4
Toxicology Letters 202 (2011) 129–132 Contents lists available at ScienceDirect Toxicology Letters journal homepage: www.elsevier.com/locate/toxlet Drug–drug interactions by azole antifungals: Beyond a dogma of CYP3A4 enzyme activity inhibition Zdenek Dvorak Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 11, 783 71 Olomouc, Czech Republic article info Article history: Received 30 December 2010 Received in revised form 26 January 2011 Accepted 28 January 2011 Available online 17 February 2011 Keywords: Azoles Cytochrome CYP3A4 Pregnane X receptor Glucocorticoid receptor Drug interactions abstract Azoles antifungals are widespread used drugs for various medicinal indications. These drugs are well known for their numerous drug–drug interactions, which are believed to occur via inhibition of CYP3A4 enzymatic activity and consequently altering pharmacokinetic of co-administered drugs. In the current communication a complex view on the molecular interactions between azoles antimycotics and CYP3A4 is presented. Beside inhibition of CYP3A4 catalytic activity, azoles influence transcriptional activity of preg- nane X receptor (PXR) and consequently expression of drug-metabolizing enzymes, including CYP3A4. Interactions between azoles and PXR occur by multiple mechanisms, including modulation of ligand- dependent activation of PXR (agonism, antagonism) or affecting recruitment of PXR co-activators SRC-1 (steroid receptor co-activator 1) and HNF4 (hepatocyte nuclear factor 4). Miconazole and ketoconazole are antagonists of glucocorticoid receptor (GR), therefore these drugs inhibit GR-mediated expression of PXR and drug metabolizing cytochromes P450. In addition, PXR and GR are key regulators of interme- diary metabolism (e.g. carbohydrate, lipids or bile acids homeostasis) and many other cellular functions (e.g. immune response), hence, the interactions between azoles and PXR/GR are of broader physiologi- cal importance. In conclusion, while inhibition of CYP3A4 enzymatic activity by azoles is considered as primary cause of azoles drug–drug interactions, the effects of azoles on PXR and GR should be taken in account. Apart from CYP3A4, azoles influence the expression and activity of others drug-metabolizing cytochromes P450. © 2011 Published by Elsevier Ireland Ltd. 1. Azoles antifungals Azole antimycotics are large group of antifungal drugs, compris- ing imidazoles (e.g. ketoconazole and miconazole), triazoles (e.g. itraconazole and fluconazole) and thiazoles (e.g. abafungin). These drugs are used to treat primary fungal infections, but also as pro- phylactic or supporting treatment of immune suppressed patients (perioperative states, anticancer chemotherapy, HIV, organ trans- plants etc.). The mechanism of action of azoles is inhibition of fungal cytochrome P450 CYP51A1 (lanosterol-14a-demethylase) that cat- alyzes conversion of lanosterol to ergosterol (Heeres et al.). There are numerous reports on the drug–drug interactions caused by azoles; e.g. with bortezomib (Venkatakrishnan et al., 2009), bosutinib (Dvorak and PavekAbbas et al.), statins (Molden et al., 2008), venlafaxine (Lindh et al., 2003), tricyclic antidepres- sants (Spina et al., 1997b), carbamazepine (Spina et al., 1997a), Abbreviations: CAR, constitutive androstane receptor; GR, glucocorticoid recep- tor; HNF4, hepatocyte nuclear factor 4 alpha; PXR, pregnane X receptor; RXR, retinoid X receptor alpha; SRC-1, steroid receptor co-activator 1. Tel.: +420 58 5634903; fax: +420 58 5634905. E-mail address: [email protected] antivirotics (Kaeser et al., 2009) and many others (Gubbins and Heldenbrand). The majority of drug–drug interactions are caused by inhibition of drug-metabolizing enzymes. In case of azoles, the inhibition of CYP3A4 (but also CYP2C9) enzymatic activity is con- sidered as the main source of drug–drug interactions by azoles. Indeed, azole antifungals are potent inhibitors of human CYP3A4, as revealed by several studies (Lamb et al., 2000; Monostory et al., 2004; Sakaeda et al., 2005; Wang et al., 2007; Youdim et al., 2008) (Fig. 1(1)). In the current communication, several other mechanisms by which azole antifungals influence human drug-metabolizing enzymes are described. 2. Pregnane X receptor An orphan nuclear receptor, pregnane X receptor (PXR; NR1I2) is key transcriptional regulator of drug-metabolizing enzymes, including CYP3A4 (Lehmann et al., 1998). In its active form, PXR is in complex with heterodimerization partner retinoid X receptor alpha (RXR; NR2B1) (Pascussi et al., 2008) and with several co- activators, such as steroid receptor co-activator 1 (SRC-1) (Watkins et al., 2003) and hepatocyte nuclear factor 4 alpha (HNF4)(Jover et al., 2001, 2009; Tirona and Kim, 2005). 0378-4274/$ – see front matter © 2011 Published by Elsevier Ireland Ltd. doi:10.1016/j.toxlet.2011.01.027

