Expert Opinion on Drug Metabolism & Toxicology

ISSN: 1742-5255 (Print) 1744-7607 (Online) Journal homepage: http://www.tandfonline.com/loi/iemt20

Opportunities and challenges in using human hepatocytes in cytochrome P450 induction assays Zdenek Dvorak To cite this article: Zdenek Dvorak (2015): Opportunities and challenges in using human hepatocytes in cytochrome P450 induction assays, Expert Opinion on Drug Metabolism & Toxicology, DOI: 10.1517/17425255.2016.1125881 To link to this article: http://dx.doi.org/10.1517/17425255.2016.1125881

Accepted author version posted online: 27 Nov 2015.

Submit your article to this journal

Article views: 20

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=iemt20 Download by: [University of California, San Diego]

Date: 04 December 2015, At: 00:32

PUBLISHER: TAYLOR & FRANCIS JOURNAL: EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY DOI: 10.1517/17425255.2016.1125881

OPPORTUNITIES AND CHALLENGES IN USING HUMAN HEPATOCYTES IN CYTOCHROME P450

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

INDUCTION ASSAYS

Zdenek Dvorak Department of Cell Biology and Genetics Faculty of Science, Palacky University Olomouc Slechtitelu 27, Olomouc 783 71 Czech Republic Tel: +420-58-563-4901

Fax: +420-58-563-4901

Email: [email protected]

ABSTRACT Introduction: Identification of inducers of xenobiotic-metabolizing cytochrome P450s is of topical interest. The issue mainly concerns three sectors: (i) pre-clinical testing of drug candidates and testing existing drugs and their combinations; (ii) food safety applications with regards to additives, contaminants and adulterants; (iii) environmental applications, comprising detection and identification of endocrine disruptors. Areas covered: A literature search was performed using the PubMed database, covering state-of-the-art of human hepatocyte (HHs) culture use, and their exploitation for identification of P450s inducers. A list of CYPs inducers identified by HHs is provided.

1

Expert opinion: Primary cultures of HHs had long been considered as a gold standard for induction assays of xenobiotic-metabolizing enzymes. Owing to several shortcomings of HHs, alternative approaches such as immortalization of HHs, use of cell lines, generation of clonal cell lines from HHs, use of iPS, cells from humanized animals etc. were employed. While yielding particular advantage, overall, alternatives to HHs still remain an avenue for discrete applications or technical situations. Thus, HHs remain the most suitable model for

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

complex CYP induction studies. The summary may be effectively expressed by SWOT analysis.

Keywords: drug-drug interactions; drug-metabolizing enzymes; gene induction; human hepatocytes

Article highlights box: •

Human hepatocytes HHs are the gold standard for induction assays of xenobiotic metabolizing P450s



HHs provide the opportunity to study induction of phase II enzymes and xenobiotics transporters



HHs have complex signaling and metabolizing pathways, providing highly physiological model



HHs allow studying the effects of metabolites and maternal drugs simultaneously



HHs avoid interspecies differences, which are important in induction process

2

1. INTRODUCTION Metabolism of drugs and other xenobiotics in humans occurs dominantly in liver, but also in several extrahepatic tissues including intestine, lungs, skin, placenta etc. An integral part of xenobiotics metabolism is so called Phase I., which is also referred as to oxido-reductive & hydrolytic phase. The purpose of Phase I. is to increase the polarity of xenobiotic by the

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

means of introduction of polar group in the molecule, conversion of less polar group to more polar one, or by unmasking the polar group by hydrolysis. The major role in Phase I. oxidoreductive reactions plays a superfamily of drug-metabolizing enzymes cytochromes P450 (CYPs). Biotransformation of xenobiotics is inseparable from a phenomenon called drug-drug interactions, which means that ultimate effect of drug #1 is influenced by the presence of drug #2. Clinically relevant pharmacokinetic drug-drug interactions involving cytochromes P450 occur by two main mechanisms: (i) The inhibition of particular CYP or the competition for CYP by the drug [1, 2]; (ii) The induction of particular CYP by the drug = the topic of the current paper [3]. The induction of drug-metabolizing CYPs by xenobiotics, i.e. de novo protein synthesis, is mediated through several ligand-activated transcriptional factors, including: (i) Xenoreceptors: aryl hydrocarbon receptor (AhR), pregnane X receptor (PXR), constitutive androstane receptor (CAR); (ii) Steroid receptors: glucocorticoid receptor (GR), estrogen receptor (ER); (iii) Nuclear receptors: vitamin D receptor (VDR), retinoic acid receptors (RARs), retinoic X receptors (RXRs), hepatocyte nuclear factor 4 1 (HNF4 1), peroxisome proliferator-activated receptors (PPARs). Ligand-receptor complex binds to its cognate DNAbinding sequence in CYP promoter and gene transcription is triggered resulting in classical axis CYP mRNA – CYP protein - functional CYP enzyme. It is worth to emphasize that 3

xenoreceptors mediate transcriptional regulation of drug-metabolizing CYPs, but also of many other enzymes and genes involved in intermediary cell metabolism, such as CYPs participating in metabolism of steroid hormones (CYP17A1) cholesterol (CYP46A1), eicosanoids (CYP4A11, CYP4F2), vitamin D (CYP24A1) or retinoids (CYP26A1). Vice versa, steroid and nuclear receptors are important transcriptional regulators of drugmetabolizing CYPs [4-7].

