast majority of considered to be the primary CYPs involved in the the nonsynonymous variants are rare and occur at metabolism of all-trans-retinoic acid, the active form of minor allele frequencies of 0.01 or less in all investi- vitamin A (retinol) (Leo et al., 1989; Nadin and Murray, gated populations. 1999; Marill et al., 2000). All-trans-retinoic acid is involved in gene transcription, cell division, and differ- A. Population Genetics entiation (Tzimas and Nau, 2001; Marill et al., 2003; Three alleles, known as CYP2C8*2, *3, and *4, Duester, 2008). According to more recent data, however, account for the majority of nonsynonymous variability CYP26A1 and CYP3A4 are the primary determinants of of CYP2C8 in humans. Their frequencies differ signif- all-trans-retinoic acid metabolism in humans, whereas icantly both between and within continental popula- the role of CYP2C8 is of smaller importance (Thatcher tions (Table 5, Fig. 6). et al., 2010). In vitro, CYP2C8 also catalyzes the The CYP2C8*2 allele (c.805A.T, p.Cys266Phe) oc- metabolism of other retinoids, including 9-cis-retinoic curs mostly in individuals with a sub-Saharan African TABLE 5. 188 Nonsynonymous and essential splice site nucleotide changes in the CYP2C8 gene Nucleotide positions are given in relation to the full length CYP2C8 mRNA sequence (NM_000770.3) and amino acid positions in relation to the respective protein sequence (NP_000761.3). Variant allele frequency data are from the 1000 genomes project for populations with African, European, South Asian, East Asian and American ancestry (www.1000genomes.org, 1000 Genomes Project Consortium, 2012). SIFT and PolyPhen predictions of the possible impact of amino acid substitutions on protein function were obtained using the Variant Effect Predictor (Kumar et al., 2009; Adzhubei et al., 2010; McLaren et al., 2010). *-allele nomenclature was retrieved from the Human Cytochrome P450 (CYP) Allele Nomenclature Database (www.cypalleles.ki.se).

Enzyme Activity

*-allele dbSNP ID Location Nucleotide Change Amino Acid Change Variant Allele Frequency In Silico Prediction

African European South Asian East Asian American SIFT PolyPhen In Vivo In Vitro rs142470035 Exon 1 c.1A.G p.Met1? 0.0045 0 0 0 0 deleterious probably damaging rs373001219 Exon 1 c.7C.A p.Pro3Thr —— — — —tolerated benign rs530027098 Exon 1 c.40A.T p.Met14Leu 0 0 0.001 0 0 tolerated benign rs202131138 Exon 1 c.63A.T p.Arg21Ser —— — — —deleterious benign rs267602643 Exon 1 c.77G.A p.Arg26Lys —— — — —tolerated benign rs375170154 Exon 1 c.149T.C p.Ile50Thr —— — — —tolerated benign rs113939225 Exon 1 c.167A.G p.Asn56Ser —— — — —tolerated benign ↔ rs376132046 Exon 2 c.199G.A p.Val67Met —— — — —deleterious possibly damaging rs17851796 Exon 2 c.244G.A p.Ala82Thr 0 0 0 0.001 0 deleterious benign rs17851796 Exon 2 c.244G.T p.Ala82Ser 0 0 0 0.001 0 deleterious benign rs201449274 Exon 2 c.263T.C p.Ile88Thr 0 0 0 0 0.0014 deleterious benign rs372299895 Exon 2 c.268A.G p.Asn90Asp —— — — —tolerated benign rs578254206 Exon 2 c.293G.T p.Gly98Val 0 0 0.001 0 0 deleterious probably damaging rs199931273 Intron 2 c.331+2T.C —— — — — — — rs369552457 Exon 3 c.345C.A p.Ser115Arg —— — — —deleterious probably damaging rs201739495 Exon 3 c.368T.A p.Ile123Asn —— — — —deleterious possibly damaging rs377386087 Exon 3 c.370C.T p.Arg124Trp —— — — —deleterious probably damaging aka tal. et Backman rs369591911 Exon 3 c.371G.A p.Arg124Gln —— — — —deleterious probably damaging rs188111115 Exon 3 c.373C.T p.Arg125Cys 0.0008 0 0 0 0 deleterious possibly damaging rs139650638 Exon 3 c.389C.A p.Thr130Asn 0 0 0.001 0 0 deleterious possibly damaging rs139650638 Exon 3 c.389C.T p.Thr130Ile 0 0 0.001 0 0 tolerated probably damaging *3 rs11572080 Exon 3 c.416G.A p.Arg139Lys 0.0083 0.1183 0.0297 0.001 0.0994 tolerated benign ↑↓↑ rs540288649 Exon 3 c.430C.G p.Arg144Gly 0 0 0.0102 0 0 deleterious probably damaging rs200057634 Exon 3 c.449A.G p.His150Arg —— — — —tolerated benign rs201561213 Exon 3 c.472A.G p.Lys158Glu —— ———deleterious benign *5 rs72558196 Exon 3 c.475delA p.Thr159ProfsTer19 —— — — — — — none rs576554998 Exon 4 c.497C.T p.Pro166Leu —— — — —deleterious probably damaging *6 rs142886225 Exon 4 c.511G.A p.Gly171Ser 0 0 0 0.006 0 tolerated benign ↔ rs553407481 Exon 4 c.516T.A p.Cys172Ter —— — — — — — rs141350682 Exon 4 c.525C.A p.Cys175Ter —— — — — — — rs113008582 Exon 4 c.526A.G p.Asn176Asp —— — — —deleterious probably damaging rs201219972 Exon 4 c.536G.C p.Cys179Ser —— — — —tolerated possibly damaging rs41286886 Exon 4 c.541G.A p.Val181Ile 0 0.0109 0 0 0.0029 tolerated benign rs147150224 Exon 4 c.544G.A p.Val182Ile —— — — —tolerated benign *7 rs72558195 Exon 4 c.556C.T p.Arg186Ter —— — — — — — none *8 rs543793530 Exon 4 c.557G.A p.Arg186Gln 0 0 0.001 0 0 deleterious probably damaging ↓ rs201899315 Exon 4 c.581T.A p.Phe194Tyr —— — — —deleterious benign rs201045618 Exon 4 c.602T.A p.Phe201Tyr 0 0 0 0.001 0 deleterious possibly damaging rs146962089 Exon 4 c.635G.A p.Trp212Ter 0.0023 0 0 0 0 —— rs148974310 Exon 5 c.643G.A p.Val215Ile —— — — —tolerated benign *13 N.A. Exon 5 c.669T.G p.Ile223Met —— — — —tolerated benign ↔ rs569886323 Exon 5 c.703A.G p.Lys235Glu 0 0 0 0.001 0 tolerated benign *14 rs188934928 Exon 5 c.712G.C p.Ala238Pro 0 0 0 0.001 0 tolerated benign ↓ rs537006401 Exon 5 c.713C.T p.Ala238Val —— — — —tolerated benign rs200358471 Exon 5 c.716T.C p.Leu239Pro —— — — —tolerated benign rs536085663 Exon 5 c.721C.T p.Arg241Ter 0.0008 0 0 0 0 —— rs11572102 Exon 5 c.730A.G p.Ile244Val 0.0068 0 0 0 0 tolerated benign *9 N.A. Exon 5 c.740A.G p.Lys247Arg —— — — —tolerated benign ↔ (continued) TABLE 5.—Continued

Enzyme Activity

*-allele dbSNP ID Location Nucleotide Change Amino Acid Change Variant Allele Frequency In Silico Prediction

African European South Asian East Asian American SIFT PolyPhen In Vivo In Vitro rs141120323 Exon 5 c.767A.G p.Asp256Gly —— — — —deleterious probably damaging rs527793637 Exon 5 c.781C.T p.Arg261Trp 0 0 0.001 0 0 deleterious benign rs370459834 Exon 5 c.782G.T p.Arg261Leu —— — — —deleterious benign *4 rs1058930 Exon 5 c.792C.G p.Ile264Met 0.0038 0.0577 0.0072 0 0.0187 deleterious probably damaging ↓↑ rs551515028 Exon 5 c.793G.A p.Asp265Asn 0.0008 0 0 0 0 deleterious probably damaging rs377675927 Exon 5 c.797G.T p.Cys266Phe —— — — —deleterious probably damaging *2 rs11572103 Exon 5 c.805A.T p.Ile269Phe 0.1891 0.004 0.0123 0 0.0115 deleterious probably damaging ↓↔ rs373613215 Exon 5 c.816G.C p.Glu272Asp —— — — —tolerated benign *11 rs78637571 Exon 6 c.820G.T p.Glu274Ter 0 0 0 0.003 0 —— none rs140599093 Exon 6 c.821A.G p.Glu274Gly —— — — —deleterious benign rs370806022 Exon 6 c.848A.G p.Asn283Ser —— — — —tolerated benign . rs537326361 Exon 6 c.955G A p.Val319Ile 0 0 0 0 0.0014 tolerated benign Interactions and Metabolism Drug in CYP2C8 of Role rs146806199 Exon 7 c.992T.C p.Ile331Thr 0.0015 0 0.0041 0 0 deleterious probably damaging rs148442781 Exon 7 c.1028G.T p.Ser343Ile —— — — —tolerated benign rs199691080 Exon 7 c.1060G.A p.Glu354Lys —— — — —deleterious probably damaging rs373461548 Exon 7 c.1063A.T p.Ile355Phe —— — — —deleterious probably damaging rs45438799 Exon 7 c.1081C.T p.Leu361Phe 0 0.001 0 0 0 tolerated possibly damaging rs77147096 Exon 7 c.1093G.A p.Gly365Ser 0.0098 0 0 0 0 tolerated benign rs147133669 Exon 7 c.1096G.A p.Val366Met 0.0008 0.001 0 0 0.0014 deleterious benign rs375271607 Exon 7 c.1144C.T p.Pro382Ser —— — — —deleterious probably damaging *10 N.A. Exon 7 c.1149G.T p.Lys383Asn —— — — —deleterious probably damaging ↔ rs143386810 Exon 8 c.1150G.A p.Gly384Ser 0 0.001 0.001 0 0 deleterious possibly damaging rs553009747 Exon 8 c.1154C.T p.Thr385Ile 0 0 0.001 0 0 deleterious probably damaging rs267602641 Exon 8 c.1165G.A p.Ala389Thr —— — — —tolerated benign rs72558194 Exon 8 c.1169T.C p.Leu390Ser —— — — —tolerated benign rs74454169 Exon 8 c.1171C.A p.Leu391Met —— — — —deleterious probably damaging rs201421851 Exon 8 c.1178C.G p.Ser393Cys 0 0 0 0 0.0014 deleterious probably damaging rs190807911 Exon 8 c.1180G.A p.Val394Met 0 0 0 0.001 0 deleterious probably damaging rs201301235 Exon 8 c.1187A.C p.His396Pro —— — — —deleterious possibly damaging rs186285658 Exon 8 c.1189G.A p.Asp397Asn 0 0 0 0.002 0 deleterious possibly damaging rs113669182 Exon 8 c.1193A.G p.Asp398Gly —— — — —tolerated benign *3 rs10509681 Exon 8 c.1196A.G p.Lys399Arg 0.0083 0.1183 0.0297 0.001 0.0994 tolerated benign ↑↓↑ rs181982392 Exon 8 c.1198G.T p.Glu400Ter 0 0 0 0.001 0 —— rs66501115 Exon 8 c.1210C.G p.Pro404Ala —— — — —deleterious possibly damaging ↓ rs150733212 Exon 8 c.1225C.T p.Pro409Ser —— — — —deleterious probably damaging rs374605743 Exon 8 c.1246A.C p.Asn416His —— — — —deleterious benign rs141209951 Exon 8 c.1250G.T p.Gly417Val 0.0015 0 0 0 0 deleterious probably damaging rs552247471 Exon 8 c.1252A.T p.Asn418Tyr 0 0 0.001 0 0 deleterious probably damaging rs371330493 Exon 8 c.1273T.C p.Phe425Leu 0.0008 0 0 0 0 deleterious probably damaging rs148348784 Exon 8 c.1276A.G p.Met426Val —— — — —tolerated benign ↔ rs372999683 Exon 9 c.1313A.G p.Glu438Gly —— — — —deleterious probably damaging rs143038562 Exon 9 c.1324C.T p.Arg442Cys —— — — —deleterious possibly damaging rs138495387 Exon 9 c.1325G.A p.Arg442His —— — — —deleterious possibly damaging rs369600584 Exon 9 c.1327A.C p.Met443Leu —— — — —deleterious benign *12 rs3832694 Exon 9 c.1382_1384delTTG p.Val461del —— — — — — — rs61757318 Exon 9 c.1413delA p.Val472LeufsTer23 —— — — — — — ↔ rs529725725 Exon 9 c.1414G.A p.Val472Ile 0 0 0 0 0.0029 tolerated benign rs376016142 Exon 9 c.1441C.T p.Pro481Ser —— — — —deleterious probably damaging rs140481138 Exon 9 c.1466C.T p.Pro489Leu —— — — —deleterious probably damaging

↔, unchanged activity; ↓, reduced activity; ↑, increased activity; N.A., not available; SIFT, sorting intolerant from tolerant 189 190 Backman et al.

Fig. 6. Global distribution of CYP2C8*2, CYP2C8*3, and CYP2C8*4 alleles. Color intensity indicates allele frequency. References are given in the text. ancestry. In sub-Saharan African populations, its allele (Martis et al., 2013). The allele is also relatively frequency ranges from about 0.10 in a Fulani population common in the mixed Brazilian population with a inBurkinaFasoto0.37inaMbutipygmypopulationin frequency of 0.06, New York area Hispanic population Congo (Cavaco et al., 2005; Rower et al., 2005; Parikh with a frequency of 0.02, and North and South Indian et al., 2007; Kudzi et al., 2009; Speed et al., 2009; populations, with frequencies of 0.03 and 0.01, respec- Paganotti et al., 2011, 2012; 1000 Genomes Project tively (Suarez-Kurtz et al., 2012; Martis et al., 2013; Consortium, 2012; Arnaldo et al., 2013; Marwa et al., Minhas et al., 2013; Arun Kumar et al., 2015). The 2014). In an African-American population in the New CYP2C8*2 allele is very rare or absent in East Asian York area, the allele frequency of CYP2C8*2 was 0.10 and European populations, with the exception of an Role of CYP2C8 in Drug Metabolism and Interactions 191 allele frequency of 0.01 in a Portuguese European Muthiah et al., 2005; Speed et al., 2009; 1000 Genomes sample (Nakajima et al., 2003; Muthiah et al., 2005; Project Consortium, 2012; Staehli Hodel et al., 2013; Cavaco et al., 2006; Pechandova et al., 2012; Vargens Wu et al., 2013). The allele is rare in individuals with et al., 2012; Martis et al., 2013; Wu et al., 2013). a sub-Saharan African ancestry, with a frequency of The CYP2C8*3 allele is a haplotype consisting of below 0.01 in all investigated sub-Saharan African two nonsynonymous variants (c.416G.A, p.Arg139Lys populations and 0.01 in an African American population and c.1196A.G, p.Lys399Arg), which appear to be in (Cavaco et al., 2005; Rower et al., 2005; Kudzi et al., a complete or nearly complete linkage disequilibrium 2009; 1000 Genomes Project Consortium, 2012; Arnaldo in all investigated populations (1000 Genomes Project et al., 2013; Martis et al., 2013). Consortium, 2012). The linkage disequilibrium extends In addition to the common variants, rare nonsynon- also beyond the CYP2C8 gene, as evidenced by a strong ymous CYP2C8 variants exist in all continental pop- correlation between the CYP2C8*3 and CYP2C9*2 ulations (Table 5). A number of the rare CYP2C8 (rs1799853; c.430C.T, p.Arg144Cys) alleles in the variants can be predicted to result in a loss-of-function Swedish population (Yasar et al., 2002). The highest because of premature termination of protein syn- allele frequencies of CYP2C8*3 are seen in individuals thesis. The c.635G.A (p.Trp212Ter) and c.721C.T with a European ancestry. In European populations, (p.Arg241Ter) variants have a combined allele frequency the allele frequency of CYP2C8*3 ranges from 0.069 in of 0.003 in sub-Saharan African populations, and the Faroe Islanders to 0.198 in a Portuguese population, c.820G.T (p.Glu274Ter) and c.1198G.T (p.Glu400Ter) with an apparent north-to-south cline from lower to variants have a combined allele frequency of 0.004 in higher frequencies (Fig. 6; Yasar et al., 2002; Halling East Asians (1000 Genomes Project Consortium, 2012). et al., 2005; Cavaco et al., 2006; Speed et al., 2009; 1000 Other predicted loss-of-function CYP2C8 variants were Genomes Project Consortium, 2012; Pechandova et al., not found in the 1000 Genomes Project populations, 2012; Suarez-Kurtz et al., 2012). The allele is also quite and data are too scarce to estimate their population common in European American and North American frequencies. Hispanic populations, with frequencies of 0.09 and 0.08, respectively (Martis et al., 2013). In the mixed Brazilian B. Functional Studies and Ecuadorian populations, its frequency is 0.08 and The functional effects of CYP2C8 variants have been 0.07, and in a Chilean mestizo population it is 0.06 investigated using recombinantly expressed variant (Roco et al., 2012; Suarez-Kurtz et al., 2012; Vicente proteins and HLM with different CYP2C8 genotypes. et al., 2014). There is wide variability in the frequency Recombinant CYP2C8.2 has been quite consistently of CYP2C8*3 in sub-Saharan African populations, even associated with an about 50% decrease in the intrinsic within a country (Cavaco et al., 2005; Rower et al., 2005; clearance for paclitaxel 6a-hydroxylation, compared Parikh et al., 2007; Kudzi et al., 2009; Arnaldo et al., with CYP2C8.1 (Dai et al., 2001; Gao et al., 2010; 2013; Staehli Hodel et al., 2013; Marwa et al., 2014; Yu et al., 2013b). In addition, the intrinsic clearance Paganotti et al., 2014). For example, the frequency of of amodiaquine has been reduced by 80–90% in CYP2C8*3 was found to be 0.00 in individuals in central CYP2C8.2 and that of repaglinide by 20% compared Tanzania and as high as 0.10 in the Mwanza region of with CYP2C8.1 (Parikh et al., 2007; Yu et al., 2013b). Tanzania (Staehli Hodel et al., 2013; Marwa et al., Similarly, the intrinsic clearances of arachidonic acid 2014). and tanshinol borneol ester appeared to be lower by The CYP2C8*4 (c.792C.G, p.Ile264Met) allele has CYP2C8.2 than by CYP2C8.1, but the differences were its highest frequencies in European populations, with not statistically significant (Dai et al., 2001; Liu et al., the allele frequency ranging from 0.04 in a Spanish 2010a). On the other hand, the intrinsic clearances for population to 0.07 in the Irish (Cavaco et al., 2006; cerivastatin M-23 and M-1 metabolite formation and R- Speed et al., 2009; 1000 Genomes Project Consortium, and S-ibuprofen hydroxylations have been nonsignificantly 2012; Pechandova et al., 2012). Its frequency was 0.03 higher in CYP2C8.2 than in CYP2C8.1 (Kaspera et al., in a European American population and a mixed Bra- 2010; Yu et al., 2013b). Both the SIFT or Polyphen in silico zilian population (Suarez-Kurtz et al., 2012; Martis prediction algorithms suggest that the amino acid change in et al., 2013). In Peruvian, Colombian, Puerto Rican, and CYP2C8.2 is deleterious for CYP2C8 activity (Table 5). North American Hispanic populations, the frequency In several studies, the intrinsic clearance for pacli- ranges from 0.01 to 0.02 (1000 Genomes Project Con- taxel 6a-hydroxylation by recombinant CYP2C8.3 has sortium, 2012; Martis et al., 2013). The CYP2C8*4 allele been between 30 and 85% lower than by CYP2C8.1 (Dai is found with a frequency of about 0.03–0.04 in Indian et al., 2001; Soyama et al., 2001; Gao et al., 2010; Yu individuals and 0.01 in the Pakistani (1000 Genomes et al., 2013b). Other studies employing recombinant Project Consortium, 2012; Minhas et al., 2013). In East CYP2C8.3 have shown increased intrinsic clearance Asian populations, the frequency of CYP2C8*4 is for repaglinide and cerivastatin but reduced intrinsic generally 0.01 or less, but a frequency of 0.02 was seen clearance for R- and S-ibuprofen and nearly abolished in an Uighur Chinese population (Nakajima et al., 2003; intrinsic clearance for amodiaquine (Kaspera et al., 192 Backman et al.

2010; Parikh et al., 2007; Yu et al., 2013b). The intrinsic et al., 2005; Hanioka et al., 2010). In one study, the clearances of arachidonic acid to 11,12- and 14,15- p.Ala238Pro and p.Ile223Met variants were associated and tanshinol borneol ester with reduced amiodarone metabolism (Hanioka et al., have also been significantly lower by CYP2C8.3 than by 2011). One study demonstrated lack of CYP2C8 protein CYP2C8.1 (Dai et al., 2001; Liu et al., 2010a). However, expression in association with the p.Glu274Ter (*11) in a recent study expressing CYP2C8.3 together with nonsense variant (Yeo et al., 2011). cytochrome P450 reductase and cytochrome b5, the intrinsic clearance for paclitaxel 6a-hydroxylation was C. Effects on Drug Metabolism in Humans about twofold higher, that for amodiaquine about two- In contrast to previous in vitro studies suggesting fold higher, that for rosiglitazone about 2.5-fold higher, a reduced CYP2C8 activity in association with the and that for cerivastatin about 4.5-fold higher compared CYP2C8*3 allele (Dai et al., 2001; Bahadur et al., with CYP2C8.1 (Kaspera et al., 2011). A stronger 2002), the first pharmacokinetic study in humans binding affinity of ligands to CYP2C8.3 together with an showed that the CYP2C8*3 allele was associated with increase in heme spin change during binding of ligands reduced plasma concentrations of repaglinide (Niemi and redox partners were suggested to partly explain et al., 2003c). In this and later studies, individuals with the increased catalytic activity (Kaspera et al., 2011). the CYP2C8*1/*3 genotype have had an approximately In one study, HLM heterozygous for the CYP2C8*3 40–50% lower AUC of a subtherapeutic dose of repagli- allele showed lowered paclitaxel 6a-hydroxylase activ- nide than individuals with the CYP2C8*1/*1 genotype ity and in another study no change in amodiaquine (Niemi et al., 2005b,c). However, this finding has not N-deethylation compared with microsomes homozy- been fully replicated in studies with higher repaglinide gous for CYP2C8*1 (Bahadur et al., 2002; Kaspera doses (Bidstrup et al., 2006; Tomalik-Scharte et al., et al., 2011). Studies employing HLM heterozygous or 2011), suggesting that the effect of CYP2C8*3 allele on homozygous for CYP2C8*3 have shown increased in- repaglinide pharmacokinetics may be dose dependent. trinsic clearance of pioglitazone and imatinib (Muschler Similarly to repaglinide, the CYP2C8*3 allele has et al., 2009; Khan et al., 2015). In silico predictions been associated with apparently increased clearance of suggest that neither of the amino acid changes in the rosiglitazone and pioglitazone CYP2C8.3 affect CYP2C8 activity (Table 5). Taken (Kirchheiner et al., 2006; Aquilante et al., 2008, 2013a; together, in vitro evidence concerning the functional Tornio et al., 2008b). The AUCs of rosiglitazone or pioglit- effects of CYP2C8*3 suggests some degree of a azone have been about 20–40% lower in CYP2C8*3 carriers substrate-specific effect but is discrepant for some than in noncarriers, with an apparent gene-dose effect substrates in that both decreased and increased activ- (Kirchheiner et al., 2006; Aquilante et al., 2008, 2013a; ities have been reported. Tornio et al., 2008b). Furthermore, in a study in patients In three studies, paclitaxel 6a-hydroxylation intrinsic with type 2 diabetes mellitus, the CYP2C8*3 allele has clearance was reduced by about 70% by recombinant been associated with significantly lower trough rosigli- CYP2C8.4 compared with CYP2C8.1 (Singh et al., 2008; tazone concentrations and an impaired lowering of Gao et al., 2010; Yu et al., 2013b). Similarly, the intrinsic glycosylated hemoglobin (HbA1c) during rosiglitazone clearances of repaglinide and R-andS-ibuprofen have treatment (Stage et al., 2013). In one study in African been 20, 50, and 53% lower by CYP2C8.4 than by American subjects, the CYP2C8*2 allele had no impact CYP2C8.1, respectively (Yu et al., 2013b). On the other on parent pioglitazone pharmacokinetics but was asso- hand, the intrinsic clearances of cerivastatin to M-23 and ciated with impaired metabolism of pioglitazone to its M-1 were about 2- to 2.5-fold higher by CYP2C8.4 than by M3 metabolite (Aquilante et al., 2013c). CYP2C8.1 (Kaspera et al., 2010). The intrinsic clearance Although CYP2C8*2 has been associated with signif- of tanshinol borneol ester was not significantly different icantly impaired amodiaquine metabolism in vitro between CYP2C8.4 and CYP2C8.1 (Liu et al., 2010a). One (Parikh et al., 2007), the allele has not been clearly study suggests that the amino acid change in CYP2C8.4 associated with amodiaquine efficacy or toxicity (Adjei disrupts heme binding and results in an inactive protein et al., 2008). However, more recent evidence suggests that (Singh et al., 2008). HLM heterozygous for CYP2C8*4 CYP2C8 genetic variability can influence the occurrence showed a nonsignificant tendency for lower paclitaxel of amodiaquine or chloroquine resistance in malaria 6a-hydroxylase activity (Bahadur et al., 2002). In silico parasites (Paganotti et al., 2011; Cavaco et al., 2013). predictions suggest that the amino acid change in Studies in cancer patients have suggested that the CYP2C8.4 is deleterious for CYP2C8 activity (Table 5). CYP2C8*3 allele can slightly impair the clearance of In vitro studies employing recombinant CYP2C8 paclitaxel (Henningsson et al., 2005; Bergmann et al., have shown reduced paclitaxel 6a-hydroxylase activity 2011). Some studies have also suggested that the in association with the p.Arg186Gln, p.Ala238Pro (*14), CYP2C8*3 allele or other CYP2C8 variants may be risk and p.Pro404Ala variants but no change in activity due factors for paclitaxel-induced neurotoxicity or myelo- to the p.Gly171Ser, p.Ile223Met (*13), p.Lys247Arg, suppression and affect the benefit-to-risk ratio of and p.Lys383Asn variants (Soyama et al., 2001; Hichiya paclitaxel therapy (Green et al., 2011; Leskelä et al., Role of CYP2C8 in Drug Metabolism and Interactions 193

2011; Hertz et al., 2012; Hertz et al., 2014; Lee et al., neither montelukast nor affected the phar- 2015). Further studies are required to clarify the role macokinetics of CYP2C8 substrate drugs in vivo of CYP2C8 genetic variants in affecting paclitaxel (Jaakkola et al., 2006b; Kajosaari et al., 2006b; Kim response. et al., 2007). The lack of in vivo effect is likely explained Some studies have reported significantly increased by their extensive (.99%) plasma concentrations and apparently reduced clear- (FDA, 1998; Dekhuijzen and Koopmans, 2002). Also ance of racemic ibuprofen and its enantiomers in the inhibition of CYP2C8 by candesartran cilexetil association with the CYP2C8*3 allele (Garcia-Martin (prodrug of candesartan), clotrimazole, and mometa- et al., 2004; Martinez et al., 2005; Karazniewicz-Łada sone furoate are probably not clinically relevant. The et al., 2009). On the other hand, one study reported antifungal clotrimazole and anti-inflammatory mome- enhanced clearance of R-ibuprofen in association with tasone furoate are topically applied and are therefore CYP2C8*3 (Lopez-Rodriguez et al., 2008). Because unlikely to cause interactions because of low systemic ibuprofen is a substrate of CYP2C9, it is likely that concentrations (Walsky et al., 2005a). In the systemic the discrepancies are due to the strong linkage disequi- circulation, candesartan cilexetil is cleaved to candesartan, librium between CYP2C8*3 and CYP2C9*2 and re- and, consequently, the likelihood of a drug interaction duced ibuprofen clearance in CYP2C8*3 carriers is in elicited by its prodrug is low. Furthermore, predictions fact due to the CYP2C9*2 allele. suggested a relatively weak potential for drug-drug Although CYP2C8 is not known to be involved in interactions due to CYP2C8 inhibition by felodipine. No pharmacokinetics, an intronic SNV in drug interaction studies between felodipine and CYP2C8 (rs1934951) has been associated with zole- CYP2C8 substrates have been reported. dronic acid-induced osteonecrosis of the jaw in patients Trimethoprim, an antimicrobial agent, is a competi- treated for multiple myeloma (Sarasquete et al., 2008). tive inhibitor of CYP2C8 in vitro (Wen et al., 2002), with In a more recent study, this SNV was associated with a Ki value typically around 10–30 mM in HLM (Table 6). the mandibular localization of bisphosphonate-induced The inhibition of CYP2C8 by trimethoprim seems to be osteonecrosis (Balla et al., 2012). However, there was no rather selective, because it does not inhibit CYP1A2, significant relationship between the variant and the CYP2C9, CYP2C19, CYP2D6, CYP2E1, or CYP3A4 at development of bisphosphonate-induced osteonecrosis concentrations below 100 mM. In healthy subjects, tri- of the jaw in men with prostate cancer (English et al., methoprim has moderately increased the plasma expo- 2010) or in patients with multiple myeloma (Such et al., sure to several CYP2C8 substrate drugs (section VI.). 2011). A meta-analysis found no significantly increased The quercetin is one of the earliest in vitro susceptibility to bisphosphonate-induced osteonecrosis inhibitors of CYP2C8 detected. In studies of paclitaxel of the jaw in rs1934951 carriers when all cancer types metabolism, it was observed that quercetin, unlike were pooled, but suggested a significant association in CYP3A4 inhibitors, inhibited paclitaxel 6a-hydroxylation multiple myeloma patients (Zhong et al., 2013). (Harris et al., 1994; Kumar et al., 1994). Because it was shown that the 6a-hydroxylation of paclitaxel is V. In Vitro Inhibition and Induction of mediated by CYP2C8, it was evident that quercetin is Cytochrome P450 2C8 an inhibitor of this enzyme (Rahman et al., 1994). Quercetin inhibits CYP2C8 competitively with a Ki of A. Reversible Inhibition 0.03–20 mM (Table 7) and is classified as a "preferred" 1. Drugs That Act as Inhibitors of Cytochrome P450 probe in vitro inhibitor of CYP2C8 by the FDA (http:// 2C8. Several drugs, drug metabolites, and other com- www.fda.gov/Drugs/DevelopmentApprovalProcess/ pounds have been found to inhibit CYP2C8 activity DevelopmentResources/DrugInteractionsLabeling/ reversibly in vitro (Tables 6 and 7). In an in vitro ucm093664.htm). However, quercetin is not selective screening of 209 commonly used drugs, 48 compounds for CYP2C8; it also inhibits CYP1A2, CYP2C9, exhibited greater than 50% inhibition of recombinant CYP2C19, CYP2D6, and CYP3A4 with IC50 values of CYP2C8 activity at an inhibitor concentration of 30 mM 3.1–47 mM (Obach, 2000; Zou et al., 2002). In vivo, (Walsky et al., 2005a). Montelukast, candesartan cilex- quercetin at steady state did not affect the pharmaco- etil, zafirlukast, clotrimazole, felodipine, and mometa- kinetics of rosiglitazone (Kim et al., 2005a). sone furoate inhibited CYP2C8 with concentrations The lipid-lowering drug gemfibrozil is a moderate, supporting half of the maximal inhibition (IC50)of direct competitive inhibitor of CYP2C8 in vitro (Wen #3 mM in recombinant CYP2C8 and HLM. In another et al., 2001; Prueksaritanont et al., 2002; Wang et al., study, the inhibition of CYP2C8 by montelukast was 2002), with a Ki range between 9.3 and 270 mM found to be competitive and selective, with reversible (Table 6). Gemfibrozil also inhibits CYP2C9 and inhibition constants (Ki) ranging from 0.0092 to 0.15 mM, CYP2C19 with Ki values of 5.8 and 24 mM, respectively, depending on the protein concentration used in the and CYP1A2 with a Ki of 82 mM (Wen et al., 2001). incubation (Walsky et al., 2005b). However, despite Moreover, it inhibits several drug transporters in vitro, their strong inhibitory effect on CYP2C8 in vitro, most notably OATP1B1 (lowest reported Ki =4mM) TABLE 6 194 Drugs, drug metabolites, and some other compounds that act as reversible CYP2C8 inhibitors

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References

mM(mg/ml) mM(mg/ml) mM 2-Oxo-clopidogrel Drug metabolite 33.9 rCYP2C8 DBF Hagihara et al., 2008 4.1 rCYP2C8 Ceri-1 Floyd et al., 2012 4.2 rCYP2C8 Ceri-23 Floyd et al., 2012 32.0 HLM Amo Tornio et al., 2014 2,4-Dichloroaniline Drug metabolite 99.4 HLM Rosi-OH Wu et al., 2014 4-Hydroxyospemifene Drug metabolite 27.7 HLM Amo FDA, 2013i; Turpeinen et al., 2013 4’-Hydroxyospemifene Drug metabolite 7 HLM Amo FDA, 2013i; Turpeinen et al., 2013 7-Epi-paclitaxel Paclitaxel epimer 2.1 HLM Pacli Zhang et al., 2009b 7-O-succinyl macrolactin A Antibiotic 20.5 HLM Rosi-OH Bae et al., 2014 17b-Estradiol (estradiol) Hormonal replacement 21.5 rCYP2C8 Amo Walsky et al., 2005a therapy 6.6 Competitive HLM Amo VandenBrink et al., 2011 23.8 Competitive HLM Monte VandenBrink et al., 2011 17.7 Competitive HLM Pacli VandenBrink et al., 2011 8.9 Competitive HLM Repa VandenBrink et al., 2011 23.8 Competitive HLM Rosi VandenBrink et al., 2011 19 HLM Amo Nirogi et al., 2014 Abiraterone Anticancer, CYP17A1 1.6 HLM n/a 0.65 0.002 0.81 ,0.01 EMA, 2012a inhibitor

Abiraterone acetate Anticancer, CYP17A1 1.3 HLM n/a EMA, 2012a al. et Backman inhibitor Acotiamide (Z-338) Antidyspeptic, 121 Competitive HLM DBF Furuta et al., 2004, acetylcholinesterase inhibitor Afatinib Anticancer, PKI 94.83 HLM Pacli 0.078 0.428 ,0.01 ,0.01 Wang et al., 2014a Alisertib (MLN8237) Anticancer, PKI 16.3 n/a n/a Pusalkar et al., 2014 Alitretinoin (9-cis-retinoic Anticancer, retinoid 17.6 20.2 HLM Taza 0.28 0.01 Attar et al., 2003 acid) Amlodipine Antihypertensive, CCB 10.7 rCYP2C8 Amo 0.0319 0.07 ,0.01 ,0.01 Walsky et al., 2005a 6.4 rCYP2C8 Ceri-1 0.01 ,0.01 Floyd et al., 2012 4.0 rCYP2C8 Ceri-23 0.02 ,0.01 Floyd et al., 2012 9.4 HLM Amo ,0.01 ,0.01 Nirogi et al., 2014 Amodiaquine Antimalarial 11.7 HLM Monte 0.047 ,0.01 VandenBrink et al., 2011 .100 HLM Pacli ,0.01 VandenBrink et al., 2011 1.9 HLM Repa 0.02 VandenBrink et al., 2011 11.0 HLM Rosi ,0.01 VandenBrink et al., 2011 Anastrozole Anticancer, aromatase 48 10 Competitive HLM Tolbu 0.16 0.60 0.02 0.01 Grimm and Dyroff, 1997 inhibitor Anidulafungin Antifungal 12 HLM Amo 3.07 0.16 0.51 0.08 Damle et al., 2009 Anti-Parkinson, dopamine 1–10 rCYP2C8 DBF 4 ,0.01 8.00 ,0.08 Salminen et al., 2011 agonist Apremilast Antipsoriatic, PDE4 inhibitor 56.1 HLM Pacli 0.764 0.32 0.03 ,0.01 FDA, 2014e Antiviral, protease inhibitor 2.1 n/a n/a 7.66 0.14 3.65 0.51 FDA, 2015b Atorvastatin (acid, parent) Antihyperlipidemic, 38.4 15.9 Mixed HLM Pacli 0.098 0.02 ,0.01 ,0.01 Tornio et al., 2005 HMG-CoA reductase inhibitor 38.4 HLM Pacli ,0.01 ,0.01 Sakaeda et al., 2006 21.9 HLM Amo ,0.01 ,0.01 Jenkins et al., 2011 55.7 rCYP2C8 Fluo ,0.01 ,0.01 Schelleman et al., 2014 Atorvastatin acyl-b-D Drug metabolite 45 HLM Amo Jenkins et al., 2011 glucuronide (G2) (continued) TABLE 6—Continued

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References Atorvastatin lactone Antihyperlipidemic, HMG- 28.8 HLM Pacli Sakaeda et al., 2006 CoA reductase inhibitor Atrazine Pesticide 31.3 HLM Amo Abass et al., 2009 Axitinib Anticancer, PKI 0.5 HLM Pacli 0.16 0.01 0.32 ,0.01 FDA, 2012f 0.11 HLM Amo 2.90 0.03 Filppula et al., 2014 0.17 HLM Pacli 0.94 ,0.01 Wang et al., 2014a AZD2624 Antipsychotic, neurokinin-3 83.3 HLM Amo 0.7 0.02 Li et al., 2010 receptor and tachykinin receptor 3 antagonist Azilsartan medoxomil Antihypertensive, prodrug 3.5 HLM Pacli FDA, 2011f Belinostat Anticancer, histone 100 HLM n/a 100 0.071 2.00 0.14 FDA, 2014c deacetylase inhibitor Belinostat 3-ASBA (M24) Drug metabolite 49.1 HLM n/a FDA, 2014c Belinostat acid (M26) Drug metabolite 22.1 HLM n/a FDA, 2014c Belinostat amide Drug metabolite 30.8 HLM n/a FDA, 2014c Interactions and Metabolism Drug in CYP2C8 of Role Belinostat PX106507 Drug metabolite 13.8 HLM n/a FDA, 2014c Benzbromarone Antihyperuricemic, XO 0.055 Competitive HLM Amo VandenBrink et al., 2011 inhibitor 0.38 Competitive HLM Monte VandenBrink et al., 2011 0.95 Competitive HLM Pacli VandenBrink et al., 2011 0.15 Competitive HLM Repa VandenBrink et al., 2011 0.36 Competitive HLM Rosi VandenBrink et al., 2011 (2)-N-3-benzyl- derivative 34 HLM Pacli Cai et al., 2004 phenobarbital Antihyperlipidemic, PPARa 74 HLM Pacli 39.5 0.05 1.07 0.05 Fujino et al., 2003a agonist 9.7 Competitive HLM Pacli 4.07 0.20 Kajosaari et al., 2005a Bisphenol A Bisphenol 97 Noncompetitive rCYP2C8 Ami Niwa et al., 2000 BTFM gemfibrozil Gemfibrozil analog 13 HLM Amo Jenkins et al., 2011 BTFM gemfibrozil acyl- Gemfibrozil acyl-b-D- 37 HLM Amo Jenkins et al., 2011 b-D-glucuronide glucuronide analog Cabozantinib Anticancer, PKI 5.0 rCYP2C8 n/a 3.27 ,0.003 1.31 ,0.01 FDA, 2012c 6.4 4.6 Noncompetitive HLM Amo 0.71 ,0.01 FDA, 2012c 3.8 4.6 Noncompetitive HLM Amo 0.71 ,0.01 Lacy et al., 2015; Nguyen et al., 2015 Canagliflozin Antidiabetic, SGLT2 75 n/a n/a 7.60 0.017 0.20 ,0.01 FDA, 2013e inhibitor Canagliflozin glucuronide Drug metabolite 64 n/a Amo FDA, 2013e (M7) Candesartan Antihypertensive, ARB 36.2 rCYP2C8 Amo 0.19 0.002 0.01 ,0.01 Walsky et al., 2005a Candesartan cilexetil Antihypertensive, prodrug 0.496 rCYP2C8 Amo 0.41 ,0.01 1.65 0.02 Walsky et al., 2005a 3.04 HLM Amo 0.27 ,0.01 Walsky et al., 2005a Pesticide 34.0 HLM Amo Abass et al., 2009 Antihypertensive 16.6 rCYP2C8 Amo 0.258 0.05 0.03 ,0.01 Walsky et al., 2005a Cefuroxime axetil Antibiotic, prodrug 11.1 rCYP2C8 Amo 19.6 0.67 3.53 2.37 Walsky et al., 2005a Celecoxib Anti-inflammatory, NSAID 15.9 rCYP2C8 Amo 1.85 0.03 0.23 ,0.01 Walsky et al., 2005a 4.9 Competitive HLM Amo 0.38 0.01 VandenBrink et al., 2011 7.9 Competitive HLM Monte 0.23 ,0.01 VandenBrink et al., 2011 54.4 Competitive HLM Pacli 0.03 ,0.01 VandenBrink et al., 2011 3.1 Competitive HLM Repa 0.60 0.02 VandenBrink et al., 2011 5.1 Competitive HLM Rosi 0.36 0.01 VandenBrink et al., 2011 9.9 rCYP2C8 Ceri-1 0.37 0.01 Floyd et al., 2012 5.4 rCYP2C8 Ceri-23 0.69 0.02 Floyd et al., 2012 Ceritinib Anticancer, PKI 0.6d 4.86d HLM Amo 1.81 0.028 0.37 0.01 FDA, 2014i d 0.6 HLM Pacli 6.03 0.17 FDA, 2014i 195 (continued) TABLE 6—Continued 196

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References Cerivastatin (acid, parent) Antihyperlipidemic, HMG- 34.4 HLM Pacli 0.0085 ,0.01 ,0.01 ,0.01 Fujino et al., 2004 CoA reductase inhibitor 30.0 31.7 Mixed HLM Pacli ,0.01 ,0.01 Tornio et al., 2005 29.8 HLM Pacli ,0.01 ,0.01 Sakaeda et al., 2006 4.2 Competitive HLM Amo ,0.01 ,0.01 VandenBrink et al., 2011 4.6 Competitive HLM Monte ,0.01 ,0.01 VandenBrink et al., 2011 77.4 Competitive HLM Pacli ,0.01 ,0.01 VandenBrink et al., 2011 4.4 Competitive HLM Repa ,0.01 ,0.01 VandenBrink et al., 2011 13.4 Competitive HLM Rosi ,0.01 ,0.01 VandenBrink et al., 2011 Cerivastatin lactone Antihyperlipidemic, HMG- 44.3 HLM Pacli Sakaeda et al., 2006 CoA reductase inhibitor Pesticide 22.2 HLM Amo Abass et al., 2009 Antipsychotic 20 HLM Amo 0.470 0.05 0.05 ,0.01 Nirogi et al., 2014 Cimetidine Antiulcerative, H2RA 250 HLM Pacli 12 0.81 0.10 0.08 Monsarrat et al., 1997 Antihyperlipidemic, PPARa 441 HLM Pacli 83.0 0.01 0.38 ,0.01 Fujino et al., 2003a agonist Clofazimine Antilepric 14.1 HLM Pacli Shimokawa et al., 2015 Clopidogrel Antithrombotic, platelet 10.2 rCYP2C8 Amo 0.06 0.02 0.01 ,0.01 Walsky et al., 2005a aggregation inhibitor 33.2 rCYP2C8 DBF ,0.01 ,0.01 Hagihara et al., 2008 2.8 rCYP2C8 Ceri-1 0.04 ,0.01 Floyd et al., 2012 3.4 rCYP2C8 Ceri-23 0.04 ,0.01 Floyd et al., 2012 53.6 HLM Amo ,0.01 ,0.01 Tornio et al., 2014 Clopidogrel carboxylic acid Drug metabolite .50.0 rCYP2C8 DBF Hagihara et al., 2008 107 rCYP2C8 Ceri-1 Floyd et al., 2012 al. et Backman 136 rCYP2C8 Ceri-23 Floyd et al., 2012 Clopidogrel active Drug metabolite .50.0 rCYP2C8 DBF Hagihara et al., 2008 metabolite Clotrimazole Antifungal 2.5 Noncompetitive rCYP2C8 Torse 0.008 ,0.01 Ong et al., 2000 0.725 rCYP2C8 Amo 0.02 Walsky et al., 2005a 0.776 HLM Amo 0.02 Walsky et al., 2005a 0.12 Competitive HLM Amo 0.07 VandenBrink et al., 2011 0.27 Competitive HLM Monte 0.03 VandenBrink et al., 2011 0.22 Competitive HLM Pacli 0.04 VandenBrink et al., 2011 0.19 Competitive HLM Repa 0.04 VandenBrink et al., 2011 1.9 Competitive HLM Rosi ,0.01 VandenBrink et al., 2011 0.803 HLM Pacli 0.02 Lee et al., 2012a Antiviral, pharmacokinetic 30.1 HLM Pacli 2.38 0.03 0.16 ,0.01 FDA, 2012j inhibitor CP-778875 Antihyperlipidemic, PPARa 1.83 HLM Amo Kalgutkar et al., 2013 agonist Cyclosporine Immunosuppressant, 79 rCYP2C8 Pacli 1.11 0.07 0.03 ,0.01 Yoshida et al., 2012 calcineurin inhibitor CYP3cide (PF-04981517) Pharmacokinetic inhibitor 78 HLM Pacli Walsky et al., 2012 Dabrafenib Anticancer, PKI 7.7–8.2 HLM Rosi 2.85 0.003 0.74 ,0.01 Lawrence et al., 2014 Antihyperlipidemic, 1.5 HLM Pacli 9.70 12.93 Derks et al., 2009 cholesterylester transfer protein inhibitor Danazol Hypoestrogenic, 1.95 HLM Pacli 0.21 0.22 Lee et al., 2012a hyperandrogenic, ethisterone derivative Dasabuvir (ABT-333) Antiviral, NSB5 inhibitor ;17 Competitive n/a n/a 2.09 0.005 ;0.25 ,0.01 FDA, 2014k Dasatinib Anticancer, PKI 12 3.6 HLM Pacli 0.13 0.04 0.04 ,0.01 FDA, 2006a 38.6 HLM Pacli ,0.01 ,0.01 Kim et al., 2013b 6.31 HLM Pacli 0.02 ,0.01 Wang et al., 2014a (continued) TABLE 6—Continued