Drug–drug interactions by azole antifungals: Beyond a dogma of CYP3A4 enzyme activity inhibition

Embed Size (px)

Citation preview

Da

ZD

a

ARRAA

KACPGD

1

iidp(pca

c2es

tr

0d

Toxicology Letters 202 (2011) 129–132

Contents lists available at ScienceDirect

Toxicology Letters

journa l homepage: www.e lsev ier .com/ locate / tox le t

rug–drug interactions by azole antifungals: Beyond a dogma of CYP3A4 enzymectivity inhibition

denek Dvorak ∗

epartment of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 11, 783 71 Olomouc, Czech Republic

r t i c l e i n f o

rticle history:eceived 30 December 2010eceived in revised form 26 January 2011ccepted 28 January 2011vailable online 17 February 2011

eywords:zolesytochrome CYP3A4regnane X receptorlucocorticoid receptor

a b s t r a c t

Azoles antifungals are widespread used drugs for various medicinal indications. These drugs are wellknown for their numerous drug–drug interactions, which are believed to occur via inhibition of CYP3A4enzymatic activity and consequently altering pharmacokinetic of co-administered drugs. In the currentcommunication a complex view on the molecular interactions between azoles antimycotics and CYP3A4 ispresented. Beside inhibition of CYP3A4 catalytic activity, azoles influence transcriptional activity of preg-nane X receptor (PXR) and consequently expression of drug-metabolizing enzymes, including CYP3A4.Interactions between azoles and PXR occur by multiple mechanisms, including modulation of ligand-dependent activation of PXR (agonism, antagonism) or affecting recruitment of PXR co-activators SRC-1(steroid receptor co-activator 1) and HNF4� (hepatocyte nuclear factor 4�). Miconazole and ketoconazoleare antagonists of glucocorticoid receptor (GR), therefore these drugs inhibit GR-mediated expression of

rug interactions PXR and drug metabolizing cytochromes P450. In addition, PXR and GR are key regulators of interme-diary metabolism (e.g. carbohydrate, lipids or bile acids homeostasis) and many other cellular functions(e.g. immune response), hence, the interactions between azoles and PXR/GR are of broader physiologi-cal importance. In conclusion, while inhibition of CYP3A4 enzymatic activity by azoles is considered asprimary cause of azoles drug–drug interactions, the effects of azoles on PXR and GR should be taken inaccount. Apart from CYP3A4, azoles influence the expression and activity of others drug-metabolizing

cytochromes P450.

. Azoles antifungals

Azole antimycotics are large group of antifungal drugs, compris-ng imidazoles (e.g. ketoconazole and miconazole), triazoles (e.g.traconazole and fluconazole) and thiazoles (e.g. abafungin). Theserugs are used to treat primary fungal infections, but also as pro-hylactic or supporting treatment of immune suppressed patientsperioperative states, anticancer chemotherapy, HIV, organ trans-lants etc.). The mechanism of action of azoles is inhibition of fungalytochrome P450 CYP51A1 (lanosterol-14a-demethylase) that cat-lyzes conversion of lanosterol to ergosterol (Heeres et al.).