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

Unlike pharmacodynamic drug-drug interactions, pharmacokinetic ones are not principally foreseeable and appropriate experimental tools are needed to predict and prevent them. The existing experimental approaches are extensive. The first issue is the choice of species. While induction studies on xenobiotics vs P450s were carried out in many species, including rodents (mouse, rat, guinea pig), aquatic organisms (zebrafish, rainbow trout, catfish), pig, cat, dog, chicken, camel etc., for the purposes of studies related to drug-drug interactions, environmental exposure or food safety, the human models remain the systems of choice. The reasons are multiple, including interspecies differences in CYP nomenclature, substrate specificity or transcriptional regulation. The second issue is the complexity of the experimental models. The model with highest degree of integrity for human studies is a man, which is exploitable in late stages of clinical studies. The opposite model, regarding the complexity, is perhaps isolated biomacromolecule (e.g. nuclear receptor), often only ligandbinding sequence, and the interaction between xenobiotic and the molecule is either directly measured, or studied in silico. Between these two extremes in complexity, the large number of experimental models exists, including perfused organs, tissue cultures, tissue slices, cell suspensions or adherent cells in various spatial arrangements etc. The choice of model according to its integrity depends on multiple parameters, including the number of tested compounds, the time schedule of experiments, or the purpose of the study. Human hepatocytes (HHs) are highly complex experimental system, representing closely liver

4

physiology and pathophysiology. On the other hand, it is still classical in vitro model, raising minimal ethical issues. The following section will deal with the model of human hepatocytes for induction studies of cytochrome P450 by xenobiotics.

2. BODY 2.1. Source of human hepatocytes

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

The only source of human hepatocytes is the liver from alive human (i.e. not post mortem). Acquisition of liver tissue raises ethical issues that are addressed in compliance with national laws [8, 9]. Liver tissue for preparation of human hepatocytes is obtained from surplus tissue obtained from small liver biopsies or partial hepatectomies carried out for medical purposes unrelated to human hepatocytes research. The weakness of such a source is that liver tissue may be contaminated or afflicted by a pathology for which the medical intervention was carried out (e.g. cancer, cirrhosis, hepatitis, alcoholic liver disease etc.). Another source of liver tissue is from multiorgan donors, i.e. from patients diagnosed for brain death, still having organs such as kidney, heart, liver, lung etc. vital and in condition suitable for transplantation. In case that there was not found recipient for liver from multiorgan donor, the organ could be used for research purposes. With increasing capabilities in terms of transportation, networking and communication between medical centers, the livers from transplant patients for isolation of human hepatocytes are scarce. On the other hand, such a liver is healthy, without pathological burden.

2.2. Storage and maintenance of human hepatocytes A specific feature of human hepatocytes is that they are terminally differentiated cells, which do not proliferate in vitro. Therefore, once isolated, a great challenge is to store human hepatocytes. Enormous efforts were expended for development and optimization of

5

cryopreservation procedures. Nowadays, cryopreserved human hepatocytes for induction studies cytochrome P450 are commercially available [10, 11]. The disadvantage of cryopreserved hepatocytes is their limited amount for experiments, because cryotube contains usually about 2 mL of cell suspension, giving limited number of attached viable cells. Novel research directions deal with long-term cold preservation of human hepatocytes, which are intended also for liver cell biotherapy [12]. The second shortcoming of human hepatocytes in

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

primary culture is their dedifferentiation, which means that they lose their hepatospecific functions with increasing time in culture. So called short-term cultures of human hepatocytes are exploitable for induction studies up to 10-14 days upon their isolation. Several culture protocols were developed to allow human hepatocytes in primary culture remain fully functional for long periods of time, allowing induction studies over 2 months in culture [13].

2.3. Experimental models of human hepatocytes Hepatocytes in stirred suspensions may be used only for short-term, often metabolic, experiments, lasting few hours. The majority of cytochrome P450 induction assays in human hepatocytes is carried out in the monolayers, so called primary cultures of human hepatocytes, which are attached usually on collagen-coated plastic ware. Various spatial arrangements were developed and invented in laboratories, having particular advantages over primary cultures, but often compensated by loss of comfort during experimentation and some trouble shootings. For instance, induction studies were performed in sandwich cultures (mimics passage of xenobiotics through extra-cellular matrix) [14, 15], in various 3D arrangements including in matrigel (mimics spatial situation in liver) [16, 17], and in various arrangements using immobilized cells – bioreactors (mimics blood stream flow) [18].

6

2.4. Alternative models to human hepatocytes Various approaches were applied in order to design and developed alternative models to “classical primary human hepatocytes: (i) In vitro, isolated hepatocytes do not longer retain their proliferative capacity, but cell growth can be boosted by immortalization of hepatocytes. Well-defined immortalization genes can be artificially overexpressed in hepatocytes or the