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References N-debutyldronedarone Drug metabolite 24.4 36.6 Noncompetitive HLM Pacli FDA, 2009a (SR35021) N-deethyl sunitinib Drug metabolite 52 HLM Pacli FDA, 2006b (SU12662) N-demethylimatinib Drug metabolite 99 HLM Pacli FDA, 2001 31.3 12.8 Mixed HLM Amo Filppula et al., 2012 N-demethyltoremifene Drug metabolite 2.1 HLM Pacli Kim et al., 2011b Deferasirox , iron chelating 100 HLM Pacli 0.46 0.01 ,0.01 ,0.01 FDA, 2005a agent Deferasirox metabolite Drug metabolite 160 HLM Pacli FDA, 2005a CGP82813A Dexamethasone Anti-inflammatory, 12.0 rCYP2C8 Amo 8.15 1.36 Walsky et al., 2005a glucucorticoid Diclofenac Anti-inflammatory, NSAID 54 HLM Amo 6.28 ,0.005 0.23 ,0.01 Jenkins et al., 2011 Diclofenac acyl glucuronide Drug metabolite 14 HLM Amo Jenkins et al., 2011 Interactions and Metabolism Drug in CYP2C8 of Role Diethyldithiocarbamate Alcoholic detergent 129.5 rCYP2C8 Dia Sai et al., 2000 464.1 rCYP2C8 Phena Sai et al., 2000 Diethylstilbestrol Synthetic estrogen 8.0 Competitive HLM Pacli Qu et al., 2011 Diltiazem Antihypertensive, CCB 25 rCYP2C8 Ceri-1 0.335 0.22 0.03 ,0.01 Floyd et al., 2012 124 rCYP2C8 Ceri-23 ,0.01 ,0.01 Floyd et al., 2012 Doxorubicin Anticancer 2 HLM Pacli 1.63 0.24 1.63 0.39 Monsarrat et al., 1997 64.8 Competitive HLM Pacli 0.03 ,0.01 Bun et al., 2003 90 Noncompetitive HLM Luci 0.02 ,0.01 Masek et al., 2011 Duloxetine Antidepressant, SNRI 180 HLM Pacli 0.079 0.05 ,0.01 ,0.01 FDA, 2008b 60 HLM Amo ,0.01 ,0.01 Paris et al., 2009 Antiviral, NNRTI 4.0 rCYP2C8 Amo 12.6 0.005 6.30 0.03 Parikh et al., 2007 6.05 Competitive rCYP2C8 Amo 2.08 0.01 Xu and Desta, 2013 4.78 Competitive HLM Amo 2.64 0.01 Xu and Desta, 2013 Eltrombopag Antihemorrhagic, c-mpl 24.8 HLM Pacli 29 ,0.01 2.34 0.02 FDA, 2008c receptor agonist Enzalutamide Anticancer, antiandrogen 10 5.5 Mixed HLM Amo 35.7 0.02 6.50 0.13 FDA, 2012k Enzalutamide M1 Drug metabolite 20 HLM Amo FDA, 2012k Enzalutamide M2 Drug metabolite 28 HLM Amo FDA, 2012k Erlotinib Anticancer, PKI 6.17 5.8 Competitive HLM Pacli 6.06 0.10 1.05 0.10 Dong et al., 2011 9.5 HLM Pacli 1.28 0.13 Kim et al., 2013b 4.02 HLM Pacli 1.51 0.15 Wang et al., 2014a Esomeprazole (S- Antiulcerative, PPI 31.0 HLM Amo 4.5 0.05 0.29 0.02 Zvyaga et al., 2012 omeprazole) Ethionamide Antituberculosis 110 HLM Pacli 12.99 0.70 0.24 0.17 Shimokawa et al., 2015 Antiviral, NNRTI 19.6 Noncompetitive HLM Pacli 2.2 0.01 0.11 ,0.01 FDA, 2008a Exemestane Anticancer, aromatase 13.5 rCYP2C8 Amo 0.060 0.10 ,0.01 ,0.01 Walsky et al., 2005a inhibitor Febuxostat Antihyperuricemic, XO 20 n/a n/a 16.78 0.007 0.84 ,0.01 Naik et al., 2012 inhibitor Felodipine Antihypertensive, CCB 0.726 rCYP2C8 Amo 0.0073 0.004 0.02 ,0.01 Walsky et al., 2005a 1.20 HLM Amo 0.01 ,0.01 Walsky et al., 2005a Pesticide 4.3 HLM Amo Abass et al., 2009 Antihyperlipidemic, 288 HLM Pacli 23.83 ,0.01 0.17 ,0.01 Fujino et al., 2003a PPARa agonist 92.6 Competitive HLM Pacli 0.26 ,0.01 Kajosaari et al., 2005a 2.39 rCYP2C8 Amo 19.94 0.20 Walsky et al., 2005a 4.8 rCYP2C8 Fluo 9.93 0.10 Schelleman et al., 2014 Fluoxymesterone Androgenic, 3-oxoandrosten 16 rCYP2C8 Ceri-1 Floyd et al., 2012 (4) derivative

16 rCYP2C8 Ceri-23 Floyd et al., 2012 197 (continued) TABLE 6—Continued 198

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References Anti-inflammatory, 0.58 HLM Pacli 0.00023 0.01 ,0.01 ,0.01 FDA, 2013c glucocorticoid Fluticasone M10 Drug metabolite 80 HLM Pacli FDA, 2013c metabolite Fluvastatin (acid, parent) Antihyperlipidemic, HMG- 20 HLM Pacli 0.461 0.01 0.05 ,0.01 Fischer et al., 1999 CoA reductase inhibitor 36.7 18.9 Mixed HLM Pacli 0.02 ,0.01 Tornio et al., 2005 15.1 rCYP2C8 Amo 0.06 ,0.01 Walsky et al., 2005a 70.2 HLM Pacli 0.01 ,0.01 Sakaeda et al., 2006 Fluvastatin lactone Antihyperlipidemic, HMG- 55.4 HLM Pacli Sakaeda et al., 2006 CoA reductase inhibitor Gefitinib Anticancer, PKI 31.0 HLM Pacli 0.8 0.10 0.05 ,0.01 Kim et al., 2013b 12.3 HLM Amo 0.13 0.01 Filppula et al., 2014 8.69 HLM Pacli 0.09 ,0.01 Wang et al., 2014a Gemfibrozil Antihyperlipidemic, PPARa 49 Hep Ceri-23 100 ,0.03 4.08 0.12 Prueksaritanont agonist et al., 2002 87 Competitive HLM Pacli 1.15 0.03 Prueksaritanont et al., 2002 78 rCYP2C8 Ceri-1 2.56 0.08 Wang et al., 2002 68 rCYP2C8 Ceri-23 2.94 0.09 Wang et al., 2002 .250 273 Competitive HLM Ceri-1 ,0.37 ,0.01 Wang et al., 2002 95 69 Competitive HLM Ceri-23 1.45 0.04 Wang et al., 2002 91 75–76 Competitive HLM Pacli 1.33 0.04 Wang et al., 2002 48 Mixed HLM Pacli 4.17 0.13 Fujino et al., 2003a 55.4 Mixed HLM Pacli 1.81 0.05 Fujino et al., 2003b al. et Backman 36.8 rCYP2C8 Ceri-1 5.44 0.16 Shitara et al., 2004 29.7 rCYP2C8 Ceri-23 6.73 0.20 Shitara et al., 2004 119 69.0 Noncompetitive HLM Rosi-OH 1.45 0.04 Hruska et al., 2005 30.4 Competitive HLM Pacli 3.29 0.10 Kajosaari et al., 2005a 75.6 rCYP2C8 Amo 2.65 0.08 Walsky et al., 2005a 59 HLM Pio 3.39 0.10 Jaakkola et al., 2006c 120 HLM Pacli 1.67 0.05 Ogilvie et al., 2006 107 HLM Monte 1.87 0.06 Karonen et al., 2010 63 HLM Monte-4 3.18 0.10 Karonen et al., 2010 120 HLM Amo 1.67 0.05 Jenkins et al., 2011 10.2 Competitive HLM Amo 9.80 0.29 VandenBrink et al., 2011 13.5 Competitive HLM Monte 7.41 0.22 VandenBrink et al., 2011 .100 Competitive HLM Pacli 1.00 0.03 VandenBrink et al., 2011 9.3 Competitive HLM Repa 10.75 0.32 VandenBrink et al., 2011 36.1 Competitive HLM Rosi 2.77 0.08 VandenBrink et al., 2011 14 rCYP2C8 Ceri-23 7.14 0.21 Floyd et al., 2012 Genistein Anticancer, PKI 2.5 HLM Pacli Burnett et al., 2011 Glipizide Antidiabetic, sulfonylurea 338.2 rCYP2C8 Fluo 1.04 0.016 ,0.01 ,0.01 Schelleman et al., 2014 Glyburide (glibenclamide) Antidiabetic, sulfonylurea 10.8 rCYP2C8 Amo 0.214 0.002 0.04 ,0.01 Walsky et al., 2005a 4.3 rCYP2C8 Ceri-1 0.10 ,0.01 Floyd et al., 2012 6.7 rCYP2C8 Ceri-23 0.06 ,0.01 Floyd et al., 2012 Glyphosate Pesticide 82.0 HLM Amo Abass et al., 2009 Hydroxymethyl-ivacaftor Drug metabolite 17.7 0.39 Competitive HLM Amo FDA, 2012g (M1) Ibrutinib Anticancer, PKI 12.03 HLM Pacli 0.37 0.127 0.03 ,0.01 FDA, 2013d Ibrutinib metabolite PCI- Drug metabolite (7.84) HLM Pacli FDA, 2013d 45227 Iclaprim Antibiotic (91.5) rCYP2C8 n/a Hall et al., (2007) ID951551 Acotiamide analog 17 HLM DBF Furuta et al., 2004 Idelalisib Anticancer, PKI 13 HLM Pacli 4.6 ,0.16 0.71 0.11 FDA, 2014h (continued) TABLE 6—Continued

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References Idelalisib metabolite GS- Drug metabolite 39.8 HLM Pacli 9.4 ,0.12 0.47 0.06 FDA, 2014h 563117 Imatinib Anticancer, PKI 15.7 8.4 Mixed HLM Amo 5.27 0.05 0.63 0.03 Filppula et al., 2012 25.9 HLM Pacli 0.41 0.02 Kim et al., 2013b 11.28 HLM Pacli 0.47 0.02 Wang et al., 2014a Antiobstructive, LABA 30 HLM Pacli 0.00125 0.049 ,0.01 ,0.01 FDA, 2011a Sedative, GABAA receptor 30 15 HLM Pacli Madan et al., 2007 modulator Indomethacin Anti-inflammatory, NSAID 88 HLM Amo 6.7 0.10 0.15 0.02 Jenkins et al., 2011 Indomethacin acyl-b-D- Drug metabolite 26 HLM Amo Jenkins et al., 2011 glucuronide Ipriflavone M1 Drug metabolite 9.9 HLM Pacli Moon et al., 2007 Ipriflavone M2 Drug metabolite 10.2 HLM Pacli Moon et al., 2007 Ipriflavone M4 Drug metabolite 31.8 HLM Pacli Moon et al., 2007 Ipriflavone M5 Drug metabolite 2.5 HLM Pacli Moon et al., 2007 Interactions and Metabolism Drug in CYP2C8 of Role Irbesartan Antihypertensive, ARB 9.73 rCYP2C8 Amo 3.0 0.10 0.62 0.06 Walsky et al., 2005a 18 rCYP2C8 Ceri-1 0.33 0.03 Floyd et al., 2012 16 rCYP2C8 Ceri-23 0.38 0.04 Floyd et al., 2012 Isotretinoin (13-cis-retinoic Antiacne, retinoid 15.1 66.2 HLM Taza 0.69 ,0.01 0.01 ,0.01 Attar et al., 2003 acid) Isradipine Antihypertensive, CCB 5.00 HLM Pacli 0.030 0.03 0.01 ,0.01 Lee et al., 2012a Itraconazole Antifungal 31 rCYP2C8 Pacli 0.9 0.002 0.06 ,0.01 Yoshida et al., 2012 Ivacaftor Antifibrotic 3.8 3.4 Mixed HLM Amo 13.89 ,0.02 4.09 0.08 FDA, 2012g Ivacaftor metabolite (M6) Drug metabolite 63.1 HLM Amo FDA, 2012g Ketoconazole Antifungal 25 HLM Pacli 3.2 0.01 0.26 ,0.01 Monsarrat et al., 1997 2.5 Noncompetitive rCYP2C8 Torse 1.28 0.01 Ong et al., 2000 4.0 rCYP2C8 Dia 1.60 0.02 Sai et al., 2000 8.9 rCYP2C8 Phena 0.72 ,0.01 Sai et al., 2000 6-9 HLM Pacli 1.07 0.01 Dierks et al., 2001 11.8 Noncompetitive HLM Pacli 0.27 ,0.01 Bun et al., 2003 87.7 HLM Amo 0.07 ,0.01 Turpeinen et al., 2005 5.51 rCYP2C8 Amo 1.16 0.01 Walsky et al., 2005a 4 rCYP2C8 Amo 1.60 0.02 O’Donnell et al., 2007 2.45 HLM Pacli 2.61 0.03 Lee et al., 2012a 1.7 HLM Amo 3.77 0.04 Nirogi et al., 2015 Ketoprofen acyl-b-D- Drug metabolite 26 HLM Amo Jenkins et al., 2011 glucuronide KR-32570 Antiarrhythmic 30 HLM Pacli Kim et al., 2006 KR-60436 Antiulcerative, PPI 30 HLM Pacli Ji et al., 2005 Lansoprazole Antiulcerative, PPI 55 rCYP2C8 Ceri-1 0.671 0.03 0.02 ,0.01 Floyd et al., 2012 19 rCYP2C8 Ceri-23 0.07 ,0.01 Floyd et al., 2012 5.75 HLM Pacli 0.23 ,0.01 Lee et al., 2012a Lapatinib Anticancer, PKI 0.60 Competitive HLM Pacli 4.2 ,0.01 7.00 ,0.07 FDA, 2007e 1.43 HLM Pacli 2.94 ,0.03 Wang et al., 2014a Antiasthmatic, PGD2 6.5 n/a n/a Schwartz et al., 2009 receptor antagonist Laromustine Anticancer, alkylating agent .750 Noncompetitive HLM Amo Nassar et al., 2009 (VNP40101M) Lasofoxifene Antiosteoporotic, SERM 8.1 HLM Amo 0.0050 ,0.01 Moller et al., 2006 Lenvatinib Anticancer, PKI 10.1 10.1 HLM Pacli 1.546 0.02 0.15 ,0.01 FDA, 2015a Lestaurtinib (CEP-701) Anticancer, PKI 9.5 HLM Amo Filppula et al., 2014 Levothyroxine Hormonal replacement 3.30 rCYP2C8 Amo Walsky et al., 2005a therapy 5.4 rCYP2C8 Ceri-1 Floyd et al., 2012

4.6 rCYP2C8 Ceri-23 Floyd et al., 2012 199 (continued) TABLE 6—Continued 200

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References Loperamide Antidiarrheal, opioid 24 HLM Amo 0.0042 ,0.01 Nirogi et al., 2014 Antiviral, protease inhibitor 4.1 rCYP2C8 Amo 15.6 0.02 7.61 0.15 Parikh et al., 2007 Loratadine 3.36 rCYP2C8 Amo 0.0088 0.03 ,0.01 ,0.01 Walsky et al., 2005a 2.95 HLM Pacli ,0.01 ,0.01 Lee et al., 2012a Antiobesity, 5-HT2C receptor .200 HLM Pacli 0.434 0.30 ,0.01 ,0.01 FDA, 2012b agonist Lorcaserin sulfamate (M1) Drug metabolite .200 HLM Pacli FDA, 2012b Losartan Antihypertensive, ARB 12.9 rCYP2C8 Amo 0.64 0.013 0.10 ,0.01 Walsky et al., 2005a 40.7 Competitive rCYP2C8 Pacli 0.02 ,0.01 Mukai et al., 2014 (lactone, Antihyperlipidemic, HMG- 14.7 8.4 Mixed HLM Pacli 0.100 0.05 0.01 ,0.01 Tornio et al., 2005 parent) CoA reductase inhibitor 9.10 rCYP2C8 Amo 0.02 ,0.01 Walsky et al., 2005a 79.9 HLM Pacli ,0.01 ,0.01 Sakaeda et al., 2006 5.6 Competitive HLM Amo 0.02 ,0.01 VandenBrink et al., 2011 9.3 Competitive HLM Monte 0.01 ,0.01 VandenBrink et al., 2011 18.8 Competitive HLM Pacli ,0.01 ,0.01 VandenBrink et al., 2011 2.8 Competitive HLM Repa 0.04 ,0.01 VandenBrink et al., 2011 4.2 Competitive HLM Rosi 0.02 ,0.01 VandenBrink et al., 2011 27.5 rCYP2C8 Fluo ,0.01 ,0.01 Schelleman et al., 2014 Lovastatin acid Antihyperlipidemic, HMG- 54.9 48.9 Mixed HLM Pacli 0.011 0.05 ,0.01 ,0.01 Tornio et al., 2005 CoA reductase inhibitor 74.6 HLM Pacli ,0.01 ,0.01 Sakaeda et al., 2006 Macitentan Antihypertensive, ERA 21 HLM Pacli 0.29 ,0.01 0.03 ,0.01 FDA, 2013k Macitentan metabolite M6 Drug metabolite 23 HLM Pacli FDA, 2013k (ACT-132577) al. et Backman Macrolactin A Antibiotic 26.4 HLM Rosi-OH Bae et al., 2014 Pesticide 31.0 HLM Amo Abass et al., 2009 Medroxyprogesterone Progestin 4.79 rCYP2C8 Amo 0.123 0.14 0.05 ,0.01 Walsky et al., 2005a 0.76 Competitive HLM Amo 0.16 0.02 VandenBrink et al., 2011 7.5 Competitive HLM Monte 0.02 ,0.01 VandenBrink et al., 2011 8.2 Competitive HLM Pacli 0.02 ,0.01 VandenBrink et al., 2011 1.9 Competitive HLM Repa 0.07 ,0.01 VandenBrink et al., 2011 6.6 Competitive HLM Rosi 0.02 ,0.01 VandenBrink et al., 2011 Anti-inflammatory, NSAID 14.9 HLM Amo 41.44 ,0.10 5.56 0.56 Jenkins et al., 2011 Mefenamic acyl-b-D- Drug metabolite 8.5 HLM Amo Jenkins et al., 2011 glucuronide Mertansine (DM1) Antibody-drug linker 11 Competitive HLM Pacli Davis et al., 2012 Methoxsalen (8- Antipsoriatic ;10 HLM Pacli 2.36 ;0.47 Dierks et al., 2001 methoxypsoralen) Methyl belinostat Drug metabolite 13.8 HLM n/a FDA, 2014c Methylprednisolone Anti-inflammatory, 25.4 rCYP2C8 Amo 0.475 0.22 0.04 ,0.01 Walsky et al., 2005a glucucorticoid Sedative, benzodiazepine 18 Noncompetitive rCYP2C8 Torse 0.34 0.02 0.02 ,0.01 Ong et al., 2000 12.4 rCYP2C8 Amo 0.06 ,0.01 Walsky et al., 2005a MMB4 DMS Antidote, agonist 82.9 126 Noncompetitive rCYP2C8 Luci Hong et al., 2013 furoate Anti-inflammatory, 0.813 rCYP2C8 Amo 0.00012 0.02 ,0.01 ,0.01 Walsky et al., 2005a glucucorticoid 0.327 HLM Amo ,0.01 ,0.01 Walsky et al., 2005a Montelukast Antiasthmatic, LTRA 0.00922 rCYP2C8 Amo 0.89 ,0.01 193.06 1.93 Walsky et al., 2005a 0.0196 HLM Amo 90.82 0.91 Walsky et al., 2005a 0.0092 Competitive rCYP2C8 Amo 96.74 0.97 Walsky et al., 2005b 0.019 Competitive rCYP2C8 Rosi 46.84 0.47 Walsky et al., 2005b 0.020–2.0 0.014 Competitive HLM Amo 63.57 0.64 Walsky et al., 2005b 0.15 Competitive HLM Pacli 5.93 0.06 Walsky et al., 2005b 0.11 Competitive HLM Rosi 8.09 0.08 Walsky et al., 2005b (continued) TABLE 6—Continued

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References 0.18 HLM Pio 9.89 0.10 Jaakkola et al., 2006c 0.009–0.01 rCYP2C8 Amo 197.78 1.98 O’Donnell et al., 2007 0.022 0.013 HLM Amo 68.46 0.69 Perloff et al., 2009 0.0081 Competitive HLM Amo 109.88 1.10 VandenBrink et al., 2011 0.026 Competitive HLM Pacli 34.23 0.34 VandenBrink et al., 2011 0.016 Competitive HLM Repa 55.63 0.56 VandenBrink et al., 2011 0.21 Competitive HLM Rosi 4.24 0.04 VandenBrink et al., 2011 1.2 rCYP2C8 Ceri-1 1.48 0.02 Floyd et al., 2012 0.02 rCYP2C8 Ceri-23 89.00 0.89 Floyd et al., 2012 0.05–0.10 HLM Amo 35.60 0.36 Kozakai et al., 2012 0.14 Competitive HLM Pacli 6.36 0.06 Kim et al., 2013b 0.16 Competitive HLM Amo 5.56 0.06 Kim et al., 2013b 2.67 HLM Pacli 0.67 ,0.01 Zheng et al., 2013 0.27 n/a n/a 6.59 0.07 Korzekwa, 2014 159 Hep Amo 0.01 ,0.01 Kosugi et al., 2014 Interactions and Metabolism Drug in CYP2C8 of Role 2.9 HLM Rosi-OH 0.61 ,0.01 Zheng et al., 2014 0.101 HLM Pacli 17.62 0.18 FDA, 2014j 0.14 HLM Amo 12.71 0.13 FDA, 2014j 0.010–0.75 HLM Amo 178.00 1.78 Nirogi et al., 2015 Antihypertensive, b1 55 Noncompetitive HLM Pacli 3.65 0.0887 0.07 ,0.01 FDA, 2007a receptor blocker Antidepressant 23.2 rCYP2C8 Amo 4.70 ,0.01 0.41 ,0.01 Walsky et al., 2005a Netupitant 50.43 HLM Pacli 0.75 0.005 0.03 ,0.01 FDA, 2014b Netupitant hydroxylation Drug metabolite 26.95 HLM Pacli FDA, 2014b metabolite M3 Netupitant N- Drug metabolite 4.74 HLM Pacli FDA, 2014b demethylation metabolite M1 Nicardipine Antihypertensive, CCB 7.1 HLM Pacli 0.17 0.02 0.02 ,0.01 Nakamura et al., 2005 1.56 HLM Pacli 0.22 ,0.01 Lee et al., 2012a Nifedipine Antihypertensive, CCB 9.66 rCYP2C8 Amo 0.14 0.04 0.03 ,0.01 Walsky et al., 2005a 20-23 rCYP2C8 Amo 0.01 ,0.01 O’Donnell et al., 2007 13.53 rCYP2C8 Pacli 0.02 ,0.01 Gao et al., 2010 2.4 Competitive HLM Amo 0.06 ,0.01 VandenBrink et al., 2011 6.3 Competitive HLM Monte 0.02 ,0.01 VandenBrink et al., 2011 9.5 Competitive HLM Pacli 0.02 ,0.01 VandenBrink et al., 2011 1.5 Competitive HLM Repa 0.09 ,0.01 VandenBrink et al., 2011 5.8 Competitive HLM Rosi 0.02 ,0.01 VandenBrink et al., 2011 3.5 HLM Amo 0.08 ,0.01 Nirogi et al., 2014 Nilotinib Anticancer, PKI ,1 0.236 Competitive HLM Pacli 4.3 0.02 18.22 0.36 FDA, 2007c 0.61 Competitive rCYP2C8 Amo 7.04 0.14 Kim et al., 2013b 0.10 Competitive rCYP2C8 Pacli 43.00 0.86 Kim et al., 2013b 0.7 0.15 Competitive HLM Amo 28.67 0.53 Kim et al., 2013b 0.4 0.9 Competitive HLM Pacli 4.78 0.10 Kim et al., 2013b 7.5 HLM Rosi-OH 1.15 0.02 Kim et al., 2013b 0.10 HLM Pacli 43.00 0.86 Wang et al., 2014a Nintedanib Anticancer, PKI .50 HLM Pacli 0.028 0.022 ,0.01 ,0.01 FDA, 2014d Nystatin Antifungal 12.7 rCYP2C8 Amo Walsky et al., 2005a Ombitasvir (ABT-267) Antiviral, NS5A inhibitor 7.4 n/a n/a 0.070 ,0.01 0.02 ,0.01 FDA, 2014k Orphenadrine 278.8 rCYP2C8 Dia Sai et al., 2000 249.1 rCYP2C8 Phena Sai et al., 2000 265 HLM Amo Nirogi et al., 2015 Orteronel (TAK-700) Anticancer, antiandrogen 27.7 HLM n/a Lu et al., 2012 Ospemifene Antidyspareunia, SERM 36.4 HLM Amo 3.16 ,0.01 0.17 ,0.01 FDA, 2013i; Turpeinen

et al., 2013 201 (continued) TABLE 6—Continued 202

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References Oxybutynin 4.50 rCYP2C8 Amo 0.031 ,0.01 0.01 ,0.01 Walsky et al., 2005a Paclitaxel (taxol) Anticancer, taxane 14.9 30.0 Competitive HLM Taza 0.85 0.12 0.03 ,0.01 Attar et al., 2003 13.3 rCYP2C8 Amo 0.13 0.02 Walsky et al., 2005a 23–24 rCYP2C8 Amo 0.07 ,0.01 O’Donnell et al., 2007 5.4 Competitive HLM Amo 0.16 0.02 VandenBrink et al., 2011 89.8 Competitive HLM Monte ,0.01 ,0.01 VandenBrink et al., 2011 6.5 Competitive HLM Repa 0.13 0.016 VandenBrink et al., 2011 12.0 Competitive HLM Rosi 0.07 ,0.01 VandenBrink et al., 2011 Pasireotide Hormonal therapy, ;50 HLM Pacli 0.015 0.12 ,0.01 ,0.01 FDA, 2012h somastatin analog Pazopanib Anticancer, PKI 10 HLM Pacli 132 ,0.01 26.40 0.26 FDA, 2009d 3.72 HLM Pacli 35.48 0.36 Wang et al., 2014a PF-562,271 Anticancer, PKI 23 HLM n/a Rong et al., 2008 Phenthoate Pesticide 10.3 HLM Amo Abass et al., 2009 Pioglitazone Antidiabetic, PPAR-g agonist 9.38 1.69 Competitive HLM Pacli 3.8 ,0.01 2.25 0.02 Sahi et al., 2003 11.7 rCYP2C8 Amo 0.65 ,0.01 Walsky et al., 2005a 6.6 Competitive HLM Amo 0.58 ,0.01 VandenBrink et al., 2011 7.1 Competitive HLM Monte 0.54 ,0.01 VandenBrink et al., 2011 37.6 Competitive HLM Pacli 0.10 ,0.01 VandenBrink et al., 2011 3.8 Competitive HLM Repa 1.00 0.01 VandenBrink et al., 2011 6.1 Competitive HLM Rosi 0.62 ,0.01 VandenBrink et al., 2011 14 rCYP2C8 Ceri-1 0.54 ,0.01 Floyd et al., 2012 16 rCYP2C8 Ceri-23 0.48 ,0.01 Floyd et al., 2012 Pitavastatin (acid, parent) Antihyperlipidemic, HMG- 57.0 HLM Pacli 0.0296 0.005 ,0.01 ,0.01 Sakaeda et al., 2006 CoA reductase inhibitor al. et Backman Pitavastatin lactone Antihyperlipidemic, HMG- 50.5 HLM Pacli 0.0190 0.005 ,0.01 ,0.01 Sakaeda et al., 2006 CoA reductase inhibitor Ponatinib Anticancer, PKI 6.1 3.05 n/a n/a 0.161 0.0008 0.05 ,0.01 FDA, 2012e Prasugrel Antithrombotic, platelet .45.2 rCYP2C8 DBF 1.37 ,0.061 Hagihara et al., 2008 aggregation inhibitor Prasugrel active Drug metabolite .45.2 rCYP2C8 DBF Hagihara et al., 2008 metabolite (R–138727) Prasugrel thiolactone (R– Drug metabolite .50.0 rCYP2C8 DBF Hagihara et al., 2008 95913) (acid, parent) Antihyperlipidemic, HMG- .100 .50 Mixed HLM Pacli 0.085 0.57 ,0.01 ,0.01 Tornio et al., 2005 CoA reductase inhibitor .100 HLM Pacli ,0.01 ,0.01 Sakaeda et al., 2006 Pravastatin lactone Antihyperlipidemic, HMG- 99.3 HLM Pacli Sakaeda et al., 2006 CoA reductase inhibitor Pesticide 84.0 HLM Amo Abass et al., 2009 Sedative, antihistamine 23 HLM Amo 0.07 0.07 ,0.01 ,0.01 Nirogi et al., 2014 Propoxyphene Analgesic, opioid 32 rCYP2C8 Ceri-1 Floyd et al., 2012 18 rCYP2C8 Ceri-23 Floyd et al., 2012 Prothionamide Antituberculosis 57.6 HLM Pacli Shimokawa et al., 2015 Pyrimethamine Antimalarial 45.1 rCYP2C8 Amo 4.74 0.13 0.21 0.027 Parikh et al., 2007 Antipsychotic 20 HLM Amo 0.314 0.17 0.03 ,0.01 Nirogi et al., 2014 Antiarrhythmic 98.5 rCYP2C8 Dia 4 0.13 0.08 0.01 Sai et al., 2000 135.4 rCYP2C8 Phena 0.06 ,0.01 Sai et al., 2000 50 HLM Pacli 0.16 0.02 Dierks et al., 2001 Quinine Antimalarial 11 Competitive rCYP2C8 Torse 29 0.15 2.64 0.40 Ong et al., 2000 R483 Antidiabetic, PPAR-g agonist 5 HLM Pacli Weber et al., 2005 Rabeprazole Antiulcerative, PPI 12.0 rCYP2C8 Amo 0.9 0.037 0.15 ,0.01 Walsky et al., 2005a Ranitidine Antiulcerative, H2RA 10,000 HLM Pacli 1.31 0.85 ,0.01 ,0.01 Monsarrat et al., 1997 3.1 HLM Amo 0.85 0.72 Nirogi et al., 2014 Regorafenib Anticancer, PKI 1.7 0.6 n/a n/a 8.1 0.005 13.50 0.04 FDA, 2012i (continued) TABLE 6—Continued

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References Regorafenib M2 Drug metabolite 1.0 n/a n/a FDA, 2012i Regorafenib M5 Drug metabolite 1.3 n/a n/a FDA, 2012i Repaglinide Antidiabetic, meglitinide 27.1 Competitive HLM Amo 0.104 0.026 ,0.01 ,0.01 VandenBrink et al., 2011 analog 11.1 Competitive HLM Monte ,0.01 ,0.01 VandenBrink et al., 2011 .100 Competitive HLM Pacli ,0.01 ,0.01 VandenBrink et al., 2011 23.0 Competitive HLM Rosi ,0.01 ,0.01 VandenBrink et al., 2011 Retinoic acid, all-trans Antiacne, retinoid 27.0 Competitive HLM Pacli 1.15 ,0.05 0.04 ,0.01 Rahman et al., 1994 (tretinoin) Rifampin (rifampicin) Antibiotic 30.2 Competitive HLM Pacli 8 0.40 0.27 0.11 Kajosaari et al., 2005a Rifapentine Antituberculosis 115 HLM Pacli 34.21 0.02 0.59 0.01 Shimokawa et al., 2015 Antiviral, NNRTI 13.2–19.1 10.0 HLM Pacli 0.5 ,0.01 0.05 ,0.01 FDA, 2011d Antiviral, protease inhibitor 3.03 rCYP2C8 Amo 15 0.02 9.90 0.20 Walsky et al., 2005a 1–2 rCYP2C8 Amo 30.00 0.60 O’Donnell et al., 2007 5.5 HLM Pacli 5.46 0.11 FDA, 2012j Interactions and Metabolism Drug in CYP2C8 of Role Rofecoxib Anti-inflammatory, NSAID 95 rCYP2C8 Ceri-1 1.02 0.13 0.02 ,0.01 Floyd et al., 2012 14 rCYP2C8 Ceri-23 0.15 0.02 Floyd et al., 2012 Rose bengal Xanthene dye 53 Hep Amo Kazmi et al., 2014

Rosiglitazone Antidiabetic, PPAR-g agonist 18 HLM Pacli 1.7 0.002 0.19 ,0.01 Baldwin et al., 1999 9.58 5.59 Competitive HLM Pacli 0.30 ,0.01 Sahi et al., 2003 24.1–26.3 HLM Pacli 0.13 ,0.01 Kim et al., 2005b 10.8 rCYP2C8 Amo 0.13 ,0.01 Walsky et al., 2005a 5.2 Competitive HLM Amo 0.33 ,0.01 VandenBrink et al., 2011 4.1 Competitive HLM Monte 0.42 ,0.01 VandenBrink et al., 2011 28.6 Competitive HLM Pacli 0.06 ,0.01 VandenBrink et al., 2011 1.4 Competitive HLM Repa 1.21 ,0.01 VandenBrink et al., 2011 3.0 rCYP2C8 Ceri-1 1.13 ,0.01 Floyd et al., 2012 2.7 rCYP2C8 Ceri-23 1.26 ,0.01 Floyd et al., 2012 (acid, Antihyperlipidemic, HMG- .100 .50 Mixed HLM Pacli 0.0046 0.12 ,0.01 ,0.01 Tornio et al., 2005 parent) CoA reductase inhibitor .100 HLM Pacli ,0.01 ,0.01 Sakaeda et al., 2006 Rosuvastatin lactone Antihyperlipidemic, HMG- 9.8 HLM Pacli Fujino et al., 2004 CoA reductase inhibitor 32.5 HLM Pacli Sakaeda et al., 2006 Antiasthmatic, b-2- agonist 1.87 rCYP2C8 Amo 0.0040 ,0.01 Walsky et al., 2005a Sanguinarine Anticancer 10.2 8.9 Noncompetitive HLM Pacli Qi et al., 2013 Antiviral, protease inhibitor 1.8 rCYP2C8 Amo 1.41 0.02 1.57 0.031 Parikh et al., 2007 Saracatinib Anticancer, PKI 201.8 HLM Amo Filppula et al., 2014 Sarizotan Antipsychotic 18.2d Competitive HLM Pacli 0.91 0.05 Gallemann et al., 2010 Satraplatin (JM-216) Anticancer 1–3 0.9 Noncompetitive HLM Pacli Ando et al., 1998 (R-roscovitine) Anticancer, PKI 119 rCYP2C8 DBF 10 0.10 0.17 0.02 McClue and Stuart, 2008 Sertraline Antidepressant, SSRI 25.5 rCYP2C8 Amo 0.484 0.02 0.04 ,0.01 Walsky et al., 2005a 350 HLM Pacli ,0.01 ,0.01 FDA, 2008b .100 Competitive HLM Amo ,0.01 ,0.01 VandenBrink et al., 2011 9.0 Competitive HLM Monte 0.05 ,0.01 VandenBrink et al., 2011 .100 Competitive HLM Pacli ,0.01 ,0.01 VandenBrink et al., 2011 7.8 Competitive HLM Repa 0.06 ,0.01 VandenBrink et al., 2011 8.1 Competitive HLM Rosi 0.06 ,0.01 VandenBrink et al., 2011 29.7 HLM Pacli 0.03 ,0.01 Erve et al., 2013 15 HLM Amo 0.06 ,0.01 Nirogi et al., 2014 Simeprevir (TMC435) Antiviral, protease inhibitor (36.8) HLM n/a 14.5 ,0.001 0.79 ,0.01 FDA, 2013h Simvastatin (lactone, Antihyperlipidemic, 9.6 7.1 HLM Pacli 0.096 0.06 0.01 ,0.01 Tornio et al., 2005 parent) HMG-CoA reductase

inhibitor 203 (continued) TABLE 6—Continued 204

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References 5.39 rCYP2C8 Amo 0.04 ,0.01 Walsky et al., 2005a 44.1 HLM Pacli ,0.01 ,0.01 Sakaeda et al., 2006 8.3 HLM Amo 0.02 ,0.01 Jenkins et al., 2011 7.5 Competitive HLM Amo 0.01 ,0.01 VandenBrink et al., 2011 5.7 Competitive HLM Monte 0.02 ,0.01 VandenBrink et al., 2011 12.3 Competitive HLM Pacli ,0.01 ,0.01 VandenBrink et al., 2011 1.1 Competitive HLM Repa 0.09 ,0.01 VandenBrink et al., 2011 3.3 Competitive HLM Rosi 0.03 ,0.01 VandenBrink et al., 2011 3.70 HLM Pacli 0.05 ,0.01 Lee et al., 2012a 28 rCYP2C8 Fluo ,0.01 ,0.01 Schelleman et al., 2014 Simvastatin acid Antihyperlipidemic, HMG- 66.5 41.1 Mixed HLM Pacli Tornio et al., 2005 CoA reductase inhibitor 51.5 HLM Pacli Sakaeda et al., 2006 76.5 rCYP2C8 Fluo Schelleman et al., 2014 Simvastatin acyl-b-D- Drug metabolite 3.8 HLM Amo Jenkins et al., 2011 glucuronide SIPI5357 Antidepressant, Serotonin- 89.23 HLM Pacli Fan et al., 2015 -dopamine reuptake inhibitor Sitaxentan Antihypertensive, ERA 1.58 HLM Pacli 22.42 28.38 Erve et al., 2013 Anticancer, PKI 1-2 rCYP2C8 Amo 21.5 0.01 21.50 0.22 FDA, 2005b 2.4 n/a n/a 8.96 0.09 Flaherty et al., 2011 1.59 HLM Pacli 13.52 0.14 Wang et al., 2014a Spironolactone Diuretic 6.99 rCYP2C8 Amo 0.444 ,0.10 0.13 0.01 Walsky et al., 2005a Antiepileptic 37.1 35 Noncompetitive rCYP2C8 Carba 28.17 0.76 Cazali et al., 2003 al. et Backman Sulfaphenazole Antimicrobial 63 Competitive rCYP2C8 DTP Mancy et al., 1996 0.42 rCYP2C8 R-ibu-2 Hamman et al., 1997 0.55 rCYP2C8 R-ibu-3 Hamman et al., 1997 0.36 rCYP2C8 S-ibu-2 Hamman et al., 1997 0.38 rCYP2C8 S-ibu-3 Hamman et al., 1997 505 rCYP2C8 Torse Miners et al., 2000 172.0 rCYP2C8 Dia Sai et al., 2000 .50 HLM Pacli Dierks et al., 2001 Sunitinib Anticancer, PKI 28 HLM Pacli 0.21 0.05 ,0.01 ,0.01 FDA, 2006b 91.51 HLM Pacli ,0.01 ,0.01 Wang et al., 2014a Sunitinib metabolite Drug metabolite 52 HLM Pacli FDA, 2006b Su012662 Sedative, 15 HLM Pacli 0.96 ,0.01 0.13 ,0.01 FDA, 2014j antagonist Suvorexant M9 Drug metabolite 37 HLM Amo FDA, 2014j (L-002015883) Tamoxifen Anticancer, SERM 3.34 rCYP2C8 Amo 0.323 ,0.02 0.19 ,0.01 Walsky et al., 2005a 3–10 rCYP2C8 Amo 0.22 ,0.01 O’Donnell et al., 2007 3.1 Competitive HLM Amo 0.10 ,0.01 VandenBrink et al., 2011 2.1 Competitive HLM Monte 0.15 ,0.01 VandenBrink et al., 2011 12.2 Competitive HLM Pacli 0.03 ,0.01 VandenBrink et al., 2011 10.1 Competitive HLM Repa 0.03 ,0.01 VandenBrink et al., 2011 2.6 Competitive HLM Rosi 0.12 ,0.01 VandenBrink et al., 2011 14.3 HLM Pacli 0.06 ,0.01 Lee et al., 2012a 2.3 HLM Amo 0.28 ,0.01 Nirogi et al., 2014 Tanespimycin Anticancer 29 HLM Pacli 17.34 ,0.10 1.20 0.12 Gan et al., 2012 Tasimelteon Circadian regulator .100 HLM Amo 0.80 ;0.10 0.02 ,0.01 FDA, 2014m Tasimelteon M12 Drug metabolite .100 HLM Amo FDA, 2014m Gastroprokinetic, 5-HT4 ;130 HLM Pacli 0.0065 0.02 ,0.01 ,0.01 Vickers et al., 2001 receptor agonist (continued) TABLE 6—Continued

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References Telithromycin Antibiotic 87 rCYP2C8 Pacli 2.7 0.30 0.06 0.02 Yoshida et al., 2012 Temsirolimus Anticancer, PKI 27 HLM Pacli 0.57 0.02 FDA, 2007d Terbinafine Antifungal ;150 HLM Pacli 3 ,0.01 Vickers et al., 1999 Terfenadine Antihistamine 5 Noncompetitive rCYP2C8 Torse 0.0033 0.03 ,0.01 ,0.01 Ong et al., 2000 11.5 rCYP2C8 Amo ,0.01 ,0.01 Walsky et al., 2005a 19.1 HLM Pacli ,0.01 ,0.01 Erve et al., 2013 Teriflunomide Immunosuppressant, drug 0.174–0.219 0.100-0.150 Competitive, HLM Pacli 0.11 0.05 1.1 0.06 FDA, 2012l metabolite mixed Ticagrelor Antithrombotic, platelet .50 HLM Pacli 1.3 ,0.02 Zhou et al., 2011; FDA, aggregation inhibitor 2011b Ticagrelor metabolite AR- Drug metabolite 43 HLM Pacli FDA, 2011b C124910XX Ticlopidine Antithrombotic, platelet 100 rCYP2C8 2-TPE 3 0.02 0.06 ,0.01 Ha-Duong et al., 2001 aggregation inhibitor 43.9 rCYP2C8 DBF 0.14 ,0.01 Hagihara et al., 2008 Interactions and Metabolism Drug in CYP2C8 of Role 29 HLM Amo 0.21 ,0.01 Nirogi et al., 2015 Antiviral, protease inhibitor 2.1 rCYP2C8 Amo 94.8 ,0.01 90.29 ,0.90 Parikh et al., 2007 Tolbutamide Antidiabetic, sulfonylurea 2,370 Mixed HLM Pacli 196 0.09 0.08 ,0.01 Rahman et al., 1994 Trametinib Anticancer, PKI 0.34 HLM Rosi 0.032 0.038 0.19 0.01 FDA, 2013f Tranylcypromine Antidepressant, MAOI 26–35 HLM Pacli 0.42 0.03 Dierks et al., 2001 12.1 rCYP2C8 Amo 0.07 Walsky et al., 2005a 103–113 rCYP2C8 Amo ,0.01 O’Donnell et al., 2007 11.24 rCYP2C8 Pacli 0.07 Gao et al., 2010 22.5 HLM Amo 0.04 Nirogi et al., 2015 Anti-inflammatory, 19.3 rCYP2C8 Amo Walsky et al., 2005a glucocorticoid 32.5 Competitive HLM Amo VandenBrink et al., 2011 53.1 Competitive HLM Monte VandenBrink et al., 2011 .100 Competitive HLM Pacli VandenBrink et al., 2011 42.5 Competitive HLM Repa VandenBrink et al., 2011 20.4 Competitive HLM Rosi VandenBrink et al., 2011 Sedative, benzodiazepine 25 Noncompetitive rCYP2C8 Torse 0.013 0.099 ,0.01 ,0.01 Ong et al., 2000 Trimethoprim Antimicrobial, dihydrofolate 75 rCYP2C8 Pacli 4 0.63 0.05 0.03 Wen et al., 2002 reductase inhibitor 54 32 Competitive HLM Pacli 0.13 0.08 Wen et al., 2002 51.5 29.0 Competitive HLM Rosi-OH 0.14 0.09 Hruska et al., 2005 71 HLM Pio 0.11 0.07 Jaakkola et al., 2006c 40.6 rCYP2C8 Amo 0.20 0.12 Parikh et al., 2007 34.1 Competitive rCYP2C8 Pio 0.12 0.07 Tornio et al., 2008b 38.2 Competitive HLM Pio 0.10 0.07 Tornio et al., 2008b 9.2 Competitive HLM Amo 0.43 0.27 VandenBrink et al., 2011 .100 Competitive HLM Monte ,0.04 ,0.03 VandenBrink et al., 2011 . 100 Competitive HLM Pacli ,0.04 ,0.03 VandenBrink et al., 2011 8.5 Competitive HLM Repa 0.47 0.30 VandenBrink et al., 2011 13.2 Competitive HLM Rosi 0.30 0.19 VandenBrink et al., 2011 4.5–17 HLM Amo 1.78 1.12 Dinger et al., 2014 122 Hep Amo 0.07 0.04 Kosugi et al., 2014 17.41–20.38 HLM Pacli 0.46 0.29 Peng et al., 2015 Troglitazone Antidiabetic, PPAR-g agonist 1–5 0.3 Competitive rCYP2C8 Pacli 3 ,0.01 10.00 ,0.10 Yamazaki et al., 2000 15–20 HLM Pacli 0.30 ,0.01 Yamazaki et al., 2000 2.33 2.59 Competitive HLM Pacli 1.16 0.01 Sahi et al., 2003 9.78 rCYP2C8 Pacli 0.61 ,0.01 Gao et al., 2010 Troglitazone M1 Drug metabolite 9–41 rCYP2C8 Pacli Yamazaki et al., 2000 Troglitazone M3 Drug metabolite 6-26 3.0 Competitive rCYP2C8 Pacli Yamazaki et al., 2000

39–.50 HLM Pacli Yamazaki et al., 2000 205 (continued) TABLE 6—Continued 206

Therapeutic Use Mode of Test Marker b b c c Inhibitor and/or Drug Class IC50 Ki Inhibition System Reactiona Imax fu I/Ki Iu/Ki References Troleandomycin Antibiotic 953.0 rCYP2C8 Phena 3 ,0.01 Sai et al., 2000 TSAHC Anticancer 1.0 0.81 Noncompetitive HLM Amo Im et al., 2012 Ulipristal Contraceptive, progesterone 2.6 HLM Pacli 0.037 ,0.06 0.03 ,0.01 FDA, 2010b receptor modulator UTL-5g Chemoprotective, TNF-a 61.2 HLM Rosi-OH Wu et al., 2014 inhibitor Valdecoxib Anti-inflammatory, NSAID 15.0 rCYP2C8 Amo 0.512 0.02 0.07 ,0.01 Walsky et al., 2005a Vemurafenib Anticancer, PKI 12 HLM n/a 125 ,0.01 20.9 0.21 EMA, 2012d Vidupiprant (AMG 853) Antiasthmatic, PGD2 1.8 Biphasic HLM Monte Foti et al., 2012 receptor antagonist 5.4 1.1 Competitive HLM Pacli Foti et al., 2012 6.0 Competitive HLM Rosi Foti et al., 2012 Vidupiprant acyl Drug metabolite 7.3 Biphasic HLM Monte Foti et al., 2012 glucuronide (M1) 2.7 Mixed HLM Pacli Foti et al., 2012 6.9 Mixed HLM Rosi Foti et al., 2012 Antidepressant, SSRI 1.8 0.46 Competitive HLM Pacli 0.33 0.04 0.72 0.03 FDA, 2011g Vinblastine Anticancer 100 HLM Pacli Monsarrat et al., 1997 Vincristine Anticancer 8 HLM Pacli 0.43 0.11 Monsarrat et al., 1997 Vismodegib (GDC-0449) Anticancer, SMO Antagonist 6.0 Noncompetitive HLM Pacli 16.4 0.01 2.73 0.03 Wong et al., 2009; LoRusso et al., 2013 Vorapaxar Antithrombotic, platelet 1.5 0.86 Mixed HLM n/a 0.0527 0.002 0.06 ,0.01 Chen et al., 2014, FDA, aggregation inhibitor 2014l Vortioxetine Antidepressant, SMS 9.34 HLM n/a 0.04724 0.02 ,0.01 ,0.01 FDA, 2013j Vortioxetine metabolite Lu Drug metabolite 4.24 HLM n/a FDA, 2013j al. et Backman AA34443 Zafirlukast Antiasthmatic, LTRA 0.644 rCYP2C8 Amo 0.295 ,0.01 0.92 ,0.01 Walsky et al., 2005a 0.388 HLM Amo 1.52 0.02 Walsky et al., 2005a 0.78 HLM Pio 0.76 Jaakkola et al., 2006c .100 Hep Amo ,0.01 ,0.01 Kosugi et al., 2014 0.014 HLM Amo 42.14 0.42 Nirogi et al., 2014