There are numerous reports on the drug–drug interactions

aused by azoles; e.g. with bortezomib (Venkatakrishnan et al.,009), bosutinib (Dvorak and PavekAbbas et al.), statins (Moldent al., 2008), venlafaxine (Lindh et al., 2003), tricyclic antidepres-ants (Spina et al., 1997b), carbamazepine (Spina et al., 1997a),

Abbreviations: CAR, constitutive androstane receptor; GR, glucocorticoid recep-or; HNF4�, hepatocyte nuclear factor 4 alpha; PXR, pregnane X receptor; RXR�,etinoid X receptor alpha; SRC-1, steroid receptor co-activator 1.∗ Tel.: +420 58 5634903; fax: +420 58 5634905.

E-mail address: [email protected]

378-4274/$ – see front matter © 2011 Published by Elsevier Ireland Ltd.oi:10.1016/j.toxlet.2011.01.027

© 2011 Published by Elsevier Ireland Ltd.

antivirotics (Kaeser et al., 2009) and many others (Gubbins andHeldenbrand). The majority of drug–drug interactions are causedby inhibition of drug-metabolizing enzymes. In case of azoles, theinhibition of CYP3A4 (but also CYP2C9) enzymatic activity is con-sidered as the main source of drug–drug interactions by azoles.Indeed, azole antifungals are potent inhibitors of human CYP3A4,as revealed by several studies (Lamb et al., 2000; Monostory et al.,2004; Sakaeda et al., 2005; Wang et al., 2007; Youdim et al., 2008)(Fig. 1(1)).

In the current communication, several other mechanismsby which azole antifungals influence human drug-metabolizingenzymes are described.

2. Pregnane X receptor

An orphan nuclear receptor, pregnane X receptor (PXR; NR1I2)is key transcriptional regulator of drug-metabolizing enzymes,including CYP3A4 (Lehmann et al., 1998). In its active form, PXR

is in complex with heterodimerization partner retinoid X receptoralpha (RXR�; NR2B1) (Pascussi et al., 2008) and with several co-activators, such as steroid receptor co-activator 1 (SRC-1) (Watkinset al., 2003) and hepatocyte nuclear factor 4 alpha (HNF4�) (Joveret al., 2001, 2009; Tirona and Kim, 2005).

130 Z. Dvorak / Toxicology Letters 202 (2011) 129–132

Fig. 1. Effects of azole antifungals on human CYP3A4. (1) azoles inhibit catalytic activity of CYP3A4 enzyme; (2) ketoconazole disrupts interaction between SRC-1 and PXR;( omotec anscri R in la

fl(S2oTPPttd2bi(asIieazSoaPim

3) ketoconazole disrupts interaction between HNF4� and PXR; (4) itraconazole prlotrimazole are the ligands for PXR and augment basal and rifampicin-mediated trnhibit/potentiate PXR-mediated induction of CYP3A4; (7) ketoconazole binds to PXre antagonists of GR.

The most of studies were carried out with ketoconazole. Con-icting data were obtained regarding the effects of ketoconazolebut also fluconazole and clotrimazole) on CYP3A4 expression.ome authors described that azoles induce CYP3A4 (Luo et al.,002; Svecova et al., 2008), whereas others observed inhibitionf xenobiotic-inducible expression of CYP3A4 (Wang et al., 2007).his behavior indicates partial agonism of ketoconazole againstXR. However, azoles also interfere with interactions betweenXR and its transcriptional co-activators. Huang et al. describedhat xenobiotic-mediated induction of CYP3A4 gene transcrip-ion was inhibited by ketoconazole. They found that ketoconazoleisrupted the interaction between PXR and SRC-1 (Huang et al.,007) (Fig. 1(2)). Lim et al. (2009) demonstrated that the inhi-ition of the interaction of PXR with HNF4� might be involved

n ketoconazole-mediated inhibition of CYP3A4 gene expressionFig. 1(3)). In our recent paper, we have observed additive andntagonistic interactions of azoles with rifampicin on PXR tran-criptional activity and CYP3A4 expression (Svecova et al., 2008).traconazole stimulated recruitment of SRC-1 to PXR, but it did notnteract with PXR ligand binding domain (Fig. 1(4)). Oxiconazole,conazole and miconazole were identified as potent PXR ligandsnd inducers of CYP3A4 (Fig. 1(5)). We observed that ketocona-ole suppressed rifampicin-mediated CYP3A4 transactivation andRC-1 recruitment to PXR, which is in accordance with data from

ther authors (Huang et al., 2007; Wang et al., 2007) (Fig. 1(6)). Inddition, ketoconazole diminished interaction of rifampicin withXR ligand binding domain (Fig. 1(7)). Interestingly, ketoconazolenduced CYP3A4 mRNA through CYP3A4 promoter response ele-

ents, albeit it is not an agonist of PXR (Svecova et al., 2008).