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

cells can be conditionally immortalized leading to controlled cell proliferation [19]. However, limitations of immortalized human hepatocytes as predictive tool for CYP induction studies were reported [20]. Differentiation and proliferative activity of human hepatocytes in primary culture rapidly declines with increasing time; interestingly this decline is slower in cell from juvenile donors as compared to adult ones [21]. Several studies were engaged to search for culture conditions, which could restart and maintain proliferation and differentiation state of human hepatocytes, however, without substantial success [22]. Human fetal liver cells can be easily expanded in vitro, however, they have decreased hepatic functionality [23]. Highly promising alternative to adult primary human hepatocytes are hepatic-like cells obtained from liver progenitor cells or stem cells. Non-parenchymal epithelial cells were isolated from adult human liver and then cultured at confluence in the presence of hepatocyte growth factor, epidermal growth factor and fibroblast growth factor-4. Cells entered a differentiation process toward hepatocytes and their phenotype was quantitatively compared with that of mature human hepatocytes in primary culture [24, 25]. Human induced pluripotent stem (iPS) cells can be efficiently induced to differentiate into hepatocyte-like cells [26-28], which were described as suitable models for testing hepatocellular acute toxicity of xenobiotics [29], chronic toxicity [30], liver injury [31], and which retained the xenobiotics-mediated induction of drug-metabolizing enzymes [32]. Recently, it was demonstrated that iPS cells derived hepatocyte-like cells retain donor-specific cytochrome P450 metabolism capacity and drug

7

responsiveness, therefore, reflecting the interindividual differences in that aspect [33]. The attempts regarding stability of iPS-derived hepatocytes were made [34]. Similarly like iPS, also hepatocytes derived from human embryonic stem cells (hESC) were functional in the aspects described above [28]. Liver progenitor cell line HepaRG was isolated from an Edmonson grade I differentiated liver tumor that developed macronodular cirrhosis resulting from long-term chronic HCV infection [35]. Basal activities of P450s and their response to

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

prototypical inducers revealed the functional resemblance of HepaRG cells to primary cultured human hepatocytes [36]. Several ongoing studies confirmed that HepaRG represent a reliable surrogate to human hepatocytes for drug metabolism and toxicity studies and for prediction of induction of drug-metabolizing P450 enzymes in vivo in humans [37, 38]. Recently, the usefulness of isolated human hepatocytes from chimeric mice was demonstrated for metabolism and induction studies of xenobiotics and HBV-infection studies [39-41].

2.5. Human hepatocytes in primary culture for xenobiotic-metabolizing P450s induction studies Standard protocols were elaborated for the treatments and cultivation of primary human hepatocytes to evaluate the capability of xenobiotic to induce drug-metabolizing P450s. It is worth mentioning several aspects regarding such the assays: (i) There is a substantial interindividual variability between primary cultures prepared from different donors. Therefore, induction studies have to be performed in several primary cultures from different donors (at least at 3 cultures; usually in 5 – 10 cultures). The interindividual differences are often due to the gene polymorphism of cytochromes P450, their transcriptional regulators, drug transporters and phase II enzymes. It implies that the race of donor, and hence relative frequency of polymorphism in this race, contribute to overall interindividual variability. There are also many physiological (age, sex, pregnancy) and pathophysiological (obesity, diabetes,

8

cancer, inflammation, infection) factors contributing to interindividual variability [42-44]. (ii) Human hepatocytes are fully metabolically competent cells; therefore, an assessment of xenobiotics capability to induce P450s involves not only the effects of maternal compound, but also the effects of its phase I and phase II metabolites. The absence of metabolites in the culture medium of various liver-derived cancer cells (e.g. HepG2), is the major difference as compared to human hepatocytes, resulting potentially in both false negative and/or false

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

positive data [45, 46]. (iii) Standard protocols were elaborated for induction studies in primary human hepatocytes. Briefly, following the seeding of hepatocytes, they are allowed to stabilize for about 48 h - 72 h. During this period, basal level of xenobiotic-inducible CYPs drops drastically; hence, the magnitude of induction as compared to control cells increase substantially. Incubation of hepatocytes with tested compounds is carried out in serum-free medium. Medium including tested compounds (usually dissolved in DMSO as vehicle) are exchanged every day during the treatment. The time of incubation depends on measured endpoint for induction. For measurement of mRNA, hepatocytes are incubated with xenobiotic either for 4 h – 12 h to assess dynamics of de novo transcription, or for 24 h – 48 h to assess gross effect of xenobiotic plus its metabolites. Expression of CYP proteins, and catalytic activity of CYP enzymes is usually evaluated after 48 h – 96 h of incubation with tested compounds. Accordingly, the data have to be properly interpreted; e.g. induction of CYP mRNA is rather of mechanistic importance, while substantial decline in CYP catalytic activity is potentially of clinical relevance [47, 48]. Indeed, a bad correlation was reported between the induction of CYP mRNA and the induction of CYP catalytic activity in HHs [49]. (iv) Identification of CYPs inducers using human hepatocytes: An extensive literature search for articles dealing with CYPs induction by xenobiotics in human hepatocytes was performed using PubMed database. Over 600 articles were carefully analyzed, and the exhaustive list of inducers and repressors of xenobiotics-metabolizing CYPs, which were identified by assays in

9

both freshly prepared and cryopreserved human hepatocytes is provided in Supplemental Table 1. The list does not include: (a) Articles that presented negative effects (no induction of CYPs) by new drugs candidates as well as existing drugs and xebobiotics; (b) Studies involving herbal extracts, environmental samples and another mixtures; (c) Comparative studies, i.e. between the models, species, cell lines etc.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