3-ASBA, 3-(anilinosulfonyl)-benzenecarboxylic acid; 5-HT, 5-hydroxytryptamine (serotonin); 5-MeO-DIPT, 5-methoxy-N,N-diisopropyltryptamine; ARB, angiotensin II receptor blocker; BTFM gemfibrozil, 5-(2,5-bis (trifluoromethyl)phenoxy)-2,2-dimethylpentanoic acid; CCB, calcium channel blocker; CP-778875; 5-(N-(4-((4-ethylbenzyl)thio)phenyl)sulfamoyl)-2-methyl benzoic acid; ERA, endothelin receptor antagonist; fu, fraction unbound in plasma; GABA, g-aminobutyric acid; H2RA, H-2 receptor antagonist; HLM, human liver microsomes; HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A; IC50 , inhibitor concentration supporting half of the maximal inhibition; Imax , peak inhibitor concentration in plasma; Ki, reversible inhibition constant; LABA, long-acting b-adrenoceptor agonist; LTRA, leukotriene receptor antagonist; MAO, monoamine oxidase; MMB4 DMS, 1,19-methylenebis [4-[(hydroxyimino)methyl]-pyridinium] dimethanesulfonate; c-mpl, myeloproliferative leukemia; n/a, not available; NNRTI, nonnucleoside reverse transcriptase inhibitor; NS, nonstructural protein; NSAID, nonsteroidal anti- inflammatory drug; PDE, phosphodiesterase; PGD2, prostaglandin D2; PKI, protein kinase inhibitor; PPAR, peroxisome proliferator-activated receptor; PPI, proton pump inhibitor; rCYP2C8, recombinant CYP2C8; SERM, selective estrogen receptor modulator; SGLT, sodium-glucose linked transporter; SMO, smoothened receptor; SMS, serotonin modulator and stimulator; SNRI, serotonin-norepinephrine reuptake inhibitor; SSRI, selective serotonin reuptake inhibitor; TNF, tumor necrosis factor; TSAHC, 4-(p-toluenesulfonylamido)-4-hydroxychalcone; XO, xanthine oxidase; UTL-5g, N-(2,4-dichlorophenyl)-5-methyl-1,2-oxazole-3-carboxamide. a2-TPE, 2-aroylthiophen 5-hydyroxylation; Ami, aminopyrine N-demethylation; Amo, amodiaquine N-deethylation; Carba, carbamazepine 10,11-epoxidation; Ceri-1, cerivastatin demethylation (M-1); Ceri-23, cerivastatin 6-hydroxylation (M-23); DBF, dibenzylfluorescein (fluorescent reaction); Dia, diazepam N-demethylation; DTP, 2,3-dichloro-4(2-thenoyl) 5-hydroxylation; Fluo, fluorometric reaction; R-ibu-2, R-ibuprofen 2-hydroxylation; R-ibu-3, R-ibuprofen 3-hydroxylation; S-ibu-2, S-ibuprofen 2-hydroxylation; S-ibu-3, S-ibuprofen 3-hydroxylation; Luci, luciferin-6 methyl ether demethylation (luminogenic reaction); Monte, montelukast 36-hydroxylation; Monte-4, formation of montelukast metabolite 4; Pacli, paclitaxel 6a-hydroxylation; Phena, phenanthrene hydroxylation; Pio, pioglitazone hydroxylation (M-IV); Repa, repaglinide 39-hydroxylation; Rosi, rosiglitazone N-demethylation; Rosi- OH, rosiglitazone p-hydroxylation; Taza, tazarotenic acid sulfoxidation; Tolbu, tolbutamide methyl hydroxylation; Torse, torsemide methylhydroxylation. bThis information is primarily based on information from the UW Metabolism and Transport Drug Interaction Database (DIDB), Copyright University of Washington 1999-2015 (DIDB accessed May-September, 2015), and secondarily on information from Martindale: The Complete Drug Reference. London: Pharmaceutical Press (electronic version), Truven Health Analytics (Healthcare), Greenwood Village, Colorado. Available at: http://www. micromedexsolutions.com/ (Martindale accessed June-September, 2015). In case several values were reported for Imax and fu, the highest values were selected. c When experimentally determined Ki was not available, Ki was calculated as IC50 /2. An I/Ki . 1.0 indicates that a clinically relevant inhibition is likely, I/Ki =0.1–1 indicates that a clinically relevant inhibition is possible, I/Ki , 0.1 indicates that a clinically relevant inhibition is unlikely. dUnbound value. Role of CYP2C8 in Drug Metabolism and Interactions 207

TABLE 7 Natural and endogenous compounds that act as reversible CYP2C8 inhibitors

Mode of Test Marker Inhibitor Description IC50 Ki Inhibition System Reactiona References

mM(mg/ml) mM(mg/ml) 3-Isomangostin Constituent of 0.64 0.66 Competitive HLM Pacli Foti et al., 2009 mangosteen 6-Gingerol Constituent of ginger (6.5) HLM Amo Mukkavilli et al., 2014 root 6-Prenylnaringenin Prenylflavonoid 1.9 HLM Amo Yuan et al., 2014 6-Shogaol Constitutent of ginger (0.8) HLM Amo Mukkavilli et al., 2014 root 8-Desoxygartanin Constituent of 1.85 2.80 Competitive HLM Pacli Foti et al., 2009 mangosteen 8-Gingerol Constituent of ginger (0.7) HLM Amo Mukkavilli et al., 2014 root 8-Prenylnaringenin Prenylflavonoid 0.6 HLM Amo Yuan et al., 2014 (flavaprenin) 9-Hydroxycalabaxanthone Constituent of 14.1 HLM Pacli Foti et al., 2009 mangosteen 10-Gingerol Constituent of ginger (0.7) HLM Amo Mukkavilli et al., 2014 root 11-Keto-b-boswellic acid Triterpene 9.5 HLM Pacli Frank and Unger, 2006 Acetyl-a-boswellic acid Triterpene 65.8 HLM Pacli Frank and Unger, 2006 Acetyl-b-boswellic acid Triterpene 33.4 HLM Pacli Frank and Unger, 2006 Acetyl-11-keto-b-boswellic Triterpene 10.1 HLM Pacli Frank and Unger, 2006 acid Allocryptopine Alkaloid 10-100 rCYP2C8 DBF Salminen et al., 2011 Arachidonic acid Endogenous 7 Competitive rCYP2C8 Pacli Yamazaki and Shimada, compound 1999 b-Boswellic acid Triterpene 8.7 HLM Pacli Frank and Unger, 2006 Bo-yang-hwan-o-tang Oriental herbal (17,209) HLM Pacli Lee et al., 2012b medicine BST204 Dry extract of ginseng (17.4) HLM Rosi Zheng et al., 2014 Capnoidine Alkaloid 10-100 rCYP2C8 DBF Salminen et al., 2011 Cedrol Sesquiterpene 41.0 HLM Amo Jeong et al., 2014 Corybulbine Alkaloid 10-100 rCYP2C8 DBF Salminen et al., 2011 Corycavamine Alkaloid 10-100 rCYP2C8 DBF Salminen et al., 2011 Corycavidine Alkaloid 10-100 rCYP2C8 DBF Salminen et al., 2011 Alkaloid 10-100 rCYP2C8 DBF Salminen et al., 2011 Corypalmine Alkaloid 10-100 rCYP2C8 DBF Salminen et al., 2011 Cranberry (powder) Natural product (24.7) HLM Amo Albassam et al., 2015 (24.0) HLM Pio Albassam et al., 2015 Cryptopine Alkaloid 10-100 rCYP2C8 DBF Salminen et al., 2011 b-Cryptoxanthin Carotenoid pigment 13.8 HLM Pacli Zheng et al., 2013 Cudratricusxanthone Constituent of 4.69 2.2 Noncompetitive HLM Pacli Sim et al., 2015 Cudrania tricuspidata Curcumin Diarylheptanoid 129.7 rCYP2C8 DBF Mach et al., 2010 DA-9801 Extract (449.9) HLM Amo Ji et al., 2013 Dioscorea nipponica Extract (237.1) HLM Amo Ji et al., 2013 Diosmetin Flavonoid 3.13 Mixed HLM Pacli Quintieri et al., 2011 Ellagic acid Constitutent of guava (13.99) rCYP2C8 DBF Kaneko et al., 2013 leaf (2)-Epigallocatechin-3- 10.9 6.8 Competitive HLM Amo Misaka et al., 2013 gallate Eupatilin Flavone 104.9 101.9 Competitive HLM Amo Ji et al., 2010 Feverfew herb Natural product (104–126) rCYP2C8 Pacli Unger and Frank, 2004 Fisetin Flavonol 10.8 1.3–6.0 Mixed HLM Pacli Václavíková et al., 2003 Gartanin Constituent of 6.28 HLM Pacli Foti et al., 2009 mangosteen Ginger extract Extract (122.5) HLM Amo Mukkavilli et al., 2014 Green tea extract Extract (4.5) HLM Amo Misaka et al., 2013 Guava leaf extract Extract (18.16) rCYP2C8 DBF Kaneko et al., 2013 Guava leaf polyphenol Constitutent of guava (1.45) rCYP2C8 DBF Kaneko et al., 2013 leaf Gypenosides Oriental herbal (20.06) HLM Pacli He et al., 2013 medicine Hesperedin Flavonoid 274.7 rCYP2C8 Tolbu Pang et al., 2012 Hesperetin Flavonoid 68.5 HLM Pacli Quintieri et al., 2011 168.4 rCYP2C8 Tolbu Pang et al., 2012 Hibiscus sabdariffa Extract (424) HLM Amo Johnson et al., 2013 extract Honey Natural product (102.9) (50.5) Competitive rCYP2C8 Amo Muthiah et al., 2012 Constituent of 8.9 4.9 Competitive HLM Amo Jeong et al., 2013 Magnolia Horsetail Natural product (93.0) HLM Amo Sevior et al., 2010 (continued) 208 Backman et al.

TABLE 7—Continued

Mode of Test Marker Inhibitor Description IC50 Ki Inhibition System Reactiona References Hops extract Extract 0.8 HLM Amo Yuan et al., 2014 Hunnemannine Alkaloid 1–10 rCYP2C8 DBF Salminen et al., 2011 Hyperforin Constituent of 56 HLM Amo Hokkanen et al., 2011 St. John’s Wort Hypoxis hemerocallidea Extract (192) HLM Pacli Fasinu et al., 2013a extract Isocorybulbine Alkaloid 10–100 rCYP2C8 DBF Salminen et al., 2011 Isoxanthohumol Prenylflavonoid 0.2 HLM Amo Yuan et al., 2014 Jaceosidin Flavone 106.4 109.4 Competitive HLM Amo Ji et al., 2010 Labisia pumila extracts Extracts (2.39–352.3) (0.70-33.9) Noncompetitive, rCYP2C8 DBF Pan et al., 2012 mixed b-Lapachone Quinone 3.8 HLM Pacli Kim et al., 2013a Liquorice extract Extract (14.36–17.06) HLM Amo Li et al., 2015 Liquorice root Natural product (22.6) HLM Amo Sevior et al., 2010 Flavonoid 82.0 rCYP2C8 Tolbu Pang et al., 2012 a-Mangostin Constituent of 0.88 0.64 Competitive HLM Pacli Foti et al., 2009 mangosteen b-Mangostin Constituent of 8.39 HLM Pacli Foti et al., 2009 mangosteen Mecambridine Alkaloid 10–100 rCYP2C8 DBF Salminen et al., 2011 Milk thistle extract Extract (8.35) Mixed HLM Pacli Doehmer et al., 2011 Morin Flavonol 17.3 7.3-12.3 Mixed HLM Pacli Václavíková et al., 2003 Alkaloid 1–10 rCYP2C8 DBF Salminen et al., 2011 a-Naphthoflavone Flavone derivative 0.36 HLM Amo Nirogi et al., 2015 Obovatol Constituent of 11.1 HLM Amo Joo et al., 2013 Magnolia Quercetin Flavonoid 1.29 Competitive rCYP2C8 Pacli Rahman et al., 1994 1.14 Competitive HLM Pacli Rahman et al., 1994 7 HLM Pacli Dierks et al., 2001 1.96-2.35 Competitive rCYP2C8 Amo Li et al., 2002 1.56 Competitive HLM Amo Li et al., 2002 2.47 rCYP2C8 DBF Yamamoto et al., 2002 4.07 19.7 HLM Taza Attar et al., 2003 10.1 Competitive HLM Pacli Bun et al., 2003 20.6 HLM DBF Ghosal et al., 2003 3.3 HLM Pacli Cai et al., 2004 6.3 HLM Pacli Donato et al., 2004 2.9 THLE DBF Donato et al., 2004 29.5 THLE Pacli Donato et al., 2004 3.9–6.2 rCYP2C8 Pacli Unger and Frank, 2004 3.33 rCYP2C8 Amo Walsky and Obach, 2004 3.06 HLM Amo Walsky and Obach, 2004 7.19–8.47 HLM Pacli Kim et al., 2005b 57.8 HLM Amo Turpeinen et al., 2005 3.94 rCYP2C8 Amo Walsky et al., 2005a 3.3–5.6 rCYP2C8 Amo O’Donnell et al., 2007 1.6 rCYP2C8 DBF McClue and Stuart, 2008 5.28–5.38 rCYP2C8 Pacli Gao et al., 2010 1.33 rCYP2C8 Bomcc Liu et al., 2010a 0.029 HLM Amo Teng et al., 2010 0.49 Competitive HLM Amo VandenBrink et al., 2011 0.52 Competitive HLM Monte VandenBrink et al., 2011 3.0 Competitive HLM Pacli VandenBrink et al., 2011 0.61 Competitive HLM Repa VandenBrink et al., 2011 0.61 Competitive HLM Rosi VandenBrink et al., 2011 4.20 2.07 Competitive rCYP2C8 Amo Muthiah et al., 2012 1.39 HLM Pacli Lee et al., 2012a 46.0 rCYP2C8 Tolbu Pang et al., 2012 18.7 HLM Pacli Bymaster et al., 2013 (0.59) rCYP2C8 DBF Kaneko et al., 2013 (0.3702) HLM Amo Mukkavilli et al., 2014 24.5 rcCYP2C8 DBF Pan et al., 2014 12.3 HLM Rosi-OH Wu et al., 2014 3.40–5.92 HLM Amo Li et al., 2015 1.2 HLM Amo Nirogi et al., 2015 Piperlonguminine Alkaloid 76.2 HLM Amo Song et al., 2014b (continued) Role of CYP2C8 in Drug Metabolism and Interactions 209

TABLE 7—Continued

Mode of Test Marker Inhibitor Description IC50 Ki Inhibition System Reactiona References Reserveratrol Stilbenoid 26.5 16.2–20.7 HLM Pacli Václavíková et al., 2003 Saw palmetto Natural product (8) HLM Amo Sevior et al., 2010 (15.4) HLM Amo Albassam et al., 2015 (9.6) HLM Pio Albassam et al., 2015 Scoulerine Alkaloid 10-100 rCYP2C8 DBF Salminen et al., 2011 Star fruit (averrhoa Fruit juice 2.2b HLM Pacli Zhang et al., 2007b carambola) juice Sutherlandia frutescens Herb (22.4) HLM Pacli Fasinu et al., 2013b Tanshinol borneol ester Combination of the 105 rCYP2C8 Bomcc Liu et al., 2010b natural compounds danshensu and borneol Thalictricavine Alkaloid 10–100 rCYP2C8 DBF Salminen et al., 2011 Thelephoric acid Antioxidant 24.6 HLM Amo Song et al., 2014a Tiliroside Flavonoid 12.1 9.4 Competitive HLM Pacli Sun et al., 2010 Tualang honey Natural product (102.9) (50.5) Competitive rCYP2C8 Amo Muthiah et al., 2012 Natural product (523.3) HLM Amo Sevior et al., 2010 Xanthohumol Natural product 1.1 HLM Amo Yuan et al., 2014

HLM, human liver microsomes; IC50, inhibitor concentration supporting half of the maximal inhibition; Ki, reversible inhibition constant; n/a, not available; rcCYP2C8, reconstituted CYP2C8; rCYP2C8, recombinant CYP2C8; THLE, immortalized human liver epithelial cells. aAmo, amodiaquine N-deethylation; Bomcc, flurogenic substrate; DBF, dibenzylfluorescein; Monte, montelukast 36-hydroxylation; Pacli, paclitaxel 6a-hydroxylation; Pio, pioglitazone hydroxylation (M-IV); Repa, repaglinide 39-hydroxylation; Rosi, rosiglitazone N-demethylation; Rosi-OH, rosiglitazone p-hydroxylation; Taza, tazarotenic acid sulfoxidation; Tolbu, tolbutamide methyl hydroxylation. b% (v/v).

and organic anion transporter (OAT) 3 (Ki = 6.8 mM) small/moderate. For instance, axitinib inhibits CYP2C8 (Schneck et al., 2004; Shitara et al., 2004; Nakagomi- in vitro with a Ki of 0.2–0.5 mM (I/Ki = 0.3–0.9), but it did Hagihara et al., 2007). The strong interactions between not alter paclitaxel plasma concentrations in patients gemfibrozil and CYP2C8 substrate drugs observed in (FDA, 2012f; Wang et al., 2014a). Similarly, cabozanti- vivo are mainly due to its glucuronide metabolite nib is a noncompetitive inhibitor of CYP2C8 in vitro (Ogilvie et al., 2006). Gemfibrozil 1-O-b glucuronide (Ki =4.6mM, I/Ki = 0.7), but the in vivo pharmacokinetics affects CYP2C8 by mechanism-based inhibition (sec- of rosiglitazone was not affected by cabozantinib (FDA, tions V.B and VI.B). 2012c; Nguyen et al., 2015). The inhibition of CYP2C8 In vitro, the thiazolidinedione drugs pioglitazone, by pazopanib (Ki of 3.7 mM, I/Ki = 35) may be of clinical rosiglitazone, and troglitazone are potent, competitive relevance (FDA, 2009d; Tan et al., 2014; Wang et al., inhibitors of CYP2C8 with IC50 and Ki values of ,40 mM 2014b). Nilotinib is a strong competitive CYP2C8 in- (Table 6). However, e.g., pioglitazone does not affect hibitor in vitro (Ki = 0.1–0.9 mM, I/Ki = 4.8–43), but it CYP2C8 in vivo, likely because of its extensive protein also induces CYP2C8 (FDA, 2007c). Hence, a clinical binding (Kajosaari et al., 2006a). interaction study with a CYP2C8 probe substrate has The antiviral agents atazanavir and efavirenz inhibit been recommended by the FDA to evaluate the in vivo CYP2C8 in vitro with Ki values of 2.1 and 4.8 mM, effect on CYP2C8 activity by nilotinib. Similarly, an respectively [inhibitor concentration (I) to Ki ratios interaction study with a CYP2C8 substrate drug has (I/Ki) = 3.7 and 6.3, respectively] (Table 6). In vivo, also been recommended for regorafenib, which inhibits atazanavir has slightly affected the pharmacokinetics CYP2C8 with a Ki value of 0.6 mM in vitro (I/Ki = 13.5) of rosiglitazone (FDA, 2015b). According to predictions, (FDA, 2012i). In addition, sorafenib seems to be a strong efavirenz may increase the area under the plasma CYP2C8 inhibitor in vitro with Ki values , 3 mM (I/Ki = concentration-time curve (AUC) of CYP2C8 substrates 9–22) (Table 6), but the effect of sorafenib on CYP2C8 by more than fourfold at steady state, and such effects in vivo has not been evaluated. have been observed in vivo (German et al., 2007). Also several other anticancer agents exhibit inhibi- The immunosuppressant teriflunomide inhibits tion of CYP2C8 in vitro (Table 6). For instance, the CYP2C8 with a very low Ki of 0.10–0.15 mM (FDA, androgen receptor antagonist enzalutamide is both a 2012a). Thus, its estimated I/Ki ratio of 1.1 indicates substrate and inhibitor of CYP2C8 in vitro (Ki = 5.5 mM, that interactions between teriflunomide and CYP2C8 I/Ki = 6.5) (FDA, 2012k). Vismodegib, an oral hedgehog substrate drugs are likely, in agreement with in vivo pathway inhibitor, inhibits CYP2C8 in vitro with a Ki of findings (section IV.C.2). 6.0 mM (I/Ki = 2.7), but vismodegib at steady state did Numerous protein kinase inhibitors inhibit CYP2C8 not affect the pharmacokinetics of rosiglitazone (Wong to various degrees in vitro (Table 6). However, for the et al., 2009; LoRusso et al., 2013). most part, their in vivo inhibitory effects on CYP2C8 The iron chelator deferasirox inhibits CYP2C8 with have not been studied. For those whose inhibition has an IC50 of 100 mM (I/Ki ,0.01) (FDA, 2005a), but it has been examined in a clinical setting, it seems to be rather increased repaglinide AUC by 2.3-fold in vivo (Skerjanec 210 Backman et al. et al., 2010). Febuxostat, a xanthine oxidase inhibitor, mechanism-based inhibition or quasi-irreversible in- inhibits CYP2C8 in vitro with a Ki of 20 mM, suggesting hibition, leading to clinically important drug-drug that the inhibition may be of clinical relevance (I/Ki = interactions (Figs. 2 and 5; Table 8; Ogilvie et al., 0.8). However, febuxostat at steady state had no effect 2006; Tornio et al., 2014). Very recently, also the acyl on the concentrations of a single dose of rosiglitazone in glucuronide of deleobuvir, an HCV protease inhibitor, vivo (Naik et al., 2012). Similarly, rosiglitazone phar- was found to be a very potent mechanism-based in- macokinetics was not affected by the platelet aggregation hibitor of CYP2C8 (Sane et al. 2015). In addition, there inhibitor vorapaxar in vivo (Ki =0.86mM, I/Ki =0.06) is in vitro evidence suggesting that the carbamoyl (Chen et al., 2014; FDA, 2014l). glucuronide metabolite of Lu AA34893 may affect Sulfaphenazole, ketoconazole, diethyldithiocarba- CYP2C8 in a similar manner (Kazmi et al., 2010). Of mate, methoxsalen (8-methoxypsoralen), and tranylcy- interest, parent clopidogrel and gemfibrozil do not seem promine, commonly used as in vitro inhibitors of to be metabolized by CYP2C8. For example, clopidogrel CYP2C9, CYP3A4, CYP2E1, CYP2A6, and CYP2C19, is mainly eliminated by carboxylesterase 1, whereas its respectively, also inhibit CYP2C8 in vitro (Table 6). For activation is dependent on CYP2C19 and CYP3A4 instance, sulfaphenazole is a strong competitive in- (Mega et al., 2009; Simon et al., 2009; Holmberg et al., hibitor of CYP2C9 with a Ki of 0.3 mM, whereas its Ki 2014; Tarkiainen et al., 2015). for CYP2C8 inhibition is 0.4–63 mM (Mancy et al., 1996; The inhibitory effect of gemfibrozil on CYP2C8 is Hamman et al., 1997). based principally on its metabolite, gemfibrozil 1-O-b 2. Natural Compounds. A range of natural com- glucuronide, which is formed mainly by UGT2B7 in pounds have been tested for CYP2C8 inhibition in vitro, hepatocytes (Shitara et al., 2004; Ogilvie et al., 2006; and inhibition parameters have been determined for Mano et al., 2007). The metabolite acts as a mechanism- several of them (Table 7). In a CYP inhibition screening based inhibitor of CYP2C8, with inhibitor concentra- of 10 herbal products commercially available in Aus- tion supporting half of the maximal rate of enzyme tralia, horsetail (Equisetum arvense) affected CYP2C8 inactivation (KI)andmaximalrateofinactivation(kinact) with an IC50 of 93.0 mg/ml (Sevior et al., 2010). The values of 20–52 mM and 0.21 1/min in vitro (Ogilvie authors suggested that the inhibition of CYP2C8 by et al., 2006; Baer et al., 2009). Similarly, clopidogrel acyl horsetail, which is used for treatment of urinary tract 1-b-D-glucuronide causes a metabolism-dependent in- , cystitis, and prostate problems, may be hibition of CYP2C8 with KI and kinact values of 9.9 mM clinically relevant (Sevior et al., 2010). In another in and 0.047 1/min (Tornio et al., 2014). The in vivo vitro study, six herbal supplements inhibited CYP2C8 consequences of the inhibitory effects of these metabo- to various degree, but the inhibition by cranberry lites are discussed in section VI. In an in vitro study powder (IC50 = 24.7 mg/ml) and saw palmetto (IC50 = by Jenkins et al. (2011), the acyl glucuronides of ator- 15.4 mg/ml) were suggested to potentially be of clinical vastatin, dehydroketoprofen, diclofenac, ibuprofen, significance (Albassam et al., 2015). indomethacin, rac-ketoprofen, mefenamic acid, R- and Among five CYP enzymes tested, CYP2C8 was most S-naproxen, and simvastatin did not affect CYP2C8 by sensitive to inhibition by green tea extract in HLM metabolism-dependent inhibition. (IC50 =4.5mg/ml) (Misaka et al., 2013). The major catechin Several other metabolism-dependent inhibitors of in green tea, (2)-epigallocatechin-3-gallate, inhibited CYP2C8havebeenreportedintheliterature(Table8). CYP2C8 with a Ki of 6.8 mM, indicating that green tea However, the clinical importance of their interaction intake may affect CYP2C8 in vivo. It has been reported potential is unknown. that 15% of Japanese older than 40 years of age consume more than 1.8 l of green tea daily, correspond- C. Induction ing to a daily epigallocatechin-3-gallate intake of 540– Increased expression of CYP2C8 protein in hepato- 720 mg (Misaka et al., 2013). cytes due to enzyme-inducing drugs/xenobiotics is an important mechanism of drug-drug interactions that B. Metabolism-dependent Inhibition can lead to markedly increased clearance of CYP2C8 Metabolism-dependent inhibitors are compounds substrates, resulting in reduced efficacy and therapeu- that are metabolized to metabolites or reactive inter- tic failure. Several drug-responsive nuclear receptors, mediates that cause time-dependent enzyme inhibition. including CAR, PXR, VDR, and GR, can mediate the Metabolism-dependent inhibition may be either direct, transcriptional activation of the CYP2C8 gene by quasi-irreversible, or irreversible (mechanism-based recognizing the respective responsive elements within inhibition). Mechanism-based inhibitors inactivate the 59-flanking promoter region of the gene (Chen and their victim enzymes permanently, and enzyme activity Goldstein, 2009). After activation of nuclear receptors can only be regained by de novo synthesis of the by their ligands/activators (in particular, enzyme in- enzyme (Lin and Lu, 1998). Interestingly, two glucuro- ducing drugs), the nuclear receptors enter the nu- nide metabolites, gemfibrozil 1-O-b glucuronide and cleus, bind to their responsive elements in the DNA, clopidogrel acyl 1-b-D-glucuronide, affect CYP2C8 by recruit coactivators that affect chromatin structure, TABLE 8 Metabolism-dependent CYP2C8 inhibitors in vitro

Therapeutic Use a b Test Marker Inhibitor and/or Drug Class Mode of Inhibition Preinc. IC50 IC50 Shift KI kinact System Reactionc References mM ratio mM 1/min 17a-Ethinylestradiol Contraceptive, hormone derivative 8.3 1.9 HLM Pacli Chang et al., 2009 Amiodarone Antiarrhythmic 1.5 0.079 rCYP2C8 Pacli Polasek et al., 2004 51.2 0.029 HLM Pacli Polasek et al., 2004 Bosutinib Anticancer, PKI 16.9 2.6 54.8 0.018 HLM Amo Filppula et al., 2014 Clopidogrel acyl Drug metabolite 12.0 4.7 9.9 0.047 HLM Amo Tornio et al., 2014 1-b-D-glucuronide Desethylamiodarone Drug metabolite 0.67 3.3 4.4 0.009 HLM Amo Obach et al., 2007 Demethyldabrafenib Drug metabolite 30a 1.6 HLM Rosi Lawrence et al., 2014 Fluoxetine Antidepressant, SSRI Quasi-irreversible 294 0.083 rCYP2C8 Pacli Polasek et al., 2004 Gemfibrozil 1-O-b Drug metabolite Irreversible 1.8 13.3 20-52 0.21 HLM Pacli Ogilvie et al., 2006 glucuronide 4.51 0.106 Hep n/a Negishi et al., 2007 29 0.072 rCYP2C8 Amo Baer et al., 2009 Interactions and Metabolism Drug in CYP2C8 of Role 0.46 98 HLM Amo Perloff et al., 2009 3.0 HLM Monte Karonen et al., 2010 4.5 HLM Monte-4 Karonen et al., 2010 0.26 0.015 HLM Amo Teng et al., 2010 1.4 16 HLM Amo Jenkins et al., 2011 10.1 0.041 HLM Amo VandenBrink et al., 2011 21.3 0.050 HLM Monte VandenBrink et al., 2011 35 0.022 HLM Pacli VandenBrink et al., 2011 33.6 0.082 HLM Pio VandenBrink et al., 2011 18.4 0.035 HLM Repa VandenBrink et al., 2011 48.5 0.071 HLM Rosi VandenBrink et al., 2011 10.34–25.4 0.104–0.25 HLM Amo Korzekwa et al., 2014 Gemfibrozil d6-1-O-b Drug metabolite, Deuterated 29 0.033 rCYP2C8 Amo Baer et al., 2009 glucuronide Isoniazid Antituberculosis Quasi-irreversible 374 0.042 rCYP2C8 Pacli Polasek et al., 2004 170 0.012 HLM Pacli Polasek et al., 2004 Lu AA34893 carbamoyl Drug Metabolite 8.5 8.4 HLM n/a Kazmi et al., 2010 glucuronide O-methylgemfibrozil Gemfibrozil Acyl-b-D-glucuronide Analog 17 3.2 HLM Amo Jenkins et al., 2011 acyl-b-D-glucuronide Antidepressant, TCA Quasi-irreversible 49.9 0.036 rCYP2C8 Pacli Polasek et al., 2004 Phenelzine Antidepressant, MAOI 1.2 0.243 rCYP2C8 Pacli Polasek et al., 2004 54.3 0.17 HLM Pacli Polasek et al., 2004 Raloxifene Antiosteoperotic, SERM .2 0.26 0.1 rCYP2C8 Pacli VandenBrink et al., 2012 Thujopsene Sesquiterpene 29.8 3.3 HLM Amo Jeong et al., 2014 Toremifene Anticancer, SERM 5.0 1.9 HLM Pacli Kim et al., 2011b Verapamil Antihypertensive, CCB Quasi-irreversible 17.5 0.065 rCYP2C8 Pacli Polasek et al., 2004

CCB, calcium channel blocker; Hep, hepatocytes: HLM, human liver microsomes; IC50 , inhibitor concentration supporting half of the maximal inhibition; KI, inactivation constant; kinact , maximal rate of inactivation; MAOI, monoamine oxidase inhibitor; n/a, not available; PKI, protein kinase inhibitor; rCYP2C8, recombinant CYP2C8, SERM, selective estrogen receptor modulator; SSRI, selective serotonin reuptake inhibitor; TCA, tricyclic antidepressant. a Preinc. IC50 depicts IC50 after preincubation of inhibitor for 30 min with NADPH before addition of substrate, except in the case of demethyldabrafenib where the preincubation time was 20 min. b IC50 shift = reversible IC50 /preinc. IC50 .AnIC50 shift $1.5-fold is indicative of metabolism-dependent inhibition. cAmo, amodiaquine N-deethylation; Monte, montelukast 36-hydroxylation; Monte-4, formation of montelukast M4; Pacli, paclitaxel 6a-hydroxylation; Pio; pioglitazone hydroxylation (M-IV); Repa, repaglinide 39-hydroxylation; Rosi, rosiglitazone N-demethylation. 211 212 Backman et al. and increase the transcription of the target genes 2001; Rae et al., 2001; Raucy et al., 2002; Madan et al., (Handschin and Meyer, 2003). Apart from this general 2003; Ferguson et al., 2005). Moreover, VDR may mechanism, certain compounds, such as phenobarbital be involved in the induction of CYP2C8 by lithocholic and CITCO ([6-(4-chlorophenyl)imidazo[2,1-b][1,3] acid in HepG2 cells (Makishima et al., 2002; Yajima thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime), et al., 2014), and the PPAR-alpha-agonist WY14,643 seem to cause induction by increasing the nuclear (4-chloro-6-(2,3-xylidino)-2-pyrimidinylthioacetic acid) translocation of CAR, which is constitutively active has induced CYP2C8 mRNA in HepaRG cells (Thomas (Zelko et al., 2001). Other nuclear receptors and tran- et al., 2015). In addition to the above compounds, scriptional factors, such as HNF4a, HNF3g, C/EBPa, certain other drugs have weakly induced CYP2C8 in and RORs, can regulate the constitutive expression of vitro with unknown induction mechanisms, including CYP2C genes, but these factors are probably not di- tasimelteon and crizotinib (FDA, 2011e,m; EMA, 2012c; rectly involved in induction of CYP2C8 (Ferguson et al., TGA, 2014). 2005; Chen and Goldstein, 2009; Rana et al., 2010). Yet, In humans in vivo, rifampin has markedly reduced at least HNF4a seems to be required for upregulation by the plasma exposure to several CYP2C8 substrates the PXR agonist rifampin (Rana et al., 2010). (Jaakkola et al., 2006a; Niemi et al., 2000; Niemi In experimental in vitro studies, several compounds et al., 2004a; Park et al., 2004), and it is consequently and ligands of the different nuclear receptors have been the preferred CYP2C8 inducer drug for use in clinical able to induce CYP2C8 (Table 9). On the basis of studies studies (section VII). Yet, the strength of the CYP2C8 in cultured human hepatocytes, CYP2C8 is the most inducing effect of rifampin (or any other PXR ligands) inducible member of the CYP2C subfamily (Gerbal- has been difficult to estimate, because the strong Chaloin et al., 2001; Feidt et al., 2010). Regarding CYP3A4-inducing effect of rifampin is likely to partially inducibility of CYP2C8, the PXR-receptor seems to be explain its effects on the clearance of CYP2C8 sub- the most important nuclear receptor, because typical strates. Of note, one of the strongest clinical inducers of PXR ligands/activators strongly induce CYP2C8 in vitro CYP enzymes, carbamazepine, which is also a weak (Ferguson et al., 2005; Chen and Goldstein, 2009) and PXR activator, seems to be poorly characterized with can cause induction of CYP2C8 also in vivo, whereas regard to CYP2C8 both in vitro and in vivo. ligands of the other nuclear receptors cause only moder- ate induction of CYP2C8 in vitro (Ferguson et al., 2005; VI. Clinical Drug Interactions Mediated via Chen and Goldstein, 2009) and have not been shown to Cytochrome P450 2C8 markedly induce CYP enzymes in vivo in humans. PXR activators, such as phenobarbital, hyperforin (an in- A. General Aspects gredient of St. John’s wort), and rifampin, have increased The CYP2C8 enzyme is involved in many drug-drug CYP2C8 expression at mRNA, protein, and activity interactions in humans, including interactions based levels several-fold in vitro (Dussault et al., 2001; on either inhibition or induction of CYP2C8. However, Gerbal-Chaloin et al., 2001; Rae et al., 2001; Nishimura its exact role in interactions is difficult to determine, et al., 2002; Raucy et al., 2002; Madan et al., 2003; because there are no fully selective in vivo inhibitors Ferguson et al., 2005; Komoroski et al., 2005; Thomas or inducers of CYP2C8 and all known CYP2C8 sub- et al., 2015). In addition, certain other compounds, strates are metabolized, at least to a small degree, also by including ritonavir, , cyclophosphamide, lith- other enzymes. Furthermore, the activities of OATP1B1, ocholic acid, and paclitaxel can induce CYP2C8 pre- P-glycoprotein or other membrane transporters can sumably by a PXR-mediated mechanism in vitro (Chang affect the pharmacokinetics of many CYP2C8 substrate et al., 1997; Dussault et al., 2001; Synold et al., 2001; drugs, and some inhibitors of CYP2C8 inhibit these Ferguson et al., 2005; Dixit et al., 2007). It should be transporters, too. Because drug metabolizing enzymes noted that in one study, rifampin induced CYP2C8 and transporters may influence drug metabolism in mRNA in only three of the eight commercially available concert, the isolated role of CYP2C8 in many drug-drug cryopreserved hepatocyte lots tested (Yajima et al., interactions can be very difficult to dissect in vivo. 2014), suggesting that cryopreserved hepatocytes may The clinical significance of pharmacokinetic drug- not be a reliable system for studying CYP2C8 induction. drug interactions depends both on the therapeutic index Apart from PXR, induction of CYP2C8 can be exper- of victim drug and on the extent of pharmacokinetic imentally achieved at least via CAR- and GR-mediated changes, in addition to various patient-related clinical and possibly also via VDR- and PPAR-alpha-mediated factors. Of the pharmacokinetic parameters, at least the mechanisms (Ferguson et al., 2005; Chen and Goldstein, plasma AUC, peak concentration (Cmax), time to max- 2009). The CAR- phenytoin and CITCO have imum concentration (tmax), and elimination half-life (t1/2) markedly induced CYP2C8 expression in human hepa- values are generally required for the characterization tocytes (Ferguson et al., 2005). In addition, dexameth- of an interaction. Here, to be brief, we usually report asone (GR agonist) can modestly increase CYP2C8 only fold-changes of the mean AUC values caused expression in in vitro systems (Gerbal-Chaloin et al., by interactions. Of note, e.g., interindividual genetic Role of CYP2C8 in Drug Metabolism and Interactions 213

TABLE 9 Some inducers of CYP2C8 in vitro

Therapeutic Use CYP2C8 Protein in CYP2C8 Activity Inducer and/or Drug Class CYP2C8 mRNA Hepatocytes in Hepatocytes References CITCO 2.5-fold in primary hepatocytes Ferguson et al., 2005 1-fold in primary hepatocytes Thomas et al., 2015 Clofibric acid Antihyperlipidemic 2-to 3-fold in primary Prueksaritanont hepatocytes et al., 2005 Cyclophosphamide Anticancer, alkylating agent + Chang et al., 1997 Dexamethasone Anti-inflammatory, + Chang et al., 1997 glucucortidcoid 3-fold in primary hepatocytes 2-fold Gerbal-Chaloin et al., 2001 5-fold in primary hepatocytes 4-fold Raucy et al., 2002 ,2-fold in primary hepatocytes Ferguson et al., 2005 Fenofibric acid Antihyperlipidemic 2- to 6-fold in primary Prueksaritanont hepatocytes et al., 2005 Gemfibrozil Antihyperlipidemic, 1.1- to 5-fold in primary Prueksaritanont PPARa agonist hepatocytes et al., 2005 Hyperforin Constituent of St. John’s + in primary hepatocytes Dussault et al., 2001 wort 5-fold in primary hepatocytes Ferguson et al., 2005 + Komoroski et al., 2005 Idelalisib Anticancer, PKI 3.9-fold in human hepatocytes FDA, 2014h Idelalisib metabolite Drug metabolite 1.4-fold in human hepatocytes FDA, 2014h GS-563117 Ifosfamide Anticancer, alkylating agent + Chang et al., 1997 Lithocholic acid Bile acid ,2-fold in primary hepatocytes Ferguson et al., 2005 Nelfinavir Antiviral, protease inhibitor 5-fold in primary hepatocytes 2-fold Dixit et al., 2007 Nilotinib Anticancer, PKI No induction of CYP2C8 .2-fold FDA, 2007c mRNA Paclitaxel Anticancer, taxane 4-fold in primary hepatocytes Ferguson et al., 2005 + in primary hepatocytes Synold et al., 2001 Phenobarbital Antiepileptic, + Chang et al., 1997 3-fold in primary hepatocytes 3-fold Gerbal-Chaloin et al., 2001 7-fold in primary hepatocytes Raucy et al., 2002 3- to 6-fold Madan et al., 2003 2-fold in primary hepatocytes Ferguson et al., 2005 Phenytoin Antiepileptic 2-fold in primary hepatocytes Ferguson et al., 2005 Progesterone Hormonal replacement 1.4- to 9.2-fold in primary Choi et al., 2013 therapy hepatocytes Rifampin Antibiotic + Chang et al., 1997 (rifampicin) 6-fold in primary hepatocytes 3-fold Gerbal-Chaloin et al., 2001 6.5-fold in primary hepatocytes Rae et al., 2001 + in primary hepatocytes Dussault et al., 2001 + in primary hepatocytes Synold et al., 2001 7- to 12-fold in primary 6-fold (1- to 17-fold) Raucy et al., 2002 hepatocytes 4- to 8-fold in primary Madan et al., 2003 hepatocytes 3-fold in primary hepatocytes 3- to 10-fold Ferguson et al., 2005 4- to 9-fold in primary Prueksaritanont hepatocytes et al., 2005 7-fold in primary hepatocytes 4-fold Dixit et al., 2007 6.5-fold in primary hepatocytes Rana et al., 2010 0.7- to 3-old in primary Yajima et al., 2014 hepatocytes 5-fold in HepaRG cells Thomas et al., 2015 Ritonavir Antiviral, protease inhibitor + in primary hepatocytes Dussault et al., 2001 + in primary hepatocytes Synold et al., 2001 7-fold in primary hepatocytes 2-fold Dixit et al., 2007 SR12813 Antihyperlipidemic, + in primary hepatocytes Synold et al., 2001 HMG-CoA reductase inhibitor Tasimelteon Circadian regulator 4.4-fold FDA, 2014m WY14,643 Antihyperlipidemic,PPARa 4-fold in HepaRG cells Thomas et al., 2015 agonist

CITCO, [6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime; HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A; PKI, protein kinase inhibitor, HepaRG, hepatocyte-like cells from the human hepatoma HepaRG cell line; mRNA, messenger RNA; PPAR, peroxisome proliferator-activated receptor; SR12813, tetraethyl 2-(3,5-di-tert-butyl-4-hydroxyphenyl)ethenyl-1,1-bisphosphonate; WY14,643, 4-Chloro-6-(2,3-xylidino)-2-pyrimidinylthioacetic acid. 214 Backman et al. variation in the activity of drug metabolizing enzymes B. Gemfibrozil as Prototypical Inhibitor and transporters can cause a considerable variability in 1. In Vitro Versus In Vivo. In vitro, the parent the extent of drug interactions. In particular, it is gemfibrozil is a moderately potent competitive inhibitor important to note that in an individual patient the of CYP2C9 (Ki value of 5.8 mM; Wen et al., 2001), but it is exposure to a victim drug can change much more than over 10 times less potent as inhibitor of CYP2C8 (K value the generally reported mean change. i of 75 mM, Wang et al., 2002; K 87 mM, Prueksaritanont Recognition of the role of CYP2C8 as an important i et al., 2002; Table 6). Gemfibrozil in concentrations up to oxidative enzyme in drug metabolism has led to changes in 1,000 mM has no effect on CYP3A4 activity (midazolam product information of many drugs, resulting in better 19-hydroxylation) (Backman et al., 2000), but it is rather predictability of their interactions and improved safety. In potent as an inhibitor of the OATP1B1 transporter, with a some cases, CYP2C8-mediated drug interactions and the K value ranging in different studies from 4 to 31.7 mM resultant adverse effects have forced the manufacturers i to withdraw drugs from clinical use or to add contraindi- (Schneck et al., 2004; Yamazaki et al., 2005; Hirano et al., cations or limitations to their use. In general, special 2006; Nakagomi-Hagihara et al., 2007). attentionisneeded,ifadrugwithnarrowsafetymarginis In healthy volunteers, gemfibrozil (600 mg twice daily) extensively metabolized by CYP2C8 or if a drug is a strong slightly (by 23%) increased the AUC of a CYP2C9 CYP2C8 inhibitor like gemfibrozil and clopidogrel. In substrate drug glimepiride (Niemi et al., 2001) but did addition, it should be recognized that rifampin and some not increase the exposure to racemic warfarin (Lilja other potent enzyme inducers can markedly reduce plasma et al., 2005). Gemfibrozil even caused a small but 2 concentrations and effects of CYP2C8 substrate drugs. statistically significant decrease ( 11%) in the AUC of As gemfibrozil is a well-characterized inhibitor of the CYP2C9 substrate S-warfarin. These results strongly CYP2C8 that has increased the plasma concentrations suggest that gemfibrozil is not a meaningful inhibitor of several drugs (Fig. 7; Table 10), we first present a of CYP2C9 in vivo in humans. detailed description of it as an in vivo inhibitor in the In vivo gemfibrozil is glucuronidated by the UGT 2B7 following part. The other clinically relevant CYP2C8 enzyme to gemfibrozil 1-O-b-glucuronide. The benzylic inhibitors, including clopidogrel, trimethoprim, efavirenz, oxidation of the glucuronide by CYP2C8 leads to haem and teriflunomide (Niemi et al., 2004b; German alkylation and irreversible inactivation of CYP2C8 et al., 2007; FDA, 2012a; Tornio et al., 2014), are dealt (Baer et al., 2009; Jenkins et al., 2011). In HLM, the with in the next part, where we focus on the CYP2C8- kinact value of CYP2C8 by gemfibrozil 1-O-b-glucuronide inhibition-mediated drug interactions of different has been 0.21 1/min and KI 20–52 mM (Ogilvie et al., therapeutic CYP2C8 substrate drugs. Thereafter, we 2006). The glucuronide metabolite is also a competitive present CYP2C8 induction-mediated drug interactions inhibitor of OATP1B1 (OATP2) transporter (Ki 24 mM; and their clinical relevance. Shitara et al., 2004). On the basis of clinical studies on

Fig. 7. Effects of gemfibrozil on the exposure (area under the plasma drug concentration-time curve) to different CYP2C8 substrate drugs. The effect of gemfibrozil on drug exposures may also include inhibition of OATP1B1 (cerivastatin acid, lovastatin acid, paritaprevir, repaglinide, and simvastatin acid). Dasabuvir and paritaprevir were administered as a dasabuvir-paritaprevir-ritonavir combination. A fold increase of 1 refers to no effect of gemfibrozil on drug exposure. References are given in Table 10. Role of CYP2C8 in Drug Metabolism and Interactions 215

TABLE 10 Drug-drug interactions caused by CYP2C8-inhibiting drugs in humans

Inhibitor Dosing Substrate AUC Change References fold Atazanavir 400 mg once daily for 6 days, substrate on day 6 Rosiglitazone 1.4 FDA, 2015b Clopidogrel 75–300 mg for 3 days, substrate on days 1 and 3 Repaglinide 3.9–5.1a Tornio et al., 2014 Deferasirox 30 mg/kg once daily for 3 days, substrate on day 4 Repaglinide 2.3b Skerjanec et al., 2010 Efavirenz 400 mg once daily for 12 days, substrate on day 9 Amodiaquine 1.8 Soyinka et al., 2013 Active metabolite: 0.7 Soyinka et al., 2013 N-desethylamodiaquine Enzalutamide 160 mg once daily for 97 days, substrate on day 42 Pioglitazone n.s. (1.2) Gibbons et al., 2015 Active metabolite: 0.63 Gibbons et al., 2015 hydroxypioglitazone (M-IV) Gemfibrozil 600 mg twice daily for 6 days, substrate on day 6 Alogliptin 1.1b FDA, 2013g Active metabolite: M-I 1.9b FDA, 2013g 600 mg twice daily for 7 days, substrate on day 4 Brivaracetam 1.0 Nicolas et al., 2012 600 mg twice daily for 3 days, substrate on day 3 Cerivastatin (acid) 5.6a Backman et al., 2002 Cerivastatin lactone 4.4 Backman et al., 2002 600 mg twice daily for 4 days, substrate on day 4 Dabrafenib 1.5 Suttle et al., 2015 600 mg twice daily for 5 days, substrate on day 4 Daprodustat (GSK1278863A) 18.6 Johnson et al., 2014 600 mg twice daily for 5 days, substrate on day 3 Dasabuvir (ABT-333)b 11.3 Menon et al., 2015 Active metabolite: 0.22 Menon et al., 2015 dasabuvir M1 600 mg twice daily for 21 days, substrate on day 4 Enzalutamide 4.3 Gibbons et al., 2015 Active metabolite: 0.75 Gibbons et al., 2015 N-demethylenzalutamide 600 mg twice daily for 7 days, substrate for 7 days 1.4b Reyderman et al., 2004 600 mg twice daily for 3 days, substrate on day 3 R-Ibuprofen 1.3 Tornio et al., 2007 600 mg twice daily for 6 days, substrate on day 3 Imatinib n.s. Filppula et al., 2013b Active metabolite: 0.5 Filppula et al., 2013b N-demethylimatinib 600 mg twice daily for 5 days, substrate on day 3 Loperamide 2.2 Niemi et al., 2006 600 mg twice daily for 3 days, substrate on day 3 Montelukast 4.6 Karonen et al., 2010 Active metabolite: 36- 0.6c Karonen et al., 2010 hydroxymontelukast (M6) Montelukast 4.3 Karonen et al., 2011 Active metabolite: 36- 0.6c Karonen et al., 2011 hydroxymontelukast (M6 600 mg twice daily for 5 days, substrate on day 3 Paritaprevir (ABT-450)d 1.4a Menon et al., 2015 600 mg twice daily for 3 days, substrate on day 3 Pioglitazone 3.4 Deng et al., 2005 Active metabolite: n.s. Deng et al., 2005 hydroxypioglitazone (M-IV) Active metabolite: n.s. Deng et al., 2005 ketopioglitazone (M-III) 600 mg twice daily for 4 days, substrate on day 3 Pioglitazone 3.2 Jaakkola et al., 2005 Active metabolite: 0.6 Jaakkola et al., 2005 ketopioglitazone (M-III) Active metabolite: 0.6 Jaakkola et al., 2005 hydroxypioglitazone (M-IV) 600 mg twice daily for 4 days, substrate on day 3 Pioglitazone 4.3 Aquilante et al., 2013a 600 mg twice daily for 3 days, substrate on day 3 Repaglinide 8.1a Niemi et al., 2003b 600 mg twice daily for 3 days, substrate on day 3 7.3-8.3a Kalliokoski et al., 2008b 600 mg twice daily for 3 days, substrate on day 3 7.0a Tornio et al., 2008a 600 mg twice daily for 3 days, substrate on days 3-6 1.0-7.6a Backman et al., 2009 A single dose of 30-900 mg 1 h prior to substrate 1.8-8.3a Honkalammi et al., 2011a intake A single dose of 600 mg 0-6 h prior to substrate 5.0-6.6a Honkalammi et al., 2011b intake 30–600 mg twice daily for 5 days, substrate on day 5 3.4-7.0a Honkalammi et al., 2012 600 mg twice daily for 4 days, substrate on day 3 Rosiglitazone 2.3 Niemi et al., 2003a 600 mg twice daily for 3 days, substrate on day 3 Simvastatin (lactone) 1.4 Backman et al., 2000 Simvastatin acid 2.9a Backman et al., 2000 600 mg twice daily for 3 days, substrate on day 4 Sitagliptin 1.5e Arun et al., 2012 600 mg twice daily for 4 days, substrate on day 3 Treprostinil 1.9 FDA, 2009b 600 mg twice daily for 8 days, substrate on day 3 R-Warfarin 0.9 Lilja et al., 2005. S-Warfarin 0.9 Lilja et al., 2005. 600 mg twice daily for 3 days, substrate on day 3 Zopiclone n.s. Tornio et al., 2006 N-demethylzopiclone 1.2 Tornio et al., 2006 N-oxide-zopiclone 2.0 Tornio et al., 2006 Teriflunomidef 14–70 mg once daily for 12 days, substrate Repaglinide 2.3a FDA, 2012a on day 12 Trimethoprim 960 mg (combination)g twice daily for 6 days, Amodiaquine 1.6 Akande et al., 2015 substrate on day 6 Active metabolite: 0.9 Akande et al., 2015 N-desethylamodiaquine 160 mg twice daily for 3 days, substrate on day 3 Cerivastatin (acid) 1.4 Backman et al., 2003 Cerivastatin lactone 1.5 Backman et al., 2003 (continued) 216 Backman et al.