s interaction between SRC-1 and PXR; (5) oxiconazole, econazole, miconazole andiptional activity of PXR; (6) ketoconazole, miconazole, fluconazole and oxiconazoleigand binding domain; (8) GR up-regulates PXR; (9) ketoconazole and miconazole

Overall, azole antimycotics differentially affect PXR tran-scriptional activity, with consequences in CYP3A4 expression.Interactions between azoles and PXR-signaling pathway are cer-tainly the cause of drug–drug interactions. Importantly, PXR isimportant regulator of metabolism of carbohydrates, lipids, bileacids and cholesterol. Therefore, interactions between azoles andPXR are of broader clinical and physiological significance.

3. Glucocorticoid receptor

Glucocorticoid receptor (GR; NR3C1) belongs to the familyof steroid/thyroid receptors. It regulates many cellular functionssuch as homeostasis, cell differentiation, proliferation, growth anddevelopment, and host defense. At molecular level, after bindingof glucocorticoid hormone (e.g. cortisol and dexamethasone), GRtranslocates to nucleus, where it forms homodimer GR�/GR� thattriggers transcriptional response. GR plays crucial role in regula-tion of drug metabolism, since it up-regulates xenoreceptors PXR(Pascussi et al., 2000, 2001) and CAR (Pascussi et al., 2003) (con-stitutive androstane receptor; NR1I3) and consequently indirectlycontrols expression of PXR- and CAR-dependent genes (Dvorak andPavek) (Fig. 1(8)).

Recently, it was demonstrated that ketoconazole and micona-zole (but not fluconazole) are antagonists of human GR in primary

cultures of human hepatocytes (Fig. 1(9)). Consistently, these twoazoles down-regulated xenoreceptors PXR and CAR, and conse-quently PXR- and CAR-dependent genes, including CYP2B6, CYP2C9and CYP3A4 were diminished (Duret et al., 2006). Inhibition of GRtranscriptional activity certainly produces many side-physiological

Letter

etit

cnRsfheee2crPCsV

4

iCtcCaCGopm

C

A

t

R

A

D

D

E

F

G

G

H

Z. Dvorak / Toxicology

ffects in addition to down-regulation of PXR and CAR. Fur-hermore, diminution of PXR and CAR will have consequencesn metabolism of carbohydrates, lipids, bile acids and choles-erol.

The role of GR in the regulation of drug-metabolizingytochromes P450 is very complex, comprising several mecha-isms. Apart from regulating the regulatory proteins (PXR, CAR,XRs etc.), GR is involved in direct (cis) and indirect (trans) tran-criptional regulation of P450s (Dvorak and Pavek). For instance,unctional GR response elements were identified in promoters ofuman CYP2C8 (Ferguson et al., 2005), CYP2C9 (Gerbal-Chaloint al., 2002), CYP2C19 (Chen et al., 2003) and CYP3A5 (Schuetzt al., 1996) genes. Trans-regulation of human CYP2A6 (Onicat al., 2008) and CYP3A4 (El-Sankary et al., 2002; Pavek et al.,007) by GR, was demonstrated, involving HNF4� in the pro-ess. Therefore, antagonizing GR by ketoconazole and miconazoleesults in down-regulation of drug-metabolizing cytochromes450 CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP3A4 andYP3A5. Glucocorticoids also positively regulate CYP1A2 expres-ion, by mechanisms not yet understood (Monostory et al., 2009;rzal et al., 2008).