3. EXPERT OPINION Primary cultures of human hepatocytes had long been considered as a gold standard for induction assays of xenobiotic-metabolizing enzymes. Owing to several shortcomings of HHs, alternative approaches such as immortalization of HHs, use of cell lines, generation of clonal cell lines from HHs, use of iPS, cells from humanized animals etc. were employed. While yielding particular advantage, overall, alternatives to HHs still remain an avenue for discrete applications or technical situations. The extensive efforts are made concerning the use of HHs for other purposes than induction studies, in particular, in liver biotherapy and in (patho-)physiological studies. Thus, human hepatocytes still remain to be the most suitable model for complex CYPs induction studies. The summary may be effectively expressed by SWOT analysis. 3.1 Strengths Human hepatocytes are per se the model that overcomes complications with interspecies variability, without need to extrapolate the data from animals to man. HHs are highly physiological system in comparison with other cellular liver systems derived from cancer or transformed cells. HHs possess complex and integrate biotransformation machinery, including phase I and phase II enzymes, and full panel of drug transporters. Therefore HHs are reliable and efficient model for performing metabolic studies. 3.2 Weaknesses

10

Significant drawback in use of HHs is their scarcity in both time and quantity. With improvement of networks between transplant centers and with patient-friendly surgery techniques, the availability of HHs diminished. It is also inter-individual variability between HHs obtained from different patients, which brings necessity of experimentation in several HHs cultures, thus, making use of HHs time- and material-demanding. 3.3 Opportunities

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

A great challenge in the field of experimentation with HHs is the optimization and improvement of storage and maintenance conditions. In particular, techniques allowing cryopreservation of significant amount of HHs would resolve the weakness given by scarcity of HHs. While current approaches are developed for cryopreservation of millions of HHs in milliliters of volume, really new-epoque-making invention would be the technique allowing freeze/thaw billions of HHs in one batch. Improvement of networking between clinics and researchers may be considered not only as weakness leading to diminished number of liver available for research purposes, but contrary it may result in more effective exploitation of liver sources, and increase of available experimental material. 3.4 Threats Considerable risks for the use of human liver for isolation of HHs stem from ethical considerations, including the protection of personal data regarding genomic experiments. These treats are strongly bound to legislation of individual states, hence, making current limitations and future risks dependent on the country of interest.

Declaration of Interest The author was supported by grants from the Czech Science Foundation GACR 303/12/G163 and Ministry of Health of the Czech Republic NT/13591. The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. 11

Bibliography Papers of special note have been highlighted as: * of interest ** of considerable interest

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

1.

Tian, D. and Z. Hu, CYP3A4-mediated pharmacokinetic interactions in cancer therapy. Curr Drug Metab, 2014. 15: p. 808-17 2. Sevrioukova, I.F. and T.L. Poulos, Current Approaches for Investigating and Predicting Cytochrome P450 3A4-Ligand Interactions. Adv Exp Med Biol, 2015. 851: p. 83-105 3. Sinz, M.W., Evaluation of pregnane X receptor (PXR)-mediated CYP3A4 drug-drug interactions in drug development. Drug Metab Rev, 2013. 45: p. 3-14 4. Matoulkova, P., P. Pavek, J. Maly, et al., Cytochrome P450 enzyme regulation by glucocorticoids and consequences in terms of drug interaction. Expert Opin Drug Metab Toxicol, 2014. 10: p. 425-35 5. Pavek, P. and Z. Dvorak, Xenobiotic-induced transcriptional regulation of xenobiotic metabolizing enzymes of the cytochrome P450 superfamily in human extrahepatic tissues. Curr Drug Metab, 2008. 9: p. 129-43 6. Moreau, A., M.J. Vilarem, P. Maurel, et al., Xenoreceptors CAR and PXR activation and consequences on lipid metabolism, glucose homeostasis, and inflammatory response. Mol Pharm, 2008. 5: p. 35-41 7. Pascussi, J.M., S. Gerbal-Chaloin, C. Duret, et al., The tangle of nuclear receptors that controls xenobiotic metabolism and transport: crosstalk and consequences. Annu Rev Pharmacol Toxicol, 2008. 48: p. 1-32 *Shows mutual relationships between xenobiotics and endobiotics cell signaling. 8. Hewitt, N.J., M.J. Lechon, J.B. Houston, et al., Primary hepatocytes: current understanding of the regulation of metabolic enzymes and transporter proteins, and pharmaceutical practice for the use of hepatocytes in metabolism, enzyme induction, transporter, clearance, and hepatotoxicity studies. Drug Metab Rev, 2007. 39: p. 159-234 9. Lloyd, T.D., S. Orr, and A.R. Dennison, A survey of consumer attitudes to the supply and use of human hepatocytes in the United Kingdom. Altern Lab Anim, 2003. 31: p. 483-8 10. Yajima, K., Y. Uno, N. Murayama, et al., Evaluation of 23 lots of commercially available cryopreserved hepatocytes for induction assays of human cytochromes P450. Drug Metab Dispos, 2014. 42: p. 867-71 11. Kamiguchi, N., E. Aoyama, T. Okuda, et al., A 96-well plate assay for CYP4503A induction using cryopreserved human hepatocytes. Drug Metab Dispos, 2010. 38: p. 1912-6 12. Duret, C., D. Moreno, B. Anangi, et al., Cold-preservation of human adult hepatocytes for liver cell therapy. Cell Transplant, 2015 13. Ferrini, J.B., L. Pichard, J. Domergue, et al., Long-term primary cultures of adult human hepatocytes. Chem Biol Interact, 1997. 107: p. 31-45 *Describes experimental conditions allowing HHs culture in long time. 14. Kono, Y., S. Yang, and E.A. Roberts, Extended primary culture of human hepatocytes in a collagen gel sandwich system. In Vitro Cell Dev Biol Anim, 1997. 33: p. 467-72 15. Badolo, L., B. Jensen, C. Sall, et al., Evaluation of 309 molecules as inducers of CYP3A4, CYP2B6, CYP1A2, OATP1B1, OCT1, MDR1, MRP2, MRP3 and BCRP in cryopreserved human hepatocytes in sandwich culture. Xenobiotica, 2015. 45: p. 177-87 16. Xia, L., X. Hong, R.B. Sakban, et al., Cytochrome P450 induction response in tethered spheroids as a three-dimensional human hepatocyte in vitro model. J Appl Toxicol, 2015 17. Gross-Steinmeyer, K., P.L. Stapleton, J.H. Tracy, et al., Influence of Matrigel-overlay on constitutive and inducible expression of nine genes encoding drug-metabolizing enzymes in primary human hepatocytes. Xenobiotica, 2005. 35: p. 419-38 18. Vinci, B., C. Duret, S. Klieber, et al., Modular bioreactor for primary human hepatocyte culture: medium flow stimulates expression and activity of detoxification genes. Biotechnol J, 2011. 6: p. 554-64 12