TABLE 10—Continued

Inhibitor Dosing Substrate AUC Change References 960 mg (combination)g twice daily for 3 days, substrate Loperamide 1.9 Kamali and Huang, 1996 on day 2 160 mg twice daily for 6 days, substrate on day 3 Pioglitazone 1.4 Tornio et al., 2008b Active metabolite: 1.1 Tornio et al., 2008b hydroxypioglitazone (M-IV) 160 mg twice daily for 3 days, substrate on day 3 Repaglinide 1.6 Niemi et al., 2004b 160 mg twice daily for 4 days, substrate on day 3 Rosiglitazone 1.4 Niemi et al., 2004a 200 mg twice daily for 5 days, substrate on day 5 1.3 Hruska et al., 2005

AUC, area under the plasma concentration-time curve; n.s. not statistically significant. aInhibition of OATP1B1 may also be involved. bThe role of CYP2C8 in the interaction is limited or unclear. c AUC0–7 hour. dGiven as a dasabuvir-paritaprevir-ritonavir combination. eInhibition of OAT3 may also be involved. fOf note, teriflunomide is the active metabolite of leflunomide. Hence, coadministration of leflunomide with CYP2C8 substrate drugs may also cause interactions. gTrimethoprim was given in a combination with sulfaphenazole (cotrimoxazole). the dose/time dependency of the effect of gemfibrozil on inhibition by gemfibrozil have been studied in healthy the pharmacokinetics of repaglinide and statistical volunteers using the gemfibrozil-repaglinide interac- models of enzyme and transporter inhibition, it has tion as a model. Single 600-mg doses of gemfibrozil been estimated that the in vitro mechanism-based ingested 0, 1, 3, or 6 hours before repaglinide (0.25 mg) inhibition of CYP2C8 by gemfibrozil 1-O-b-glucuronide increased the geometric mean AUC of repaglinide 5.0-, manifests into a strong and long-lasting inhibition 6.3-, 6.6-, and 5.4-fold, respectively (Fig. 8). The Cmax of of CYP2C8 at typical clinical doses of gemfibrozil the CYP2C8-mediated repaglinide M4-metabolite was (Backman et al., 2009; Honkalammi et al., 2011a,b). In 1.0-, 0.10-, 0.06-, and 0.09-fold compared with control addition, the OATP1B1 inhibitory effect of the glucuro- phase, respectively (Honkalammi et al., 2011b). These nide can lead to an up to ;50% transient inhibition of results indicate that the strong inactivation of CYP2C8 OATP1B1 in vivo. These effects are the main explana- occurs rapidly, being evident already within 1 hour after tion to the effects of gemfibrozil on CYP2C8 and oral dosing of gemfibrozil. OATP1B1 substrates (Honkalammi et al., 2012). Sim- When repaglinide was ingested 1, 24, 48, or 96 hours ilar estimations have been obtained with physiologi- after discontinuation of a gemfibrozil treatment (600 mg cally based pharmacokinetic modeling in a recent twice daily for 3 days), the AUC of repaglinide was publication (Varma et al., 2015). 7.6-, 2.9-, 1.4-, and 1.0-fold compared with the control 2. Gemfibrozil Dose Versus CYP2C8 Inhibition. phase, respectively (Backman et al., 2009). These Single oral doses of gemfibrozil, i.e., 30, 100, 300, or 900 findings confirmed and extended the previous findings, mg ingested 1 hour before repaglinide, increased the which had shown that the inhibitory effect of gemfibro- AUC of repaglinide in a dose-dependent manner 1.8-, zil persists for at least 12 hours after its ingestion 4.5-, 6.7-, and 8.3-fold compared with placebo, respectively (Tornio et al., 2008a). As the half-lives of gemfibrozil (Fig. 8; Honkalammi et al., 2011a). Also after multiple and its glucuronide are very short (about 1–2 hours), doses of gemfibrozil (30, 100, or 600 mg twice daily for these findings convincingly demonstrate that the effect 5 days), the exposure to repaglinide increased dose of gemfibrozil on repaglinide pharmacokinetics is based dependently, but the greatest AUC increase did not on irreversible mechanism-based inhibition of CYP2C8. exceed that observed after the single 900 mg gemfibrozil A several-fold increase in repaglinide AUC was evident dose (Honkalammi et al., 2012). Thus, the maximum even at very low plasma concentrations of gemfibrozil inhibition of CYP2C8 can be achieved by a single 900-mg 1-O-b-glucuronide, which are less than 1% of its peak dose of gemfibrozil (Fig. 9). Gemfibrozil in doses of 100 concentrations. The results also showed that full mg twice daily at steady state causes an about 95% CYP2C8 activity recovers gradually within 3–4 days inhibition of CYP2C8 (Fig. 9), and in doses of 10 mg twice after cessation of the clinically used therapeutic doses of daily, it causes an about 50% inhibition (Honkalammi 600 mg twice daily gemfibrozil. et al., 2011a). The fraction of a small 0.25-mg dose of 4. Quantification of CYP2C8-Mediated Drug Interac- repaglinide metabolized by CYP2C8 is about 80–90%. tions in Humans. Interactions caused by a combina- However, because repaglinide is metabolized to some tion of two or more drugs, which inhibit, in addition to extent also by CYP3A4, the relative role of CYP2C8 and CYP2C8, also some other crucial enzyme or transporter, CYP3A4 in the biotransformation of repaglinide can can increase exposure to a CYP2C8 substrates much depend on its dose and plasma concentrations as well more than is the sum of their separate effects causing a as on individual pharmacogenetic factors (Bidstrup et al., classic potentiation phenomenon. Thus, e.g., exposure 2003; Kajosaari et al., 2005a; Säll et al., 2012). to repaglinide is increased only slightly by itraconazole 3. Onset and Duration of CYP2C8 Inhibition by alone (1.4-fold), greatly by gemfibrozil alone (8.1-fold), Gemfibrozil. The onset and duration of CYP2C8 and drastically (19.4-fold) by their combination (Fig. 10; Role of CYP2C8 in Drug Metabolism and Interactions 217

Fig. 8. Effects of gemfibrozil on the exposure (area under the plasma drug concentration-time curve) to repaglinide (Repa) and its metabolites M1, M2, and M4. Fold changes in drug exposure compared with the control phase when repaglinide was taken 1 hour after a single 30-, 100-, 300-, or 900-mg dose of gemfibrozil (A) or 1 hour after the last dose of 30, 100, or 600 mg gemfibrozil twice daily (B). Fold changes in drug exposure as compared with the control phase, when a single 600-mg dose of gemfibrozil was taken simultaneously or 1, 3, or 6 hours before repaglinide intake (C) or when the last dose of gemfibrozil was taken 0, 1, 3, 6, 12, 24, 48, or 96 hours before repaglinide intake (D). For M4, AUC0-3 hour data are presented. For all other compounds, AUC0-‘ data are presented. A fold change of 1 refers to no effect of gemfibrozil on exposure. References are given in the text.

Niemi et al., 2003b). The quantitative rationalization of also the blood glucose lowering effect of repaglinide gemfibrozil-drug interactions and consideration of (Niemi et al., 2003b). This pioneering study clearly transporter-enzyme interplay have been dealt quite indicated that the extent of interaction caused by a recently by Varma et al. (2015). combination of two drugs can greatly exceed the sum of their separate effects. On the basis of these results and C. Inhibition-Mediated Drug Interactions and Their clinical observations of serious hypoglycemic episodes Clinical Significance in diabetic patients, The European Agency for the 1. Repaglinide. Interactions of the oral antidiabetic Evaluation of Medicinal Products gave “EMEA public drug repaglinide have been studied extensively, and it statement on repaglinide contraindication of concomi- is a recommended model substrate drug for CYP2C8 in- tant use of repaglinide and gemfibrozil” (21.05.2003). teraction studies (EMA, 2012b; http://www.fda.gov/Drugs/ Also the U.S. Food and Drug Administration warned DevelopmentApprovalProcess/DevelopmentResources/ against repaglinide-gemfibrozil interaction. The effect DrugInteractionsLabeling/ucm093664.htm). In healthy of gemfibrozil on repaglinide exposure was later volunteers, gemfibrozil (600 mg twice daily for 3 days) confirmed and characterized in several studies as de- raised the AUC of repaglinide 8.1-fold, itraconazole scribed in previous paragraphs (sections VI.B.1–3). The raised it 1.4-fold, and their combination raised it gemfibrozil-repaglinide interaction is mainly mediated 19.4-fold (Niemi et al., 2003b). Gemfibrozil alone and via inhibition of CYP2C8 and OATP1B1 by the gemfi- in combination with itraconazole considerably enhanced brozil 1-O-b-glucuronide. 218 Backman et al.

contraindicated, e.g., in Canada (http://healthycanadians. gc.ca/recall-alert-rappel-avis/hc-sc/2015/54454a-eng.php). The immunosuppressant teriflunomide (70 mg once daily for 4 days, followed by 14 mg once daily for 8 days) increased the AUC of repaglinide by 2.3-fold in healthy male subjects (FDA, 2012a) compared with when repaglinide was given alone. In three subjects the AUC was raised by 3.2- to 3.6-fold. Teriflunomide inhibits both CYP2C8 and OATP1B1, and the contri- bution of each mechanism to the increase in repaglinide exposure has not been established (FDA, 2012a). Of note, teriflunomide is the active metabolite of lefluno- mide and its plasma concentrations following lefluno- mide administration are equal to those observed when it is given alone. Hence, leflunomide may also be a clinically relevant CYP2C8 inhibitor. Care is warranted if inhibitors of CYP2C8 are combined with repaglinide. In particular, combination of strong CYP2C8 inhibitors, such as gemfibrozil and clopidogrel, with repaglinide should be avoided. Blood glucose levels and symptoms of hypoglycemia should be monitored closely and the doses modified as needed. The interactions with repaglinide are likely to be stronger in CYP2C8*3 carriers than in CYP2C8*1 homozygotes (Tornio et al., 2008a). Fig. 9. Predicted effects of different gemfibrozil doses on CYP2C8- 2. Other Oral Antidiabetic Drugs. The European mediated drug metabolism. Predicted fold increase in the AUC0–‘ of a drug metabolized by CYP2C8, when the fraction of the substrate drug product information of Actos (pioglitazone) stated ear- metabolized by CYP2C8 (fm,CYP2C8) varies between 50 and 99% (A), lier (e.g. 2004) that metabolism of pioglitazone occurs and the CYP2C8 activity remaining (B) after a gemfibrozil dose ranging predominantly via CYP3A4 and CYP2C9 (Jaakkola from 0 to 600 mg twice daily in steady-state conditions. Modified from Honkalammi et al. (2012). AUC, area under the concentration-time curve. et al., 2005), whereas the U.S. label stated that the major CYP isoforms involved were CYP2C8 and CYP3A4 (FDA, 1999). The in vitro study of Jaakkola et al. The antimicrobial drug trimethoprim (160 mg twice (2006c) showed that pioglitazone is metabolized mainly daily for 3 days) raised in healthy volunteers the AUC of repaglinide by 1.6-fold compared with placebo (Niemi et al., 2004b). Symptomatic hypoglycemia developed in a diabetic patient 5 days after addition of trimethoprim/ sulfamethoxazole therapy to his previously well-tolerated repaglinide (1 mg three times daily) treatment (Roustit et al., 2010). The 300-mg loading dose of clopidogrel raised the AUC of repaglinide by 5.1-fold, and the following daily 75-mg doses of clopidogrel raised the AUC by 3.9-fold in healthy volunteers (Tornio et al., 2014). The increase in repaglinide AUC caused by clopidogrel was highest in subjects with the CYP2C8*1/*4 genotype (Tornio et al., 2014). The clopidogrel-repaglinide interaction is medi- ated by formation of the clopidogrel acyl-b-D-glucuronide, which is a potent time-dependent inhibitor of CYP2C8. On the basis of this short-term study, it has been extrapolated that the daily treatment with 75 mg of clopidogrel causes a continuous, 60–85% inhibition of hepatic CYP2C8 under steady-state conditions during chronic clopidogrel use. The pharmacokinetic interaction Fig. 10. Effects of gemfibrozil (Gem), itraconazole (Itra), or their of clopidogrel and repaglinide resulted in an enhanced combination (Gem + itra) on the exposure (area under the plasma drug concentration-time curve) to repaglinide, loperamide, montelukast, and blood glucose-lowering effect of repaglinide. The con- pioglitazone. A fold increase of 1 refers to no effect of inhibitors on drug comitant use of repaglinide and clopidogrel is now exposure. References are given in Table 10 and in the text. Role of CYP2C8 in Drug Metabolism and Interactions 219 by CYP2C8 and to lesser extent by CYP3A4, whereas CYP2C8 to active desethylamodiaquine, which has a CYP2C9 is not significantly involved in pioglitazone long half-life of 9–18 days. In healthy subjects, tri- elimination. In healthy volunteers, gemfibrozil raised methoprim and efavirenz have been reported to in- the mean AUC of pioglitazone 3.2-fold (range 2.3-fold crease the AUC of amodiaquine by 1.6- and 1.8-fold, to 6.5-fold) and its elimination half-life 2.7-fold, but respectively, and to reduce that of desethylamodiaquine itraconazole had no effect on pioglitazone and did not by 12 and 26%, respectively (Soyinka et al., 2013; alter the effect of gemfibrozil on its pharmacokinetics Akande et al., 2015). In an earlier study, efavirenz (Jaakkola et al., 2005). In two other studies, gemfibrozil raised the AUC of amodiaquine in two healthy subjects increased the mean AUC of pioglitazone 3.4-fold (Deng by 2- to 4-fold and decreased the AUC of desethylamo- et al., 2005) and 4.3-fold (range 1.3-fold to 12.1-fold) diaquine by 24 and 8.5% (German et al., 2007). In both of (Aquilante et al., 2013a). CYP2C8 genotype influences these subjects, marked elevation of hepatic transami- therelativechangeinpioglitazoneexposureafter nase levels occurred several weeks after stopping the 3 gemfibrozil administration. Thus, CYP2C8*3 carriers days’ combined use, forcing premature discontinuation had a greater mean increase by gemfibrozil in pioglita- of the interaction study. The dramatic, delayed hepato- zone AUC (5.2-fold) compared with CYP2C8*1 homozy- toxicity warrants great care in combination of any gotes (3.3-fold) (Aquilante et al., 2013a). Trimethoprim CYP2C8 inhibitor with amodiaquine. (160 mg twice daily) raised in healthy volunteers the 4. Statins. Cerivastatin was initially considered as a AUC of pioglitazone by 1.4-fold and had opposite effects safe because of its dual biotransformation routes, on pioglitazone pharmacokinetics compared with the mediated both via CYP3A4 and CYP2C8 (Mück, 1998; effects of CYP2C8*3 allele during the placebo phase 2000). However, it soon became obvious that cerivasta- (Tornio et al., 2008b). tin greatly increased the incidence of fatal rhabdomyol- The AUC of rosiglitazone was raised in healthy ysis, particularly when taken along with gemfibrozil volunteers by gemfibrozil by 2.3-fold (Niemi et al., (Staffa et al., 2002). Consequently, cerivastatin was 2003a). In another study, trimethoprim (160 mg twice withdrawn from the market in 2001, only 3 years after daily) increased rosiglitazone AUC by 1.4-fold and re- its launch. Despite the “dual metabolic pathway” and duced the formation of N-demethylrosiglitazone (Niemi supposed "low propensity for drug interactions" (Mück et al., 2004a). The effect of trimethoprim (200 mg twice et al., 1998; Mück, 2000), the elimination of cerivastatin daily) on rosiglitazone pharmacokinetics was confirmed relied predominantly on CYP2C8. In healthy volun- by Hruska et al. (2005), who also demonstrated the teers, gemfibrozil (600 mg twice daily) raised the AUC competitive inhibition of rosiglitazone p-hydroxylation of the parent cerivastatin (acid) by 5.6-fold, the AUC by trimethoprim in vitro. Atazanavir (400 mg once daily) of cerivastatin lactone by 4.4-fold, and that of the increased the AUC of a single dose of rosiglitazone by CYP3A4-dependent metabolite M-1 by 4.35-fold, 1.4-fold (FDA, 2015b). whereas gemfibrozil decreased the AUC of the The AUC of nateglinide was increased only by 1.5-fold CYP2C8-dependent metabolite M-23 by 78% (Fig. 11; by 3 days’ pretreatment with therapeutic doses of both Backman et al., 2002). The increased exposure to gemfibrozil and itraconazole (Niemi et al., 2005a). Thus, cerivastatin, to its lactone, and to M-1 and the reduced neither CYP2C8 nor CYP3A4 has a substantial signif- formation of the CYP2C8-dependent metabolite re- icance to the pharmacokinetics of nateglinide. Gem- vealed the strong CYP2C8 inhibitory effect of gemfi- fibrozil increased also the AUC of the dipeptidyl brozil. In addition to irreversible inhibition of CYP2C8 peptidase inhibitor sitagliptin by 1.5-fold (Arun et al., by gemfibrozil 1-O-b-glucuronide, inhibition of the 2012). However, the gemfibrozil-sitagliptin interaction hepatic OATP1B1 may contribute to the gemfibrozil- seems to be mainly mediated by inhibition of the renal cerivastatin interaction (Ogilvie et al., 2006; Shitara OAT3, with a minor contribution by CYP2C8. et al., 2004; Tamraz et al., 2013). If gemfibrozil, clopidogrel, or other inhibitors of Interestingly, about 10 years after the withdrawal of CYP2C8 will be combined with pioglitazone or rosiglita- cerivastatin, it was found that in addition to gemfibro- zone, blood glucose levels, symptoms of hypoglycemia, and zil, also concomitant use of clopidogrel was strongly other potential adverse effects (e.g., fluid retention) should associated with cerivastatin-induced rhabdomyolysis, be monitored closely and the doses be modified as needed. with an odds ratio of ;30 (48 when gemfibrozil users As shown for the gemfibrozil-pioglitazone and gemfibrozil- were excluded) (Floyd et al., 2012). Recently, Tornio repaglinide interactions (Tornio et al., 2008a; Aquilante et al. (2014) showed that glucuronidation converts et al., 2013a), interactions may be stronger in CYP2C8*3 clopidogrel to a strong time-dependent inhibitor of carriers than in CYP2C8*1 homozygotes. CYP2C8, clopidogrel acyl-b-D-glucuronide. The forma- 3. Amodiaquine. Although amodiaquine N-deethylation tion of this metabolite leads to uninterrupted inhibition is a widely used marker reaction for CYP2C8 activity of CYP2C8 during clopidogrel treatment and explains in vitro, the sensitivity of amodiaquine to CYP2C8 the increased risk of rhabdomyolysis during concomi- inhibition is poorly characterized in humans. Amodia- tant use of cerivastatin and clopidogrel (Tornio et al., quine is rapidly and extensively metabolized by 2014). Also trimethoprim increased the AUC of 220 Backman et al.

Fig. 11. Effects of gemfibrozil on the plasma concentrations of cerivastatin, its lactone, and M-1 and M-23 metabolites after administration of cerivastatin 0.3 mg with gemfibrozil 600 mg or placebo twice daily for 3 days (modified from Backman et al., 2002). cerivastatin (by 1.4-fold) and its lactone (1.5-fold) Gemfibrozil raises the AUC of nearly all statin acids, (Backman et al., 2003). Because cerivastatin has been including simvastatin acid, lovastatin acid, atorva- withdrawn from the market, its interactions are no statin, pravastatin, rosuvastatin, and pitavastatin more of direct clinical relevance. However, they are (Neuvonen et al., 2006). However, the role of CYP2C8 in examples of clinically important challenges in drug some gemfibrozil-statin interactions seems to be limited or development and have been of paramount importance nonexistent. They are mainly mediated by inhibition in understanding the significance of CYP2C8 in drug of OATP1B1, OAT3, or other transporters (Shitara metabolism. et al., 2004; Neuvonen, 2010; Niemi et al., 2011). Interestingly, cerivastatin and repaglinide have 5. Anticancer Drugs. Most anticancer drugs have a pharmacokinetic similarities. Both drugs are sub- narrow therapeutic range. Although paclitaxel is a well- strates of CYP2C8, CYP3A4, and OATP1B1. The established CYP2C8 probe in vitro, its interactions with CYP3A4 inhibitor itraconazole has raised their AUC CYP2C8 inhibitors and inducers have not been widely only slightly, i.e., by 1.4-fold (repaglinide), 1.15-fold studied in humans. Lapatinib and pazopanib are rela- (cerivastatin acid), and 1.8-fold (cerivastatin lactone), tively strong inhibitors of CYP2C8, and they have whereas gemfibrozil has raised their AUC values much raised the AUC of paclitaxel up to 1.8-fold (Tan et al., more, i.e., by 8.1-fold (repaglinide), by 5.6-fold (cerivas- 2014). In a case report, the only clopidogrel user in a tatin), and by 4.4-fold (cerivastatin lactone) (Kantola cohort of 93 ovarian carcinoma patients treated with et al., 1999; Backman et al., 2002; Niemi et al., 2003b). paclitaxel had the second lowest clearance of unbound Because the combination of a CYP3A4 inhibitor and a paclitaxel in the cohort. She was hospitalized three CYP2C8 inhibitor caused a drastic increase in repagli- times because of severe paclitaxel toxicity (Bergmann nide AUC (by 19.4-fold; Niemi et al., 2003b), it is et al., 2015). reasonable to assume that also the exposure to cerivas- Gemfibrozil has raised the AUC of the androgen tatin acid and to its more lipophilic lactone form have receptor antagonist enzalutamide by 4.3-fold, and itra- raised even more by gemfibrozil —or clopidogrel—if the conazole raised it by 1.4-fold compared with control patients had been using also CYP3A4 inhibiting drugs. (Gibbons et al., 2015). These results agree well with the in However, there seems to be no studies on the effect of vitro findings that CYP2C8 is the predominant enzyme in CYP2C8 and CYP3A4 inhibitor combinations on the the elimination of enzalutamide. The composite exposure plasma concentrations of cerivastatin. of enzalutamide and its active metabolite was raised by Role of CYP2C8 in Drug Metabolism and Interactions 221

2.2-fold by gemfibrozil and by 1.3-fold by itraconazole. A gemfibrozil. This should lead to similar plasma concen- reduction of the enzalutamide dose by about 50% is trations of dasabuvir as those achieved by normal recommended when gemfibrozil is used concomitantly. dasabuvir doses administered without inhibitor of There are no published studies on the effect of clopidogrel CYP2C8. Also some other new antiviral drugs are on enzalutamide pharmacokinetics. However, a close partially metabolized by CYP2C8, but their suscepti- follow up and reduction of enzalutamide dose can be bility to interact with drugs affecting CYP2C8 activity recommended also in their possible coadministration. It in humans needs further studies. should also be noted that combined inhibition of 7. Antiasthmatic Drugs. In healthy volunteers. CYP2C8 and CYP3A4 can cause a greater increase in gemfibrozil raised the AUC of montelukast 4.5-fold enzalutamide + metabolite AUC. Enzalutamide itself is and its elimination half-life 3.0-fold (Karonen et al., an inhibitor of CYP2C8 and may moderately raise the 2010). Gemfibrozil reduced the AUC of the secondary exposure to its substrate drugs, e.g., pioglitazone AUC M4 metabolite of montelukast by more than 90%. In by 20% (Gibbons et al., 2015). another study, gemfibrozil alone raised the AUC of GemfibrozildidnotaffecttheAUCofimatinib montelukast 4.3-fold, itraconazole had no significant after a single imatinib dose but reduced the AUC of effects, and the effects of the gemfibrozil-itraconazole N-demethylimatinib by 48%, indicating a significant combination on montelukast pharmacokinetics did not participation of CYP2C8 in the metabolism of imatinib in differ from those of gemfibrozil alone (Karonen et al., 2012). humans (Filppula et al., 2013b). After a single dose, These findings indicate that CYP2C8 but not CYP3A4 is imatinib seems to be mainly metabolized by CYP3A4, important in the pharmacokinetics of montelukast. In but the fraction of imatinib metabolized by CYP3A4 contrast to the effect of gemfibrozil on montelukast, the decreases after its multiple doses because of auto- pharmacokinetics of zafirlukast is not affected by gemfi- inhibition of the CYP3A4-mediated metabolism of brozil (Karonen et al., 2011), although both of these imatinib (Filppula et al., 2012, 2013a). This autoinhibi- cysteinyl leukotriene receptor antagonists are potent in tion is likely to increase the relative role of CYP2C8 vitro inhibitors of CYP2C8 (Walsky et al., 2005a). in imatinib elimination and its sensitivity to interac- Montelukast has a relatively large safety margin, and tions caused by CYP2C8 inhibitors during long-term the clinical significance of its interactions with CYP2C8 treatment. According to pharmacokinetic simulations, inhibitors seems to be limited. However, neuropsychi- imatinib exposure may raise up to twofold at steady state atric symptoms have developed in a woman with HIV if a strong CYP2C8 inhibitor is given concomitantly with when montelukast was added to her therapy imatinib (Filppula et al., 2013b). containing the CYP2C8 inhibitor efavirenz (Ibarra- In melanoma patients, gemfibrozil increased the Barrueta et al., 2014). She had used efavirenz, emtrici- AUC of the CYP3A4 and CYP2C8 substrate dabrafenib tabine, and fumarate for years by 1.5-fold and ketoconazole increased it by 1.7-fold with good tolerance until montelukast was started (Suttle et al., 2015). It is probable that a combined for . Shortly thereafter unbearable symptoms administration of CYP2C8 inhibitors and CYP3A4 appeared, consisting of disturbed sleep, vivid dreams inhibitors with dabrafenib can increase its exposure and irritability, confusion, and concentration difficulties. more than does either of these inhibitors alone. In After 2 months of concomitant use, montelukast was addition to paclitaxel, dabrafenib, and imatinib, some withdrawn and the psychiatric symptoms completely dis- other anticancer drugs are metabolized by CYP2C8 appeared. This case report indicates that adverse effects (Table 1). However, their interactions with CYP2C8 can develop when these drugs are used together, although inhibitors have not been characterized in humans. the mechanism of adverse effects is not fully clear. Considering the narrow therapeutic range of many 8. Other Substrate or Inhibitor Drugs. Gemfibrozil anticancer drugs, close follow up for possible adverse raised in healthy volunteers the AUC of loperamide effects is warranted if gemfibrozil, clopidogrel, trimeth- 2.1-fold, itraconazole raised it 3.8-fold, and the oprim, or other inhibitors of CYP2C8 are used with pacli- gemfibrozil-itraconazole combination raised lopera- taxel or other anticancer drugs metabolized by CYP2C8. mide AUC 12.6-fold compared with placebo phase 6. Antiviral Drugs. Gemfibrozil (600 mg twice daily) (Niemi et al., 2006). This finding strongly suggests has increased the AUC of the antihepatitis C drug that gemfibrozil can markedly increase the loperamide dasabuvir about 11-fold and their concomitant use is exposure in subjects who are using potent inhibitors contraindicated (Menon et al., 2015). It is reasonable to of CYP3A4, i.e., when another important metabolic assume that also other potent inhibitors of CYP2C8 route is blocked. Administration of cotrimoxazole such as clopidogrel increase greatly the exposure to (trimethoprim + sulphamethoxazole) has increased the dasabuvir, and their use together should be avoided or AUC of loperamide by 1.9-fold (Kamali and Huang, the dose of dasabuvir be reduced markedly. It can be 1996). Also some other , e.g., buprenorphine, are speculated that savings could be achieved by using CYP2C8 substrates (Table 1). However, there seem to be small amounts of expensive dasabuvir (about one-tenth no published studies on their possible interaction with of normal dose) with small doses (e.g., 100 mg) of gemfibrozil or other CYP2C8 inhibitors. 222 Backman et al.

Gemfibrozil raised the AUC of the prolyl hydroxylase their AUC, and diminish their clinical efficacy. Both inhibitor agent daprodustat (GSK1278863) 18.6-fold CYP2C8 and CYP3A4 are involved in the biotransfor- (Johnson et al., 2014). This result together with in vitro mation of many drugs, which can also be substrates of studies indicates the crucial significance of CYP2C8 in various transporters (Table 1). Because both CYP2C8 and its pharmacokinetics. CYP2C8 inhibitors should not be CYP3A4 enzymes and some transporters can be highly used with this erythropoiesis- agent or its dose inducible, the importance of CYP2C8 in many rifampin needs to be reduced very greatly. On the other hand, at interactions is difficult to determine exactly (Niemi et al., least theoretically, it could be possible to take advantage of 2000). Apart from rifampin, there are very few clinical this interaction in a product containing very small doses of studies concerning the effects of other CYP enzyme daprodustat and gemfibrozil. inducers on the pharmacokinetics of CYP2C8 substrates. In healthy volunteers, gemfibrozil raised only slightly 1. Rifampin (Rifampicin). Rifampin (600 mg/day), the AUC of R-ibuprofen, by 1.3-fold, after the ingestion given for several days, has decreased the plasma of racemic ibuprofen (Tornio et al., 2007). In vitro exposure to repaglinide by 31–80% depending on the CYP2C8 participates in the metabolism of zopiclone time interval from the last rifampin dose to repaglinide (Becquemont et al., 1999). In humans, however, gemfi- ingestion (Table 11; Niemi et al., 2000; Hatorp et al., brozil did not increase the AUC of the parent zopiclone 2003; Bidstrup et al., 2004). The time interval affects but moderately (2-fold and 1.2-fold) increased the AUC the extent of interaction because rifampin is also of its potentially active metabolites (Tornio et al., 2006). a competitive inhibitor of OATP1B1, CYP2C8 and Also many other drugs are substrates of CYP2C8 in CYP3A4 (Kajosaari et al., 2005a; Varma et al., 2013). vitro, but their concomitant administration with gemfi- Interestingly, intake of St John’s Wort for 14 days has brozil has not appreciably increased their AUC, suggesting had no significant effect of the pharmacokinetics of that the CYP2C8-mediated biotransformation is of lim- repaglinide (Fan et al., 2011). ited significance to their total clearance (Table 1). Rifampin has also reduced the concentrations of the Many compounds are moderate inhibitors of CYP2C8 thiazolidinediones pioglitazone and rosiglitazone. Ri- in vitro, but their concomitant ingestion with repagli- fampin caused a substantial (54%) decrease in the AUC nide or other CYP2C8 substrates does not raise expo- of pioglitazone and increased the ratios of metabolite sure to these substrates in humans. The reason for the M-IV to pioglitazone and of M-III to pioglitazone in urine apparent discrepancy between the in vitro and in vivo by 98 and 95% (Jaakkola et al., 2006a). Similarly, results can be, for example, their low potency as rifampin reduced the mean AUC of rosiglitazone by CYP2C8 inhibitors or their high protein binding in vivo 54% and increased the formation of N-demethylrosigli- (e.g., montelukast). tazone (Niemi et al., 2004a). In Korean men, rifampin Some parent drugs such as gemfibrozil and clopidog- decreased rosiglitazone AUC by 65% (Park et al., 2004). rel are relatively weak inhibitors of CYP2C8 in vitro, Addition of tuberculosis treatment, containing rifam- but they are metabolized in vivo to glucuronide metab- pin, to treatment of a woman with type 2 diabetes olites, which are potent CYP2C8 inhibitors. In general, caused her to lose glycemic control, demonstrating poten- negative interaction results with gemfibrozil in vivo tial clinical significance of the rifampin-rosiglitazone exclude a clinically meaningful interaction mediated interaction (Pimazoni, 2009). by CYP2C8 inhibition. On the other hand, increased exposure to a victim drug by gemfibrozil does not yet VII. Points to Consider When Investigating indicate that CYP2C8 has a significant role in its Cytochrome P450 2C8-Mediated Drug metabolism because there may be other mechanisms Metabolism and Interactions mediating the observed interaction. Patients often concomitantly use different drugs Studies focusing on drug metabolism and metabolic that together inhibit several CYP enzymes, e.g., drug-drug interactions are an essential part of modern CYP1A2, CYP2C8, CYP2C9, CYP2B6, CYP2D6, or drug development, from early preclinical phases to CYP3A4. The combined inhibition of two or more of the clinical development phase and beyond. By using these enzymes often results in patients in a stronger specific and sensitive research methods, it is possible interaction than is caused by inhibition of a single to get a detailed and accurate view of potential issues enzyme in healthy volunteer studies. This aspect related to variability in drug metabolism already during together with other causes of interindividual variation the preclinical and early clinical phases of development. should be taken into consideration when the results of Methods to investigate CYP2C8 in vitro and in clinical experimental interaction studies in healthy volunteers studies have evolved markedly even during the last are translated into the clinic. decade.

D. Induction-Mediated Drug Interactions A. In Vitro Rifampin (rifampicin) can markedly increase the 1. General Aspects. Comprehensive in vitro studies clearance of many CYP2C8 substrate drugs, decrease to investigate the roles of different CYP enzymes in the Role of CYP2C8 in Drug Metabolism and Interactions 223

TABLE 11 Drug-drug interactions caused by the CYP2C8-inducing drug rifampin (rifampicin) in humans

Time Interval from the Inducer Dosing Previous Rifampin Dose to Substrate AUC Decrease References Substrate Ingestion

hours % 600 mg once daily for 6 days 13 Pioglitazone 54 Jaakkola et al., 2006a - Active metabolite: ketopioglitazone (M-III) 39 Jaakkola et al., 2006a - Active metabolite: hydroxypioglitazone (M-IV) 34 Jaakkola et al., 2006a 600 mg once daily for 5 days 12.5 Repaglinide 57 Niemi et al., 2000 600 mg once daily for 7 days 1 31 Hatorp et al., 2003 600 mg once daily for 7 days 0 48 Bidstrup et al., 2004 24 80 Bidstrup et al., 2004 600 mg once daily for 5 days 13 Rosiglitazone 54 Niemi et al., 2004a 600 mg once daily for 6 days 12 65 Park et al., 2004

AUC, area under the plasma concentration-time curve. metabolism of a (new) drug and to uncover its potential montelukast, rosiglitazone, pioglitazone, and cerivasta- for causing inhibition or induction of drug metabolism tin, each having its specific strengths and weaknesses are typically conducted already during the early pre- (Tables 12 and 13). clinical phases of drug development. The results from Paclitaxel 6-a-hydroxylation is the prototypical these studies are then used for in vitro-in vivo extra- marker reaction for CYP2C8 (Rahman et al., 1994; polations, to anticipate factors affecting the clearance Sonnichsen et al., 1995). It is highly selective for of the drug as well as its potential to act as a perpetrator CYP2C8, but the metabolic turnover is fairly low, often of pharmacokinetic drug interactions, i.e., to affect the leading to relatively long incubation times, which may clearance of other drugs. The prerequisite for accurate lead to significant inhibitor metabolism/depletion dur- extrapolations is that in vitro investigations are con- ing the incubation (Table 13). This may partly explain ducted with care and are sufficiently comprehensive, why paclitaxel seems to be less sensitive to competitive avoiding the many pitfalls of in vitro studies, under- CYP2C8 inhibitors than most other CYP2C8 marker standing the many limitations of the different ap- substrates (VandenBrink et al., 2011). In particular, proaches, and covering complex issues, such as the long incubation times should be avoided when studying potential for autoinhibition or -induction. Yet it should the potential for time-dependent or mechanism-based be understood that accurate extrapolations are not inhibition in systems based on a preincubation step, possible without some clinical pharmacokinetic data because inactivation proceeding during the incubation at the relevant dose of the investigational drug. may decrease the sensitivity of the experimental system The general aspects as well as the potential pitfalls of to detect inactivation. in vitro studies and extrapolations are well covered by many Amodiaquine metabolism to N-desethylamodiaquine excellent review articles and guidelines (Houston and is probably the second most used CYP2C8 marker Galetin, 2008; Pelkonen et al., 2008; Grimm et al., 2009; reaction. It is well characterized and highly selective EMA, 2012b; Pelkonen, 2015; http://www.fda.gov/Drugs/ for CYP2C8 and has a high turnover (Li et al., 2002), DevelopmentApprovalProcess/DevelopmentResources/ allowing for short incubation times. Overall, it seems to DrugInteractionsLabeling/ucm093664.htm). Therefore, have no major drawbacks in in vitro use. this review focuses on issues that are directly related Few years ago, montelukast, a selective competitive to CYP2C8, i.e., in vitro methods used for measurement inhibitor of CYP2C8, was shown to be a potential of CYP2C8 activity (e.g., to test the potential of the CYP2C8 marker substrate, because its 36-hydroxylation investigational drug to inhibit CYP2C8 activity) and (M6 formation) is mediated primarily by CYP2C8 with reaction phenotyping (does CYP2C8 metabolize the a minor contribution by CYP2C9 (Filppula et al., 2011). drug) and in vivo studies to characterize the drug In a successive study, montelukast 36-hydroxylation interaction potential of the new drug (either as a proved to be a sensitive and useful reaction to in- perpetrator or victim drug). vestigate CYP2C8 inhibition in vitro (VandenBrink 2. Assessment of CYP2C8 Activity In Vitro. et al., 2011). One of the weaknesses of montelukast Specific assessment of CYP2C8 activity is necessary, in is that it is highly susceptible to microsomal protein particular when studying the potential of a drug to cause binding, necessitating careful standardization of incuba- inhibition of CYP2C8 but also when using a panel of HLM tion conditions (Walsky et al., 2005b). for reaction phenotyping using the correlation approach. Of the other potential marker reactions, cerivastatin An ideal in vitro probe substrate is selective/specific, has 6-hydroxylation (M-23 formation) seems to be highly a sufficient turnover, follows Michaelis-Menten kinetics, specific for CYP2C8 (Wang et al., 2002; Shitara et al., and is not sensitive to experimental conditions. There are 2004). In addition, the hydroxylations of rosiglitazone several useful, selective probe substrates to study (p-hydroxylation) (Baldwin et al., 1999) and pioglita- CYP2C8 in vitro, including paclitaxel, amodiaquine, zone (M-IV formation; Jaakkola et al., 2006c) seem to be 224 Backman et al.

TABLE 12 CYP2C8 substrate, inhibitor, and inducer probes recommended for drug-drug interaction studies

EMAa FDAb Helsinki DDI Group Probe In Vitro In Vivo In Vitro In Vivo In Vitro In Vivo Substrate Paclitaxel Amodiaquine Paclitaxelc Repaglinide Amodiaquine Repaglinide Amodiaquine Repaglinide Amodiaquine Rosiglitazone Paclitaxel Montelukast Rosiglitazone Montelukast Pioglitazone Cerivastatin (acid) Rosiglitazone (Pioglitazone) (Dasabuvir) Inhibitor Montelukast Gemfibrozil Montelukastc Gemfibrozil Montelukast Gemfibrozil Quercetinc Gemfibrozil Clopidogrel 1-O-b glucuronide Trimethoprim Clopidogrel acyl (Trimethoprim) 1-b-D-glucuronide Gemfibrozil (Trimethoprim) Rosiglitazone Pioglitazone Inducer None recommended None recommended Rifampin (rifampicin) Rifampin (rifampicin) Rifampin (rifampicin) Rifampin (rifampicin)

DDI, drug-drug interaction; EMA, European Medicines Agency; FDA, Food and Drug Administration. aEMA, 2012b. bhttp://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/ucm093664.htm (Accessed September 15, 2015). cPreferred. relatively, albeit not completely, selective for CYP2C8. One of the most widely used chemical CYP2C8 Finally, the most used in vivo CYP2C8 probe drug inhibitors is quercetin (Rahman et al., 1994). However, repaglinide, although sometimes recommended as an in it is neither very selective for CYP2C8 nor very strong vitro probe (Kajosaari et al., 2005a; VandenBrink et al., and therefore, it can barely be recommended as a 2011), is challenging to use in vitro, e.g., because of a diagnostic inhibitor. Today, there are several more need for extremely low substrate concentrations and selective alternatives available, including trimetho- lack of commercially available metabolite standards prim, montelukast, and gemfibrozil 1-O-b-glucuronide. (Table 13). The IC50 of trimethoprim for CYP2C8 is approxi- 3. In Vitro Methods to Estimate the Contribution mately 50 mM, i.e., it is not a very strong inhibitor, but of CYP2C8 in the Metabolism of a Drug. The basic its IC50 for other CYP enzymes is at least one order of methods used for estimating the contributions of CYP magnitude greater, making it a relatively selective enzymes to the metabolism of a drug, i.e., the so-called inhibitor (Wen et al., 2002). Montelukast, on the other reaction phenotyping, are the use of diagnostic inhibi- hand, is a potent and highly selective competitive tors in a complete natural system, such as HLM, and the inhibitor of CYP2C8, with an IC50 as low as 0.01 mM, use of recombinant expressed enzymes. In both ap- when a low microsomal protein concentration is used, proaches, knowledge of clinically relevant concentra- whereas its IC50 for other CYP enzymes is at least two tions of the drug is a prerequisite for estimation of the orders of magnitude greater (Walsky et al., 2005b). The contributions of the different CYP enzymes in vivo. The major drawback of montelukast seems to be its non- advantage of HLM is the natural composition of the specific microsomal protein binding, whereby increas- system, allowing relatively straightforward estimation ing the microsomal protein concentration by 80-fold of the contributions. However, this approach requires yields an about 100-fold decrease in its inhibition human material collected according to high ethical potency (Walsky et al., 2005b). standards and is entirely dependent on the strength The mechanism-based CYP2C8 inactivator gemfibrozil and specificity of the inhibitors. On the other hand, 1-O-b-glucuronide is another appealing CYP2C8 inhibi- although recombinant expressed enzymes can be tor. With a 30-minute preincubation, its IC50 for CYP2C8 regarded as a specific tool, in vivo extrapolations of is about 2 mM, whereas its IC50 values for CYP1A2, recombinant enzyme results require the use of en- CYP2B6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 are zyme source and batch specific conversion factors more than 300 mM, suggesting an even better selectivity (preferably based on enzyme activity), complicating than that of montelukast (Ogilvie et al., 2006). Moreover, the extrapolations. it is unlikely to be markedly affected by microsomal Recombinant expressed human CYP2C8 is commer- protein concentration. Whether clopidogrel acyl-b-D- cially available at least as bacterial cell- and insect cell- glucuronide is a similarly selective CYP2C8 inactivator based products. During the last decade, both chemical remains to be investigated (Tornio et al., 2014). inhibitors and inhibitory antibodies have become avail- able that are both CYP2C8 specific and strong. In the B. In Vivo following, we review the documentation regarding 1. General Aspects. Current guidelines recommend chemical CYP2C8 inhibitors. the conduct of clinical drug-drug interaction studies on Role of CYP2C8 in Drug Metabolism and Interactions 225

the basis of in vitro studies on the CYP inhibitory effects of the drug and its main circulating metabolites (potential perpetrator), as well as on the basis of the results of the in vitro reaction phenotyping studies of the drug and its main metabolic pathways (victim). A rational selection for the first CYP-specific clinical studies is to focus on the enzyme that is inhibited most (lowest IC50/Ki) by the drug or its metabolite, prefer- ably using the highest clinically used dose of the drug, also a substrate ofand OATP1B1 CYP3A4 half-life, not easily available in all countries and OAT3 and on the enzyme that is considered the most Long half-life Also CYP2B6 inhibitor Lack of documentation Weak inhibitor Moderate inhibitor of OATP1B1 important in its own metabolism. For the first type of studies, a sensitive and selective in vivo probe substrate is used, and for the second type of studies, a strong and selective in vivo probe inhibitor is needed. In Vivo For CYP2C8, there are several alternative probe substrates and a few inhibitors that can be used in clinical trials. The contribution of CYP2C8 enzyme to the total clearance of its substrates varies greatly (Table 1). Most CYP2C8 substrates are partially metabolized also substrate of OATP1B1 OATP1B1 or CYP3A4

Strength, not an inhibitor of by other enzymes, are substrates of some membrane Sensitivity, short half-life Reduces blood glucose levels, Relative sensitivity, safety Medium long half-life Relative sensitivity, safety, not a Safety, not a substrate of OATP1B1 Only moderate sensitivity, long Strong, well-documented inducer Nonselective Sensitive Selectivity Strength, selectivity transporters, or are excreted in urine or feces in un- changed form. Thus, the significance of CYP2C8 in interactions cannot be calculated directly from changes in victim drug AUC. If the CYP2C8 substrate drug (Rifampicin) is also a substrate of transporters or other CYP Repaglinide Montelukast Pioglitazone Rosiglitazone Rifampin Dasabuvir Clopidogrel Trimethoprim Gemfibrozil enzymes, their contribution needs to be considered in the interaction, as exemplified in the dissection of the gemfibrozil-repaglinide interaction (Honkalammi et al., 2011a, 2012). For example, gemfibrozil is in vivo TABLE 13 an inhibitor of CYP2C8 as well as of OATP1B1 and OAT3, and it can increase the AUC of certain drugs (e.g., pravastatin), which are not substrates of CYP2C8 but are substrates for OATP1B1 or OAT3 (Kyrklund et al., 2003). On the other hand, CYP2C8 inhibitors usually increase the AUC of CYP2C8 substrates less than they diminish the CYP2C8-specific metabolic solubility issues for very low substrateissues concentration, with protein binding contribution by CYP3A4, requirement for low substrate concentration a preincubation microsomal protein binding requires a preincubation routes, because the CYP2C8-independent elimination Strengths and weaknesses of the recommended probe compounds Low/intermediate turnover, Nonselective Selectivity not documented, requires Availability of reference compounds Lack of metabolite standards, Selectivity not well documented, Low/intermediate turnover Low/intermediate turnover routes remain unaffected. CYP2C8-mediated drug interactions are often stud-

In Vitro ied in healthy volunteers in a randomized crossover manner by administering a potential substrate drug with and without a probe inhibitors of CYP2C8, such as the recommended probe inhibitor gemfibrozil (Table 12). To better simulate real clinical situations turnover documentation inducer in which patients often are using several different drugs Selectivity, high Selectivity, extensive Selectivity Contribution by CYP2C9, requirement Selectivity Selectivity Potency, selectivityPotency, well-documented Also a substrate of CYP2C8, Selectivity Selectivity Potency concomitantly, substrate drugs can be administered in multiple-phase studies, given alone, with an inhibitor of b CYP2C8 (gemfibrozil), with an inhibitor of another relevant CYP enzyme (e.g., with CYP3A4 inhibitor

Probe Strengths Weaknessesitraconazole), Probe and Strengths together with a Weaknesses combination of -D-glucuronide

b inhibitors. However, there are only a few studies in (Rifampicin) glucuronide 1- Amodiaquine Paclitaxel Montelukast Repaglinide Cerivastatin (acid) Montelukast Pioglitazone Gemfibrozil 1-O- Rosiglitazone Rifampin Clopidogrel acyl which the effects of multiple inhibitors (e.g., inhibitors of CYP2C8 and CYP3A4) on the pharmacokinetics of their substrate drugs have been studied both separately and together (Niemi et al., 2003b, 2006; Jaakkola et al., Substrate Inhibitor Inducer 2005; Karonen et al., 2012). 226 Backman et al.