. Conclusion

Azoles antifungals are one of the most frequently used drugsn human pharmacotherapy. They are potent inhibitors of humanYP3A4. For these reasons, azoles are subject of tight control inerms of drug–drug interactions. The majority of clinical trials arearried out with respect to inhibitory potency of azoles againstYP3A4 enzyme. In the current contribution, it is showed, thatpart from inhibition of CYP3A4 catalytic activity, azoles influenceYP3A4 activity by multiple mechanisms, including antagonizingR and interfering with PXR transcriptional activity. The interactionf azoles with PXR and GR will certainly results in more com-lex side effects of azoles than in inhibition of CYP3A4-mediatedetabolism of co-administered drugs.

onflict of interest statement

The author declares no conflict of interest.

cknowledgements

Our laboratory is supported by the grants from the Czech Scien-ific Agency GACR503/10/0579 and GACR304/10/0149.

eferences

bbas, R., Hug, B.A., Leister, C., Burns, J., Sonnichsen, D., 2010. Effect of ketoconazoleon the pharmacokinetics of oral bosutinib in healthy subjects. J. Clin. Pharmacol.

uret, C., Daujat-Chavanieu, M., Pascussi, J.M., Pichard-Garcia, L., Balaguer, P.,Fabre, J.M., Vilarem, M.J., Maurel, P., Gerbal-Chaloin, S., 2006. Ketoconazole andmiconazole are antagonists of the human glucocorticoid receptor: consequenceson the expression and function of the constitutive androstane receptor and thepregnane X receptor. Mol. Pharmacol. 70 (1), 329–339.

vorak, Z., Pavek, P., 2010. Regulation of drug-metabolizing cytochrome P450enzymes by glucocorticoids. Drug Metab. Rev. 42 (4), 621–635.

l-Sankary, W., Bombail, V., Gibson, G.G., Plant, N., 2002. Glucocorticoid-mediatedinduction of CYP3A4 is decreased by disruption of a protein: DNA interactiondistinct from the pregnane X receptor response element. Drug Metab. Dispos.30 (9), 1029–1034.

erguson, S.S., Chen, Y., LeCluyse, E.L., Negishi, M., Goldstein, J.A., 2005. HumanCYP2C8 is transcriptionally regulated by the nuclear receptors constitutiveandrostane receptor, pregnane X receptor, glucocorticoid receptor, and hepaticnuclear factor 4alpha. Mol. Pharmacol. 68 (3), 747–757.

erbal-Chaloin, S., Daujat, M., Pascussi, J.M., Pichard-Garcia, L., Vilarem, M.J., Mau-rel, P., 2002. Transcriptional regulation of CYP2C9 gene. Role of glucocorticoidreceptor and constitutive androstane receptor. J. Biol. Chem. 277 (1), 209–217.

ubbins, P.O., Heldenbrand, S., 2010. Clinically relevant drug interactions of currentantifungal agents. Mycoses 53 (2), 95–113.

eeres, J., Meerpoel, L., Lewi, P.C., 2010. Molecules 15 (6), 4129–4188.

s 202 (2011) 129–132 131

Huang, H., Wang, H., Sinz, M., Zoeckler, M., Staudinger, J., Redinbo, M.R., Teotico,D.G., Locker, J., Kalpana, G.V., Mani, S., 2007. Inhibition of drug metabolism byblocking the activation of nuclear receptors by ketoconazole. Oncogene 26 (2),258–268.

Chen, Y., Ferguson, S.S., Negishi, M., Goldstein, J.A., 2003. Identification of con-stitutive androstane receptor and glucocorticoid receptor binding sites in theCYP2C19 promoter. Mol. Pharmacol. 64 (2), 316–324.

Jover, R., Bort, R., Gomez-Lechon, M.J., Castell, J.V., 2001. Cytochrome P450 regulationby hepatocyte nuclear factor 4 in human hepatocytes: a study using adenovirus-mediated antisense targeting. Hepatology 33 (3), 668–675.

Jover, R., Moya, M., Gomez-Lechon, M.J., 2009. Transcriptional regulation ofcytochrome p450 genes by the nuclear receptor hepatocyte nuclear factor 4-alpha. Curr. Drug Metab. 10 (5), 508–519.

Kaeser, B., Zandt, H., Bour, F., Zwanziger, E., Schmitt, C., Zhang, X., 2009. Drug–druginteraction study of ketoconazole and ritonavir-boosted saquinavir. Antimicrob.Agents Chemother. 53 (2), 609–614.

Lamb, D.C., Kelly, D.E., Baldwin, B.C., Kelly, S.L., 2000. Differential inhibition of humanCYP3A4 and Candida albicans CYP51 with azole antifungal agents. Chem. Biol.Interact. 125 (3), 165–175.