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

19.

Ramboer, E., B. De Craene, J. De Kock, et al., Strategies for immortalization of primary hepatocytes. J Hepatol, 2014. 61: p. 925-43 20. Hariparsad, N., B.A. Carr, R. Evers, et al., Comparison of immortalized Fa2N-4 cells and human hepatocytes as in vitro models for cytochrome P450 induction. Drug Metab Dispos, 2008. 36: p. 1046-55 21. Walldorf, J., H. Aurich, H. Cai, et al., Expanding hepatocytes in vitro before cell transplantation: donor age-dependent proliferative capacity of cultured human hepatocytes. Scand J Gastroenterol, 2004. 39: p. 584-93 22. Yoon, J.H., G.Y. Gwak, G.H. Woo, et al., Augmentation of butyrate-induced differentiation of human hepatocyte by cyclin E over-expression. Int J Artif Organs, 2005. 28: p. 44-50 23. Deurholt, T., L. ten Bloemendaal, A.A. Chhatta, et al., In vitro functionality of human fetal liver cells and clonal derivatives under proliferative conditions. Cell Transplant, 2006. 15: p. 811-22 24. Duret, C., S. Gerbal-Chaloin, J. Ramos, et al., Isolation, characterization, and differentiation to hepatocyte-like cells of nonparenchymal epithelial cells from adult human liver. Stem Cells, 2007. 25: p. 1779-90 25. Gerbal-Chaloin, S., C. Duret, E. Raulet, et al., Isolation and Culture of Adult Human Liver Progenitor Cells: In Vitro Differentiation to Hepatocyte-Like Cells. Hepatocytes: Methods and Protocols, 2010. 640: p. 247-60 26. Song, Z.H., J. Cai, Y.X. Liu, et al., Efficient generation of hepatocyte-like cells from human induced pluripotent stem cells. Cell Research, 2009. 19: p. 1233-42 27. Si-Tayeb, K., F.K. Noto, M. Nagaoka, et al., Highly Efficient Generation of Human Hepatocyte-Like Cells from Induced Pluripotent Stem Cells (vol 51, pg 297, 2010). Hepatology, 2010. 51: p. 109428. Ulvestad, M., P. Nordell, A. Asplund, et al., Drug metabolizing enzyme and transporter protein profiles of hepatocytes derived from human embryonic and induced pluripotent stem cells. Biochem Pharmacol, 2013. 86: p. 691-702 29. Medine, C.N., B. Lucendo-Villarin, C. Storck, et al., Developing High-Fidelity Hepatotoxicity Models From Pluripotent Stem Cells. Stem Cells Translational Medicine, 2013. 2: p. 505-9 30. Holmgren, G., A.K. Sjogren, I. Barragan, et al., Long-term chronic toxicity testing using human pluripotent stem cell-derived hepatocytes. Drug Metab Dispos, 2014. 42: p. 1401-6 31. Szkolnicka, D., S.L. Farnworth, B. Lucendo-Villarin, et al., Accurate prediction of druginduced liver injury using stem cell-derived populations. Stem Cells Transl Med, 2014. 3: p. 141-8 32. Kondo, Y., T. Iwao, K. Nakamura, et al., An Efficient Method for Differentiation of Human Induced Pluripotent Stem Cells into Hepatocyte-like Cells Retaining Drug Metabolizing Activity. Drug Metabolism and Pharmacokinetics, 2014. 29: p. 237-43 33. Takayama, K., Y. Morisaki, S. Kuno, et al., Prediction of interindividual differences in hepatic functions and drug sensitivity by using human iPS-derived hepatocytes. Proceedings of the National Academy of Sciences of the United States of America, 2014. 111: p. 16772-7 34. Villarin, B.L., K. Cameron, D. Szkolnicka, et al., Polymer Supported Directed Differentiation Reveals a Unique Gene Signature Predicting Stable Hepatocyte Performance. Adv Healthc Mater, 2015. 4: p. 1820-5 35. Parent, R., M.J. Marion, L. Furio, et al., Origin and characterization of a human bipotent liver progenitor cell line. Gastroenterology, 2004. 126: p. 1147-56 **The first report on HepaRG cells. 36. Aninat, C., A. Piton, D. Glaise, et al., Expression of cytochromes P450, conjugating enzymes and nuclear receptors in human hepatoma HepaRG cells. Drug Metabolism and Disposition, 2006. 34: p. 75-83 37. Kanebratt, K.P. and T.B. Andersson, HepaRG cells as an in vitro model for evaluation of cytochrome p450 induction in humans. Drug Metabolism and Disposition, 2008. 36: p. 137-45 38. McGinnity, D.F., G. Zhang, J.R. Kenny, et al., Evaluation of Multiple in Vitro Systems for Assessment of CYP3A4 Induction in Drug Discovery: Human Hepatocytes, Pregnane X Receptor Reporter Gene, and Fa2N-4 and HepaRG Cells. Drug Metabolism and Disposition, 2009. 37: p. 1259-68 13