2. In Vivo Cytochrome P450 2C8 Probe Substrates. However, there is very little clinical documentation for its The crucial characteristics of an in vivo probe substrate use as a probe drug. Moreover, the safety of amodiaquine are its selectivity and sensitivity. In an ideal case, at as an in vivo probe drug in drug-drug interactions studies least 80% of the substrate is metabolized by the enzyme seems to be questionable (German et al., 2007). of interest, allowing for an interpretation based on the Of the other potential in vivo probe substrates of AUC of the parent drug. In some cases, the use of an CYP2C8, the two thiazolidinediones pioglitazone and enzyme-specific metabolite to parent ratio may be rosiglitazone are the best documented. As pointed out used to increase sensitivity and specificity, but with in the previous section, CYP2C8 is the main enzyme an additional caveat because of potential variability in mediating their primary hydroxylation reactions in metabolite elimination. The feasibility of an in vivo vitro (Baldwin et al., 1999; Jaakkola et al., 2006c). probe substrate also depends heavily on its safety, in Accordingly, the typical dosing of gemfibrozil 600 mg particular when large increases in its systemic concen- twice daily, which has been estimated to cause over 95% trations can be anticipated. Moreover, the pharma- inhibition of CYP2C8 (Fig. 9; Honkalammi et al., 2012), cokinetic characteristics of the probe may affect its increased the AUC of rosiglitazone about 2.3-fold and suitability. For example, a probe substrate with a that of pioglitazone 3.2-4.3-fold, simultaneously reduc- significant first-pass metabolism and short elimination ing the concentrations of their hydroxyl metabolites half-life may be able to catch even transient changes in (Niemi et al., 2003a; Deng et al., 2005; Jaakkola et al., enzyme activity, which may be necessary when study- 2005; Aquilante et al., 2013a). Unlike repaglinide, they ing inhibitors with a short half-life or time-dependent are insensitive to OATP1B1 function (Kalliokoski et al., changes in enzyme activity. 2008a). However, they have a long half-life, necessitat- The antidiabetic agent repaglinide is overall the most ing an up to 72-hour blood sampling period for a full studied and best documented in vivo probe substrate of pharmacokinetic analysis. On the basis of its better CYP2C8, and consequently both the European Medi- availability and sensitivity to CYP2C8 inhibition, pio- cines Agency (EMA) and FDA recommend its use as a glitazone is slightly preferable over rosiglitazone as a CYP2C8 probe (Table 12). Although in vitro studies CYP2C8 probe. are not fully consistent with the major in vivo role of The leukotriene receptor antagonist montelukast is CYP2C8 in the total metabolism of repaglinide (Gan another sensitive CYP2C8 substrate that could be used et al., 2010; Säll et al., 2012; Varma et al., 2013, 2015), as a CYP2C8 probe. Gemfibrozil has increased its AUC repaglinide seems to be very sensitive to inhibitors of almost fivefold and markedly reduced the formation of CYP2C8 activity, such as gemfibrozil, trimethoprim, its 36-hydroxylated metabolite (Karonen et al., 2010). and clopidogrel (Niemi et al., 2003b, 2004b; Tornio et al., On the other hand, montelukast is also partially 2014). On the basis of detailed mechanistic drug-drug metabolized by CYP3A4 in vitro (Filppula et al., 2011; interaction studies with the strong CYP2C8 inactivator VandenBrinketal.,2011).However,thestrong gemfibrozil, it has been estimated that the contribution CYP3A4 inhibitor itraconazole has had no effect on of CYP2C8 to repaglinide (0.25 mg) metabolism is about montelukast concentrations (Karonen et al., 2012), 85%, indicating that the AUC of repaglinide can be indicating that the role of CYP3A4 in montelukast increased up to an average of sevenfold by strong metabolism is minor. Moreover, montelukast is not CYP2C8 inhibition (Honkalammi et al., 2012). The known to be a substrate for OATP1B1. half-life of repaglinide is also relatively short (1 hour), In addition to the above substrates, there are some which allows for a full pharmacokinetic study within 1 other CYP2C8 substrate drugs that could be used as in day and can be beneficial when a measure of CYP2C8 vivo markers on the basis of their sensitivity to interact activity within a narrow time frame is desired. The with gemfibrozil. Such drugs include, for example, weakness of repaglinide is that it is partially metabo- daprodustat and dasabuvir. However, the former is lized by CYP3A4 (Bidstrup et al., 2003; Niemi et al., not yet on the market, and the second one is expensive, 2003b; Kajosaari, 2005a) and also a substrate of and more documentation is needed before they can be OATP1B1 (Niemi et al., 2005b). Thus, e.g., the effect of recommended as probe substrates. gemfibrozil on repaglinide pharmacokinetics is par- 3. In Vivo Cytochrome P450 2C8 Probe Inhibitors. tially mediated by inhibition of OATP1B1, in addition Probe inhibitors are needed for studying the contri- to inhibition of CYP2C8 (Honkalammi et al., 2011a). bution of CYP2C8 in the metabolism of a new drug, as Moreover, as it increases insulin secretion from pancre- well as for documenting the risk of drug-drug interac- atic b cells, there is a risk of hypoglycemia, particularly tions affecting the drug in vivo. Among clinically used when it is given to healthy subjects. Thus, the smallest CYP2C8 inhibitors, gemfibrozil is the strongest known. possible dose (e.g., 0.25 mg) of repaglinide should be Its CYP2C8 inhibitory effect is also highly selective due used, and meals, close follow up, and blood glucose to the specific mechanism that is mediated via specific monitoring be arranged when repaglinide is used. CYP2C8 inactivation by the glucuronide metabolite of Theoretically, the antimalarial agent amodiaquine and gemfibrozil (Ogilvie et al., 2006). In vitro, parent its N-deethylation could be useful in vivo CYP2C8 probes. gemfibrozil inhibits CYP2C9 activity with a fairly low Role of CYP2C8 in Drug Metabolism and Interactions 227

Ki (about 6 mM), but its inhibitory effects on the other inhibitor documented so far, increasing the AUC of main CYP enzymes are much weaker (Backman et al., repaglinide about fivefold (Tornio et al., 2014). Clopi- 2000; Wen et al., 2001; Wang et al., 2002). In clinical dogrel is obviously also a useful diagnostic CYP2C8 studies, gemfibrozil at a dose of 600 mg twice daily has inhibitor, but it is not fully selective and has not been not increased the concentrations of the CYP2C9 sub- extensively documented so far. In addition to strongly strate warfarin (Lilja et al., 2005) or had any effect that inhibiting CYP2C8, clopidogrel is also a moderate could be due to inhibition of CYP3A4 on the concentra- inhibitor of CYP2B6 (Turpeinen et al., 2005). Further- tions of the parent lactone forms of simvastatin and more, it has been suspected of causing CYP2C19 in- lovastatin (Backman et al., 2000; Kyrklund et al., 2001). hibition (Nishiya et al., 2009). However, it seems to have On the other hand, gemfibrozil has drastically, up to practically no effect on CYP3A4 or OATP1B1 activities 18.6-fold, increased the AUCs of CYP2C8 substrate in vivo (Tornio et al., 2014; Itkonen et al., 2015). drugs (Fig. 7; Table 10), suggesting that with regard to CYP enzymes, the inhibitory effect of gemfibrozil is VIII. Conclusions and Future Prospects highly selective for CYP2C8. The CYP2C8 inhibitory effect of gemfibrozil is strong, CYP2C8 is one of the main oxidative drug metabo- rapid, and long lasting. In studies using repaglinide as lizing enzymes in the liver. Its expression and function the CYP2C8 probe substrate, subtherapeutic doses of have been studied in detail, and for example, it has gemfibrozil have considerably elevated the concentra- been estimated that its in vivo turnover half-life is tions of repaglinide (Honkalammi et al., 2011a, 2012), about 22 hours in humans. The CYP2C8 gene contains and it has been estimated that the clinically used 9 exons and shares 74% sequence homology with gemfibrozil dosing (600 mg twice daily) inhibits CYP2C9. More than 100 nonsynonymous CYP2C8 CYP2C8 activity by about 99% and that one-tenth of SNVs are known to date, but only some of them are this dose would already lead to more than 90% in- associated with functional variability. Interethnic hibition of CYP2C8 (Fig. 9). Although CYP2C8 inhi- and geographical differences exist in the frequency bition by gemfibrozil is based on time-dependent of variants. For example, the low-activity variant inactivation of the enzyme by the primary glucuronide CYP2C8*2 (c.805A.T) is common in Africans but rare metabolite of gemfibrozil, CYP2C8 inhibition occurs in Caucasians and Asians. CYP2C8*3 (c.416G.A) and rapidly after gemfibrozil dosing. When repaglinide CYP2C8*4 (c.792C.G), on the other hand, are com- was given 0, 1, 3, or 6 hours after a single 600 mg dose mon in Caucasians but rare or absent in Africans of gemfibrozil, the AUC of repaglinide was increased and Asians (Fig. 6). The interethnic characterization 5.0-, 6.3-, 6.6-, and 5.4-fold, respectively, indicating that and functional activity of variants deserve further strong inhibition of CYP2C8 can be achieved almost studies. immediately after a single dose of gemfibrozil (Fig. 8, Studies on the role of CYP2C8 in drug metabolism Honkalammi et al., 2011b). It has also been demon- have demonstrated that it is the most important strated that the CYP2C8 inhibitory effect of gemfibrozil enzyme for the elimination of several drugs, such as persists virtually unchanged throughout the typical cerivastatin, montelukast, repaglinide, pioglitazone, 12-hour dosing interval of gemfibrozil (Tornio et al., and rosiglitazone, whose metabolism had been earlier 2008a). Thus, gemfibrozil can have a strong effect on thought to be attributed mainly to other enzymes. CYP2C8 substrates, irrespective of their half-life or CYP2C8 is crucial also for the biotransformation of time of daily dosing relative to gemfibrozil administra- daprodustat (GSK12788693), enzalutamide, dasabuvir, tion (Fig. 9; Table 10), making it an ideal in vivo probe and many other recently developed new drugs, and inhibitor of CYP2C8. The only caveat with gemfibrozil is overall, it contributes to the elimination of more than that it is also a moderate inhibitor of OATP1B1 and 100 drugs. CYP2C8 has a large active site cavity, and OAT3 and can therefore also increase the concentra- it can accommodate and also metabolize certain acyl tions of some drugs that are not or only partially glucuronides, such as desloratadine, diclofenac, and metabolized by CYP2C8 (Kyrklund et al., 2001, 2003; sipoglitazar glucuronides. Backman et al., 2002; Schneck et al., 2004; Neuvonen In vitro, there are several marker reactions for the et al., 2006; Whitfield et al., 2011). assessment of CYP2C8 activity, including paclitaxel Compared with gemfibrozil, all other clinically docu- 6-a-hydroxylation, amodiaquine deethylation, montelu- mented CYP2C8 inhibitors seem to be suboptimal. kast 36-hydroxylation, cerivastatin 6-hydroxylation Trimethoprim is relatively selective for CYP2C8, but (M-23 formation), rosiglitazone parahydroxylation, and as expected from its in vitro inhibitory effects (Wen pioglitazone M-IV formation. Each of these reactions et al., 2002), it is only a weak CYP2C8 inhibitor at has its strengths and weaknesses. The use of clinically clinically feasible doses (Niemi et al., 2004a,b; Hruska relevant drug concentrations in vitro is a prerequisite et al., 2005; Tornio et al., 2008b), and therefore it can for the estimation of the contribution of different only be regarded as a confirmatory CYP2C8 inhibitor in CYP enzymes in vivo. Gemfibrozil 1-O-b-glucuronide vivo. Clopidogrel is the second strongest CYP2C8 is potent and selective as a diagnostic inhibitor of 228 Backman et al.

CYP2C8-mediated metabolism in vitro. Quercetin and Authorship Contributions trimethoprim are relatively weak and unselective as Participated in research design: Backman, Filppula, Niemi, and CYP2C8 inhibitors, whereas montelukast is potent Neuvonen. Wrote or contributed to the writing of the manuscript: Backman, and selective, but suffers from nonspecific protein Filppula, Niemi, and Neuvonen. binding. Also clopidogrel acyl 1-b-D-glucuronide may be a suitable in vitro inhibitor, but further documentation References is needed. 1000 Genomes Project Consortium, Abecasis GR, Auton A, Brooks LD, DePristo MA, Durbin RM, Handsaker RE, Kang HM, Marth GT, and McVean GA; (2012) An Many drugs are CYP2C8 inhibitors or inducers. integrated map of genetic variation from 1,092 human genomes. Nature 491:56–65. Gemfibrozil is in vivo, unlike in vitro, a potent, irre- Abass K, Turpeinen M, and Pelkonen O (2009) An evaluation of the cytochrome P450 inhibition potential of selected pesticides in human hepatic microsomes. J Environ versible inhibitor of CYP2C8 via formation of gemfi- Sci Health B 44:553–563. brozil 1-O-b-glucuronide, and it is widely used as a Achour B, Barber J, and Rostami-Hodjegan A (2014) Expression of hepatic drug- metabolizing cytochrome p450 enzymes and their intercorrelations: a meta-anal- probe inhibitor. Also clopidogrel acyl 1-b-D-glucuronide ysis. Drug Metab Dispos 42:1349–1356. Adjei GO, Kristensen K, Goka BQ, Hoegberg LC, Alifrangis M, Rodrigues OP, causes metabolism-dependent inactivation of CYP2C8, and Kurtzhals JA (2008) Effect of concomitant artesunate administration and cy- indicating that glucuronides may contribute as CYP2C8 tochrome P4502C8 polymorphisms on the pharmacokinetics of amodiaquine in Ghanaian children with uncomplicated malaria. Antimicrob Agents Chemother 52: inhibitors to drug-drug interactions. Also efavirenz, 4400–4406. trimethoprim, and several protein kinase inhibitors Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS, and Sunyaev SR (2010) A method and server for predicting damaging missense are inhibitors of CYP2C8. mutations. Nat Methods 7:248–249. Agúndez JA, García-Martín E, and Martínez C (2009) Genetically based impairment in In vivo studies in humans on the role of CYP2C8 CYP2C8- and CYP2C9-dependent NSAID metabolism as a risk factor for gastroin- are challenged by the lack of suitable selective probe testinal bleeding: is a combination of pharmacogenomics and metabolomics required to improve personalized medicine? Expert Opin Drug Metab Toxicol 5:607–620. substrates, inhibitors, and inducers. Although repagli- Akande AA, Olugbenga SJ, Adebanjoi AJ, Toyin AS, and Ogobna OC (2015) Effects of nide, pioglitazone, rosiglitazone, and montelukast are co-trimoxazole co-administration on the pharmacokinetics of amodiaquine in healthy volunteers. Int J Pharm Pharm Sci 7:9; 272–276. useful probe substrates, they all have their pros and Albassam AA, Mohamed ME, and Frye RF (2015) Inhibitory effect of six herbal extracts on CYP2C8 enzyme activity in human liver microsomes. Xenobiotica 45: cons, as discussed in the text before. With regard to 406–412. CYP enzymes, gemfibrozil is a selective inhibitor of Albrecht W, Unger A, Nussler AK, and Laufer S (2008) In vitro metabolism of 2-[6-(4-chlorophenyl)-2,2-dimethyl-7-phenyl-2,3-dihydro-1H-pyrrolizin-5-yl] acetic acid CYP2C8 in vivo. Already very small doses of gemfibrozil, (licofelone, ML3000), an inhibitor of cyclooxygenase-1 and -2 and 5-lipoxygenase. Drug i.e., about 10% of its usual therapeutic dose, rapidly Metab Dispos 36:894–903. Ando A, Oshida K, Fukuyama S, Watanabe A, Hashimoto H, and Miyamoto Y (2012) cause a strong and long-lasting inactivation of CYP2C8. Identification of human cytochrome P450 enzymes involved in the metabolism of a novel к-opioid receptor agonist, nalfurafine hydrochloride. Biopharm Drug Dispos However, gemfibrozil is also an inhibitor of OATP1B1 33:257–264. and OAT3 transporters, which challenges interpreta- Ando Y, Shimizu T, Nakamura K, Mushiroda T, Nakagawa T, Kodama T, and Kamataki T (1998) Potent and non-specific inhibition of cytochrome P450 by tion of the interaction mechanisms if the CYP2C8 JM216, a new oral platinum agent. Br J Cancer 78:1170–1174. substrates are also substrates for these transporters. Aquilante CL, Bushman LR, Knutsen SD, Burt LE, Rome LC, and Kosmiski LA (2008) Influence of SLCO1B1 and CYP2C8 gene polymorphisms on rosiglitazone Rifampin is a very unselective albeit strong inducer of pharmacokinetics in healthy volunteers. Hum Genomics 3:7–16. Aquilante CL, Kosmiski LA, Bourne DW, Bushman LR, Daily EB, Hammond KP, CYP2C8. Hopley CW, Kadam RS, Kanack AT, and Kompella UB (2013a) Impact of the If a drug is significantly metabolized by CYP2C8 and CYP2C8 *3 polymorphism on the drug-drug interaction between gemfibrozil and pioglitazone. Br J Clin Pharmacol 75:217–226. CYP3A4, its concomitant administration with inhibi- Aquilante CL, Niemi M, Gong L, Altman RB, and Klein TE (2013b) PharmGKB tors of both of these enzymes, e.g., with gemfibrozil and summary: very important pharmacogene information for cytochrome P450, family 2, subfamily C, polypeptide 8. Pharmacogenet Genomics 23:721–728. itraconazole, can cause a much stronger interaction Aquilante CL, Wempe MF, Spencer SH, Kosmiski LA, Predhomme JA, and Sidhom MS (2013c) Influence of CYP2C8*2 on the pharmacokinetics of pioglitazone in than is the sum of their separate effects. Thus, the drug- healthy African-American volunteers. Pharmacotherapy 33:1000–1007. drug interaction studies performed by using inhibitors Arbus C, Benyamina A, Llorca PM, Baylé F, Bromet N, Massiere F, Garay RP, and Hameg A (2007) Characterization of human cytochrome P450 enzymes in- of one enzyme only may greatly underestimate the volved in the metabolism of cyamemazine. Eur J Pharm Sci 32:357–366. true risks as shown by the clinically very important Arnaldo P, Thompson RE, Lopes MQ, Suffys PN, and Santos AR (2013) Frequencies of cytochrome P450 2B6 and 2C8 allelic variants in the mozambican population. CYP2C8-mediated interactions affecting repaglinide Malays J Med Sci 20:13–23. Arun KP, Meda VS, Kucherlapati VSP, Dubala A, Deepalakshmi M, Anand Vijaya- or cerivastatin. At least theoretically, small doses of Kumar PR, Elango K, and Suresh B (2012) Pharmacokinetic drug interaction be- gemfibrozil or other inhibitors of CYPC8 could be tween gemfibrozil and sitagliptin in healthy Indian male volunteers. Eur J Clin Pharmacol 68:709–714. used as a booster to optimize the pharmacokinetics Arun Kumar AS, Kumar SS, Umamaheswaran G, Kesavan R, Balachandar J, of CYP2C8 substrate drugs or to prevent formation of and Adithan C (2015) Association of CYP2C8, CYP2C9 and CYP2J2 gene poly- morphisms with myocardial infarction in South Indian population. Pharmacol Rep potentially toxic metabolites via CYP2C8-mediated 67:97–101. Attar M, Dong D, Ling KH, and Tang-Liu DD (2003) Cytochrome P450 2C8 and reaction. flavin-containing monooxygenases are involved in the metabolism of tazarotenic acid in humans. Drug Metab Dispos 31:476–481. Backman JT, Honkalammi J, Neuvonen M, Kurkinen KJ, Tornio A, Niemi M, Acknowledgments and Neuvonen PJ (2009) CYP2C8 activity recovers within 96 hours after gemfi- brozil dosing: estimation of CYP2C8 half-life using repaglinide as an in vivo probe. The authors thank Dr. Tommi Nyrönen for producing the docking Drug Metab Dispos 37:2359–2366. simulations and three-dimensional artwork in Fig. 2. Backman JT, Kajosaari LI, Neuvonen M, and Neuvonen PJ (2003) Trimethoprim Backman, Niemi, and Neuvonen have filed a patent application increases the plasma concentrations of cerivastatin by inhibiting its CYP2C8- mediated metabolism, Abstract in 8th European ISSX Meeting, Dijon, France concerning use of gemfibrozil as a pharmacokinetic enhancer. (April 27-May 1, 2003). Some of the information in Tables 1, 3 and 6 is based on the UW Backman JT, Kyrklund C, Kivistö KT, Wang JS, and Neuvonen PJ (2000) Plasma Metabolism and Transport Drug Interaction Database (DIDB), concentrations of active simvastatin acid are increased by gemfibrozil. Clin – – Pharmacol Ther 68:122 129. Copyright University of Washington 1999 2015, as specified in the Backman JT, Kyrklund C, Neuvonen M, and Neuvonen PJ (2002) Gemfibrozil greatly footnotes to the tables. increases plasma concentrations of cerivastatin. Clin Pharmacol Ther 72:685–691. Role of CYP2C8 in Drug Metabolism and Interactions 229

Bae SH, Kwon MJ, Park JB, Kim D, Kim DH, Kang JS, Kim CG, Oh E, and Bae SK Cameron MD, Wright J, Black CB, and Ye N (2007) In vitro prediction and in vivo (2014) Metabolic drug-drug interaction potential of macrolactin A and 7-O-succinyl verification of enantioselective human tofisopam metabolite profiles. Drug Metab macrolactin A assessed by evaluating cytochrome P450 inhibition and induction Dispos 35:1894–1902. and UDP-glucuronosyltransferase inhibition in vitro. Antimicrob Agents Chemo- Capozzi ME, McCollum GW, and Penn JS (2014) The role of cytochrome P450 ther 58:5036–5046. in retinal angiogenesis. Invest Ophthalmol Vis Sci 55:4253–4260. Baer BR, DeLisle RK, and Allen A (2009) Benzylic oxidation of gemfibrozil-1-O-beta- Cardoso JO, Oliveira RV, Lu JB, and Desta Z (2015) In vitro metabolism of mon- glucuronide by P450 2C8 leads to heme alkylation and irreversible inhibition. telukast by cytochrome P450s (CYPs) and UDP-glucuronosyltransferases Chem Res Toxicol 22:1298–1309. (UGTs). Drug Metab Dispos 10.1124/dmd.115.065763. Bahadur N, Leathart JB, Mutch E, Steimel-Crespi D, Dunn SA, Gilissen R, Houdt Cavaco I, Mårtensson A, Fröberg G, Msellem M, Björkman A, and Gil JP (2013) JV, Hendrickx J, Mannens G, and Bohets H (2002) CYP2C8 polymorphisms in CYP2C8 status of patients with malaria influences selection of Plasmodium fal- Caucasians and their relationship with paclitaxel 6alpha-hydroxylase activity in ciparum pfmdr1 alleles after amodiaquine-artesunate treatment. J Infect Dis 207: human liver microsomes. Biochem Pharmacol 64:1579–1589. 687–688. Balla B, Vaszilko M, Kósa JP, Podani J, Takács I, Tóbiás B, Nagy Z, Lazáry A, Cavaco I, Piedade R, Gil JP, and Ribeiro V (2006) CYP2C8 polymorphism among the and Lakatos P (2012) New approach to analyze genetic and clinical data in Portuguese. Clin Chem Lab Med 44:168–170. bisphosphonate-induced osteonecrosis of the jaw. Oral Dis 18:580–585. Cavaco I, Strömberg-Nörklit J, Kaneko A, Msellem MI, Dahoma M, Ribeiro VL, Bajpai P, Srinivasan S, Ghosh J, Nagy LD, Wei S, Guengerich FP, and Avadhani NG Bjorkman A, and Gil JP (2005) CYP2C8 polymorphism frequencies among malaria (2014) Targeting of splice variants of human cytochrome P450 2C8 (CYP2C8) to patients in Zanzibar. Eur J Clin Pharmacol 61:15–18. mitochondria and their role in arachidonic acid metabolism and respiratory dys- Cazali N, Tran A, Treluyer JM, Rey E, d’Athis P, Vincent J, and Pons G (2003) function. J Biol Chem 289:29614–29630. Inhibitory effect of stiripentol on carbamazepine and saquinavir metabolism in Baldwin SJ, Clarke SE, and Chenery RJ (1999) Characterization of the cytochrome human. Br J Clin Pharmacol 56:526–536. P450 enzymes involved in the in vitro metabolism of rosiglitazone. Br J Clin Chang JT, Staffa JA, Parks M, and Green L (2004) Rhabdomyolysis with HMG-CoA Pharmacol 48:424–432. reductase inhibitors and gemfibrozil combination therapy. Pharmacoepidemiol Ballard JE, Prueksaritanont T, and Tang C (2007) Hepatic metabolism of MK-0457, a Drug Saf 13:417–426. potent aurora kinase inhibitor: interspecies comparison and role of human cyto- Chang SY, Chen C, Yang Z, and Rodrigues AD (2009) Further assessment of 17alpha- chrome P450 and flavin-containing monooxygenase. Drug Metab Dispos 35: ethinyl estradiol as an inhibitor of different human cytochrome P450 forms in vitro. 1447–1451. Drug Metab Dispos 37:1667–1675. Barbosa-Sicard E, Markovic M, Honeck H, Christ B, Muller DN, and Schunck WH Chang SY, Li W, Traeger SC, Wang B, Cui D, Zhang H, Wen B, and Rodrigues AD (2005) Eicosapentaenoic acid metabolism by cytochrome P450 enzymes of the (2008) Confirmation that cytochrome P450 2C8 (CYP2C8) plays a minor role in (S)-(+)- CYP2C subfamily. Biochem Biophys Res Commun 329:1275–1281. and (R)-(-)-ibuprofen hydroxylation in vitro. Drug Metab Dispos 36:2513–2522. Baune B, Flinois JP, Furlan V, Gimenez F, Taburet AM, Becquemont L, Chang TK, Weber GF, Crespi CL, and Waxman DJ (1993) Differential activation of and Farinotti R (1999) Halofantrine metabolism in microsomes in man: major role cyclophosphamide and ifosphamide by cytochromes P-450 2B and 3A in human of CYP 3A4 and CYP 3A5. J Pharm Pharmacol 51:419–426. liver microsomes. Cancer Res 53:5629–5637. Becquemont L, Mouajjah S, Escaffre O, Beaune P, Funck-Brentano C, and Jaillon P Chang TK, Yu L, Maurel P, and Waxman DJ (1997) Enhanced cyclophosphamide and (1999) Cytochrome P-450 3A4 and 2C8 are involved in zopiclone metabolism. Drug ifosfamide activation in primary human hepatocyte cultures: response to cyto- Metab Dispos 27:1068–1073. chrome P-450 inducers and autoinduction by oxazaphosphorines. Cancer Res 57: Bergmann TK, Brasch-Andersen C, Gréen H, Mirza M, Pedersen RS, Nielsen F, 1946–1954. Skougaard K, Wihl J, Keldsen N, and Damkier P (2011) Impact of CYP2C8*3 on Chang Y, Fang WB, Lin SN, and Moody DE (2011) Stereo-selective metabolism of paclitaxel clearance: a population pharmacokinetic and pharmacogenomic study in methadone by human liver microsomes and cDNA-expressed cytochrome P450s: a 93 patients with ovarian cancer. Pharmacogenomics J 11:113–120. reconciliation. Basic Clin Pharmacol Toxicol 108:55–62. Bergmann TK, Filppula AM, Launiainen T, Nielsen F, Backman JT, and Brosen K Chang Y, Moody DE, and McCance-Katz EF (2006) Novel metabolites of buprenor- (2015) Neurotoxicity and low paclitaxel clearance associated with concomitant phine detected in human liver microsomes and human urine. Drug Metab Dispos clopidogrel therapy in a 60 year old Caucasian woman with ovarian carcinoma. 34:440–448. Br J Clin Pharmacol doi: 10.1111/bcp.12795. Chen C and Wang DW (2015) Cytochrome P450-CYP2 family- role in Bidstrup TB, Bjørnsdottir I, Sidelmann UG, Thomsen MS, and Hansen KT (2003) inflammation and cancer. Adv Pharmacol 74:193–221. CYP2C8 and CYP3A4 are the principal enzymes involved in the human in vitro Chen X, Kosoglou T, Statkevich P, Kumar B, Li J, Dockendorf MF, Wang G, Lowe RS, biotransformation of the insulin secretagogue repaglinide. Br J Clin Pharmacol 56: Jiang J, and Liu H (2014) Pharmacokinetics of vorapaxar and its metabolite fol- 305–314. lowing oral administration in healthy Chinese and American subjects. Int J Clin Bidstrup TB, Damkier P, Olsen AK, Ekblom M, Karlsson A, and Brøsen K (2006) The Pharmacol Ther 52:889–899. impact of CYP2C8 polymorphism and grapefruit juice on the pharmacokinetics of Chen Y, Coulter S, Jetten AM, and Goldstein JA (2009) Identification of human repaglinide. Br J Clin Pharmacol 61:49–57. CYP2C8 as a retinoid-related orphan nuclear receptor target gene. J Pharmacol Bidstrup TB, Stilling N, Damkier P, Scharling B, Thomsen MS, and Brøsen K (2004) Exp Ther 329:192–201. Rifampicin seems to act as both an inducer and an inhibitor of the metabolism of Chen Y and Goldstein JA (2009) The transcriptional regulation of the human CYP2C repaglinide. Eur J Clin Pharmacol 60:109–114. genes. Curr Drug Metab 10:567–578. Blanco JG, Harrison PL, Evans WE, and Relling MV (2000) Human cytochrome P450 Cherstniakova SA, Bi D, Fuller DR, Mojsiak JZ, Collins JM, and Cantilena LR maximal activities in pediatric versus adult liver. Drug Metab Dispos 28:379–382. (2001) Metabolism of vanoxerine, 1-[2-[bis(4-fluorophenyl)methoxy]ethyl]-4-(3- Boberg M, Angerbauer R, Fey P, Kanhai WK, Karl W, Kern A, Ploschke J, phenylpropyl)piperazine, by human cytochrome P450 enzymes. Drug Metab Dispos and Radtke M (1997) Metabolism of cerivastatin by human liver microsomes in 29:1216–1220. vitro. Characterization of primary metabolic pathways and of cytochrome P450 Chiba M, Xu X, Nishime JA, Balani SK, and Lin JH (1997) Hepatic microsomal isozymes involved. Drug Metab Dispos 25:321–331. metabolism of montelukast, a potent leukotriene D4 receptor antagonist, in hu- Bogman K, Silkey M, Chan SP, Tomlinson B, and Weber C (2010) Influence of mans. Drug Metab Dispos 25:1022–1031. CYP2C19 genotype on the pharmacokinetics of R483, a CYP2C19 substrate, in Choi SY, Koh KH, and Jeong H (2013) Isoform-specific regulation of cytochromes healthy subjects and type 2 diabetes patients. Eur J Clin Pharmacol 66: P450 expression by estradiol and progesterone. Drug Metab Dispos 41:263–269. 1005–1015. Churchill FC, Patchen LC, Campbell CC, Schwartz IK, Nguyen-Dinh P, Bonierbale E, Valadon P, Pons C, Desfosses B, Dansette PM, and Mansuy D (1999) and Dickinson CM (1985) Amodiaquine as a prodrug: importance of metabolite(s) Opposite behaviors of reactive metabolites of tienilic acid and its isomer toward in the antimalarial effect of amodiaquine in humans. Life Sci 36:53–62. liver proteins: use of specific anti-tienilic acid-protein adduct antibodies and the Cizkova K, Konieczna A, Erdosova B, and Ehrmann J (2014) Time-dependent ex- possible relationship with different hepatotoxic effects of the two compounds. Chem pression of cytochrome p450 epoxygenases during human prenatal development. Res Toxicol 12:286–296. Organogenesis 10:53–61. Bort R, Gómez-Lechón MJ, Castell JV, and Jover R (2004) Role of hepatocyte nuclear Cresteil T, Monsarrat B, Alvinerie P, Tréluyer JM, Vieira I, and Wright M (1994) factor 3 gamma in the expression of human CYP2C genes. Arch Biochem Biophys Taxol metabolism by human liver microsomes: identification of cytochrome P450 426:63–72. isozymes involved in its biotransformation. Cancer Res 54:386–392. Bu HZ, Zhao P, Kang P, Pool WF, and Wu EY (2006) Identification of enzymes Cresteil T, Monsarrat B, Dubois J, Sonnier M, Alvinerie P, and Gueritte F (2002) responsible for primary and sequential oxygenation reactions of capravirine in Regioselective metabolism of taxoids by human CYP3A4 and 2C8: structure- human liver microsomes. Drug Metab Dispos 34:1798–1802. activity relationship. Drug Metab Dispos 30:438–445. Bun SS, Ciccolini J, Bun H, Aubert C, and Catalin J (2003) Drug interactions of Dai D, Zeldin DC, Blaisdell JA, Chanas B, Coulter SJ, Ghanayem BI, and Goldstein paclitaxel metabolism in human liver microsomes. J Chemother 15:266–274. JA (2001) Polymorphisms in human CYP2C8 decrease metabolism of the anti- Burnett BP, Pillai L, Bitto A, Squadrito F, and Levy RM (2011) Evaluation of CYP450 cancer drug paclitaxel and arachidonic acid. Pharmacogenetics 11:597–607. inhibitory effects and steady-state pharmacokinetics of genistein in combination Daikh BE, Lasker JM, Raucy JL, and Koop DR (1994) Regio- and stereoselective with cholecalciferol and citrated zinc bisglycinate in postmenopausal women. Int J epoxidation of arachidonic acid by human cytochromes P450 2C8 and 2C9. J Womens Health 3:139–150. Pharmacol Exp Ther 271:1427–1433. Busse D, Cosme J, Beaune P, Kroemer HK, and Eichelbaum M (1995) Cytochromes of Daily EB and Aquilante CL (2009) Cytochrome P450 2C8 pharmacogenetics: a review the P450 2C subfamily are the major enzymes involved in the O-demethylation of of clinical studies. Pharmacogenomics 10:1489–1510. verapamil in humans. Naunyn Schmiedebergs Arch Pharmacol 353:116–121. Damle BD, Dowell JA, Walsky RL, Weber GL, Stogniew M, and Inskeep PB (2009) In Bymaster FP, Chao P, Schulze H, Tran PV, and Marshall RD (2013) Bio- vitro and in vivo studies to characterize the clearance mechanism and potential pharmaceutical characterization, metabolism, and brain penetration of the triple cytochrome P450 interactions of anidulafungin. Antimicrob Agents Chemother 53: reuptake inhibitor amitifadine. Drug Metab Lett 7:23–33. 1149–1156. Cai X, Wang RW, Edom RW, Evans DC, Shou M, Rodrigues AD, Liu W, Dean DC, Davis JA, Rock DA, Wienkers LC, and Pearson JT (2012) In vitro characterization of and Baillie TA (2004) Validation of (-)-N-3-benzyl-phenobarbital as a selective in- the drug-drug interaction potential of catabolites of antibody-maytansinoid con- hibitor of CYP2C19 in human liver microsomes. Drug Metab Dispos 32:584–586. jugates. Drug Metab Dispos 40:1927–1934. 230 Backman et al.

Dekhuijzen PN and Koopmans PP (2002) Pharmaokinetic profile of zafirlukast. Clin English BC, Baum CE, Adelberg DE, Sissung TM, Kluetz PG, Dahut WL, Price DK, Pharmacokinet 41:105–114. and Figg WD (2010) A SNP in CYP2C8 is not associated with the development of Delaforge M, Pruvost A, Perrin L, and André F (2005) Cytochrome P450-mediated bisphosphonate-related osteonecrosis of the jaw in men with castrate-resistant oxidation of glucuronide derivatives: example of estradiol-17beta-glucuronide oxi- prostate cancer. Ther Clin Risk Manag 6:579–583. dation to 2-hydroxy-estradiol-17beta-glucuronide by CYP 2C8. Drug Metab Dispos Erve JC, Gauby S, Maynard JW Jr, Svensson MA, Tonn G, and Quinn KP (2013) 33:466–473. Bioactivation of sitaxentan in liver microsomes, hepatocytes, and expressed human Delozier TC, Kissling GE, Coulter SJ, Dai D, Foley JF, Bradbury JA, Murphy E, P450s with characterization of the glutathione conjugate by liquid chromatography Steenbergen C, Zeldin DC, and Goldstein JA (2007) Detection of human CYP2C8, tandem mass spectrometry. Chem Res Toxicol 26:926–936. CYP2C9, and CYP2J2 in cardiovascular tissues. Drug Metab Dispos 35:682–688. Fan G, Cao Y, and Yang J (2015) In vitro inhibition and induction of human cyto- Deng LJ, Wang F, and Li HD (2005) Effect of gemfibrozil on the pharmacokinetics of chrome P450 enzymes by SIPI5357, a potential antidepressant. Biopharm Drug pioglitazone. Eur J Clin Pharmacol 61:831–836. Dispos 10.1002/bdd.1947. Derks M, Fowler S, and Kuhlmann O (2009) In vitro and in vivo assessment of the Fan L, Zhou G, Guo D, Liu YL, Chen WQ, Liu ZQ, Tan ZR, Sheng D, Zhou HH, effect of dalcetrapib on a panel of CYP substrates. Curr Med Res Opin 25:891–902. and Zhang W (2011) The pregnane X receptor agonist St John’s Wort has no effects Desai PB, Duan JZ, Zhu YW, and Kouzi S (1998) Human liver microsomal metabolism on the pharmacokinetics and pharmacodynamics of repaglinide. Clin Pharmaco- of paclitaxel and drug interactions. Eur J Drug Metab Pharmacokinet 23:417–424. kinet 50:605–611. Desta Z, Soukhova N, Mahal SK, and Flockhart DA (2000) Interaction of cisapride Fasinu PS, Gutmann H, Schiller H, Bouic PJ, and Rosenkranz B (2013a) The po- with the human cytochrome P450 system: metabolism and inhibition studies. Drug tential of Hypoxis hemerocallidea for herb-drug interaction. Pharm Biol 51: Metab Dispos 28:789–800. 1499–1507. Desta Z, Soukhova N, Morocho AM, and Flockhart DA (2001) Stereoselective me- Fasinu PS, Gutmann H, Schiller H, James AD, Bouic PJ, and Rosenkranz B (2013b) tabolism of cisapride and enantiomer-enantiomer interaction in human cytochrome The potential of Sutherlandia frutescens for herb-drug interaction. Drug Metab P450 enzymes: major role of CYP3A. J Pharmacol Exp Ther 298:508–520. Dispos 41:488–497. Desta Z, Ward BA, Soukhova NV, and Flockhart DA (2004) Comprehensive evalua- FDA (1998) Centre for drug evaluation and research: Clinical pharmacology and tion of tamoxifen sequential biotransformation by the human cytochrome P450 biopharmaceutics review(s): Singulair. Available from: http://www.accessdata.fda. system in vitro: prominent roles for CYP3A and CYP2D6. J Pharmacol Exp Ther gov/drugsatfda_docs/nda/98/020829s000_Singular_Clin_Pharm_Biopharm.pdf – 310:1062 1075. (Accessed September 15, 2015). Dierks EA, Stams KR, Lim HK, Cornelius G, Zhang H, and Ball SE (2001) A method FDA (1999) Actos prescribing information. Available from: http://www.accessdata. for the simultaneous evaluation of the activities of seven major human drug- fda.gov/drugsatfda_docs/label/1999/21073lbl.pdf (Accessed September 15, 2015). metabolizing cytochrome P450s using an in vitro cocktail of probe substrates and FDA (2001) Centre for drug evaluation and research: Clinical pharmacology and fast gradient liquid chromatography tandem mass spectrometry. Drug Metab biopharmaceutics review(s): Gleevec. Available from: http://www.accessdata.fda. – Dispos 29:23 29. gov/drugsatfda_docs/nda/2001/21-335_Gleevec_biopharmr_P1.pdf (Accessed Sep- Dinger J, Meyer MR, and Maurer HH (2014) Development of an in vitro cytochrome tember 15, 2015). P450 cocktail inhibition assay for assessing the inhibition risk of drugs of abuse. FDA (2004) Centre for drug evaluation and research: Clinical pharmacology and – Toxicol Lett 230:28 35. biopharmaceutics review(s): Tarceva. Available from: http://www.accessdata.fda. Dixit V, Hariparsad N, Li F, Desai P, Thummel KE, and Unadkat JD (2007) Cyto- gov/drugsatfda_docs/nda/2004/21-743_Tarceva_biopharmr.pdf (Accessed Septem- chrome P450 enzymes and transporters induced by anti-human immunodeficiency ber 15, 2015). virus protease inhibitors in human hepatocytes: implications for predicting clinical FDA (2005a) Centre for drug evaluation and research: Clinical pharmacology and – drug interactions. Drug Metab Dispos 35:1853 1859. biopharmaceutics review(s): Exjade. Available from: http://www.accessdata.fda. Doecke CJ, Sansom LN, and McManus ME (1990) Phenytoin 4-hydroxylation by gov/drugsatfda_docs/nda/2005/021882_s000_Exjade_BioPharmr.pdf (Accessed Sep- rabbit liver P450IIC3 and identification of orthologs in human liver microsomes. – tember 15, 2015). Biochem Biophys Res Commun 166:860 866. FDA (2005b) Centre for drug evaluation and research: Clinical pharmacology and Doehmer J, Weiss G, McGregor GP, and Appel K (2011) Assessment of a dry extract biopharmaceutics review: Nevaxar. Available from: http://www.accessdata.fda.gov/ from milk thistle (Silybum marianum) for interference with human liver drugsatfda_docs/nda/2005/021923_s000_Nexavar_BioPharmR.pdf (Accessed Sep- cytochrome-P450 activities. Toxicol In Vitro 25:21–27. tember 15, 2015). Donato MT, Jiménez N, Castell JV, and Gómez-Lechón MJ (2004) Fluorescence- FDA (2006a) Centre for drug evaluation and research: Clinical pharmacology and based assays for screening nine cytochrome P450 (P450) activities in intact cells biopharmaceutics review(s): Sprycel. Available from: http://www.accessdata.fda. expressing individual human P450 enzymes. Drug Metab Dispos 32:699–706. gov/drugsatfda_docs/nda/2006/021986s000_Sprycel__ClinPharmR.pdf (Accessed Sep- Dong PP, Fang ZZ, Zhang YY, Ge GB, Mao YX, Zhu LL, Qu YQ, Li W, Wang LM, tember 15, 2015). and Liu CX (2011) Substrate-dependent modulation of the catalytic activity of FDA (2006b) Centre for drug evaluation and research: Clinical pharmacology and CYP3A by erlotinib. Acta Pharmacol Sin 32:399–407. biopharmaceutics review: Sutent. Available from: http://www.accessdata.fda.gov/ Duester G (2008) Retinoic acid synthesis and signaling during early organogenesis. drugsatfda_docs/nda/2006/021938_S000_Sutent_BioPharmR.pdf (Accessed Sep- Cell 134:921–931. Dussault I, Lin M, Hollister K, Wang EH, Synold TW, and Forman BM (2001) tember 15, 2015). Peptide mimetic HIV protease inhibitors are ligands for the orphan receptor SXR. FDA (2007a) Centre for drug evaluation and research: Clinical pharmacology and J Biol Chem 276:33309–33312. biopharmaceutics review(s): Bystolic. Available from: www.accessdata.fda.gov/ Dutheil F, Dauchy S, Diry M, Sazdovitch V, Cloarec O, Mellottée L, Bièche I, drugsatfda_docs/nda/2007/021742s000_ClinPharmR_P1.pdf (Accessed September Ingelman-Sundberg M, Flinois JP, de Waziers I, Beaune P, Declèves X, Duyckaerts 15, 2015). C, and Loriot MA (2009) Xenobiotic-metabolizing enzymes and transporters in the FDA (2007b) Centre for drug evaluation and research: Clinical pharmacology and normal : regional and cellular mapping as a basis for putative roles in biopharmaceutics review(s): Neupro. Available from: http://www.accessdata.fda. cerebral function. Drug Metab Dispos 37:1528–1538. gov/drugsatfda_docs/nda/2007/021829s000_ClinPharmR_P2.pdf (Accessed Sep- Eagling VA, Howe JL, Barry MJ, and Back DJ (1994) The metabolism of zidovudine tember 15, 2015). by human liver microsomes in vitro: formation of 39-amino-39-deoxythymidine. FDA (2007c) Centre for drug evaluation and research: Clinical pharmacology and Biochem Pharmacol 48:267–276. biopharmaceutics review(s): Tasigna. Available from: http://www.accessdata.fda. Eckland DA and Danhof M (2000) Clinical pharmacokinetics of pioglitazone. Exp Clin gov/drugsatfda_docs/nda/2007/022068s000_ClinPharmR.pdf (Accessed September Endocrinol Diabetes 108 (Suppl 2):S234–S242. 15, 2015). Elliot DJ, Suharjono, Lewis BC, Gillam EM, Birkett DJ, Gross AS, and Miners JO FDA (2007d) Centre for drug evaluation and research: Clinical pharmacology and (2007) Identification of the human cytochromes P450 catalysing the rate-limiting biopharmaceutics review(s): Torisel. Available from: http://www.accessdata.fda. pathways of gliclazide elimination. Br J Clin Pharmacol 64:450–457. gov/drugsatfda_docs/nda/2007/022088s000_ClinPharmR.pdf (Accessed September Enayetallah AE, French RA, Thibodeau MS, and Grant DF (2004) Distribution of 15, 2015). soluble epoxide and of cytochrome P450 2C8, 2C9, and 2J2 in human FDA (2007e) Centre for drug evaluation and research: Clinical pharmacology and tissues. J Histochem Cytochem 52:447–454. biopharmaceutics review(s): Tykerb. Available from: http://www.accessdata.fda. EMA (2012a) Assessment report: Zytiga. Available from: http://www.ema.europa.eu/ gov/drugsatfda_docs/nda/2007/022059s000_ClinPharmR.pdf (Accessed September docs/en_GB/document_library/EPAR_-_Assessment_Report_-_Variation/human/002321/ 15, 2015). WC500137814.pdf (Accessed September 15, 2015). FDA (2008a) Centre for drug evaluation and research: Clinical pharmacology and EMA (2012b) Guideline on the investigation of drug interactions. Available from: biopharmaceutics review(s): Intelence. Available from: http://www.accessdata.fda. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2012/ gov/drugsatfda_docs/nda/2008/022187s000_ClinPharmR_P5.pdf (Accessed Sep- 07/WC500129606.pdf (Accessed September 15, 2015). tember 15, 2015). EMA (2012c) Summary of product characteristics: Xalkori. Available from: http://ec.europa. FDA (2008b) Centre for drug evaluation and research: Clinical pharmacology and eu/health/documents/community-register/2012/20121023124213/anx_124213_en.pdf biopharmaceutics review(s): Pristiq. Available from: http://www.accessdata.fda. (Accessed September 15, 2015). gov/drugsatfda_docs/nda/2008/021992s000_ClinPharmR_P2.pdf (Accessed Sep- EMA (2012d) Zelboraf, Procedural steps taken and scientific information after the tember 15, 2015). authorisation. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/ FDA (2008c) Centre for drug evaluation and research: Clinical pharmacology and EPAR_-_Procedural_steps_taken_and_scientific_information_after_authorisation/human/ biopharmaceutics review(s): Promacta. Available from: http://www.accessdata.fda. 002409/WC500137805.pdf (Accessed September 15, 2015). gov/drugsatfda_docs/nda/2008/022291s000_ClinPharmR_P1.pdf (Accessed Sep- EMA (2014) Summary of product characteristics: Simbrinza. Available from: http://ec.europa. tember 15, 2015). eu/health/documents/community-register/2014/20140718129060/anx_129060_en.pdf FDA (2009a) Centre for drug evaluation and research: Clinical pharmacology and (Accessed September 15, 2015). biopharmaceutics review(s): Multaq. Available from: http://www.accessdata.fda. Engel G, Hofmann U, Heidemann H, Cosme J, and Eichelbaum M (1996) Antipyrine gov/drugsatfda_docs/nda/2009/022425s000_ClinPharm_P1.pdf (Accessed Septem- as a probe for human oxidative drug metabolism: identification of the cytochrome ber 15, 2015). P450 enzymes catalyzing 4-hydroxyantipyrine, 3-hydroxymethylantipyrine, and FDA (2009b) Centre for drug evaluation and research: Clinical pharmacology and norantipyrine formation. Clin Pharmacol Ther 59:613–623. biopharmaceutics review(s): Tyvaso. Available from: http://www.accessdata.fda. Role of CYP2C8 in Drug Metabolism and Interactions 231