Lehmann, J.M., McKee, D.D., Watson, M.A., Willson, T.M., Moore, J.T., Kliewer, S.A.,1998. The human orphan nuclear receptor PXR is activated by compounds thatregulate CYP3A4 gene expression and cause drug interactions. J. Clin. Invest. 102(5), 1016–1023.

Lim, Y.P., Kuo, S.C., Lai, M.L., Huang, J.D., 2009. Inhibition of CYP3A4 expressionby ketoconazole is mediated by the disruption of pregnane X receptor, steroidreceptor coactivator-1, and hepatocyte nuclear factor 4alpha interaction. Phar-macogenet. Genomics 19 (1), 11–24.

Lindh, J.D., Annas, A., Meurling, L., Dahl, M.L., Al-Shurbaji, A., 2003. Effect of ketocona-zole on venlafaxine plasma concentrations in extensive and poor metabolisersof debrisoquine. Eur. J. Clin. Pharmacol. 59 (5–6), 401–406.

Luo, G., Cunningham, M., Kim, S., Burn, T., Lin, J., Sinz, M., Hamilton, G., Rizzo, C., Jolley,S., Gilbert, D., Downey, A., Mudra, D., Graham, R., Carroll, K., Xie, J., Madan, A.,Parkinson, A., Christ, D., Selling, B., LeCluyse, E., Gan, L.S., 2002. CYP3A4 inductionby drugs: correlation between a pregnane X receptor reporter gene assay andCYP3A4 expression in human hepatocytes. Drug Metab. Dispos. 30 (7), 795–804.

Molden, E., Skovlund, E., Braathen, P., 2008. Risk management of simvastatin oratorvastatin interactions with CYP3A4 inhibitors. Drug Saf. 31 (7), 587–596.

Monostory, K., Hazai, E., Vereczkey, L., 2004. Inhibition of cytochrome P450enzymes participating in p-nitrophenol hydroxylation by drugs known asCYP2E1 inhibitors. Chem. Biol. Interact. 147 (3), 331–340.

Monostory, K., Pascussi, J.M., Kobori, L., Dvorak, Z., 2009. Hormonal regulation ofCYP1A expression. Drug Metab. Rev. 41 (4), 547–572.

Onica, T., Nichols, K., Larin, M., Ng, L., Maslen, A., Dvorak, Z., Pascussi, J.M., Vilarem,M.J., Maurel, P., Kirby, G.M., 2008. Dexamethasone-mediated up-regulation ofhuman CYP2A6 involves the glucocorticoid receptor and increased binding ofhepatic nuclear factor 4{alpha} to the proximal promoter. Mol. Pharmacol. 73(2), 451–460.

Pascussi, J.M., Busson-Le Coniat, M., Maurel, P., Vilarem, M.J., 2003. Transcriptionalanalysis of the orphan nuclear receptor constitutive androstane receptor (NR1I3)gene promoter: identification of a distal glucocorticoid response element. Mol.Endocrinol. 17 (1), 42–55.

Pascussi, J.M., Drocourt, L., Fabre, J.M., Maurel, P., Vilarem, M.J., 2000. Dexametha-sone induces pregnane X receptor and retinoid X receptor-alpha expression inhuman hepatocytes: synergistic increase of CYP3A4 induction by pregnane Xreceptor activators. Mol. Pharmacol. 58 (2), 361–372.

Pascussi, J.M., Drocourt, L., Gerbal-Chaloin, S., Fabre, J.M., Maurel, P., Vilarem, M.J.,2001. Dual effect of dexamethasone on CYP3A4 gene expression in human hep-atocytes. Sequential role of glucocorticoid receptor and pregnane X receptor.Eur. J. Biochem. 268 (24), 6346–6358.

Pascussi, J.M., Gerbal-Chaloin, S., Duret, C., Daujat-Chavanieu, M., Vilarem, M.J., Mau-rel, P., 2008. The tangle of nuclear receptors that controls xenobiotic metabolismand transport: crosstalk and consequences. Annu. Rev. Pharmacol. Toxicol. 48,1–32.