39. 40. 41. 42.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

43. 44. 45. 46.

47. 48.

49.

Yamasaki, C., M. Kataoka, Y. Kato, et al., In vitro evaluation of cytochrome P450 and glucuronidation activities in hepatocytes isolated from liver-humanized mice. Drug Metab Pharmacokinet, 2010. 25: p. 539-50 Kakuni, M., C. Yamasaki, A. Tachibana, et al., Chimeric mice with humanized livers: a unique tool for in vivo and in vitro enzyme induction studies. Int J Mol Sci, 2014. 15: p. 58-74 Ishida, Y., C. Yamasaki, A. Yanagi, et al., Novel robust in vitro hepatitis B virus infection model using fresh human hepatocytes isolated from humanized mice. Am J Pathol, 2015. 185: p. 1275-85 Rogue, A., C. Lambert, C. Spire, et al., Interindividual Variability in Gene Expression Profiles in Human Hepatocytes and Comparison with HepaRG Cells. Drug Metabolism and Disposition, 2012. 40: p. 151-8 Gomez-Lechon, M.J., J.V. Castell, and M.T. Donato, Hepatocytes--the choice to investigate drug metabolism and toxicity in man: in vitro variability as a reflection of in vivo. Chem Biol Interact, 2007. 168: p. 30-50 Goyak, K.M., M.C. Johnson, S.C. Strom, et al., Expression profiling of interindividual variability following xenobiotic exposures in primary human hepatocyte cultures. Toxicol Appl Pharmacol, 2008. 231: p. 216-24 Wilkening, S., F. Stahl, and A. Bader, Comparison of primary human hepatocytes and hepatoma cell line HEPG2 with regard to their biotransformation properties. Drug Metabolism and Disposition, 2003. 31: p. 1035-42 Gerets, H.H.J., K. Tilmant, B. Gerin, et al., Characterization of primary human hepatocytes, HepG2 cells, and HepaRG cells at the mRNA level and CYP activity in response to inducers and their predictivity for the detection of human hepatotoxins. Cell Biology and Toxicology, 2012. 28: p. 69-87 Daujat, M., L. Pichard, I. Fabre, et al., Induction Protocols for Cytochromes P450iiia Invivo and in Primary Cultures of Animal and Human Hepatocytes. Methods in Enzymology, 1991. 206: p. 345-53 Richert, L., C. Abadie, A. Bonet, et al., Inter-laboratory evaluation of the response of primary human hepatocyte cultures to model CYP inducers - A European Centre for Validation of Alternative Methods (ECVAM) - Funded pre-validation study. Toxicology in Vitro, 2010. 24: p. 335-45 Rodriguez-Antona, C., M.T. Donato, E. Pareja, et al., Cytochrome P-450 mRNA expression in human liver and its relationship with enzyme activity. Arch Biochem Biophys, 2001. 393: p. 308-15

14

Supplemental Table 1. LIST OF COMPOUNDS IDENTIFIED AS INDUCERS OR REPRESSORS OF XENOBIOTICS-METABOLIZING CYTOCHROMES P450 IN HUMAN HEPATOCYTES CULTURES

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

*Only chemical individuals are listed – i.e. not extracts, mixtures etc.

COMPOUND Reference

CYP induced/repressed

A-792611 (HIV protease inhibitor) [1] 1α,25-di-OH-vitamin D3 [2, 3] 2-ACETYLAMINOFLUORENE [4] ADZ7325 (GABA receptor modulator) [5] AFLATOXIN B1 [6] ALACHLOR [7] ALLOCRYPTOPINE [8] 1-AMINOBENZTRIAZOL [9] AMPK activators [10] AROCLOR1260 [11] ARSENIC TRIOXIDE [12-14] ARTEMISIN [15, 16] ASTAXANTHIN [17] ATORVASTATIN [18-21] AVISIMIBE [22] BDE-99, BDE-209 (polybrominated diphenyl ethers) [23] BERGAMOTTIN [24] BENZO[k]FLUOROANTHRENE [25]

2B6, 2C8, 2C9, 3A4

15

3A4 1A 1A2, 3A4 2B6, 2C9, 3A4/5 3A4 1A1, 1A2 2B6, 3A4 2B6 2B6 1A1, 3A4 2B6, 3A4 2B6, 3A4 2B6, 2C9, 3A4 3A4 1A2, 3A4 1A1, 1A2, 2B6, 3A4 1A1, 1A2