gov/drugsatfda_docs/nda/2009/022387s000ClinPharmR.pdf (Accessed September FDA (2013a) Actos prescribing information. Available from: http://www.accessdata.fda. 15, 2015). gov/drugsatfda_docs/label/2013/021073s046lbl.pdf (Accessed September 15, 2015). FDA (2009c) Centre for drug evaluation and research: Clinical pharmacology and FDA (2013b) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Uloric. Available from: http://www.accessdata.fda.gov/ biopharmaceutics review(s): Adempas. Available from: http://www.accessdata.fda. drugsatfda_docs/nda/2009/021856s000_ClinPharmR_P1.pdf (Accessed September gov/drugsatfda_docs/nda/2013/204819Orig1s000ClinPharmR.pdf (Accessed Sep- 15, 2015). tember 15, 2015). FDA (2009d) Centre for drug evaluation and research: Clinical pharmacology and FDA (2013c) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Votrient. Available from: http://www.accessdata.fda. biopharmaceutics review(s): Breo Ellipta. Available from: http://www.accessdata. gov/drugsatfda_docs/nda/2009/022465s000_ClinPharmR.pdf (Accessed September fda.gov/drugsatfda_docs/nda/2013/204275Orig1s000ClinPharmR.pdf (Accessed Sep- 15, 2015). tember 15, 2015). FDA (2010a) Centre for drug evaluation and research: Clinical pharmacology and FDA (2013d) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Jevtana. Available from: http://www.accessdata.fda. biopharmaceutics review(s): Imbruvica. Available from: http://www.accessdata.fda. gov/drugsatfda_docs/nda/2010/201023s000ClinPharmR.pdf (Accessed September gov/drugsatfda_docs/nda/2013/205552Orig1s000ClinPharmR.pdf (Accessed Sep- 15, 2015). tember 15, 2015). FDA (2010b) Centre for drug evaluation and research: Pharmacology review(s): FDA (2013e) Centre for drug evaluation and research: Clinical pharmacology and Ella. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/nda/2010/ biopharmaceutics review(s): Invokana. Available from: http://www.accessdata.fda. 022474s000PharmR.pdf (Accessed September 15, 2015). gov/drugsatfda_docs/nda/2013/204042Orig1s000ClinPharmR.pdf (Accessed Sep- FDA (2011a) Centre for drug evaluation and research: Clinical pharmacology and tember 15, 2015). biopharmaceutics review(s): Arcapta Neohaler. Available from: www.accessdata. FDA (2013f) Centre for drug evaluation and research: Clinical pharmacology and fda.gov/drugsatfda_docs/nda/2011/022383Orig1s000ClinPharmR.pdf (Accessed Sep- biopharmaceutics review(s): Mekinist. Available from: http://www.accessdata.fda. tember 15, 2015). gov/drugsatfda_docs/nda/2013/204114Orig1s000ClinPharmR.pdf (Accessed Sep- FDA (2011b) Centre for drug evaluation and research: Clinical pharmacology and tember 15, 2015). biopharmaceutics review(s): Brilinta. Available from: http://www.accessdata.fda. FDA (2013g) Centre for drug evaluation and research: Clinical pharmacology and gov/drugsatfda_docs/nda/2011/022433Orig1s000ClinPharmR.pdf (Accessed Sep- biopharmaceutics review(s): Nesina. Available from: http://www.accessdata.fda. tember 15, 2015). gov/drugsatfda_docs/nda/2013/022271Orig1s000ClinPharmR.pdf (Accessed Sep- FDA (2011c) Centre for drug evaluation and research: Clinical pharmacology and tember 15, 2015). biopharmaceutics review(s): Edarbi. Available from: http://www.accessdata.fda. FDA (2013h) Centre for drug evaluation and research: Clinical pharmacology and bio- gov/drugsatfda_docs/nda/2011/200796Orig1s000ClinPharmR.pdf (Accessed Sep- pharmaceutics review(s): Olysio. Available from: http://www.accessdata.fda.gov/drug- tember 15, 2015). satfda_docs/nda/2013/205123Orig1s000ClinPharmR.pdf (Accessed September 15, 2015). FDA (2011d) Centre for drug evaluation and research: Clinical pharmacology and FDA (2013i) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Edurant. Available from: http://www.accessdata.fda. biopharmaceutics review(s): Osphena. Available from: www.accessdata.fda.gov/ gov/drugsatfda_docs/nda/2011/202022Orig1s000ClinPharmR.pdf (Accessed Sep- drugsatfda_docs/nda/2013/203505Orig1s000ClinPharmR.pdf (Accessed September tember 15, 2015). 15, 2015). FDA (2011e) Centre for drug evaluation and research: Clinical pharmacology and FDA (2013j) Centre for drug evaluation and research: Pharmacology review(s): biopharmaceutics review(s): Xalkori. Available from: http://www.accessdata.fda. Brintellix. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/nda/ gov/drugsatfda_docs/nda/2011/202570Orig1s000ClinPharmR.pdf (Accessed Sep- 2013/204447Orig1s000ClinPharmR.pdf (Accessed September 15, 2015). tember 15, 2015). FDA (2013k) Centre for drug evaluation and research: Pharmacology review(s): FDA (2011f) Centre for drug evaluation and research: Pharmacology review(s): Opsumit. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/nda/ Edarbi. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/ 2013/204410Orig1s000PharmR.pdf (Accessed September 15, 2015). 200796Orig1s000PharmR.pdf (Accessed September 15, 2015). FDA (2014a) Avandia, prescribing information. Available from: http://www.accessdata. FDA (2011g) Centre for drug evaluation and research: Pharmacology review(s): fda.gov/drugsatfda_docs/label/2014/021071s047s048s049lbl.pdf (Accessed September Viibryd. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/ 15, 2015). 022567Orig1s000ClinPharmR.pdf (Accessed September 15, 2015). FDA (2014b) Centre for drug evaluation and research: Clinical pharmacology and FDA (2012a) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Akynzeo. Available from: http://www.accessdata.fda. biopharmaceutics review(s): Aubagio. Available from: http://www.accessdata.fda. gov/drugsatfda_docs/nda/2014/205718Orig1s000ClinPharmR.pdf (Accessed Sep- gov/drugsatfda_docs/nda/2012/202992Orig1s000ClinpharmR.pdf (Accessed Sep- tember 15, 2015). tember 15, 2015). FDA (2014c) Centre for drug evaluation and research: Clinical pharmacology and FDA (2012b) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Beleodaq. Available from: http://www.accessdata.fda. biopharmaceutics review(s): Belviq. Available from: http://www.accessdata.fda.gov/ gov/drugsatfda_docs/nda/2014/206256Orig1s000ClinPharmR.pdf (Accessed Sep- drugsatfda_docs/nda/2012/022529Orig1s000ClinPharmR.pdf (Accessed September 15, 2015). tember 15, 2015). FDA (2012c) Centre for drug evaluation and research: Clinical pharmacology and FDA (2014d) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Cometriq. Available from: http://www.accessdata.fda. biopharmaceutics review(s): Ofev. Available from: http://www.accessdata.fda.gov/ gov/drugsatfda_docs/nda/2012/203756Orig1s000ClinPharmR.pdf (Accessed Sep- drugsatfda_docs/nda/2014/205832Orig1s000ClinPharmR.pdf (Accessed September tember 15, 2015). 15, 2015). FDA (2012d) Centre for drug evaluation and research: Clinical pharmacology and FDA (2014e) Centre for drug evaluation and research: Clinical pharmacology and bio- biopharmaceutics review(s): Eliquis. Available from: http://www.accessdata.fda. pharmaceutics review(s): Otezla. Available from: http://www.accessdata.fda.gov/ gov/drugsatfda_docs/nda/2012/202155Orig1s000ClinPharmR.pdf (Accessed Sep- drugsatfda_docs/nda/2014/205437Orig1s000ClinPharmR.pdf (Accessed September tember 15, 2015). 15, 2015). FDA (2012e) Centre for drug evaluation and research: Clinical pharmacology and FDA (2014f) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Iclusig. Available from: http://www.accessdata.fda. biopharmaceutics review(s): . Available from: http://www.accessdata.fda. gov/drugsatfda_docs/nda/2012/203469Orig1s000ClinPharmR.pdf (Accessed Sep- gov/drugsatfda_docs/nda/2014/203108Orig1s000ClinPharmR.pdf (Accessed Sep- tember 15, 2015). tember 15, 2015). FDA (2012f) Centre for drug evaluation and research: Clinical pharmacology and FDA (2014g) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Inlyta. Available from: http://www.accessdata.fda.gov/ biopharmaceutics review(s): Viekira Pak. Available from: http://www.accessdata. drugsatfda_docs/nda/2012/202324Orig1s000ClinPharmR.pdf (Accessed September fda.gov/drugsatfda_docs/nda/2014/206619Orig1s000ClinPharmR.pdf (Accessed 15, 2015). September 15, 2015). FDA (2012g) Centre for drug evaluation and research: Clinical pharmacology and FDA (2014h) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Kalydeco. Available from: http://www.accessdata.fda. biopharmaceutics review(s): Zydelig. Available from: http://www.accessdata.fda. gov/drugsatfda_docs/nda/2012/203188Orig1s000ClinPharmR.pdf (Accessed Sep- gov/drugsatfda_docs/nda/2014/206545Orig1s000ClinPharmR.pdf (Accessed Sep- tember 15, 2015). tember 15, 2015). FDA (2012h) Centre for drug evaluation and research: Clinical pharmacology and FDA (2014i) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Signifor. Available from: http://www.accessdata.fda. biopharmaceutics review(s): Zykadia. Available from: http://www.accessdata.fda. gov/drugsatfda_docs/nda/2012/200677Orig1s000ClinPharmR.pdf (Accessed Sep- gov/drugsatfda_docs/nda/2014/205755Orig1s000ClinPharmR.pdf (Accessed Sep- tember 15, 2015). tember 15, 2015). FDA (2012i) Centre for drug evaluation and research: Clinical pharmacology and FDA (2014j) Centre for drug evaluation and research: Clinical pharmacology and biopharmaceutics review(s): Stivarga. Available from: http://www.accessdata.fda. biopharmaceutics review: Belsomra. Available from: http://www.accessdata.fda. gov/drugsatfda_docs/nda/2012/203085Orig1s000ClinPharmR.pdf (Accessed Sep- gov/drugsatfda_docs/nda/2014/204569Orig1s000ClinPharmR.pdf (Accessed Sep- tember 15, 2015). tember 15, 2015). FDA (2012j) Centre for drug evaluation and research: Clinical pharmacology and FDA (2014k) Centre for drug evaluation and research: Pharmacology review(s): biopharmaceutics review(s): Stribild. Available from: http://www.accessdata.fda. Viekira Pak. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/nda/ gov/drugsatfda_docs/nda/2012/203100Orig1s000ClinPharmR.pdf (Accessed Sep- 2014/206619Orig1s000PharmR.pdf (Accessed September 15, 2015). tember 15, 2015). FDA (2014l) Centre for drug evaluation and research: Pharmacology review(s): FDA (2012k) Centre for drug evaluation and research: Clinical pharmacology and Zontivity. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/nda/ biopharmaceutics review(s): Xtandi. Available from: http://www.accessdata.fda. 2014/204886Orig1s000PharmR.pdf (Accessed September 15, 2015). gov/drugsatfda_docs/nda/2012/203415Orig1s000ClinPharmR.pdf (Accessed Sep- FDA (2014m) Hetlioz, prescribing information. Available from: http://www.accessdata. tember 15, 2015). fda.gov/drugsatfda_docs/nda/2014/205677Orig1s000ClinPharmR.pdf (Accessed Sep- FDA (2012l) Centre for drug evaluation and research: Pharmacology review(s): tember 15, 2015). Aubagio. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/ FDA (2015a) Centre for drug evaluation and research: Clinical pharmacology and 202992Orig1s000PharmR.pdf (Accessed September 15, 2015). biopharmaceutics review(s): Lenvima. Available from: http://www.accessdata.fda. 232 Backman et al.

gov/drugsatfda_docs/nda/2015/206947Orig1s000ClinPharmR.pdf (Accessed Sep- identification, and evaluation of cytochrome p450 inhibition. Drug Metab Dispos tember 15, 2015). 38:905–916. FDA (2015b) Reyataz prescribing information. Available from: http://www.accessdata. Gan J, Chen W, Shen H, Gao L, Hong Y, Tian Y, Li W, Zhang Y, Tang Y, and Zhang H fda.gov/drugsatfda_docs/label/2015/021567s037,206352s002lbl.pdf (Accessed Sep- (2010) Repaglinide-gemfibrozil drug interaction: inhibition of repaglinide glucur- tember 15, 2015). onidation as a potential additional contributing mechanism. Br J Clin Pharmacol Feidt DM, Klein K, Hofmann U, Riedmaier S, Knobeloch D, Thasler WE, Weiss TS, 70:870–880. Schwab M, and Zanger UM (2010) Profiling induction of cytochrome p450 enzyme Gan J, Liu-Kreyche P, and Humphreys WG (2012) In vitro assessment of cytochrome activity by statins using a new liquid chromatography-tandem mass spectrometry P450 inhibition and induction potential of tanespimycin and its major metabolite, cocktail assay in human hepatocytes. Drug Metab Dispos 38:1589–1597. 17-amino-17-demethoxygeldanamycin. Cancer Chemother Pharmacol 69:51–56. Feng R, Zhou X, Tan XS, Or PM, Hu T, Fu J, Ma JY, Huang M, He CY, and Shi JG Gao Y, Liu D, Wang H, Zhu J, and Chen C (2010) Functional characterization of five (2014) In vitro identification of cytochrome P450 isoforms responsible for the me- CYP2C8 variants and prediction of CYP2C8 genotype-dependent effects on in vitro tabolism of 1-hydroxyl-2,3,5-trimethoxy-xanthone purified from Halenia elliptica and in vivo drug-drug interactions. Xenobiotica 40:467–475. D. Don. Chem Biol Interact 210:12–19. García-Martín E, Martínez C, Ladero JM, and Agúndez JA (2006) Interethnic and Ferguson SS, Chen Y, LeCluyse EL, Negishi M, and Goldstein JA (2005) Human intraethnic variability of CYP2C8 and CYP2C9 polymorphisms in healthy indi- CYP2C8 is transcriptionally regulated by the nuclear receptors constitutive viduals. Mol Diagn Ther 10:29–40. androstane receptor, pregnane X receptor, glucocorticoid receptor, and hepatic García-Martín E, Martínez C, Tabarés B, Frías J, and Agúndez JA (2004) In- nuclear factor 4alpha. Mol Pharmacol 68:747–757. terindividual variability in ibuprofen pharmacokinetics is related to interaction of Filppula AM, Laitila J, Neuvonen PJ, and Backman JT (2011) Reevaluation of the cytochrome P450 2C8 and 2C9 amino acid polymorphisms. Clin Pharmacol Ther microsomal metabolism of montelukast: major contribution by CYP2C8 at clini- 76:119–127. cally relevant concentrations. Drug Metab Dispos 39:904–911. Gerbal-Chaloin S, Daujat M, Pascussi JM, Pichard-Garcia L, Vilarem MJ, Filppula AM, Laitila J, Neuvonen PJ, and Backman JT (2012) Potent mechanism- and Maurel P (2002) Transcriptional regulation of CYP2C9 gene. Role of gluco- based inhibition of CYP3A4 by imatinib explains its liability to interact with corticoid receptor and constitutive androstane receptor. J Biol Chem 277:209–217. CYP3A4 substrates. Br J Pharmacol 165:2787–2798. Gerbal-Chaloin S, Pascussi JM, Pichard-Garcia L, Daujat M, Waechter F, Fabre JM, Filppula AM, Neuvonen M, Laitila J, Neuvonen PJ, and Backman JT (2013a) Carrère N, and Maurel P (2001) Induction of CYP2C genes in human hepatocytes Autoinhibition of CYP3A4 leads to important role of CYP2C8 in imatinib metab- in primary culture. Drug Metab Dispos 29:242–251. olism: variability in CYP2C8 activity may alter plasma concentrations and re- German P, Greenhouse B, Coates C, Dorsey G, Rosenthal PJ, Charlebois E, Lindegardh sponse. Drug Metab Dispos 41:50–59. N, Havlir D, and Aweeka FT (2007) Hepatotoxicity due to a drug interaction between Filppula AM, Neuvonen PJ, and Backman JT (2014) In vitro assessment of time- amodiaquine plus artesunate and efavirenz. Clin Infect Dis 44:889–891. dependent inhibitory effects on CYP2C8 and CYP3A activity by fourteen protein Ghosal A, Chowdhury SK, Tong W, Hapangama N, Yuan Y, Su AD, and Zbaida S kinase inhibitors. Drug Metab Dispos 42:1202–1209. (2006) Identification of human liver cytochrome P450 enzymes responsible for the Filppula AM, Tornio A, Niemi M, Neuvonen PJ, and Backman JT (2013b) Gemfi- metabolism of lonafarnib (Sarasar). Drug Metab Dispos 34:628–635. brozil impairs imatinib absorption and inhibits the CYP2C8-mediated formation of Ghosal A, Hapangama N, Yuan Y, Lu X, Horne D, Patrick JE, and Zbaida S (2003) its main metabolite. Clin Pharmacol Ther 94:383–393. Rapid determination of enzyme activities of recombinant human cytochromes P450, Fischer V, Johanson L, Heitz F, Tullman R, Graham E, Baldeck JP, and Robinson human liver microsomes and hepatocytes. Biopharm Drug Dispos 24:375–384. WT (1999) The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor flu- Gibbons JA, de Vries M, Krauwinkel W, Ohtsu Y, Noukens J, van der Walt JS, Mol R, vastatin: effect on human cytochrome P-450 and implications for metabolic drug Mordenti J, and Ouatas T (2015) Pharmacokinetic drug interaction studies with interactions. Drug Metab Dispos 27:410–416. Enzalutamide. Clin Pharmacokinet 54:1057–1069. Fischer V, Rodríguez-Gascón A, Heitz F, Tynes R, Hauck C, Cohen D, and Vickers AE Gil JP and Gil Berglund E (2007) CYP2C8 and antimalaria drug efficacy. Pharma- (1998) The multidrug resistance modulator valspodar (PSC 833) is metabolized by cogenomics 8:187–198. human cytochrome P450 3A. Implications for drug-drug interactions and phar- Gorman GS, Coward L, Kerstner-Wood C, Cork L, Kapetanovic IM, Brouillette WJ, macological activity of the main metabolite. Drug Metab Dispos 26:802–811. and Muccio DD (2007) In vitro metabolic characterization, phenotyping, and ki- Flaherty KT, Lathia C, Frye RF, Schuchter L, Redlinger M, Rosen M, and O’Dwyer netic studies of 9cUAB30, a retinoid X receptor-specific retinoid. Drug Metab PJ (2011) Interaction of sorafenib and cytochrome P450 isoenzymes in patients Dispos 35:1157–1164. with advanced melanoma: a phase I/II pharmacokinetic interaction study. Cancer Grace JM, Skanchy DJ, and Aguilar AJ (1999) Metabolism of artelinic acid to dihy- Chemother Pharmacol 68:1111–1118. droqinqhaosu by human liver cytochrome P4503A. Xenobiotica 29:703–717. Fleming I (2014) The pharmacology of the cytochrome P450 epoxygenase/soluble Gray IC, Nobile C, Muresu R, Ford S, and Spurr NK (1995) A 2.4-megabase physical epoxide hydrolase axis in the vasculature and cardiovascular disease. Pharmacol map spanning the CYP2C gene cluster on chromosome 10q24. Genomics 28: Rev 66:1106–1140. 328–332. Floyd JS, Kaspera R, Marciante KD, Weiss NS, Heckbert SR, Lumley T, Wiggins KL, Gréen H, Khan MS, Jakobsen-Falk I, Åvall-Lundqvist E, and Peterson C (2011) Tamraz B, Kwok PY, and Totah RA (2012) A screening study of drug-drug inter- Impact of CYP3A5*3 and CYP2C8-HapC on paclitaxel/carboplatin-induced mye- actions in cerivastatin users: an adverse effect of clopidogrel. Clin Pharmacol Ther losuppression in patients with ovarian cancer. J Pharm Sci 100:4205–4209. 91:896–904. Grimm SW and Dyroff MC (1997) Inhibition of human drug metabolizing cyto- Foti RS, Pearson JT, Rock DA, Wahlstrom JL, and Wienkers LC (2009) In vitro chromes P450 by anastrozole, a potent and selective inhibitor of aromatase. Drug inhibition of multiple cytochrome P450 isoforms by xanthone derivatives from Metab Dispos 25:598–602. mangosteen extract. Drug Metab Dispos 37:1848–1855. Grimm SW, Einolf HJ, Hall SD, He K, Lim HK, Ling KH, Lu C, Nomeir AA, Seibert Foti RS, Pearson JT, Wong SL, Zalikowski JA, Boudreaux MD, Prokop SP, Davis JA, E, and Skordos KW (2009) The conduct of in vitro studies to address time- Banfield C, Emery MG, and Rock DA (2012) Predicting the drug interaction po- dependent inhibition of drug-metabolizing enzymes: a perspective of the pharma- tential of AMG 853, a dual antagonist of the D-prostanoid and chemoattractant ceutical research and manufacturers of America. Drug Metab Dispos 37: receptor-homologous molecule expressed on T helper 2 cells receptors. Drug Metab 1355–1370. Dispos 40:2239–2249. Guitton J, Buronfosse T, Desage M, Flinois JP, Perdrix JP, Brazier JL, and Beaune P Frank A and Unger M (2006) Analysis of frankincense from various Boswellia species (1998) Possible involvement of multiple human cytochrome P450 isoforms in the with inhibitory activity on human drug metabolising cytochrome P450 enzymes liver metabolism of propofol. Br J Anaesth 80:788–795. using liquid chromatography mass spectrometry after automated on-line extrac- Guo J, Nikolic D, Chadwick LR, Pauli GF, and van Breemen RB (2006) Identification tion. J Chromatogr A 1112:255–262. of human hepatic cytochrome P450 enzymes involved in the metabolism of 8-pre- Frick MH, Elo O, Haapa K, Heinonen OP, Heinsalmi P, Helo P, Huttunen JK, nylnaringenin and isoxanthohumol from hops (Humulus lupulus L.). Drug Metab Kaitaniemi P, Koskinen P, and Manninen V (1987) Helsinki Heart Study: primary- Dispos 34:1152–1159. prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of Ha-Duong NT, Dijols S, Macherey AC, Goldstein JA, Dansette PM, and Mansuy D treatment, changes in risk factors, and incidence of coronary heart disease. N Engl (2001) Ticlopidine as a selective mechanism-based inhibitor of human cytochrome JMed317:1237–1245. P450 2C19. Biochemistry 40:12112–12122. Fujino H, Saito T, Tsunenari Y, Kojima J, and Sakaeda T (2004) Metabolic properties Hagihara K, Nishiya Y, Kurihara A, Kazui M, Farid NA, and Ikeda T (2008) Com- of the acid and lactone forms of HMG-CoA reductase inhibitors. Xenobiotica 34: parison of human cytochrome P450 inhibition by the thienopyridines prasugrel, 961–971. clopidogrel, and ticlopidine. Drug Metab Pharmacokinet 23:412–420. Fujino H, Shimada S, Yamada I, Hirano M, Tsunenari Y, and Kojima J (2003a) Hakkola J, Pasanen M, Purkunen R, Saarikoski S, Pelkonen O, Mäenpää J, Rane A, Studies on the interaction between and statins using human hepatic mi- and Raunio H (1994) Expression of xenobiotic-metabolizing cytochrome P450 forms crosomes. Arzneimittelforschung 53:701–707. in human adult and fetal liver. Biochem Pharmacol 48:59–64. Fujino H, Yamada I, Shimada S, and Yoneda M (2001) Simultaneous determination Hall M, Matthews A, Paul DS, Foster JR, Holding JD, Islam K, and Brandt RD(2007) of taxol and its metabolites in microsomal samples by a simple thin-layer chro- Interactions of Iclaprim with Human Cytochrome P450 Enzymes, in 8th In- matography radioactivity assay–inhibitory effect of NK-104, a new inhibitor of ternational ISSX Meeting, Sendai, Japan; (October 9–12). Available from: http://issx. HMG-CoA reductase. J Chromatogr B Biomed Sci Appl 757:143–150. confex.com/issx/intl8/webprogram/Paper6798.html (Accessed August 26, 2015). Fujino H, Yamada I, Shimada S, Hirano M, Tsunenari Y, and Kojima J (2003b) Halling J, Petersen MS, Damkier P, Nielsen F, Grandjean P, Weihe P, Lundgren S, Interaction between fibrates and statins–metabolic interactions with gemfibrozil. Lundblad MS, and Brøsen K (2005) Polymorphism of CYP2D6, CYP2C19, CYP2C9 Drug Metabol Drug Interact 19:161–176. and CYP2C8 in the Faroese population. Eur J Clin Pharmacol 61:491–497. Furuta S, Kamada E, Sugimoto T, Kawabata Y, Wu XC, Skibbe J, Usuki E, Parkinson Hamitouche S, Poupon J, Dreano Y, Amet Y, and Lucas D (2006) Ethanol oxidation A, and Kurimoto T (2004) Involvement of CYP2C8 and UGT1A9 in the metabolism of into acetaldehyde by 16 recombinant human cytochrome P450 isoforms: role of a novel gastroprokinetic agent, Z-338, in 2nd Pharmaceutical Sciences World Con- CYP2C isoforms in human liver microsomes. Toxicol Lett 167:221–230. gress, Kyoto, Japan (May 31-June 5, 2004). Available from: http://www.cerep.fr/cerep/ Hamman MA, Thompson GA, and Hall SD (1997) Regioselective and stereoselective Users/pages/News/Publications/92.pdf (Accessed June 18, 2015). metabolism of ibuprofen by human cytochrome P450 2C. Biochem Pharmacol 54: Gallemann D, Wimmer E, Höfer CC, Freisleben A, Fluck M, Ladstetter B, and Dolgos 33–41. H (2010) In vitro characterization of sarizotan metabolism: hepatic clearance, Handschin C and Meyer UA (2003) Induction of drug metabolism: the role of nuclear identification and characterization of metabolites, drug-metabolizing enzyme receptors. Pharmacol Rev 55:649–673. Role of CYP2C8 in Drug Metabolism and Interactions 233

Hanioka N, Matsumoto K, Saito Y, and Narimatsu S (2010) Functional character- Iribarne C, Berthou F, Baird S, Dréano Y, Picart D, Bail JP, Beaune P, and Ménez JF ization of CYP2C8.13 and CYP2C8.14: catalytic activities toward paclitaxel. Basic (1996) Involvement of cytochrome P450 3A4 enzyme in the N-demethylation of Clin Pharmacol Toxicol 107:565–569. methadone in human liver microsomes. Chem Res Toxicol 9:365–373. Hanioka N, Matsumoto K, Saito Y, and Narimatsu S (2011) Influence of CYP2C8*13 Itkonen MK, Tornio A, Neuvonen M, Neuvonen PJ, Niemi M, and Backman JT and CYP2C8*14 alleles on amiodarone N-deethylation. Basic Clin Pharmacol (2015) Clopidogrel has no clinically meaningful effect on the pharmacokinetics of Toxicol 108:359–362. the OATP1B1 and CYP3A4 substrate simvastatin. Drug Metab Dispos 43: Harris JW, Rahman A, Kim BR, Guengerich FP, and Collins JM (1994) Metabolism 1655–1660. of taxol by human hepatic microsomes and liver slices: participation of cytochrome Jaakkola T, Backman JT, Neuvonen M, Laitila J, and Neuvonen PJ (2006a) Effect of P450 3A4 and an unknown P450 enzyme. Cancer Res 54:4026–4035. rifampicin on the pharmacokinetics of pioglitazone. Br J Clin Pharmacol 61:70–78. Hatorp V, Hansen KT, and Thomsen MS (2003) Influence of drugs interacting with Jaakkola T, Backman JT, Neuvonen M, and Neuvonen PJ (2005) Effects of gemfi- CYP3A4 on the pharmacokinetics, pharmacodynamics, and safety of the prandial brozil, itraconazole, and their combination on the pharmacokinetics of pioglitazone. glucose regulator repaglinide. J Clin Pharmacol 43:649–660. Clin Pharmacol Ther 77:404–414. He M, Jiang J, Qiu F, Liu S, Peng P, Gao C, and Miao P (2013) Inhibitory effects of Jaakkola T, Backman JT, Neuvonen M, Niemi M, and Neuvonen PJ (2006b) Mon- gypenosides on seven human cytochrome P450 enzymes in vitro. Food Chem telukast and zafirlukast do not affect the pharmacokinetics of the CYP2C8 sub- Toxicol 57:262–265. strate pioglitazone. Eur J Clin Pharmacol 62:503–509. Henningsson A, Marsh S, Loos WJ, Karlsson MO, Garsa A, Mross K, Mielke S, Jaakkola T, Laitila J, Neuvonen PJ, and Backman JT (2006c) Pioglitazone is Viganò L, Locatelli A, and Verweij J (2005) Association of CYP2C8, CYP3A4, metabolised by CYP2C8 and CYP3A4 in vitro: potential for interactions with CYP3A5, and ABCB1 polymorphisms with the pharmacokinetics of paclitaxel. Clin CYP2C8 inhibitors. Basic Clin Pharmacol Toxicol 99:44–51. Cancer Res 11:8097–8104. Jacobsen W, Christians U, and Benet LZ (2000a) In vitro evaluation of the disposition Hertz DL, Motsinger-Reif AA, Drobish A, Winham SJ, McLeod HL, Carey LA, of A novel cysteine protease inhibitor. Drug Metab Dispos 28:1343–1351. and Dees EC (2012) CYP2C8*3 predicts benefit/risk profile in breast cancer pa- Jacobsen W, Kuhn B, Soldner A, Kirchner G, Sewing KF, Kollman PA, Benet LZ, tients receiving neoadjuvant paclitaxel. Breast Cancer Res Treat 134:401–410. and Christians U (2000b) Lactonization is the critical first step in the disposition of Hertz DL, Roy S, Jack J, Motsinger-Reif AA, Drobish A, Clark LS, Carey LA, Dees the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor atorvastatin. Drug Metab EC, and McLeod HL (2014) Genetic heterogeneity beyond CYP2C8*3 does not Dispos 28:1369–1378. explain differential sensitivity to paclitaxel-induced neuropathy. Breast Cancer Res Jacobsen W, Serkova N, Hausen B, Morris RE, Benet LZ, and Christians U (2001) Treat 145:245–254. Comparison of the in vitro metabolism of the macrolide immunosuppressants Hichiya H, Tanaka-Kagawa T, Soyama A, Jinno H, Koyano S, Katori N, Matsushima sirolimus and RAD. Transplant Proc 33:514–515. E, Uchiyama S, Tokunaga H, and Kimura H (2005) Functional characterization of Jancová P, Anzenbacherová E, Papousková B, Lemr K, Luzná P, Veinlichová A, five novel CYP2C8 variants, G171S, R186X, R186G, K247R, and K383N, found in a Anzenbacher P, and Simánek V (2007) Silybin is metabolized by cytochrome P450 Japanese population. Drug Metab Dispos 33:630–636. 2C8 in vitro. Drug Metab Dispos 35:2035–2039. Hidestrand M, Oscarson M, Salonen JS, Nyman L, Pelkonen O, Turpeinen M, Jenkins SM, Zvyaga T, Johnson SR, Hurley J, Wagner A, Burrell R, Turley W, Leet and Ingelman-Sundberg M (2001) CYP2B6 and CYP2C19 as the major enzymes JE, Philip T, and Rodrigues AD (2011) Studies to further investigate the inhibition responsible for the metabolism of selegiline, a drug used in the treatment of Par- of human liver microsomal CYP2C8 by the acyl-b-glucuronide of gemfibrozil. Drug kinson’s disease, as revealed from experiments with recombinant enzymes. Drug Metab Dispos 39:2421–2430. Metab Dispos 29:1480–1484. Jeong HU, Kong TY, Kwon SS, Hong SW, Yeon SH, Choi JH, Lee JY, Cho YY, Hirano M, Maeda K, Shitara Y, and Sugiyama Y (2006) Drug-drug interaction be- and Lee HS (2013) Effect of honokiol on cytochrome P450 and UDP- tween pitavastatin and various drugs via OATP1B1. Drug Metab Dispos 34: glucuronosyltransferase enzyme activities in human liver microsomes. Molecules 1229–1236. 18:10681–10693. Hiratsuka M, Hinai Y, Sasaki T, Konno Y, Imagawa K, Ishikawa M, and Mizugaki M Jeong HU, Kwon SS, Kong TY, Kim JH, and Lee HS (2014) Inhibitory effects of (2007) Characterization of human cytochrome p450 enzymes involved in the me- cedrol, b-cedrene, and thujopsene on cytochrome P450 enzyme activities in human tabolism of cilostazol. Drug Metab Dispos 35:1730–1732. liver microsomes. J Toxicol Environ Health A 77:1522–1532. Hokkanen J, Tolonen A, Mattila S, and Turpeinen M (2011) Metabolism of hyper- Ji HY, Kim SY, Kim DK, Jeong JH, and Lee HS (2010) Effects of eupatilin and forin, the active constituent of St. John’s wort, in human liver microsomes. Eur J jaceosidin on cytochrome p450 enzyme activities in human liver microsomes. Pharm Sci 42:273–284. Molecules 15:6466–6475. Holmberg MT, Tornio A, Neuvonen M, Neuvonen PJ, Backman JT, and Niemi M Ji HY, Lee HW, Kim HH, Choi JK, and Lee HS (2005) Characterization of human (2014) Grapefruit juice inhibits the metabolic activation of clopidogrel. Clin liver cytochrome P450 enzymes involved in the metabolism of a new H+/K+- Pharmacol Ther 95:307–313. ATPase inhibitor KR-60436. Toxicol Lett 155:103–114. Hong SP, Lusiak BD, Burback BL, and Johnson JD (2013) Evaluations of in vitro Ji HY, Liu KH, Kong TY, Jeong HU, Choi SZ, Son M, Cho YY, and Lee HS (2013) metabolism, drug-drug interactions mediated by reversible and time-dependent Evaluation of DA-9801, a new herbal drug for diabetic neuropathy, on metabolism- inhibition of CYPs, and plasma protein binding of MMB4 DMS. Int J Toxicol mediated interaction. Arch Pharm Res 36:1–5. 32(4, Suppl):75S–87S. Johansson M, Strahm E, Rane A, and Ekström L (2014) CYP2C8 and CYP2C9 mRNA Honkalammi J, Niemi M, Neuvonen PJ, and Backman JT (2011a) Dose-dependent expression profile in the human fetus. Front Genet 5:58. interaction between gemfibrozil and repaglinide in humans: strong inhibition of Johnson BM, Stier BA, and Caltabiano S (2014) Effect of food and gemfibrozil on the CYP2C8 with subtherapeutic gemfibrozil doses. Drug Metab Dispos 39:1977–1986. pharmacokinetics of the novel prolyl hydroxylase inhibitor GSK1278863. Clin Honkalammi J, Niemi M, Neuvonen PJ, and Backman JT (2011b) Mechanism-based Pharmacol Drug Develop 3:109–117. inactivation of CYP2C8 by gemfibrozil occurs rapidly in humans. Clin Pharmacol Johnson EF and Stout CD (2005) Structural diversity of human xenobiotic- Ther 89:579–586. metabolizing cytochrome P450 monooxygenases. Biochem Biophys Res Commun Honkalammi J, Niemi M, Neuvonen PJ, and Backman JT (2012) Gemfibrozil is a 338:331–336. strong inactivator of CYP2C8 in very small multiple doses. Clin Pharmacol Ther Johnson SS, Oyelola FT, Ari T, and Juho H (2013) In vitro inhibitory activities of the 91:846–855. extract of Hibiscus sabdariffa L. (family Malvaceae) on selected cytochrome P450 Houston JB and Galetin A (2008) Methods for predicting in vivo pharmacokinetics isoforms. Afr J Tradit Complement Altern Medicines 10:533–540. using data from in vitro assays. Curr Drug Metab 9:940–951. Joo J, Lee D, Wu Z, Shin JH, Lee HS, Kwon BM, Huh TL, Kim YW, Lee SJ, and Kim Hruska MW, Amico JA, Langaee TY, Ferrell RE, Fitzgerald SM, and Frye RF (2005) TW (2013) In vitro metabolism of obovatol and its effect on cytochrome P450 en- The effect of trimethoprim on CYP2C8 mediated rosiglitazone metabolism in hu- zyme activities in human liver microsomes. Biopharm Drug Dispos 34:195–202. man liver microsomes and healthy subjects. Br J Clin Pharmacol 59:70–79. Kajosaari LI, Jaakkola T, Neuvonen PJ, and Backman JT (2006a) Pioglitazone, an in Hu G, Johnson EF, and Kemper B (2010) CYP2C8 exists as a dimer in natural vitro inhibitor of CYP2C8 and CYP3A4, does not increase the plasma concentra- membranes. Drug Metab Dispos 38:1976–1983. tions of the CYP2C8 and CYP3A4 substrate repaglinide. Eur J Clin Pharmacol 62: Hu M, Krausz K, Chen J, Ge X, Li J, Gelboin HL, and Gonzalez FJ (2003) Identifi- 217–223. cation of CYP1A2 as the main isoform for the phase I hydroxylated metabolism of Kajosaari LI, Laitila J, Neuvonen PJ, and Backman JT (2005a) Metabolism of genistein and a prodrug converting enzyme of methylated isoflavones. Drug Metab repaglinide by CYP2C8 and CYP3A4 in vitro: effect of fibrates and rifampicin. Dispos 31:924–931. Basic Clin Pharmacol Toxicol 97:249–256. Huang SM, Strong JM, Zhang L, Reynolds KS, Nallani S, Temple R, Abraham S, Kajosaari LI, Niemi M, Backman JT, and Neuvonen PJ (2006b) Telithromycin, but Habet SA, Baweja RK, and Burckart GJ (2008) New era in drug interaction not montelukast, increases the plasma concentrations and effects of the cyto- evaluation: US Food and Drug Administration update on CYP enzymes, trans- chrome P450 3A4 and 2C8 substrate repaglinide. Clin Pharmacol Ther 79: porters, and the guidance process. J Clin Pharmacol 48:662–670. 231–242. Huang Z, Roy P, and Waxman DJ (2000) Role of human liver microsomal CYP3A4 Kajosaari LI, Niemi M, Neuvonen M, Laitila J, Neuvonen PJ, and Backman JT and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfa- (2005b) Cyclosporine markedly raises the plasma concentrations of repaglinide. mide. Biochem Pharmacol 59:961–972. Clin Pharmacol Ther 78:388–399. Ibarra-Barrueta O, Palacios-Zabalza I, Mora-Atorrasagasti O, and Mayo-Suarez J Kalgutkar AS, Chen D, Varma MV, Feng B, Terra SG, Scialis RJ, Rotter CJ, Fred- (2014) Effect of concomitant use of montelukast and efavirenz on neuropsychiatric erick KS, West MA, and Goosen TC (2013) Elucidation of the biochemical basis for adverse events. Ann Pharmacother 48:145–148. a clinical drug-drug interaction between atorvastatin and 5-(N-(4-((4-ethylbenzyl) Illingworth NA, Boddy AV, Daly AK, and Veal GJ (2011) Characterization of the thio)phenyl)sulfamoyl)-2-methyl benzoic acid (CP-778875), a subtype selective ag- metabolism of fenretinide by human liver microsomes, cytochrome P450 enzymes onist of the peroxisome proliferator-activated receptor alpha. Xenobiotica 43: and UDP-glucuronosyltransferases. Br J Pharmacol 162:989–999. 963–972. Im Y, Kim YW, Song IS, Joo J, Shin JH, Wu Z, Lee HS, Park KH, and Liu KH (2012) Kalliokoski A, Backman JT, Kurkinen KJ, Neuvonen PJ, and Niemi M (2008b) Ef- Effect of TSHAC on human cytochrome P450 activity, and transport mediated by fects of gemfibrozil and atorvastatin on the pharmacokinetics of repaglinide in P-glycoprotein. J Microbiol Biotechnol 22:1659–1664. relation to SLCO1B1 polymorphism. Clin Pharmacol Ther 84:488–496. Inoue S, Howgate EM, Rowland-Yeo K, Shimada T, Yamazaki H, Tucker GT, Kalliokoski A, Neuvonen M, Neuvonen PJ, and Niemi M (2008a) No significant effect and Rostami-Hodjegan A (2006) Prediction of in vivo drug clearance from in vitro of SLCO1B1 polymorphism on the pharmacokinetics of rosiglitazone and piogli- data. II: potential inter-ethnic differences. Xenobiotica 36:499–513. tazone. Br J Clin Pharmacol 65:78–86. 234 Backman et al.