Pavek, P., Cerveny, L., Svecova, L., Brysch, M., Libra, A., Vrzal, R., Nachtigal, P., Staud, F.,Ulrichova, J., Fendrich, Z., Dvorak, Z., 2007. Examination of glucocorticoid recep-tor alpha-mediated transcriptional regulation of P-glycoprotein CYP3A4, andCYP2C9 genes in placental trophoblast cell lines. Placenta 28 (10), 1004–1011.

Sakaeda, T., Iwaki, K., Kakumoto, M., Nishikawa, M., Niwa, T., Jin, J.S., Nakamura,T., Nishiguchi, K., Okamura, N., Okumura, K., 2005. Effect of micafungin oncytochrome P450 3A4 and multidrug resistance protein 1 activities, and itscomparison with azole antifungal drugs. J. Pharm. Pharmacol. 57 (6), 759–764.

Schuetz, J.D., Schuetz, E.G., Thottassery, J.V., Guzelian, P.S., Strom, S., Sun, D., 1996.Identification of a novel dexamethasone responsive enhancer in the humanCYP3A5 gene and its activation in human and rat liver cells. Mol. Pharmacol.49 (1), 63–72.

Spina, E., Arena, D., Scordo, M.G., Fazio, A., Pisani, F., Perucca E, 1997a. Eleva-tion of plasma carbamazepine concentrations by ketoconazole in patients withepilepsy. Ther. Drug Monit. 19 (5), 535–538.

Spina, E., Avenoso, A., Campo, G.M., Scordo, M.G., Caputi, A.P., Perucca E., 1997b.Effect of ketoconazole on the pharmacokinetics of imipramine and desipramine

in healthy subjects. Br. J. Clin. Pharmacol. 43 (3), 315–318.

Svecova, L., Vrzal, R., Burysek, L., Anzenbacherova, E., Cerveny, L., Grim, J., Tre-jtnar, F., Kunes, J., Pour, M., Staud, F., Anzenbacher, P., Dvorak, Z., Pavek,P., 2008. Azole antimycotics differentially affect rifampicin-induced pregnaneX receptor-mediated CYP3A4 gene expression. Drug Metab. Dispos. 36 (2),339–348.

1 Letter

T

V

V

binding promotes the specific interaction between ligand and the pregnane Xreceptor. J. Mol. Biol. 331 (4), 815–828.

32 Z. Dvorak / Toxicology

irona, R.G., Kim, R.B., 2005. Nuclear receptors and drug disposition gene regulation.J. Pharm. Sci. 94 (6), 1169–1186.

enkatakrishnan, K., Rader, M., Ramanathan, R.K., Ramalingam, S., Chen, E., Riordan,W., Trepicchio, W., Cooper, M., Karol, M., von Moltke, L., Neuwirth, R., Egorin, M.,Chatta, G., 2009. Effect of the CYP3A inhibitor ketoconazole on the pharmacoki-netics and pharmacodynamics of bortezomib in patients with advanced solidtumors: a prospective, multicenter, open-label, randomized, two-way crossover

drug–drug interaction study. Clin. Ther. 31 (Pt 2), 2444–2458.

rzal, R., Daujat-Chavanieu, M., Pascussi, J.M., Ulrichova, J., Maurel, P., Dvorak,Z., 2008. Microtubules-interfering agents restrict aryl hydrocarbon receptor-mediated CYP1A2 induction in primary cultures of human hepatocytes viac-jun-N-terminal kinase and glucocorticoid receptor. Eur. J. Pharmacol. 581 (3),244–254.

s 202 (2011) 129–132

Wang, H., Huang, H., Li, H., Teotico, D.G., Sinz, M., Baker, S.D., Staudinger,J., Kalpana, G., Redinbo, M.R., Mani, S., 2007. Activated pregnenolone X-receptor is a target for ketoconazole and its analogs. Clin. Cancer Res. 13 (8),2488–2495.

Watkins, R.E., Davis-Searles, P.R., Lambert, M.H., Redinbo, M.R., 2003. Coactivator

Youdim, K.A., Lyons, R., Payne, L., Jones, B.C., Saunders, K., 2008. An auto-mated, high-throughput, 384 well Cytochrome P450 cocktail IC50 assay usinga rapid resolution LC-MS/MS end-point. J. Pharm. Biomed. Anal. 48 (1),92–99.