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

BENZIMIDAZOLE-5-YLBENZENESULFONAMIDES [26] BIOCHANIN A [27] BISPHENOL A [28] BUTYLHYDROXYTOLUENE [29] BYAKANGELICIN [30] CAMPTOTHECIN [31] CARBAMAZEPINE [32] CARBOXYMEFLOQUINE [33] CARNOSIC ACID [34] CATECHOLAMINES [35] CHLORPYRIFOS [36] CITCO [37, 38] CLEVIDIPINE [39] CLINDAMYCIN [40] CLOFIBRATE [41] CLOTRIMAZOLE [32] COLCHICINE [42-44] CORTISOL [45] CRIZOTINIB [46] CYANIDIN [47] CYCLOPHOSPHAMIDE [48, 49] CYTOKINES / GROWTH FACTORS [45, 50-59] DABRAFENIB [60] DEXAMETHASONE [32, 38, 61-72] DICLOXACILLIN [40] 16

2B6, 3A4 2A6, 2B6, 2C9 1A1, 3A4 2B6, 3A4 3A4 3A4 2B6, 3A4 2B6, 2C8, 3A4 2B6, 3A4 3A4 1A1, 1A2, 2A6, 2B6, 3A4 2B6, 2C8 3A4 3A4 2C8, 3A4 2B6, 3A4 1A1, 1A2, 2B6, 2C8, 2C9, 3A4 3A4 3A4 1A1, 1A2 2B6, 3A4 1A1/2, 2B6, 2A6, 2C8/9, 2E1, 3A4 2B6, 3A4 1A1, 2A6, 2B6, 2C8/9, 3A4, 3A5 3A4

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

DIETHYLTOLUAMIDE [36] DIHYDROEPIANDROSTENON [72-74] DIINDOYLMETHANE [75-77] EMD 392949 (antidiabetic) [78] EFAVIRENZ [79] (-)-epigallocatechin gallate [80] ENDOSULFAN [81] ENTHOTELIN-A ANTAGON.(CI-1034) [82] EQUOL [83] ESTRADIOL [84-86] ETHANOL [87] ETRAVIRINE [88] FENOFIBRATE [41] FIPRONIL [89] FLUCLOXACILLIN [90] FLUVASTATIN [18] GEMFIBROSIL [41] GENTIOPICROSIDE [91] GINKGOLIDE A, B [92] GUGGULSTERONE [93] GW3965 (LXRα agonist) [94] HONOKIOL [95] HYPERFORIN [96-98] IBROLIPIM (NO-1886) [99, 100] IFOSFAMIDE [49]

1A1, 1A2, 2A6, 2B6, 3A4 1A1, 1A2, 2B6, 2C9 1A1, 1A2, 3A4 2C8, 3A4 3A4 1A1 2B6, 3A4 3A4 3A4 2A6, 2B6, 3A4 2E1, 3A4 3A4 2C8, 3A4 1A1, 2B6, 3A4 3A4 2B6, 3A4 2C8, 3A4 1A2, 3A4 3A4 3A4 3A4 2B6 2C9, 3A4 2C8, 3A4 2B6, 3A4

17

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

IRINOTECAN [31] ISOPENTANOL [87] ISRADIPINE [101] KETOCONAZOLE [102, 103] KHELLIN [104] KYNURENIC ACID [105] LANSOPRAZOLE [106-109] LCL161 (inhibitor of apoptosis proteins) [110] LIPOPOLYSACCHARIDE [35] LK-935 (statin) [20] LOVASTATIN [18] LY2090314 (GSK3 inhibitor) [111] MATRINE [112] MEDAZEPAM [113] MESALAZINE [114] METFORMIN [115-117] METHADONE [118, 119] METHANESULFONAMIDE C2BA-4 [120] METHOTREXATE [32] 4-METHYLBENZYLIDENE CAMPHOR [121] METOLACHLOR [7] METOLAZONE [122] METYRAPONE [123] MICONAZOLE [124] MIDAZOLAM [113]

3A4 2E1, 3A4 2B6, 2C9, 3A4 1A1, 1A2, 3A4 1A1 1A1, 1A2 1A1, 1A2, 1B1, 3A4 3A4 3A4 2B6, 2C9, 3A4 2B6, 3A4 CYP2B6 2A6, 2B6, 3A4 3A4 2B6, 3A4 2B6, 3A4 2B6, 3A4 2B6, 3A4 2B6, 3A4 2B6, 3A4 3A4 3A4 3A4 3A4 3A4

18

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

MIFEPRISTONE [125] ΜΚΧ−963(anti-aggregation platelet) [126] MOSAPRIDE [114] NAFCILLIN [40] β-NAPHTOFLAVONE [108] NELFINAVIR [127, 128] NICARDIPINE [101] NIFEDIPINE [101] NOCODAZOLE [44] NONYLPHENOL [129] OCHRATOXIN [130, 131] 4-tert-OCTYLPHENOL [121] OLTIPRAZ [132] OMEPRAZOLE [106-109, 133-135] ONCOSTATIN M [136] ORLISTAT [137] OXADIAZON [7] OXICONAZOLE [124] PANTOPRAZOLE [107] PARATHION [28] PELARGONIDIN [47] PENTACHLORODIBENZOFURAN [138] PERAZINE [139] PHENOBARBITAL [10, 32, 67, 68, 98, 134, 140-144] PHENYTOIN [32, 134, 145, 146]