Kamali F and Huang ML (1996) Increased systemic availability of loperamide after fibroblast growth factor receptors and VEGF receptors, in patients with advanced oral administration of loperamide and loperamide oxide with cotrimoxazole. Br J melanoma. Clin Cancer Res 17:7451–7461. Clin Pharmacol 41:125–128. Kim MJ, Kim H, Cha IJ, Park JS, Shon JH, Liu KH, and Shin JG (2005b) High- Kamel A, Colizza K, and Obach RS (2013) In vitro metabolism of the 5-hydroxy- throughput screening of inhibitory potential of nine cytochrome P450 enzymes in tryptamine1B receptor antagonist elzasonan. Xenobiotica 43:368–378. vitro using liquid chromatography/tandem mass spectrometry. Rapid Commun Kamdem LK, Liu Y, Stearns V, Kadlubar SA, Ramirez J, Jeter S, Shahverdi K, Ward Mass Spectrom 19:2651–2658. BA, Ogburn E, and Ratain MJ (2010) In vitro and in vivo oxidative metabolism and Kim MJ, Lee JW, Oh KS, Choi CS, Kim KH, Han WS, Yoon CN, Chung ES, Kim glucuronidation of anastrozole. Br J Clin Pharmacol 70:854–869. DH, and Shin JG (2013b) The inhibitor nilotinib selectively Kaneko K, Suzuki K, Iwadate-Iwata E, Kato I, Uchida K, and Onoue M (2013) inhibits CYP2C8 activities in human liver microsomes. Drug Metab Phar- Evaluation of food-drug interaction of guava leaf tea. Phytother Res 27:299–305. macokinet 28:462–467. Kang P, Dalvie D, Smith E, Zhou S, Deese A, and Nieman JA (2008) Bioactivation of Kim SY, Kang JY, Hartman JH, Park SH, Jones DR, Yun CH, Boysen G, and Miller flutamide metabolites by human liver microsomes. Drug Metab Dispos 36: GP (2012) Metabolism of R- and S-warfarin by CYP2C19 into four hydrox- 1425–1437. ywarfarins. Drug Metab Lett 6:157–164. Kantola T, Kivistö KT, and Neuvonen PJ (1999) Effect of itraconazole on cerivastatin Kim YW, Kim YK, Kim DK, and Sheen YY (2008) Identification of human cytochrome pharmacokinetics. Eur J Clin Pharmacol 54:851–855. P450 enzymes involved in the metabolism of IN-1130, a novel activin receptor-like Karam WG, Goldstein JA, Lasker JM, and Ghanayem BI (1996) Human CYP2C19 is kinase-5 (ALK5) inhibitor. Xenobiotica 38:451–464. a major omeprazole 5-hydroxylase, as demonstrated with recombinant cytochrome Kirchheiner J, Roots I, Goldammer M, Rosenkranz B, and Brockmöller J (2005) P450 enzymes. Drug Metab Dispos 24:1081–1087. Effect of genetic polymorphisms in cytochrome p450 (CYP) 2C9 and CYP2C8 on the Karazniewicz-Łada M, Luczak M, and Główka F (2009) Pharmacokinetic studies of pharmacokinetics of oral antidiabetic drugs: clinical relevance. Clin Pharmacokinet enantiomers of ibuprofen and its chiral metabolites in humans with different variants 44:1209–1225. of genes coding CYP2C8 and CYP2C9 isoenzymes. Xenobiotica 39:476–485. Kirchheiner J, Thomas S, Bauer S, Tomalik-Scharte D, Hering U, Doroshyenko O, Karlsson FH, Bouchene S, Hilgendorf C, Dolgos H, and Peters SA (2013) Utility of in Jetter A, Stehle S, Tsahuridu M, and Meineke I (2006) Pharmacokinetics and vitro systems and preclinical data for the prediction of human intestinal first-pass pharmacodynamics of rosiglitazone in relation to CYP2C8 genotype. Clin Phar- metabolism during drug discovery and preclinical development. Drug Metab Dispos macol Ther 80:657–667. 41:2033–2046. Kitamura A, Mizuno Y, Natsui K, Yabuki M, Komuro S, and Kanamaru H (2005) Karonen T, Filppula A, Laitila J, Niemi M, Neuvonen PJ, and Backman JT (2010) Characterization of human cytochrome P450 enzymes involved in the in vitro – Gemfibrozil markedly increases the plasma concentrations of montelukast: a pre- metabolism of perospirone. Biopharm Drug Dispos 26:59 65. viously unrecognized role for CYP2C8 in the metabolism of montelukast. Clin Klieber S, Arabeyre-Fabre C, Moliner P, Marti E, Mandray M, Ngo R, Ollier C, Brun Pharmacol Ther 88:223–230. P, and Fabre G (2014) Identification of metabolic pathways and enzyme systems Karonen T, Neuvonen PJ, and Backman JT (2011) The CYP2C8 inhibitor gemfibrozil involved in the in vitro human hepatic metabolism of dronedarone, a potent new does not affect the pharmacokinetics of zafirlukast. Eur J Clin Pharmacol 67: oral antiarrhythmic drug. Pharmacol Res Perspect 2:e00044. 151–155. Klose TS, Blaisdell JA, and Goldstein JA (1999) Gene structure of CYP2C8 and Karonen T, Neuvonen PJ, and Backman JT (2012) CYP2C8 but not CYP3A4 is im- extrahepatic distribution of the human CYP2Cs. J Biochem Mol Toxicol 13: – portant in the pharmacokinetics of montelukast. Br J Clin Pharmacol 73:257–267. 289 295. Kaspera R, Naraharisetti SB, Evangelista EA, Marciante KD, Psaty BM, and Totah Kochansky CJ, Xia YQ, Wang S, Cato B, Creighton M, Vincent SH, Franklin RB, RA (2011) Drug metabolism by CYP2C8.3 is determined by substrate dependent and Reed JR (2005) Species differences in the elimination of a peroxisome interactions with cytochrome P450 reductase and cytochrome b5. Biochem Phar- proliferator-activated receptor agonist highlighted by oxidative metabolism of its – macol 82:681–691. acyl glucuronide. Drug Metab Dispos 33:1894 1904. Kaspera R, Naraharisetti SB, Tamraz B, Sahele T, Cheesman MJ, Kwok PY, Marciante Kojima K, Nagata K, Matsubara T, and Yamazoe Y (2007) Broad but distinct role of K, Heckbert SR, Psaty BM, and Totah RA (2010) Cerivastatin in vitro metabolism by pregnane x receptor on the expression of individual cytochrome p450s in human hepatocytes. Drug Metab Pharmacokinet 22:276–286. CYP2C8 variants found in patients experiencing rhabdomyolysis. Pharmacogenet Komatsu K, Ito K, Nakajima Y, Kanamitsu Si, Imaoka S, Funae Y, Green CE, Tyson Genomics 20:619–629. CA, Shimada N, and Sugiyama Y (2000a) Prediction of in vivo drug-drug interac- Kassahun K, McIntosh I, Koeplinger K, Sun L, Talaty JE, Miller DL, Dixon R, Zajic tions between tolbutamide and various sulfonamides in humans based on in vitro S, and Stoch SA (2014) Disposition and metabolism of the cathepsin K inhibitor experiments. Drug Metab Dispos 28:475–481. odanacatib in humans. Drug Metab Dispos 42:818–827. Komatsu T, Yamazaki H, Shimada N, Nagayama S, Kawaguchi Y, Nakajima M, Kazmi F, Barbara JE, Yerino P, and Parkinson A (2015) A long-standing mystery and Yokoi T (2001) Involvement of microsomal cytochrome P450 and cytosolic solved: the formation of 3-hydroxydesloratadine is catalyzed by CYP2C8 but prior thymidine phosphorylase in 5-fluorouracil formation from tegafur in human liver. glucuronidation of desloratadine by UDP-glucuronosyltransferase 2B10 is an Clin Cancer Res 7:675–681. obligatory requirement. Drug Metab Dispos 43:523–533. Komatsu T, Yamazaki H, Shimada N, Nakajima M, and Yokoi T (2000b) Roles of Kazmi F, Haupt LJ, Horkman JR, Smith BD, Buckley DB, Wachter EA, and Singer cytochromes P450 1A2, 2A6, and 2C8 in 5-fluorouracil formation from tegafur, an JM (2014) In vitro inhibition of human liver cytochrome P450 (CYP) and UDP- anticancer prodrug, in human liver microsomes. Drug Metab Dispos 28:1457–1463. glucuronosyltransferase (UGT) enzymes by rose bengal: system-dependent effects Komoroski BJ, Parise RA, Egorin MJ, Strom SC, and Venkataramanan R (2005) – on inhibitory potential. Xenobiotica 44:606 614. Effect of the St. John’s wort constituent hyperforin on docetaxel metabolism by Kazmi F, Smith B, Hvenegaard MG, Bendahl L, Gipson A, Buckley D, Ogilvie B, human hepatocyte cultures. Clin Cancer Res 11:6972–6979. and Parkinson A (2010) Identification of a novel carbamoyl glucuronide as a Korprasertthaworn P, Polasek TM, Sorich MJ, McLachlan AJ, Miners JO, Tucker metabolism-dependent inhibitor of CYP2C8, in 9th International ISSX Meeting, GT, and Rowland A (2015) In vitro characterization of the human liver microsomal Istanbul, Turkey (September 4-8, 2010). Available from: https://issx.confex.com/ kinetics and reaction phenotyping of olanzapine metabolism. Drug Metab Dispos issx/intl9/webprogram/Paper21723.html (Accessed June 6, 2015). 43:1806–1814. Kerr BM, Thummel KE, Wurden CJ, Klein SM, Kroetz DL, Gonzalez FJ, and Levy Korzekwa K (2014) Case study 4. Predicting the drug interaction potential for in- RH (1994) Human liver carbamazepine metabolism. Role of CYP3A4 and CYP2C8 hibition of CYP2C8 by montelukast. Methods Mol Biol 1113:461–469. – in 10,11-epoxide formation. Biochem Pharmacol 47:1969 1979. Korzekwa K, Tweedie D, Argikar UA, Whitcher-Johnstone A, Bell L, Bickford S, Khan MS, Barratt DT, and Somogyi AA (2015) Impact of CYP2C8*3 polymorphism and Nagar S (2014) A numerical method for analysis of in vitro time-dependent – on in vitro metabolism of imatinib to N-desmethyl imatinib. Xenobiotica 10:1 10. inhibition data. Part 2. Application to experimental data. Drug Metab Dispos 42: Kim DK, Liu KH, Jeong JH, Ji HY, Oh SR, Lee HK, and Lee HS (2011a) In vitro 1587–1595. metabolism of magnolin and characterization of cytochrome P450 enzymes re- Kosugi Y, Hirabayashi H, Igari T, Fujioka Y, Okuda T, and Moriwaki T (2014) Risk sponsible for its metabolism in human liver microsomes. Xenobiotica 41:358–371. assessment of drug-drug interactions using hepatocytes suspended in serum dur- Kim H, Kang S, Kim H, Yoon YJ, Cha EY, Lee HS, Kim JH, Yea SS, Lee SS, and Shin ing the drug discovery process. Xenobiotica 44:336–344. JG (2006) In vitro metabolism of a new cardioprotective agent, KR-32570, in hu- Kot M and Daniel WA (2008) The relative contribution of human cytochrome P450 man liver microsomes. Rapid Commun Mass Spectrom 20:837–843. isoforms to the four caffeine oxidation pathways: an in vitro comparative study Kim IS, Kim Y, Kwak TH, and Yoo HH (2013a) Effects of b-lapachone, a new anti- with cDNA-expressed P450s including CYP2C isoforms. Biochem Pharmacol 76: cancer candidate, on cytochrome P450-mediated drug metabolism. Cancer Che- 543–551. mother Pharmacol 72:699–702. Kozakai K, Yamada Y, Oshikata M, Kawase T, Suzuki E, Haramaki Y, and Taniguchi Kim J, Peraire C, Solà J, Johanning KM, Dalton JT, and Veverka KA (2011b) Drug H (2012) Reliable high-throughput method for inhibition assay of 8 cytochrome interaction potential of toremifene and N-desmethyltoremifene with multiple cy- P450 isoforms using cocktail of probe substrates and stable isotope-labeled internal tochrome P450 isoforms. Xenobiotica 41:851–862. standards. Drug Metab Pharmacokinet 27:520–529. Kim KA, Chung J, Jung DH, and Park JY (2004) Identification of cytochrome P450 Kudo T, Ozaki Y, Kusano T, Hotta E, Oya Y, Komatsu S, Goda H, and Ito K (2015) isoforms involved in the metabolism of loperamide in human liver microsomes. Eur Effect of buffer conditions on CYP2C8-mediated paclitaxel 6a-hydroxylation and J Clin Pharmacol 60:575–581. CYP3A4-mediated triazolam a- and 4-hydroxylation by human liver microsomes. Kim KA, Park JY, Lee JS, and Lim S (2003) Cytochrome P450 2C8 and CYP3A4/5 are Xenobiotica 10.3109/00498254.2015.1071502. involved in chloroquine metabolism in human liver microsomes. Arch Pharm Res Kudzi W, Dodoo AN, and Mills JJ (2009) Characterisation of CYP2C8, CYP2C9 and 26:631–637. CYP2C19 polymorphisms in a Ghanaian population. BMC Med Genet 10:124. Kim KA, Park PW, Kim HK, Ha JM, and Park JY (2005a) Effect of quercetin on the Kumar GN, Dubberke E, Rodrigues AD, Roberts E, and Dennisen JF (1997) Identifica- pharmacokinetics of rosiglitazone, a CYP2C8 substrate, in healthy subjects. J Clin tion of cytochromes P450 involved in the human liver microsomal metabolism of the Pharmacol 45:941–946. thromboxane A2 inhibitor seratrodast (ABT-001). Drug Metab Dispos 25:110–115. Kim KA, Park PW, Kim KR, and Park JY (2007) Effect of multiple doses of mon- Kumar GN, Walle UK, and Walle T (1994) Cytochrome P450 3A-mediated human liver telukast on the pharmacokinetics of rosiglitazone, a CYP2C8 substrate, in humans. microsomal taxol 6 alpha-hydroxylation. JPharmacolExpTher268:1160–1165. Br J Clin Pharmacol 63:339–345. Kumar P, Henikoff S, and Ng PC (2009) Predicting the effects of coding non- Kim KB, Chesney J, Robinson D, Gardner H, Shi MM, and Kirkwood JM (2011c) synonymous variants on protein function using the SIFT algorithm. Nat Protoc 4: Phase I/II and pharmacodynamic study of dovitinib (TKI258), an inhibitor of 1073–1081. Role of CYP2C8 in Drug Metabolism and Interactions 235

Kumar S, Samuel K, Subramanian R, Braun MP, Stearns RA, Chiu SH, Evans DC, nucleotide polymorphisms on tanshinol borneol ester metabolism and inhibition and Baillie TA (2002) Extrapolation of diclofenac clearance from in vitro micro- potential. Drug Metab Dispos 38:2259–2265. somal metabolism data: role of acyl glucuronidation and sequential oxidative me- Liu D, Zheng X, Tang Y, Zi J, Nan Y, Wang S, Xiao C, Zhu J, and Chen C (2010b) tabolism of the acyl glucuronide. J Pharmacol Exp Ther 303:969–978. Metabolism of tanshinol borneol ester in rat and human liver microsomes. Drug Kurata N, Nishimura Y, Iwase M, Fischer NE, Tang BK, Inaba T, and Yasuhara H Metab Dispos 38:1464–1470. (1998) Trimethadione metabolism by human liver cytochrome P450: evidence for Liu K, Li F, Lu J, Liu S, Dorko K, Xie W, and Ma X (2014) Bedaquiline metabolism: the involvement of CYP2E1. Xenobiotica 28:1041–1047. enzymes and novel metabolites. Drug Metab Dispos 42:863–866. Kyrklund C, Backman JT, Kivistö KT, Neuvonen M, Laitila J, and Neuvonen PJ Lopez Garcia MP, Dansette PM, Valadon P, Amar C, Beaune PH, Guengerich FP, (2001) Plasma concentrations of active lovastatin acid are markedly increased by and Mansuy D (1993) Human-liver cytochromes P-450 expressed in yeast as tools gemfibrozil but not by bezafibrate. Clin Pharmacol Ther 69:340–345. for reactive-metabolite formation studies. Oxidative activation of tienilic acid by Kyrklund C, Backman JT, Neuvonen M, and Neuvonen PJ (2003) Gemfibrozil in- cytochromes P-450 2C9 and 2C10. Eur J Biochem 213:223–232. creases plasma pravastatin concentrations and reduces pravastatin renal clear- López-Rodríguez R, Novalbos J, Gallego-Sandín S, Román-Martínez M, Torrado J, ance. Clin Pharmacol Ther 73:538–544. Gisbert JP, and Abad-Santos F (2008) Influence of CYP2C8 and CYP2C9 poly- Lacy S, Hsu B, Miles D, Aftab D, Wang R, and Nguyen L (2015) Metabolism and morphisms on pharmacokinetic and pharmacodynamic parameters of racemic and Disposition of Cabozantinib in Healthy Male Volunteers and Pharmacologic enantiomeric forms of ibuprofen in healthy volunteers. Pharmacol Res 58:77–84. Characterization of Its Major Metabolites. Drug Metab Dispos 43:1190–1207. LoRusso PM, Piha-Paul SA, Mita M, Colevas AD, Malhi V, Colburn D, Yin M, Low Lai XS, Yang LP, Li XT, Liu JP, Zhou ZW, and Zhou SF (2009) Human CYP2C8: JA, and Graham RA (2013) Co-administration of vismodegib with rosiglitazone or structure, substrate specificity, inhibitor selectivity, inducers and polymorphisms. combined oral contraceptive in patients with locally advanced or metastatic solid Curr Drug Metab 10:1009–1047. tumors: a pharmacokinetic assessment of drug-drug interaction potential. Cancer Läpple F, von Richter O, Fromm MF, Richter T, Thon KP, Wisser H, Griese EU, Chemother Pharmacol 71:193–202. Eichelbaum M, and Kivistö KT (2003) Differential expression and function of Lu C, Suri A, and Shimoga P (2012) Application of physiologically based pharma- CYP2C isoforms in human intestine and liver. Pharmacogenetics 13:565–575. cokinetic modeling and simulation for waiver of orteronel drug-drug interaction Lawrence SK, Nguyen D, Bowen C, Richards-Peterson L, and Skordos KW (2014) The trials, in 12th European ISS Meeting; Noordwijk, Netherlands (June 17-21, 2012). metabolic drug-drug interaction profile of Dabrafenib: in vitro investigations and quan- Available from: http://issx.confex.com/issx/12euro/webprogram/Paper27322.html titative extrapolation of the P450-mediated DDI risk. Drug Metab Dispos 42:1180–1190. (Accessed June 18, 2015). Lee CA, Jones JP 3rd, Katayama J, Kaspera R, Jiang Y, Freiwald S, Smith E, Walker Lundblad MS, Stark K, Eliasson E, Oliw E, and Rane A (2005) Biosynthesis of GS, and Totah RA (2012a) Identifying a selective substrate and inhibitor pair for epoxyeicosatrienoic acids varies between polymorphic CYP2C enzymes. Biochem – the evaluation of CYP2J2 activity. Drug Metab Dispos 40:943–951. Biophys Res Commun 327:1052 1057. Lee HS, Ji HY, Park EJ, and Kim SY (2007) In vitro metabolism of eupatilin by Ma B, Subramanian R, Schrag ML, Rodrigues AD, and Tang C (2004) Cytochrome multiple cytochrome P450 and UDP-glucuronosyltransferase enzymes. Xenobiotica P450 2C8 (CYP2C8)-mediated hydroxylation of an endothelin ETA receptor an- – 37:803–817. tagonist in human liver microsomes. Drug Metab Dispos 32:473 478. Lee HS, Park EJ, Ji HY, Kim SY, Im GJ, Lee SM, and Jang IJ (2008) Identification of Macé K, Bowman ED, Vautravers P, Shields PG, Harris CC, and Pfeifer AM (1998) cytochrome P450 enzymes responsible for N -dealkylation of a new oral erecto- Characterisation of xenobiotic-metabolising enzyme expression in human bron- – genic, mirodenafil. Xenobiotica 38:21–33. chial mucosa and peripheral lung tissues. Eur J Cancer 34:914 920. Lee KC, Shorr R, Rodriguez R, Maturo C, Boteju LW, and Sheldon A (2011) For- Mach CM, Chen JH, Mosley SA, Kurzrock R, and Smith JA (2010) Evaluation of – mation and anti-tumor activity of uncommon in vitro and in vivo metabolites of liposomal curcumin cytochrome p450 metabolism. Anticancer Res 30:811 814. Madan A, Fisher A, Jin L, Chapman D, and Bozigian HP (2007) In vitro metabolism of CPI-613, a novel anti-tumor compound that selectively alters tumor energy me- – tabolism. Drug Metab Lett 5:163–182. indiplon and an assessment of its drug interaction potential. Xenobiotica 37:736 752. Lee M, Park J, Lim MS, Seong SJ, Lee J, Seo JJ, Park YK, Lee HW, and Yoon YR Madan A, Graham RA, Carroll KM, Mudra DR, Burton LA, Krueger LA, Downey AD, Czerwinski M, Forster J, and Ribadeneira MD (2003) Effects of prototypical mi- (2012b) High-throughput screening of inhibitory effects of Bo-yang-hwan-o-tang on crosomal enzyme inducers on cytochrome P450 expression in cultured human he- human cytochrome P450 isoforms in vitro using UPLC/MS/MS. Anal Sci 28: patocytes. Drug Metab Dispos 31:421–431. 1197–1201. Makishima M, Lu TT, Xie W, Whitfield GK, Domoto H, Evans RM, Haussler MR, Lee MY, Apellániz-Ruiz M, Johansson I, Vikingsson S, Bergmann TK, Brøsen K, and Mangelsdorf DJ (2002) Vitamin D receptor as an intestinal bile acid sensor. Green H, Rodríguez-Antona C, and Ingelman-Sundberg M (2015) Role of cyto- Science 296:1313–1316. chrome P450 2C8*3 (CYP2C8*3) in paclitaxel metabolism and paclitaxel-induced Mancy A, Antignac M, Minoletti C, Dijols S, Mouries V, Duong NT, Battioni P, neurotoxicity. Pharmacogenomics doi:10.2217/pgs.15.46. Dansette PM, and Mansuy D (1999) Diclofenac and its derivatives as tools for Lee TM, Huang L, Johnson MK, Lizak P, Kroetz D, Aweeka F, and Parikh S (2012c) studying human cytochromes P450 active sites: particular efficiency and regiose- In vitro metabolism of piperaquine is primarily mediated by CYP3A4. Xenobiotica – – lectivity of P450 2Cs. Biochemistry 38:14264 14270. 42:1088 1095. Mancy A, Dijols S, Poli S, Guengerich P, and Mansuy D (1996) Interaction of sulfa- Leo MA, Lasker JM, Raucy JL, Kim CI, Black M, and Lieber CS (1989) Metabolism of phenazole derivatives with human liver cytochromes P450 2C: molecular origin of retinol and retinoic acid by human liver cytochrome P450IIC8. Arch Biochem – the specific inhibitory effects of sulfaphenazole on CYP 2C9 and consequences for Biophys 269:305 312. the substrate binding site topology of CYP 2C9. Biochemistry 35:16205–16212. Leskelä S, Jara C, Leandro-García LJ, Martínez A, García-Donas J, Hernando S, Mano Y, Usui T, and Kamimura H (2007) The UDP-glucuronosyltransferase 2B7 Hurtado A, Vicario JC, Montero-Conde C, and Landa I (2011) Polymorphisms in isozyme is responsible for gemfibrozil glucuronidation in the human liver. Drug cytochromes P450 2C8 and 3A5 are associated with paclitaxel neurotoxicity. Metab Dispos 35:2040–2044. – Pharmacogenomics J 11:121 129. Marill J, Capron CC, Idres N, and Chabot GG (2002) Human cytochrome P450s Li AP and Schlicht KE (2014) Application of a higher throughput approach to derive involved in the metabolism of 9-cis- and 13-cis-retinoic acids. Biochem Pharmacol apparent Michaelis-Menten constants of isoform-selective p450-mediated bio- 63:933–943. – transformation reactions in human hepatocytes. Drug Metab Lett 8:2 11. Marill J, Cresteil T, Lanotte M, and Chabot GG (2000) Identification of human cy- Li G, Huang K, Nikolic D, and van Breemen RB (2015) High-throughput cytochrome tochrome P450s involved in the formation of all-trans-retinoic acid principal me- P450 cocktail inhibition assay for assessing drug-drug and drug-botanical inter- tabolites. Mol Pharmacol 58:1341–1348. – actions. Drug Metab Dispos 43:1670 1678 10.1124/dmd.115.065987. Marill J, Idres N, Capron CC, Nguyen E, and Chabot GG (2003) Retinoic acid me- Li L, Tu M, Yang X, Sun S, Wu X, Zhou H, Zeng S, and Jiang H (2014b) The tabolism and mechanism of action: a review. Curr Drug Metab 4:1–10. contribution of human OCT1, OCT3, and CYP3A4 to nitidine chloride-induced Martínez C, García-Martín E, Blanco G, Gamito FJ, Ladero JM, and Agúndez JA hepatocellular toxicity. Drug Metab Dispos 42:1227–1234. (2005) The effect of the cytochrome P450 CYP2C8 polymorphism on the disposition Li X, Jeso V, Heyward S, Walker GS, Sharma R, Micalizio GC, and Cameron MD of (R)-ibuprofen enantiomer in healthy subjects. Br J Clin Pharmacol 59:62–69. (2014a) Characterization of T-5 N-oxide formation as the first highly selective Martis S, Peter I, Hulot JS, Kornreich R, Desnick RJ, and Scott SA (2013) Multi- measure of CYP3A5 activity. Drug Metab Dispos 42:334–342. ethnic distribution of clinically relevant CYP2C genotypes and haplotypes. Phar- Li XQ, Björkman A, Andersson TB, Ridderström M, and Masimirembwa CM (2002) macogenomics J 13:369–377. Amodiaquine clearance and its metabolism to N-desethylamodiaquine is mediated Marwa KJ, Schmidt T, Sjögren M, Minzi OM, Kamugisha E, and Swedberg G (2014) Cy- by CYP2C8: a new high affinity and turnover enzyme-specific probe substrate. J tochrome P450 single nucleotide polymorphisms in an indigenous Tanzanian population: Pharmacol Exp Ther 300:399–407. a concern about the metabolism of artemisinin-based combinations. Malar J 13:420. Li Y, Zhou D, Ferguson SS, Dorff P, Simpson TR, and Grimm SW (2010) In vitro Masek V, Anzenbacherová E, Etrych T, Strohalm J, Ulbrich K, and Anzenbacher P assessment of metabolic drug–drug interaction potential of AZD2624, neurokinin-3 (2011) Interaction of N-(2-hydroxypropyl)methacrylamide copolymer-doxorubicin receptor antagonist, through cytochrome P(450) enzyme identification, inhibition, conjugates with human liver microsomal cytochromes P450: comparison with free and induction studies. Xenobiotica 40:721–729. doxorubicin. Drug Metab Dispos 39:1704–1710. Lilja JJ, Backman JT, and Neuvonen PJ (2005) Effect of gemfibrozil on the phar- Masimirembwa CM, Otter C, Berg M, Jönsson M, Leidvik B, Jonsson E, Johansson T, macokinetics and pharmacodynamics of racemic warfarin in healthy subjects. Br J Bäckman A, Edlund A, and Andersson TB (1999) Heterologous expression and Clin Pharmacol 59:433–439. kinetic characterization of human cytochromes P-450: validation of a pharmaceu- Lin JH and Lu AY (1998) Inhibition and induction of cytochrome P450 and the tical tool for drug metabolism research. Drug Metab Dispos 27:1117–1122. clinical implications. Clin Pharmacokinet 35:361–390. McClue SJ and Stuart I (2008) Metabolism of the trisubstituted purine cyclin- Ling J, Johnson KA, Miao Z, Rakhit A, Pantze MP, Hamilton M, Lum BL, dependent kinase inhibitor seliciclib (R-roscovitine) in vitro and in vivo. Drug and Prakash C (2006) Metabolism and of erlotinib, a small molecule Metab Dispos 36:561–570. inhibitor of epidermal growth factor receptor tyrosine kinase, in healthy male McFayden MC, Melvin WT, and Murray GI (1998) Regional distribution of individual volunteers. Drug Metab Dispos 34:420–426. forms of cytochrome P450 mRNA in normal adult human brain. Biochem Phar- Linnet K and Olesen OV (1997) Metabolism of clozapine by cDNA-expressed human macol 55:825–830. cytochrome P450 enzymes. Drug Metab Dispos 25:1379–1382. McLaren W, Pritchard B, Rios D, Chen Y, Flicek P, and Cunningham F (2010) De- Liu D, Gao Y, Wang H, Zi J, Huang H, Ji J, Zhou R, Nan Y, Wang S, and Zheng X riving the consequences of genomic variants with the Ensembl API and SNP Effect (2010a) Evaluation of the effects of cytochrome P450 nonsynonymous single- Predictor. Bioinformatics 26:2069–2070. 236 Backman et al.

Mega JL, Close SL, Wiviott SD, Shen L, Hockett RD, Brandt JT, Walker JR, Antman Nakagomi-Hagihara R, Nakai D, and Tokui T (2007) Inhibition of human organic EM, Macias W, and Braunwald E (2009) Cytochrome p-450 polymorphisms and anion transporter 3 mediated pravastatin transport by gemfibrozil and the me- response to clopidogrel. N Engl J Med 360:354–362. tabolites in humans. Xenobiotica 37:416–426. Melet A, Marques-Soares C, Schoch GA, Macherey AC, Jaouen M, Dansette PM, Sari Nakajima M, Fujiki Y, Noda K, Ohtsuka H, Ohkuni H, Kyo S, Inoue M, Kuroiwa Y, MA, Johnson EF, and Mansuy D (2004) Analysis of human cytochrome P450 2C8 and Yokoi T (2003) Genetic polymorphisms of CYP2C8 in Japanese population. substrate specificity using a substrate pharmacophore and site-directed mutants. Drug Metab Dispos 31:687–690. Biochemistry 43:15379–15392. Nakamura K, Ariyoshi N, Iwatsubo T, Fukunaga Y, Higuchi S, Itoh K, Shimada N, Menon RM, Badri PS, Wang T, Polepally AR, Zha J, Khatri A, Wang H, Hu B, Nagashima K, Yokoi T, and Yamamoto K (2005) Inhibitory effects of nicardipine to Coakley EP, and Podsadecki TJ (2015) Drug-drug interaction profile of the all-oral cytochrome P450 (CYP) in human liver microsomes. Biol Pharm Bull 28:882–885. anti-hepatitis C virus regimen of paritaprevir/ritonavir, ombitasvir, and dasabuvir. Naraharisetti SB, Lin YS, Rieder MJ, Marciante KD, Psaty BM, Thummel KE, J Hepatol 63:20–29. and Totah RA (2010) Human liver expression of CYP2C8: gender, age, and geno- Miller VP, Stresser DM, Blanchard AP, Turner S, and Crespi CL (2000) Fluorometric type effects. Drug Metab Dispos 38:889–893. high-throughput screening for inhibitors of cytochrome P450. Ann N Y Acad Sci Narimatsu S, Yonemoto R, Saito K, Takaya K, Kumamoto T, Ishikawa T, Asanuma 919:26–32. M, Funada M, Kiryu K, and Naito S (2006) Oxidative metabolism of 5-methoxy-N, Miners JO, Coulter S, Birkett DJ, and Goldstein JA (2000) Torsemide metabolism by N-diisopropyltryptamine (Foxy) by human liver microsomes and recombinant cy- CYP2C9 variants and other human CYP2C subfamily enzymes. Pharmacogenetics tochrome P450 enzymes. Biochem Pharmacol 71:1377–1385. 10:267–270. Nassar AE, King I, Paris BL, Haupt L, Ndikum-Moffor F, Campbell R, Usuki E, Minhas S, Setia N, Pandita S, Saxena R, Verma IC, and Aggarwal S (2013) Preva- Skibbe J, Brobst D, and Ogilvie BW (2009) An in vitro evaluation of the victim and lence of CYP2C8 polymorphisms in a North Indian population. Genet Mol Res 12: perpetrator potential of the anticancer agent laromustine (VNP40101M), based on 2260–2266. reaction phenotyping and inhibition and induction of cytochrome P450 enzymes. Misaka S, Kawabe K, Onoue S, Werba JP, Giroli M, Tamaki S, Kan T, Kimura J, Drug Metab Dispos 37:1922–1930. Watanabe H, and Yamada S (2013) Effects of green tea on cytochrome Nebot N, Crettol S, d’Esposito F, Tattam B, Hibbs DE, and Murray M (2010) Par- P450 2B6, 2C8, 2C19, 2D6 and 3A activities in human liver and intestinal mi- ticipation of CYP2C8 and CYP3A4 in the N-demethylation of imatinib in human – crosomes. Drug Metab Pharmacokinet 28:244–249. hepatic microsomes. Br J Pharmacol 161:1059 1069. Miyazawa M, Shindo M, and Shimada T (2002) Sex differences in the metabolism of Negishi I, Tsuda-Tsukimoto M, Kozakai K, and Kume T (2007) Utility of cryopreserved (+)- and (-)-limonene enantiomers to carveol and perillyl alcohol derivatives by human hepatocytes for assessment of mechanism-based inactivation, in 8th In- cytochrome p450 enzymes in rat liver microsomes. Chem Res Toxicol 15:15–20. ternational ISXX Meeting; Sendai, Japan (October 9-12, 2007). Available from: http:// Mizuno Y, Tani N, Komuro S, Kanamaru H, and Nakatsuka I (2003) In vitro me- issx.confex.com/issx/intl8/webprogram/Paper6588.html (Accessed June 18, 2015). tabolism of perospirone in rat, monkey and human liver microsomes. Eur J Drug Neuvonen PJ (2010) Drug interactions with HMG-CoA reductase inhibitors (statins): Metab Pharmacokinet 28:59–65. the importance of CYP enzymes, transporters and pharmacogenetics. Curr Opin – Moller RA, Fisher JM, Taylor AE, Kolluri S, Gardner MJ, Obach RS, and Walsky RL Investig Drugs 11:323 332. – (2006) Effects of steady-state lasofoxifene on CYP2D6- and CYP2E1-mediated Neuvonen PJ (2012) Towards safer and more predictable drug treatment reflections metabolism. Ann Pharmacother 40:32–37. from studies of the First BCPT Prize awardee. Basic Clin Pharmacol Toxicol 110: – Monsarrat B, Royer I, Wright M, and Cresteil T (1997) Biotransformation of taxoids 207 218. by human cytochromes P450: structure-activity relationship. Bull Cancer 84: Neuvonen PJ, Niemi M, and Backman JT (2006) Drug interactions with lipid- 125–133. lowering drugs: mechanisms and clinical relevance. Clin Pharmacol Ther 80: 565–581. Moody DE, Slawson MH, Strain EC, Laycock JD, Spanbauer AC, and Foltz RL (2002) ’ A liquid chromatographic-electrospray ionization-tandem mass spectrometric Nguyen L, Holland J, Miles D, Engel C, Benrimoh N, O Reilly T, and Lacy S (2015) Pharmacokinetic (PK) drug interaction studies of cabozantinib: Effect of CYP3A method for determination of buprenorphine, its metabolite, norbuprenorphine, and inducer rifampin and inhibitor ketoconazole on cabozantinib plasma PK and effect a coformulant, naloxone, that is suitable for in vivo and in vitro metabolism of cabozantinib on CYP2C8 probe substrate rosiglitazone plasma PK. J Clin studies. Anal Biochem 306:31–39. Pharmacol 55:1012–1023. Moon Y, Kim SY, Ji HY, Kim YK, Chae HJ, Chae SW, and Lee HS (2007) Charac- Nicolas JM, Chanteux H, Rosa M, Watanabe S, and Stockis A (2012) Effect of gem- terization of cytochrome P450s mediating ipriflavone metabolism in human liver fibrozil on the metabolism of brivaracetam in vitro and in human subjects. Drug microsomes. Xenobiotica 37:246–259. Metab Dispos 40:1466–1472. Morioka Y, Otsu M, Naito S, and Imai T (2002) Phosphonate O-deethylation of [4-(4- Niemi M, Backman JT, Granfors M, Laitila J, Neuvonen M, and Neuvonen PJ bromo-2-cyano-phenylcarbamoyl) benzyl]-phosphonic acid diethyl ester, a lipopro- (2003a) Gemfibrozil considerably increases the plasma concentrations of rosigli- tein lipase-promoting agent, catalyzed by cytochrome P450 2C8 and 3A4 in human tazone. Diabetologia 46:1319–1323. liver microsomes. Drug Metab Dispos 30:301–306. Niemi M, Backman JT, Juntti-Patinen L, Neuvonen M, and Neuvonen PJ (2005a) Mount DL, Patchen LC, Nguyen-Dinh P, Barber AM, Schwartz IK, and Churchill FC Coadministration of gemfibrozil and itraconazole has only a minor effect on the (1986) Sensitive analysis of blood for amodiaquine and three metabolites by high- pharmacokinetics of the CYP2C9 and CYP3A4 substrate nateglinide. Br J Clin performance liquid chromatography with electrochemical detection. J Chromatogr – – Pharmacol 60:208 217. A 383:375 386. Niemi M, Backman JT, Kajosaari LI, Leathart JB, Neuvonen M, Daly AK, Eichelbaum Mukai Y, Toda T, Hayakawa T, Eliasson E, Rane A, and Inotsume N (2014) Losartan M, Kivistö KT, and Neuvonen PJ (2005b) Polymorphic organic anion transporting competitively inhibits CYP2C8-dependent paclitaxel metabolism in vitro. Biol polypeptide 1B1 is a major determinant of repaglinide pharmacokinetics. Clin – Pharm Bull 37:1550 1554. Pharmacol Ther 77:468–478. Mukkavilli R, Gundala SR, Yang C, Donthamsetty S, Cantuaria G, Jadhav GR, Niemi M, Backman JT, Neuvonen M, and Neuvonen PJ (2003b) Effects of gemfi- Vangala S, Reid MD, and Aneja R (2014) Modulation of cytochrome P450 metab- brozil, itraconazole, and their combination on the pharmacokinetics and pharma- olism and transport across intestinal epithelial barrier by ginger biophenolics. codynamics of repaglinide: potentially hazardous interaction between gemfibrozil PLoS One 9:e108386. and repaglinide. Diabetologia 46:347–351. Mukoyoshi M, Nishimura S, Hoshide S, Umeda S, Kanou M, Taniguchi K, Niemi M, Backman JT, Neuvonen M, Neuvonen PJ, and Kivistö KT (2000) Rifampin and Muroga H (2008) In vitro drug-drug interaction studies with febuxostat, a decreases the plasma concentrations and effects of repaglinide. Clin Pharmacol novel non-purine selective inhibitor of xanthine oxidase: plasma protein binding, Ther 68:495–500. identification of metabolic enzymes and cytochrome P450 inhibition. Xenobiotica Niemi M, Backman JT, and Neuvonen PJ (2004a) Effects of trimethoprim and ri- 38:496–510. fampin on the pharmacokinetics of the cytochrome P450 2C8 substrate rosiglita- Mürdter TE, Kerb R, Turpeinen M, Schroth W, Ganchev B, Böhmer GM, Igel S, zone. Clin Pharmacol Ther 76:239–249. Schaeffeler E, Zanger U, and Brauch H (2012) Genetic polymorphism of cyto- Niemi M, Kajosaari LI, Neuvonen M, Backman JT, and Neuvonen PJ (2004b) The chrome P450 2D6 determines oestrogen receptor activity of the major infertility CYP2C8 inhibitor trimethoprim increases the plasma concentrations of repagli- drug clomiphene via its active metabolites. Hum Mol Genet 21:1145–1154. nide in healthy subjects. Br J Clin Pharmacol 57:441–447. Muschler E, Lal J, Jetter A, Rattay A, Zanger U, Zadoyan G, Fuhr U, Niemi M, Leathart JB, Neuvonen M, Backman JT, Daly AK, and Neuvonen PJ and Kirchheiner J (2009) The role of human CYP2C8 and CYP2C9 variants in (2003c) Polymorphism in CYP2C8 is associated with reduced plasma concentra- pioglitazone metabolism in vitro. Basic Clin Pharmacol Toxicol 105:374–379. tions of repaglinide. Clin Pharmacol Ther 74:380–387. Muthiah YD, Lee WL, Teh LK, Ong CE, and Ismail R (2005) Genetic polymorphism of Niemi M, Neuvonen PJ, and Kivistö KT (2001) Effect of gemfibrozil on the pharmacoki- CYP2C8 in three Malaysian ethnics: CYP2C8*2 and CYP2C8*3 are found in netics and pharmacodynamics of glimepiride. Clin Pharmacol Ther 70:439–445. Malaysian Indians. J Clin Pharm Ther 30:487–490. Niemi M, Pasanen MK, and Neuvonen PJ (2011) Organic anion transporting poly- Muthiah YD, Ong CE, Sulaiman SA, Tan SC, and Ismail R (2012) In-vitro inhibitory peptide 1B1: a genetically polymorphic transporter of major importance for hepatic effect of Tualang honey on cytochrome P450 2C8 activity. J Pharm Pharmacol 64: drug uptake. Pharmacol Rev 63:157–181. 1761–1769. Niemi M, Tornio A, Pasanen MK, Fredrikson H, Neuvonen PJ, and Backman JT Mück W (1998) Rational assessment of the interaction profile of cerivastatin supports (2006) Itraconazole, gemfibrozil and their combination markedly raise the plasma its low propensity for drug interactions. Drugs 56 (Suppl 1):15–23, discussion 33. concentrations of loperamide. Eur J Clin Pharmacol 62:463–472. Mück W (2000) Clinical pharmacokinetics of cerivastatin. Clin Pharmacokinet 39: Nirogi R, Kandikere V, Palacharla RC, Bhyrapuneni G, Kanamarlapudi VB, 99–116. Ponnamaneni RK, and Manoharan AK (2014) Identification of a suitable and selec- Mück W, Ochmann K, Rohde G, Unger S, and Kuhlmann J (1998) Influence of tive inhibitor towards aldehyde oxidase catalyzed reactions. Xenobiotica 44:197–204. erythromycin pre- and co-treatment on single-dose pharmacokinetics of the HMG- Nirogi R, Palacharla RC, Uthukam V, Manoharan A, Srikakolapu SR, Kalaikadhiban CoA reductase inhibitor cerivastatin. Eur J Clin Pharmacol 53:469–473. I, Boggavarapu RK, Ponnamaneni RK, Ajjala DR, and Bhyrapuneni G (2015) Nadin L and Murray M (1999) Participation of CYP2C8 in retinoic acid 4-hydroxylation Chemical inhibitors of CYP450 enzymes in liver microsomes: combining selectivity in human hepatic microsomes. Biochem Pharmacol 58:1201–1208. and unbound fractions to guide selection of appropriate concentration in pheno- Naik H, Wu JT, Palmer R, and McLean L (2012) The effects of febuxostat on the typing assays. Xenobiotica 45:95–106. pharmacokinetic parameters of rosiglitazone, a CYP2C8 substrate. Br J Clin Nishihara M, Sudo M, Kawaguchi N, Takahashi J, Kiyota Y, Kondo T, and Asahi S Pharmacol 74:327–335. (2012) An unusual metabolic pathway of sipoglitazar, a novel antidiabetic agent: Role of CYP2C8 in Drug Metabolism and Interactions 237

cytochrome P450-catalyzed oxidation of sipoglitazar acyl glucuronide. Drug Metab Pelkonen O, Turpeinen M, Hakkola J, Honkakoski P, Hukkanen J, and Raunio H Dispos 40:249–258. (2008) Inhibition and induction of human cytochrome P450 enzymes: current sta- Nishimura M, Yaguti H, Yoshitsugu H, Naito S, and Satoh T (2003) Tissue distri- tus. Arch Toxicol 82:667–715. bution of mRNA expression of human cytochrome P450 isoforms assessed by high- Peng Y, Wu H, Zhang X, Zhang F, Qi H, Zhong Y, Wang Y, Sang H, Wang G, and Sun sensitivity real-time reverse transcription PCR. Yakugaku Zasshi 123:369–375. J (2015) A comprehensive assay for nine major cytochrome P450 enzymes activities Nishimura M, Yoshitsugu H, Naito S, and Hiraoka I (2002) Evaluation of gene in- with 16 probe reactions on human liver microsomes by a single LC/MS/MS run to duction of drug-metabolizing enzymes and transporters in primary culture of hu- support reliable in vitro inhibitory drug-drug interaction evaluation. Xenobiotica man hepatocytes using high-sensitivity real-time reverse transcription PCR. 45:961–977. Yakugaku Zasshi 122:339–361. Perez-Ruixo JJ, Piotrovskij V, Zhang S, Hayes S, De Porre P, and Zannikos P (2006) Nishiya Y, Hagihara K, Kurihara A, Okudaira N, Farid NA, Okazaki O, and Ikeda T Population pharmacokinetics of tipifarnib in healthy subjects and adult cancer (2009) Comparison of mechanism-based inhibition of human cytochrome P450 patients. Br J Clin Pharmacol 62:81–96. 2C19 by ticlopidine, clopidogrel, and prasugrel. Xenobiotica 39:836–843. Perloff ES, Mason AK, Dehal SS, Blanchard AP, Morgan L, Ho T, Dandeneau A, Niwa T, Sato R, Yabusaki Y, Ishibashi F, and Katagiri M (1999) Contribution of Crocker RM, Chandler CM, and Boily N (2009) Validation of cytochrome P450 human hepatic cytochrome P450s and steroidogenic CYP17 to the N-demethylation time-dependent inhibition assays: a two-time point IC50 shift approach facilitates of aminopyrine. Xenobiotica 29:187–193. kinact assay design. Xenobiotica 39:99–112. Niwa T, Tsutsui M, Kishimoto K, Yabusaki Y, Ishibashi F, and Katagiri M (2000) Pichard L, Curi-Pedrosa R, Bonfils C, Jacqz-Aigrain E, Domergue J, Joyeux H, Inhibition of drug-metabolizing enzyme activity in human hepatic cytochrome Cosme J, Guengerich FP, and Maurel P (1995) Oxidative metabolism of lanso- P450s by bisphenol A. Biol Pharm Bull 23:498–501. prazole by human liver cytochromes P450. Mol Pharmacol 47:410–418. Obach RS (2000) Inhibition of human cytochrome P450 enzymes by constituents of Picard N, Cresteil T, Djebli N, and Marquet P (2005) In vitro metabolism study of St. John’s Wort, an herbal preparation used in the treatment of depression. J buprenorphine: evidence for new metabolic pathways. Drug Metab Dispos 33: Pharmacol Exp Ther 294:88–95. 689–695. Obach RS, Walsky RL, and Venkatakrishnan K (2007) Mechanism-based in- Picard N, Rouguieg-Malki K, Kamar N, Rostaing L, and Marquet P (2011) CYP3A5 activation of human cytochrome p450 enzymes and the prediction of drug-drug genotype does not influence everolimus in vitro metabolism and clinical pharma- interactions. Drug Metab Dispos 35:246–255. cokinetics in renal transplant recipients. Transplantation 91:652–656. O’Donnell CJ, Grime K, Courtney P, Slee D, and Riley RJ (2007) The development of Pimazoni A (2009) The impact of tuberculosis treatment on glycaemic control and the a cocktail CYP2B6, CYP2C8, and CYP3A5 inhibition assay and a preliminary significant response to rosiglitazone. BMJ Case Rep 2009 10.1136/bcr.09.2008.0994. assessment of utility in a drug discovery setting. Drug Metab Dispos 35:381–385. PMDA (2014) Japanese Pharmaceuticals and Medical Devices Agency, review report: Ogilvie BW, Zhang D, Li W, Rodrigues AD, Gipson AE, Holsapple J, Toren P, Acofide. Available from: http://www.pmda.go.jp/files/000153467.pdf (Accessed and Parkinson A (2006) Glucuronidation converts gemfibrozil to a potent, September 15, 2015). metabolism-dependent inhibitor of CYP2C8: implications for drug-drug interac- Polasek TM, Elliot DJ, Lewis BC, and Miners JO (2004) Mechanism-based in- tions. Drug Metab Dispos 34:191–197. activation of human cytochrome P4502C8 by drugs in vitro. J Pharmacol Exp Ther Ohtsuki S, Schaefer O, Kawakami H, Inoue T, Liehner S, Saito A, Ishiguro N, Kishimoto 311:996–1007. W, Ludwig-Schwellinger E, and Ebner T (2012) Simultaneous absolute protein quan- Porubsky PR, Meneely KM, and Scott EE (2008) Structures of human cytochrome tification of transporters, cytochromes P450, and UDP-glucuronosyltransferases as a P-450 2E1. Insights into the binding of inhibitors and both small molecular weight novel approach for the characterization of individual human liver: comparison and fatty acid substrates. J Biol Chem 283:33698–33707. with mRNA levels and activities. Drug Metab Dispos 40:83–92. Prakash C, Wang W, O’Connell T, and Johnson KA (2008) CYP2C8- and CYP3A- Ohyama K, Nakajima M, Nakamura S, Shimada N, Yamazaki H, and Yokoi T (2000) mediated C-demethylation of (3-[(4-tert-butylbenzyl)-(pyridine-3-sulfonyl)-amino]- A significant role of human cytochrome P450 2C8 in amiodarone N-deethylation: methyl-phenoxy)-acetic acid (CP-533,536), an EP2 receptor-selective prostaglandin an approach to predict the contribution with relative activity factor. Drug Metab E2 agonist: characterization of metabolites by high-resolution liquid chromatography- Dispos 28:1303–1310. tandem mass spectrometry and liquid chromatography/mass spectrometry-nuclear Olesen OV and Linnet K (2000) Identification of the human cytochrome P450 iso- magnetic resonance. Drug Metab Dispos 36:2093–2103. forms mediating in vitro N-dealkylation of perphenazine. Br J Clin Pharmacol 50: Projean D, Baune B, Farinotti R, Flinois JP, Beaune P, Taburet AM, and Ducharme J 563–571. (2003a) In vitro metabolism of chloroquine: identification of CYP2C8, CYP3A4, and Ong CE, Coulter S, Birkett DJ, Bhasker CR, and Miners JO (2000) The xenobiotic CYP2D6 as the main isoforms catalyzing N-desethylchloroquine formation. Drug inhibitor profile of cytochrome P4502C8. Br J Clin Pharmacol 50:573–580. Metab Dispos 31:748–754. Paganotti GM, Cosentino V, Russo G, Tabacchi F, Gramolelli S, Coluzzi M, Projean D, Morin PE, Tu TM, and Ducharme J (2003b) Identification of CYP3A4 and Romano R (2014) Absence of the human CYP2C8*3 allele in Ugandan children and CYP2C8 as the major cytochrome P450 s responsible for morphine exposed to Plasmodium falciparum malaria. Infect Genet Evol 27:432–435. N-demethylation in human liver microsomes. Xenobiotica 33:841–854. Paganotti GM, Gallo BC, Verra F, Sirima BS, Nebié I, Diarra A, Coluzzi M, Prueksaritanont T, Gorham LM, Ma B, Liu L, Yu X, Zhao JJ, Slaughter DE, Arison and Modiano D (2011) Human genetic variation is associated with Plasmodium BH, and Vyas KP (1997) In vitro metabolism of simvastatin in humans [SBT] falciparum drug resistance. J Infect Dis 204:1772–1778. identification of metabolizing enzymes and effect of the drug on hepatic P450s. Paganotti GM, Gramolelli S, Tabacchi F, Russo G, Modiano D, Coluzzi M, Drug Metab Dispos 25:1191–1199. and Romano R (2012) Distribution of human CYP2C8*2 allele in three different Prueksaritanont T, Lu P, Gorham L, Sternfeld F, and Vyas KP (2000) Interspecies African populations. Malar J 11:125. comparison and role of human cytochrome P450 and flavin-containing mono- Pan Y, Tiong KH, Abd-Rashid BA, Ismail Z, Ismail R, Mak JW, and Ong CE (2012) in hepatic metabolism of L-775,606, a potent 5-HT(1D) receptor agonist. Inhibitory effects of cytochrome P450 enzymes CYP2C8, CYP2C9, CYP2C19 and Xenobiotica 30:47–59. CYP3A4 by Labisia pumila extracts. J Ethnopharmacol 143:586–591. Prueksaritanont T, Ma B, and Yu N (2003) The human hepatic metabolism of sim- Pan Y, Tiong KH, Abd-Rashid BA, Ismail Z, Ismail R, Mak JW, and Ong CE (2014) vastatin hydroxy acid is mediated primarily by CYP3A, and not CYP2D6. Br J Clin Effect of eurycomanone on cytochrome P450 isoforms CYP1A2, CYP2A6, CYP2C8, Pharmacol 56:120–124. CYP2C9, CYP2C19, CYP2E1 and CYP3A4 in vitro. J Nat Med 68:402–406. Prueksaritanont T, Richards KM, Qiu Y, Strong-Basalyga K, Miller A, Li C, Eisen- Pang CY, Mak JW, Ismail R, and Ong CE (2012) In vitro modulatory effects of handler R, and Carlini EJ (2005) Comparative effects of fibrates on drug metab- on human cytochrome P450 2C8 (CYP2C8). Naunyn Schmiedebergs olizing enzymes in human hepatocytes. Pharm Res 22:71–78. Arch Pharmacol 385:495–502. Prueksaritanont T, Tang C, Qiu Y, Mu L, Subramanian R, and Lin JH (2002) Effects Parikh S, Ouedraogo JB, Goldstein JA, Rosenthal PJ, and Kroetz DL (2007) Amo- of fibrates on metabolism of statins in human hepatocytes. Drug Metab Dispos 30: diaquine metabolism is impaired by common polymorphisms in CYP2C8: impli- 1280–1287. cations for malaria treatment in Africa. Clin Pharmacol Ther 82:197–203. Pusalkar S, Zhou X, Li Y, Liao M, Yang J, Chong S, Venkatakrishnan K, and Paris BL, Ogilvie BW, Scheinkoenig JA, Ndikum-Moffor F, Gibson R, and Parkinson Chowdhury S (2014) Absorption, metabolism, and excretion of alisertib A (2009) In vitro inhibition and induction of human liver cytochrome p450 enzymes (MLN8237), an aurora A kinase inhibitor, in patients with advanced solid tumors by milnacipran. Drug Metab Dispos 37:2045–2054. or lymphomas, in 19th North American ISSX/29th JSSX Meeting; San Francisco, Park JY, Kim KA, Kang MH, Kim SL, and Shin JG (2004) Effect of rifampin on the CA (October 19-23, 2014). Available from: http://issx.confex.com/issx/19NA/ pharmacokinetics of rosiglitazone in healthy subjects. Clin Pharmacol Ther 75: webprogrampreliminary/Paper34303.html (Accessed August 26, 2015). 157–162. Qi XY, Liang SC, Ge GB, Liu Y, Dong PP, Zhang JW, Wang AX, Hou J, Zhu LL, Parte P and Kupfer D (2005) Oxidation of tamoxifen by human flavin-containing and Yang L (2013) Inhibitory effects of sanguinarine on human liver cytochrome monooxygenase (FMO) 1 and FMO3 to tamoxifen-N-oxide and its novel reduction P450 enzymes. Food Chem Toxicol 56:392–397. back to tamoxifen by human cytochromes P450 and hemoglobin. Drug Metab Qu YQ, Fang ZZ, Yang L, Gao ZM, Liang R, Zhu LL, Dong PP, Zhang YY, Ge GB, Dispos 33:1446–1452. and Wang LM (2011) Reversible inhibition of four important human liver cyto- Pearce RE, Gotschall RR, Kearns GL, and Leeder JS (2001) Cytochrome P450 in- chrome P450 enzymes by diethylstilbestrol. Pharmazie 66:216–221. volvement in the biotransformation of cisapride and racemic norcisapride in vitro: Quintieri L, Palatini P, Moro S, and Floreani M (2011) Inhibition of cytochrome P450 differential activity of individual human CYP3A isoforms. Drug Metab Dispos 29: 2C8-mediated drug metabolism by the flavonoid diosmetin. Drug Metab Pharma- 1548–1554. cokinet 26:559–568. Pechandova K, Buzkova H, Matouskova O, Perlik F, and Slanar O (2012) Genetic Rae JM, Johnson MD, Lippman ME, and Flockhart DA (2001) Rifampin is a selective, polymorphisms of CYP2C8 in the Czech Republic. Genet Test Mol Biomarkers 16: pleiotropic inducer of drug metabolism genes in human hepatocytes: studies with 812–816. cDNA and oligonucleotide expression arrays. J Pharmacol Exp Ther 299:849–857. Pelkonen O (2015) Drug Metabolism - From In Vitro to In Vivo, From Simple to Rahman A, Korzekwa KR, Grogan J, Gonzalez FJ, and Harris JW (1994) Selective Complex: Reflections of the BCPT Nordic Prize 2014 Awardee. Basic Clin Phar- biotransformation of taxol to 6 alpha-hydroxytaxol by human cytochrome P450 macol Toxicol 117:147–155. 2C8. Cancer Res 54:5543–5546. Pelkonen O, Myllynen P, Taavitsainen P, Boobis AR, Watts P, Lake BG, Price RJ, Rana R, Chen Y, Ferguson SS, Kissling GE, Surapureddi S, and Goldstein JA (2010) Renwick AB, Gómez-Lechón MJ, and Castell JV (2001) Carbamazepine: a ‘blind’ Hepatocyte nuclear factor 4alpha regulates rifampicin-mediated induction of assessment of CVP-associated metabolism and interactions in human liver-derived CYP2C genes in primary cultures of human hepatocytes. Drug Metab Dispos 38: in vitro systems. Xenobiotica 31:321–343. 591–599. 238 Backman et al.

Raucy JL, Mueller L, Duan K, Allen SW, Strom S, and Lasker JM (2002) Expression drug-drug interactions of sulfonylureas with fibrates and statins. Br J Clin and induction of CYP2C P450 enzymes in primary cultures of human hepatocytes. Pharmacol 78:639–648. J Pharmacol Exp Ther 302:475–482. Schneck DW, Birmingham BK, Zalikowski JA, Mitchell PD, Wang Y, Martin PD, Relling MV, Aoyama T, Gonzalez FJ, and Meyer UA (1990) Tolbutamide and Lasseter KC, Brown CD, Windass AS, and Raza A (2004) The effect of gemfibrozil mephenytoin hydroxylation by human cytochrome P450s in the CYP2C subfamily. on the pharmacokinetics of rosuvastatin. Clin Pharmacol Ther 75:455–463. J Pharmacol Exp Ther 252:442–447. Schoch GA, Yano JK, Sansen S, Dansette PM, Stout CD, and Johnson EF (2008) Rettie AE, Wienkers LC, Gonzalez FJ, Trager WF, and Korzekwa KR (1994) Im- Determinants of cytochrome P450 2C8 substrate binding: structures of complexes paired (S)-warfarin metabolism catalysed by the R144C allelic variant of CYP2C9. with montelukast, troglitazone, felodipine, and 9-cis-retinoic acid. J Biol Chem Pharmacogenetics 4:39–42. 283:17227–17237. Reyderman L, Kosoglou T, Statkevich P, Pember L, Boutros T, Maxwell SE, Affrime Schoch GA, Yano JK, Wester MR, Griffin KJ, Stout CD, and Johnson EF (2004) M, and Batra V (2004) Assessment of a multiple-dose drug interaction between Structure of human microsomal cytochrome P450 2C8. Evidence for a peripheral ezetimibe, a novel selective absorption inhibitor and gemfibrozil. Int J fatty acid binding site. J Biol Chem 279:9497–9503. Clin Pharmacol Ther 42:512–518. Scordo MG, Pengo V, Spina E, Dahl ML, Gusella M, and Padrini R (2002) Influence of Rifkind AB, Lee C, Chang TK, and Waxman DJ (1995) Arachidonic acid metabolism CYP2C9 and CYP2C19 genetic polymorphisms on warfarin maintenance dose and by human cytochrome P450s 2C8, 2C9, 2E1, and 1A2: regioselective oxygenation metabolic clearance. Clin Pharmacol Ther 72:702–710. and evidence for a role for CYP2C enzymes in arachidonic acid epoxygenation in Schwartz JI, Stroh M, Gao B, Liu F, Rosko K, Zajic S, Meehan AJ, Ruckle J, Lai E, human liver microsomes. Arch Biochem Biophys 320:380–389. and Wagner JA (2009) Effects of laropiprant, a selective prostaglandin D(2) re- Robert M, Salvà M, Segarra R, Pavesi M, Esbri R, Roberts D, and Golor G (2007) The ceptor 1 antagonist, on the pharmacokinetics of rosiglitazone. Cardiovasc Ther 27: prokinetic cinitapride has no clinically relevant pharmacokinetic interaction and 239–245. effect on QT during coadministration with ketoconazole. Drug Metab Dispos 35: Sevior DK, Hokkanen J, Tolonen A, Abass K, Tursas L, Pelkonen O, and Ahokas JT 1149–1156. (2010) Rapid screening of commercially available herbal products for the inhibition Rochat B, Zoete V, Grosdidier A, von Grünigen S, Marull M, and Michielin O (2008) of major human hepatic cytochrome P450 enzymes using the N-in-one cocktail. In vitro biotransformation of imatinib by the tumor expressed CYP1A1 and Xenobiotica 40:245–254. CYP1B1. Biopharm Drug Dispos 29:103–118. Shahabi P, Siest G, Meyer UA, and Visvikis-Siest S (2014) Human cytochrome P450 Roco A, Quiñones L, Agúndez JA, García-Martín E, Squicciarini V, Miranda C, Garay epoxygenases: variability in expression and role in inflammation-related disorders. J, Farfán N, Saavedra I, and Cáceres D (2012) Frequencies of 23 functionally Pharmacol Ther 144:134–161. significant variant alleles related with metabolism of antineoplastic drugs in the Shimokawa Y, Yoda N, Kondo S, Yamamura Y, Takiguchi Y, and Umehara K (2015) chilean population: comparison with caucasian and asian populations. Front Genet Inhibitory Potential of Twenty Five Anti-tuberculosis Drugs on CYP Activities in 3:229. Human Liver Microsomes. Biol Pharm Bull 38:1425–1429 . Rodrigues AD, Kukulka MJ, Roberts EM, Ouellet D, and Rodgers TR (1996) Shitara Y, Hirano M, Sato H, and Sugiyama Y (2004) Gemfibrozil and its glucuronide [O-methyl 14C]naproxen O-demethylase activity in human liver microsomes: evi- inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)- dence for the involvement of cytochrome P4501A2 and P4502C9/10. Drug Metab mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: – Dispos 24:126 136. analysis of the mechanism of the clinically relevant drug-drug interaction between Rong H, Maclin D, Yao L, Lin J, Yin D, and Xu H (2008) Mechanism-based inhibition cerivastatin and gemfibrozil. J Pharmacol Exp Ther 311:228–236. of human liver microsomal cytochrome P450 3A by a focal adhesion kinase (FAK) Sidharta PN, Treiber A, and Dingemanse J (2015) Clinical pharmacokinetics and inhibitor PF-562,27, in 2nd Asian Pacific ISSX Meeting; Shanghai, China (May 11- pharmacodynamics of the endothelin receptor antagonist macitentan. Clin Phar- 13, 2008). Available from: http://issx.confex.com/issx/ap08/webprogram/start.html macokinet 54:457–471. (Accessed August 25, 2015). Sim J, Choi E, Lee YM, Jeong GS, and Lee S (2015) In vitro inhibition of human Rostami-Hodjegan A and Tucker GT (2007) Simulation and prediction of in vivo drug cytochrome P450 by cudratricusxanthone A. Food Chem Toxicol 81:171–175. metabolism in human populations from in vitro data. Nat Rev Drug Discov 6: – Simon T, Verstuyft C, Mary-Krause M, Quteineh L, Drouet E, Méneveau N, Steg PG, 140 148. Ferrières J, Danchin N, and Becquemont L; French Registry of Acute ST-Elevation Roustit M, Blondel E, Villier C, Fonrose X, and Mallaret MP (2010) Symptomatic and Non-ST-Elevation Myocardial Infarction (FAST-MI) Investigators (2009) Ge- hypoglycemia associated with trimethoprim/sulfamethoxazole and repaglinide in a netic determinants of response to clopidogrel and cardiovascular events. N Engl J diabetic patient. Ann Pharmacother 44:764–767. Med 360:363–375. Rowbotham SE, Boddy AV, Redfern CP, Veal GJ, and Daly AK (2010) Relevance of Singh R, Ting JG, Pan Y, Teh LK, Ismail R, and Ong CE (2008) Functional role of nonsynonymous CYP2C8 polymorphisms to 13-cis retinoic acid and paclitaxel Ile264 in CYP2C8: mutations affect haem incorporation and catalytic activity. hydroxylation. Drug Metab Dispos 38:1261–1266. Drug Metab Pharmacokinet 23:165–174. Röwer S, Bienzle U, Weise A, Lambertz U, Forst T, Otchwemah RN, Pfützner A, Sjögren E, Svanberg P, and Kanebratt KP (2012) Optimized experimental design for and Mockenhaupt FP (2005) Short communication: high prevalence of the cyto- the estimation of enzyme kinetic parameters: an experimental evaluation. Drug chrome P450 2C8*2 mutation in Northern Ghana. Trop Med Int Health 10: Metab Dispos 40:2273–2279. 1271–1273. Skerjanec A, Wang J, Maren K, and Rojkjaer L (2010) Investigation of the phar- Rowland P, Blaney FE, Smyth MG, Jones JJ, Leydon VR, Oxbrow AK, Lewis CJ, macokinetic interactions of deferasirox, a once-daily oral iron chelator, with mid- Tennant MG, Modi S, and Eggleston DS (2006) Crystal structure of human cyto- chrome P450 2D6. J Biol Chem 281:7614–7622. azolam, rifampin, and repaglinide in healthy volunteers. J Clin Pharmacol 50: – Rowland Yeo K, Rostami Hodjegan A, and Tucker GT (2004) Abundance of cyto- 205 213. chromes P450 in human liver: a meta-analysis. Br J Clin Pharmacol 57:687–688. Smith JA, Newman RA, Hausheer FH, and Madden T (2003) Evaluation of in vitro Sahi J, Black CB, Hamilton GA, Zheng X, Jolley S, Rose KA, Gilbert D, LeCluyse EL, drug interactions with karenitecin, a novel, highly lipophilic camptothecin de- – and Sinz MW (2003) Comparative effects of thiazolidinediones on in vitro P450 rivative in phase II clinical development. J Clin Pharmacol 43:1008 1014. enzyme induction and inhibition. Drug Metab Dispos 31:439–446. Smith MT (2003) Mechanisms of troglitazone hepatotoxicity. Chem Res Toxicol 16: – Sai Y, Dai R, Yang TJ, Krausz KW, Gonzalez FJ, Gelboin HV, and Shou M (2000) 679 687. Assessment of specificity of eight chemical inhibitors using cDNA-expressed cyto- Song M, Do H, Kwon OK, Yang EJ, Bae JS, Jeong TC, Song KS, and Lee S (2014a) A chromes P450. Xenobiotica 30:327–343. Comparison of the In Vitro Inhibitory Effects of Thelephoric Acid and SKF-525A on – Sakaeda T, Fujino H, Komoto C, Kakumoto M, Jin JS, Iwaki K, Nishiguchi K, Human Cytochrome P450 Activity. Biomol Ther (Seoul) 22:155 160. Nakamura T, Okamura N, and Okumura K (2006) Effects of acid and lactone forms Song M, Hwang JY, Lee MY, Jee JG, Lee YM, Bae JS, Kim JA, Lee SH, and Lee S of eight HMG-CoA reductase inhibitors on CYP-mediated metabolism and MDR1- (2014b) In vitro inhibitory effect of piperlonguminine isolated from Piper longum – mediated transport. Pharm Res 23:506–512. on human cytochrome P450 1A2. Arch Pharm Res 37:1063 1068. Säll C, Houston JB, and Galetin A (2012) A comprehensive assessment of repaglinide Sonnichsen DS, Liu Q, Schuetz EG, Schuetz JD, Pappo A, and Relling MV (1995) metabolic pathways: impact of choice of in vitro system and relative enzyme con- Variability in human cytochrome P450 paclitaxel metabolism. J Pharmacol Exp tribution to in vitro clearance. Drug Metab Dispos 40:1279–1289. Ther 275:566–575. Salminen KA, Meyer A, Jerabkova L, Korhonen LE, Rahnasto M, Juvonen RO, Soyama A, Hanioka N, Saito Y, Murayama N, Ando M, Ozawa S, and Sawada J Imming P, and Raunio H (2011) Inhibition of human drug metabolizing cytochrome (2002) Amiodarone N-deethylation by CYP2C8 and its variants, CYP2C8*3 and P450 enzymes by plant isoquinoline . Phytomedicine 18:533–538. CYP2C8 P404A. Pharmacol Toxicol 91:174–178. Salonen JS, Nyman L, Boobis AR, Edwards RJ, Watts P, Lake BG, Price RJ, Renwick Soyama A, Saito Y, Hanioka N, Murayama N, Nakajima O, Katori N, Ishida S, Sai K, AB, Gómez-Lechón MJ, and Castell JV (2003) Comparative studies on the cyto- Ozawa S, and Sawada JI (2001) Non-synonymous single nucleotide alterations chrome p450-associated metabolism and interaction potential of selegiline between found in the CYP2C8 gene result in reduced in vitro paclitaxel metabolism. Biol human liver-derived in vitro systems. Drug Metab Dispos 31:1093–1102. Pharm Bull 24:1427–1430. Sane RS, Ramsden D, Sabo JP, Cooper C, Rowland L, Ting N, Whitcher-Johnstone A, Soyinka JO, Onyeji CO, Nathaniel TI, Odunfa OO, and Ebeshi BU (2013) Effects of and Tweedie DJ (2015) Contribution of major metabolites towards complex drug- concurrent administration of efavirenz on the disposition kinetics of amodiaquine drug interactions of deleobuvir: in vitro predictions and in vivo outcomes. Drug in healthy volunteers. J Pharm Res 6:275–279. Metab Dispos pii: dmd.115.066985. Speed WC, Kang SP, Tuck DP, Harris LN, and Kidd KK (2009) Global variation in Sansen S, Yano JK, Reynald RL, Schoch GA, Griffin KJ, Stout CD, and Johnson EF CYP2C8-CYP2C9 functional haplotypes. Pharmacogenomics J 9:283–290. (2007) Adaptations for the oxidation of polycyclic aromatic hydrocarbons exhibited Spink DC, Eugster HP, Lincoln DW 2nd, Schuetz JD, Schuetz EG, Johnson JA, by the structure of human P450 1A2. J Biol Chem 282:14348–14355. Kaminsky LS, and Gierthy JF (1992) 17 beta-estradiol hydroxylation catalyzed by Sarasquete ME, García-Sanz R, Marín L, Alcoceba M, Chillón MC, Balanzategui human cytochrome P450 1A1: a comparison of the activities induced by 2,3,7,8- A, Santamaria C, Rosiñol L, de la Rubia J, and Hernandez MT (2008) tetrachlorodibenzo-p-dioxin in MCF-7 cells with those from heterologous expres- Bisphosphonate-related osteonecrosis of the jaw is associated with polymorphisms sion of the cDNA. Arch Biochem Biophys 293:342–348. of the cytochrome P450 CYP2C8 in multiple myeloma: a genome-wide single nu- Spink DC, Hayes CL, Young NR, Christou M, Sutter TR, Jefcoate CR, and Gierthy JF cleotide polymorphism analysis. Blood 112:2709–2712. (1994) The effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on estrogen metabolism in Schelleman H, Han X, Brensinger CM, Quinney SK, Bilker WB, Flockhart DA, Li L, MCF-7 breast cancer cells: evidence for induction of a novel 17 beta-estradiol and Hennessy S (2014) Pharmacoepidemiologic and in vitro evaluation of potential 4-hydroxylase. J Steroid Biochem Mol Biol 51:251–258. Role of CYP2C8 in Drug Metabolism and Interactions 239

Srivastava PK, Yun CH, Beaune PH, Ged C, and Guengerich FP (1991) Separation of Tornio A, Niemi M, Neuvonen PJ, and Backman JT (2008b) Trimethoprim and the human liver microsomal tolbutamide hydroxylase and (S)-mephenytoin 49-hydroxylase CYP2C8*3 allele have opposite effects on the pharmacokinetics of pioglitazone. cytochrome P-450 enzymes. Mol Pharmacol 40:69–79. Drug Metab Dispos 36:73–80. Staehli Hodel EM, Csajka C, Ariey F, Guidi M, Kabanywanyi AM, Duong S, Deco- Tornio A, Pasanen MK, Laitila J, Neuvonen PJ, and Backman JT (2005) Comparison sterd LA, Olliaro P, Beck HP, and Genton B (2013) Effect of single nucleotide of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (sta- polymorphisms in cytochrome P450 isoenzyme and N-acetyltransferase 2 genes on tins) as inhibitors of cytochrome P450 2C8. Basic Clin Pharmacol Toxicol 97: the metabolism of artemisinin-based combination therapies in malaria patients 104–108. from Cambodia and Tanzania. Antimicrob Agents Chemother 57:950–958. Totah RA and Rettie AE (2005) Cytochrome P450 2C8: substrates, inhibitors, Staffa JA, Chang J, and Green L (2002) Cerivastatin and reports of fatal rhabdo- pharmacogenetics, and clinical relevance. Clin Pharmacol Ther 77:341–352. myolysis. N Engl J Med 346:539–540. Tougou K, Gotou H, Ohno Y, and Nakamura A (2004) Stereoselective glucuronidation and Stage TB, Christensen MM, Feddersen S, Beck-Nielsen H, and Brøsen K (2013) The hydroxylation of etodolac by UGT1A9 and CYP2C9 in man. Xenobiotica 34:449–461. role of genetic variants in CYP2C8, LPIN1, PPARGC1A and PPARg on the trough Tracy TS, Korzekwa KR, Gonzalez FJ, and Wainer IW (1999) Cytochrome P450 steady-state plasma concentrations of rosiglitazone and on glycosylated haemo- isoforms involved in metabolism of the enantiomers of verapamil and norver- globin A1c in type 2 diabetes. Pharmacogenet Genomics 23:219–227. apamil. Br J Clin Pharmacol 47:545–552. Störmer E, von Moltke LL, Shader RI, and Greenblatt DJ (2000) Metabolism of the Tracy TS, Marra C, Wrighton SA, Gonzalez FJ, and Korzekwa KR (1997) In- antidepressant mirtazapine in vitro: contribution of cytochromes P-450 1A2, 2D6, volvement of multiple cytochrome P450 isoforms in naproxen O-demethylation. and 3A4. Drug Metab Dispos 28:1168–1175. Eur J Clin Pharmacol 52:293–298. Suarez-Kurtz G, Genro JP, de Moraes MO, Ojopi EB, Pena SD, Perini JA, Treluyer JM, Gueret G, Cheron G, Sonnier M, and Cresteil T (1997) Developmental Ribeiro-dos-Santos A, Romano-Silva MA, Santana I, and Struchiner CJ (2012) expression of CYP2C and CYP2C-dependent activities in the human liver: in-vivo/ Global pharmacogenomics: Impact of population diversity on the distribution of in-vitro correlation and inducibility. Pharmacogenetics 7:441–452. polymorphisms in the CYP2C cluster among Brazilians. Pharmacogenomics J 12: Tse S, Leung L, Raje S, Seymour M, Shishikura Y, and Obach RS (2014) Disposition 267–276. and metabolic profiling of [(14)C]cerlapirdine using accelerator mass spectrometry. Such E, Cervera J, Terpos E, Bagán JV, Avaria A, Gómez I, Margaix M, Ibañez M, Drug Metab Dispos 42:2023–2032. Luna I, and Cordón L (2011) CYP2C8 gene polymorphism and bisphosphonate- Turpeinen M, Uusitalo J, Jalonen J, and Pelkonen O (2005) Multiple P450 substrates related osteonecrosis of the jaw in patients with multiple myeloma. Haematologica in a single run: rapid and comprehensive in vitro interaction assay. Eur J Pharm – 96:1557 1559. Sci 24:123–132. Sun DX, Lu JC, Fang ZZ, Zhang YY, Cao YF, Mao YX, Zhu LL, Yin J, and Yang L Turpeinen M, Uusitalo J, Lehtinen T, Kailajärvi M, Pelkonen O, Vuorinen J, Tapa- (2010) Reversible inhibition of three important human liver cytochrome p450 en- nainen P, Stjernschantz C, Lammintausta R, and Scheinin M (2013) Effects of – zymes by tiliroside. Phytother Res 24:1670 1675. ospemifene on drug metabolism mediated by cytochrome P450 enzymes in humans Suttle AB, Grossmann KF, Ouellet D, Richards-Peterson LE, Aktan G, Gordon MS, in vitro and in vivo. Int J Mol Sci 14:14064–14075. LoRusso PM, Infante JR, Sharma S, and Kendra K (2015) Assessment of the drug Tzimas G and Nau H (2001) The role of metabolism and toxicokinetics in retinoid interaction potential and single- and repeat-dose pharmacokinetics of the BRAF teratogenesis. Curr Pharm Des 7:803–831. – inhibitor dabrafenib. J Clin Pharmacol 55:392 400. Ufer M, Svensson JO, Krausz KW, Gelboin HV, Rane A, and Tybring G (2004) Sutton D, Butler AM, Nadin L, and Murray M (1997) Role of CYP3A4 in human Identification of cytochromes P450 2C9 and 3A4 as the major catalysts of phen- hepatic diltiazem N-demethylation: inhibition of CYP3A4 activity by oxidized dil- procoumon hydroxylation in vitro. Eur J Clin Pharmacol 60:173–182. – tiazem metabolites. J Pharmacol Exp Ther 282:294 300. Unger M and Frank A (2004) Simultaneous determination of the inhibitory potency Suzuki A, Iida I, Tanaka F, Akimoto M, Fukushima K, Tani M, Ishizaki T, and Chiba of herbal extracts on the activity of six major cytochrome P450 enzymes using K (1999) Identification of human cytochrome P-450 isoforms involved in metabo- liquid chromatography/mass spectrometry and automated online extraction. Rapid lism of R(+)- and S(-)-gallopamil: utility of in vitro disappearance rate. Drug Metab – – Commun Mass Spectrom 18:2273 2281. Dispos 27:1254 1259. Václavíková R, Horský S, Simek P, and Gut I (2003) Paclitaxel metabolism in rat and Synold TW, Dussault I, and Forman BM (2001) The orphan nuclear receptor SXR human liver microsomes is inhibited by phenolic antioxidants. Naunyn Schmie- coordinately regulates drug metabolism and efflux. Nat Med 7:584–590. debergs Arch Pharmacol 368:200–209. Tachibana S, Fujimaki Y, Yokoyama H, Okazaki O, and Sudo K (2005) In vitro Walle T, Walle UK, Kumar GN, and Bhalla KN (1995) Taxol metabolism and dis- metabolism of the calmodulin antagonist DY-9760e (3-[2-[4-(3-chloro-2-methylphenyl)- position in cancer patients. Drug Metab Dispos 23:506–512. 1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidaz olylmethyl)-1H-indazole dihydro- Walles M, Wolf T, Jin Y, Ritzau M, Leuthold LA, Krauser J, Gschwind HP, Carcache chloride 3.5 hydrate) by human liver microsomes: involvement of cytochromes p450 D, Kittelmann M, and Ocwieja M (2013) Metabolism and disposition of the in atypical kinetics and potential drug interactions. Drug Metab Dispos 33: metabotropic glutamate receptor 5 antagonist (mGluR5) mavoglurant (AFQ056) in 1628–1636. healthy subjects. Drug Metab Dispos 41:1626–1641. Tamraz B, Fukushima H, Wolfe AR, Kaspera R, Totah RA, Floyd JS, Ma B, Chu C, Walsky RL, Gaman EA, and Obach RS (2005a) Examination of 209 drugs for in- Marciante KD, and Heckbert SR (2013) OATP1B1-related drug-drug and drug- hibition of cytochrome P450 2C8. J Clin Pharmacol 45:68–78. gene interactions as potential risk factors for cerivastatin-induced rhabdomyolysis. Walsky RL and Obach RS (2004) Validated assays for human cytochrome P450 ac- Pharmacogenet Genomics 23:355–364. tivities. Drug Metab Dispos 32:647–660. Tan AR, Dowlati A, Stein MN, Jones SF, Infante JR, Bendell J, Kane MP, Levinson Walsky RL, Obach RS, Gaman EA, Gleeson JP, and Proctor WR (2005b) Selective in- KT, Suttle AB, and Burris HA 3rd (2014) Phase I study of weekly paclitaxel in – combination with pazopanib and lapatinib in advanced solid malignancies. Br J hibition of human cytochrome P4502C8 by montelukast. Drug Metab Dispos 33:413 418. Walsky RL, Obach RS, Hyland R, Kang P, Zhou S, West M, Geoghegan KF, Helal CJ, Cancer 110:2647–2654. Tarkiainen EK, Holmberg MT, Tornio A, Neuvonen M, Neuvonen PJ, Backman JT, Walker GS, and Goosen TC (2012) Selective mechanism-based inactivation of and Niemi M (2015) Carboxylesterase 1 c.428G.A single nucleotide variation in- CYP3A4 by CYP3cide (PF-04981517) and its utility as an in vitro tool for de- creases the antiplatelet effects of clopidogrel by reducing its hydrolysis in humans. lineating the relative roles of CYP3A4 versus CYP3A5 in the metabolism of drugs. – Clin Pharmacol Ther 97:650–658. Drug Metab Dispos 40:1686 1697. Teng WC, Oh JW, New LS, Wahlin MD, Nelson SD, Ho HK, and Chan EC (2010) VandenBrink BM, Davis JA, Pearson JT, Foti RS, Wienkers LC, and Rock DA (2012) Mechanism-based inactivation of cytochrome P450 3A4 by lapatinib. Mol Phar- Cytochrome p450 architecture and cysteine nucleophile placement impact – macol 78:693–703. raloxifene-mediated mechanism-based inactivation. Mol Pharmacol 82:835 842. TGA (2014) Australian Public Assessment Report for Crizotinib. Available from: VandenBrink BM, Foti RS, Rock DA, Wienkers LC, and Wahlstrom JL (2011) https://www.tga.gov.au/file/882/download (Accessed September, 2015). Evaluation of CYP2C8 inhibition in vitro: utility of montelukast as a selective – Thatcher JE, Zelter A, and Isoherranen N (2010) The relative importance of CYP26A1 CYP2C8 probe substrate. Drug Metab Dispos 39:1546 1554. in hepatic clearance of all-trans retinoic acid. Biochem Pharmacol 80:903–912. Wang B, Sanchez RI, Franklin RB, Evans DC, and Huskey SE (2004) The in- Thomas M, Bayha C, Vetter S, Hofmann U, Schwarz M, Zanger UM, and Braeuning volvement of CYP3A4 and CYP2C9 in the metabolism of 17 alpha-ethinylestradiol. A (2015) Activating and Inhibitory Functions of WNT/b-Catenin in the Induction of Drug Metab Dispos 32:1209–1212. Cytochromes P450 by Nuclear Receptors in HepaRG Cells. Mol Pharmacol 87: Wang JS and DeVane CL (2003) Involvement of CYP3A4, CYP2C8, and CYP2D6 in 1013–1020. the metabolism of (R)- and (S)-methadone in vitro. Drug Metab Dispos 31:742–747. Thum T and Borlak J (2000) Gene expression in distinct regions of the heart. Lancet Wang JS, Neuvonen M, Wen X, Backman JT, and Neuvonen PJ (2002) Gemfibrozil 355:979–983. inhibits CYP2C8-mediated cerivastatin metabolism in human liver microsomes. Tomalik-Scharte D, Fuhr U, Hellmich M, Frank D, Doroshyenko O, Jetter A, Drug Metab Dispos 30:1352–1356. and Stingl JC (2011) Effect of the CYP2C8 genotype on the pharmacokinetics and Wang MZ, Saulter JY, Usuki E, Cheung YL, Hall M, Bridges AS, Loewen G, pharmacodynamics of repaglinide. Drug Metab Dispos 39:927–932. Parkinson OT, Stephens CE, and Allen JL (2006) CYP4F enzymes are the major Tornio A, Filppula AM, Kailari O, Neuvonen M, Nyrönen TH, Tapaninen T, Neuvonen enzymes in human liver microsomes that catalyze the O-demethylation of the PJ, Niemi M, and Backman JT (2014) Glucuronidation converts clopidogrel to a antiparasitic prodrug DB289 [2,5-bis(4-amidinophenyl)furan-bis-O-methyl- strong time-dependent inhibitor of CYP2C8: a phase II metabolite as a perpetrator of amidoxime]. Drug Metab Dispos 34:1985–1994. drug-drug interactions. Clin Pharmacol Ther 96:498–507. Wang Y, Wang M, Qi H, Pan P, Hou T, Li J, He G, and Zhang H (2014a) Pathway- Tornio A, Neuvonen PJ, and Backman JT (2006) The CYP2C8 inhibitor gemfibrozil dependent inhibition of paclitaxel hydroxylation by kinase inhibitors and assess- does not increase the plasma concentrations of zopiclone. Eur J Clin Pharmacol 62: ment of drug-drug interaction potentials. Drug Metab Dispos 42:782–795. 645–651. Wang Z, Wang S, Huang M, Hu H, Yu L, and Zeng S (2014b) Characterizing the effect Tornio A, Niemi M, Neuvonen M, Laitila J, Kalliokoski A, Neuvonen PJ, of cytochrome P450 (CYP) 2C8, CYP2C9, and CYP2D6 genetic polymorphisms on and Backman JT (2008a) The effect of gemfibrozil on repaglinide pharmacokinetics stereoselective N-demethylation of fluoxetine. Chirality 26:166–173. persists for at least 12 h after the dose: evidence for mechanism-based inhibition of Vargens DD, Petzl-Erler ML, and Suarez-Kurtz G (2012) Distribution of CYP2C CYP2C8 in vivo. Clin Pharmacol Ther 84:403–411. polymorphisms in an Amerindian population of Brazil. Basic Clin Pharmacol Tornio A, Niemi M, Neuvonen PJ, and Backman JT (2007) Stereoselective interaction Toxicol 110:396–400. between the CYP2C8 inhibitor gemfibrozil and racemic ibuprofen. Eur J Clin Varma MV, Lin J, Bi YA, Kimoto E, and Rodrigues AD (2015) Quantitative Pharmacol 63:463–469. Rationalization of Gemfibrozil Drug Interactions: Consideration of Transporters- 240 Backman et al.

Enzyme Interplay and the Role of Circulating Metabolite Gemfibrozil 1-O-b-Glucuronide. Xu C and Desta Z (2013) In vitro analysis and quantitative prediction of efavirenz Drug Metab Dispos 43:1108–1118. inhibition of eight cytochrome P450 (CYP) enzymes: major effects on CYPs 2B6, Varma MV, Lin J, Bi YA, Rotter CJ, Fahmi OA, Lam JL, El-Kattan AF, Goosen TC, 2C8, 2C9 and 2C19. Drug Metab Pharmacokinet 28:362–371. and Lai Y (2013) Quantitative prediction of repaglinide-rifampicin complex drug Yajima K, Uno Y, Murayama N, Uehara S, Shimizu M, Nakamura C, Iwasaki K, interactions using dynamic and static mechanistic models: delineating differential Utoh M, and Yamazaki H (2014) Evaluation of 23 lots of commercially available CYP3A4 induction and OATP1B1 inhibition potential of rifampicin. Drug Metab cryopreserved hepatocytes for induction assays of human cytochromes P450. Drug Dispos 41:966–974. Metab Dispos 42:867–871. Varma MV, Scialis RJ, Lin J, Bi YA, Rotter CJ, Goosen TC, and Yang X (2014) Yamamoto T, Suzuki A, and Kohno Y (2002) Application of microtiter plate assay to Mechanism-based pharmacokinetic modeling to evaluate transporter-enzyme interplay evaluate inhibitory effects of various compounds on nine cytochrome P450 isoforms in drug interactions and pharmacogenetics of glyburide. AAPS J 16:736–748. and to estimate their inhibition patterns. Drug Metab Pharmacokinet 17:437–448. Watanabe K, Yamaori S, Funahashi T, Kimura T, and Yamamoto I (2007) Cyto- Yamazaki H, Inoue K, Hashimoto M, and Shimada T (1999a) Roles of CYP2A6 and – chrome P450 enzymes involved in the metabolism of and CYP2B6 in nicotine C-oxidation by human liver microsomes. Arch Toxicol 73:65 70. cannabinol by human hepatic microsomes. Life Sci 80:1415–1419. Yamazaki H, Shibata A, Suzuki M, Nakajima M, Shimada N, Guengerich FP, Wattanachai N, Polasek TM, Heath TM, Uchaipichat V, Tassaneeyakul W, Tassa- and Yokoi T (1999b) Oxidation of troglitazone to a quinone-type metabolite cata- neeyakul W, and Miners JO (2011) In vitro-in vivo extrapolation of CYP2C8- lyzed by cytochrome P-450 2C8 and P-450 3A4 in human liver microsomes. Drug – catalyzed paclitaxel 6a-hydroxylation: effects of albumin on in vitro kinetic Metab Dispos 27:1260 1266. parameters and assessment of interindividual variability in predicted clearance. Yamazaki H and Shimada T (1999) Effects of arachidonic acid, prostaglandins, ret- – inol, retinoic acid and cholecalciferol on xenobiotic oxidations catalysed by human Eur J Clin Pharmacol 67:815 824. – Waxman DJ, Lapenson DP, Aoyama T, Gelboin HV, Gonzalez FJ, and Korzekwa K cytochrome P450 enzymes. Xenobiotica 29:231 241. Yamazaki H, Suzuki M, Tane K, Shimada N, Nakajima M, and Yokoi T (2000) In (1991) Steroid hormone hydroxylase specificities of eleven cDNA-expressed human vitro inhibitory effects of troglitazone and its metabolites on drug oxidation ac- cytochrome P450s. Arch Biochem Biophys 290:160–166. tivities of human cytochrome P450 enzymes: comparison with pioglitazone and Weber C, Funk C, Frank K, MacDonald A, and Charoin J (2005) In vitro studies and rosiglitazone. Xenobiotica 30:61–70. computer simulation of interactions between R483, a novel thiazolidinedione, and Yamazaki M, Li B, Louie SW, Pudvah NT, Stocco R, Wong W, Abramovitz M, cytochrome P450 substrates. Clin Pharmacol Ther 77:P74. Demartis A, Laufer R, and Hochman JH (2005) Effects of fibrates on human or- Wen X, Wang JS, Backman JT, Kivistö KT, and Neuvonen PJ (2001) Gemfibrozil is a – ganic anion-transporting polypeptide 1B1-, multidrug resistance protein 2- and P- potent inhibitor of human cytochrome P450 2C9. Drug Metab Dispos 29:1359 1361. glycoprotein-mediated transport. Xenobiotica 35:737–753. Wen X, Wang JS, Backman JT, Laitila J, and Neuvonen PJ (2002) Trimethoprim and Yang J, He MM, Niu W, Wrighton SA, Li L, Liu Y, and Li C (2012) Metabolic sulfamethoxazole are selective inhibitors of CYP2C8 and CYP2C9, respectively. capabilities of cytochrome P450 enzymes in Chinese liver microsomes compared – Drug Metab Dispos 30:631 635. with those in Caucasian liver microsomes. Br J Clin Pharmacol 73:268–284. Venepally P, Chen D, and Kemper B (1992) Transcriptional regulatory elements for Yang J, Liao M, Shou M, Jamei M, Yeo KR, Tucker GT, and Rostami-Hodjegan A – basal expression of cytochrome P450IIC genes. J Biol Chem 267:17333 17338. (2008) Cytochrome p450 turnover: regulation of synthesis and degradation, Venkatakrishnan K, Schmider J, Harmatz JS, Ehrenberg BL, von Moltke LL, Graf methods for determining rates, and implications for the prediction of drug inter- JA, Mertzanis P, Corbett KE, Rodriguez MC, and Shader RI (2001) Relative con- actions. Curr Drug Metab 9:384–394. tribution of CYP3A to amitriptyline clearance in humans: in vitro and in vivo Yang TJ, Shou M, Korzekwa KR, Gonzalez FJ, Gelboin HV, and Yang SK (1998) Role studies. J Clin Pharmacol 41:1043–1054. of cDNA-expressed human cytochromes P450 in the metabolism of diazepam. Ventura V, Solà J, Celma C, Peraire C, and Obach R (2011) In vitro metabolism of Biochem Pharmacol 55:889–896. irosustat, a novel steroid sulfatase inhibitor: interspecies comparison, metabolite Yano JK, Wester MR, Schoch GA, Griffin KJ, Stout CD, and Johnson EF (2004) The identification, and metabolic enzyme identification. Drug Metab Dispos 39: structure of human microsomal cytochrome P450 3A4 determined by X-ray crys- 1235–1246. tallography to 2.05-A resolution. J Biol Chem 279:38091–38094. Vermeir M, Hemeryck A, Cuyckens F, Francesch A, Bockx M, Van Houdt J, Steemans Yasar U, Lundgren S, Eliasson E, Bennet A, Wiman B, de Faire U, and Rane A (2002) K, Mannens G, Avilés P, and De Coster R (2009) In vitro studies on the metabolism Linkage between the CYP2C8 and CYP2C9 genetic polymorphisms. Biochem of trabectedin (YONDELIS) in monkey and man, including human CYP reaction Biophys Res Commun 299:25–28. phenotyping. Biochem Pharmacol 77:1642–1654. Ye L, Tang L, Gong Y, Lv C, Zheng Z, Jiang Z, and Liu Z (2011) Characterization of Veronese ME, Doecke CJ, Mackenzie PI, McManus ME, Miners JO, Rees DL, Gasser metabolites and human P450 isoforms involved in the microsomal metabolism of R, Meyer UA, and Birkett DJ (1993) Site-directed mutation studies of human liver mesaconitine. Xenobiotica 41:46–58. cytochrome P-450 isoenzymes in the CYP2C subfamily. Biochem J 289:533–538. Yeo CW, Lee SJ, Lee SS, Bae SK, Kim EY, Shon JH, Rhee BD, and Shin JG (2011) Whitfield LR, Porcari AR, Alvey C, Abel R, Bullen W, and Hartman D (2011) Effect of Discovery of a novel allelic variant of CYP2C8, CYP2C8*11, in Asian populations and its gemfibrozil and fenofibrate on the pharmacokinetics of atorvastatin. J Clin clinical effect on the rosiglitazone disposition in vivo. Drug Metab Dispos 39:711–716. Pharmacol 51:378–388. Yi P, Hadden C, Kulanthaivel P, Calvert N, Annes W, Brown T, Barbuch RJ, Chaudhary Whomsley E, Brochot A, Dell’Aiera S, Delepine X, and Espié P (2007) Identification of A, Ayan-Oshodi MA, and Ring BJ (2010) Disposition and metabolism of semagacestat, a the cytochrome P450 isoforms responsible for the hydroxylation of Brivaracetam, gamma-secretase inhibitor, in humans. Drug Metab Dispos 38:554–565. in AAPS Annual Meeting and Exposition, San Diego, California (November 12-16, Yoshida K, Maeda K, and Sugiyama Y (2012) Transporter-mediated drug–drug in- 2007). AAPS J 9 (Suppl 2):T3408. teractions involving OATP substrates: predictions based on in vitro inhibition Vicente J, González-Andrade F, Soriano A, Fanlo A, Martínez-Jarreta B, and Sinués B studies. Clin Pharmacol Ther 91:1053–1064. (2014) Genetic polymorphisms of CYP2C8, CYP2C9 and CYP2C19 in Ecuadorian Yu L, Jiang Y, Wang L, Sheng R, Hu Y, and Zeng S (2013a) Metabolism of BYZX in Mestizo and Spaniard populations: a comparative study. MolBiolRep41:1267–1272. human liver microsomes and cytosol: identification of the metabolites and meta- Vickers AE, Sinclair JR, Zollinger M, Heitz F, Glänzel U, Johanson L, and Fischer V bolic pathways of BYZX. PLoS One 8:e59882. (1999) Multiple cytochrome P-450s involved in the metabolism of terbinafine Yu L, Shi D, Ma L, Zhou Q, and Zeng S (2013b) Influence of CYP2C8 polymorphisms suggest a limited potential for drug-drug interactions. Drug Metab Dispos 27: on the hydroxylation metabolism of paclitaxel, repaglinide and ibuprofen enan- – tiomers in vitro. Biopharm Drug Dispos 34:278–287. 1029 1038.  Vickers AE, Zollinger M, Dannecker R, Tynes R, Heitz F, and Fischer V (2001) In Yuan Y, Qiu X, Nikolic D, Chen SN, Huang K, Li G, Pauli GF, and van Breemen RB vitro metabolism of tegaserod in human liver and intestine: assessment of drug (2014) Inhibition of human cytochrome P450 enzymes by hops (Humulus lupulus) and hop prenylphenols. Eur J Pharm Sci 53:55–61. interactions. Drug Metab Dispos 29:1269–1276. Zeldin DC, DuBois RN, Falck JR, and Capdevila JH (1995) Molecular cloning, ex- Vincent SH, Reed JR, Bergman AJ, Elmore CS, Zhu B, Xu S, Ebel D, Larson P, Zeng pression and characterization of an endogenous human cytochrome P450 arachi- W, and Chen L (2007) Metabolism and excretion of the dipeptidyl peptidase 4 donic acid epoxygenase isoform. Arch Biochem Biophys 322:76–86. inhibitor [14C]sitagliptin in humans. Drug Metab Dispos 35:533–538. Zelko I, Sueyoshi T, Kawamoto T, Moore R, and Negishi M (2001) The peptide near Williams PA, Cosme J, Ward A, Angove HC, Matak Vinkovic D, and Jhoti H (2003) the C terminus regulates receptor CAR nuclear translocation induced by xen- Crystal structure of human cytochrome P450 2C9 with bound warfarin. Nature – – ochemicals in mouse liver. Mol Cell Biol 21:2838 2846. 424:464 468. Zhang D, Wang L, Chandrasena G, Ma L, Zhu M, Zhang H, Davis CD, Winter HR and Unadkat JD (2005) Identification of cytochrome P450 and arylamine and Humphreys WG (2007a) Involvement of multiple cytochrome P450 and UDP- N-acetyltransferase isoforms involved in sulfadiazine metabolism. Drug Metab glucuronosyltransferase enzymes in the in vitro metabolism of muraglitazar. Drug – Dispos 33:969 976. Metab Dispos 35:139–149. Winter HR, Wang Y, and Unadkat JD (2000) CYP2C8/9 mediate dapsone Zhang JW, Liu Y, Cheng J, Li W, Ma H, Liu HT, Sun J, Wang LM, He YQ, and Wang N-hydroxylation at clinical concentrations of dapsone. Drug Metab Dispos 28: Y (2007b) Inhibition of human liver cytochrome P450 by star fruit juice. J Pharm – 865 868. Pharm Sci 10:496–503. Wong H, Chen JZ, Chou B, Halladay JS, Kenny JR, La H, Marsters JC Jr, Plise E, Zhang YY, Liu Y, Zhang JW, Ge GB, Liu HX, Wang LM, Sun J, and Yang L (2009a) Rudewicz PJ, and Robarge K (2009) Preclinical assessment of the absorption, C-7 configuration as one of determinants in taxanes metabolism by human cyto- distribution, metabolism and excretion of GDC-0449 (2-chloro-N-(4-chloro-3-(pyridin- chrome P450 enzymes. Xenobiotica 39:903–914. 2-yl)phenyl)-4-(methylsulfonyl)benzamide), an orally bioavailable systemic Hedgehog Zhang YY, Liu Y, Zhang JW, Ge GB, Wang LM, Sun J, and Yang L (2009b) Char- signalling pathway inhibitor. Xenobiotica 39:850–861. acterization of human cytochrome P450 isoforms involved in the metabolism of Wu X, Zuo J, Guo T, and Yuan L (2013) CYP2C8 polymorphism frequencies among 7-epi-paclitaxel. Xenobiotica 39:283–292. Han, Uighur, Hui, and Mongolian Chinese populations. Genet Test Mol Biomarkers Zharikova OL, Fokina VM, Nanovskaya TN, Hill RA, Mattison DR, Hankins GD, 17:104–108. and Ahmed MS (2009) Identification of the major human hepatic and placental Wu J, Shaw J, Dubaisi S, Valeriote F, and Li J (2014) In vitro metabolism and drug- enzymes responsible for the biotransformation of glyburide. Biochem Pharmacol drug interaction potential of UTL-5g, a novel chemo- and radioprotective agent. 78:1483–1490. Drug Metab Dispos 42:2058–2067. Zheng YF, Bae SH, Choi EJ, Park JB, Kim SO, Jang MJ, Park GH, Shin WG, Oh E, Xiaoping L, Zhong F, and Tan X (2013) Cytochrome P450 2C8 and drug metabolism. and Bae SK (2014) Evaluation of the in vitro/in vivo drug interaction potential of Curr Top Med Chem 13:2241–2253. BST204, a purified dry extract of ginseng, and its four bioactive ginsenosides Role of CYP2C8 in Drug Metabolism and Interactions 241

through cytochrome P450 inhibition/induction and UDP-glucuronosyltransferase Zhou L, Naraharisetti SB, Liu L, Wang H, Lin YS, Isoherranen N, Unadkat JD, inhibition. Food Chem Toxicol 68:117–127. Hebert MF, and Mao Q (2010) Contributions of human cytochrome P450 enzymes Zheng YF, Bae SH, Kwon MJ, Park JB, Choi HD, Shin WG, and Bae SK (2013) to glyburide metabolism. Biopharm Drug Dispos 31:228–242. Inhibitory effects of astaxanthin, b-cryptoxanthin, canthaxanthin, lutein, and Zhu L, Liu X, Zhu L, Zhang X, Fu X, Huang J, and Yuan M (2014) Identification of zeaxanthin on cytochrome P450 enzyme activities. Food Chem Toxicol 59:78–85. human cytochrome P450 isozymes involved in the metabolism of naftopidil enan- tiomers in vitro. J Pharm Pharmacol 66:1534–1551. Zhong DN, Wu JZ, and Li GJ (2013) Association between CYP2C8 (rs1934951) Zou L, Harkey MR, and Henderson GL (2002) Effects of herbal components on cDNA- polymorphism and bisphosphonate-related osteonecrosis of the jaws in patients on expressed cytochrome P450 enzyme catalytic activity. Life Sci 71:1579–1589. – bisphosphonate therapy: a meta-analysis. Acta Haematol 129:90 95. Zvyaga T, Chang SY, Chen C, Yang Z, Vuppugalla R, Hurley J, Thorndike D, Wagner Zhou D, Andersson TB, and Grimm SW (2011) In vitro evaluation of potential drug- A, Chimalakonda A, and Rodrigues AD (2012) Evaluation of six proton pump in- drug interactions with ticagrelor: cytochrome P450 reaction phenotyping, in- hibitors as inhibitors of various human cytochromes P450: focus on cytochrome hibition, induction, and differential kinetics. Drug Metab Dispos 39:703–710. P450 2C19. Drug Metab Dispos 40:1698–1711.