3A4 3A4 1A2, 2B6, 3A4 3A4 1A1, 1A2, 1B1 2B6, 2C8, 2C9, 3A4 2B6, 2C9, 3A4 2B6, 2C9, 3A4 1A2 2B6 3A4 2B6, 3A4 2B6 1A1, 1A2, 1B1, 3A4 1A1, 1A2, 2B6, 2A6, 3A4 3A4 3A4 3A4 1A1, 3A4 1A1, 3A4 1A1, 1A2 1A1, 1A2 1A2, 2C19, 3A4 2A6, 2B6, 2C8, 2C9, 2C19, 3A4 2B6, 2C9, 3A4

19

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

Ph-morpholino-antioligomers [147] PHTALATES (DEHP, MEHP) [121, 148, 149] PIPERINE [150] PRETILACHLOR [7] PRIMAQUINE [108, 151] PROBENICID [32] PROGESTERONE [85] PROMAZINE [139] PROTOPINE [8] PYRAZOLE [152] PYRETHRINS [153] PYRETHROIDS [154-156] QUERCETIN [134] All-trans-RETINOIC ACID [157] RIFABUTIN [158-160] RIFAMPICIN [32, 40, 67, 69, 97, 98, 134, 141, 144, 158-162] RIFAPENTINE [158, 160] RITONAVIR [32, 127, 128] Ro 48-8071 (inh. 2,3-oxiskvalene:lanosterol cyclase) [163] ROSUVASTATIN [20] S16020 (anticancer) [164] SEMAGACESTAT (γ-secretase inhibitor) [165] SIMVASTATIN [18] SIPI5357 (antidepressant) [166] SP600125 (JNK inhibitor) [167] 20

3A4 2B6, 3A4 3A4 3A4 1A1, 1A2, 1B1 2B6, 3A4 2A6, 2B6, 2C8, 3A4, 3A5 1A2, 2C19, 3A4 1A1, 1A2 2A6 2B6, 3A4 1A1, 2A6, 2B6, 3A4 3A4 2C9 3A4 2A6, 2B6, 2C8/9, 2C19, 3A4, 3A7 3A4 2B6, 2C8, 2C9, 3A4 2B6, 3A4 2B6, 2C9, 3A4 1A1, 1A2 CYP3A4 2B6, 3A4 2B6 1A1, 1A2

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

SULFADIMINE [32] SULFINPYRAZONE [32] SULFISOXAZOLE [40] SULFORAPHANE [168, 169] T0901317 (LXR agonist) [170] TACE/MMP-inhibitors [171] TADALAFIL [172] TAMOXIFEN [173, 174] TAXOL [44, 175, 176] TETRACHLORODIBENZOFURAN [138] TETRACYCLINE [40] TCDD [138, 177-180] THALIDOMIDE [181] THIABENDAZOLE [29] α-TOCOPHEROL [182] TOPIRAMATE [183] TOREMIFENE [184] TROLEANDOMYCIN [32, 40, 185] TROGLITAZONE [186, 187] TSU-68 (anticancer) [188] U0126 (MEK1/2 inhibitor) [189] VALPROATE [190] VINBLASTINE [44] VINCRISTINE [44] VISNAGIN [104]

2B6, 3A4 2B6, 3A4 3A4 2A6, 3A4 2B6, 3A4 3A4 3A4 3A4 1A2, 3A4 1A1, 1A2 3A4 1A1, 1A2, 1B1 2B6, 3A4 1A2, 2B6, 3A4 3A4 3A4 3A4 2B6, 3A4 2B6, 3A4 1A1, 1A2 3A4 3A4 1A2 1A2 1A1

21

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

WARFARIN [191] WY14643 (PPARα agonist) [192, 193] YM758 (If channel inhibitor) [194]

2C9, 3A4 3A4 3A4

REFERENCES 1.

2.

3.

4.

5.

6.

7. 8.

9. 10.

11.

Healan-Greenberg, C., J.F. Waring, D.J. Kempf, et al., A human immunodeficiency virus protease inhibitor is a novel functional inhibitor of human pregnane X receptor. Drug Metab Dispos, 2008. 36: p. 500-7 Schmiedlin-Ren, P., K.E. Thummel, J.M. Fisher, et al., Induction of CYP3A4 by 1 alpha,25dihydroxyvitamin D3 is human cell line-specific and is unlikely to involve pregnane X receptor. Drug Metab Dispos, 2001. 29: p. 1446-53 Drocourt, L., J.C. Ourlin, J.M. Pascussi, et al., Expression of CYP3A4, CYP2B6, and CYP2C9 is regulated by the vitamin D receptor pathway in primary human hepatocytes. J Biol Chem, 2002. 277: p. 25125-32 Lecureur, V., A. Guillouzo, and O. Fardel, Differential regulation of mdr genes in response to 2-acetylaminofluorene treatment in cultured rat and human hepatocytes. Carcinogenesis, 1996. 17: p. 1157-60 Zhou, D., M. Sunzel, M.D. Ribadeneira, et al., A clinical study to assess CYP1A2 and CYP3A4 induction by AZD7325, a selective GABA(A) receptor modulator - an in vitro and in vivo comparison. Br J Clin Pharmacol, 2012. 74: p. 98-108 Ayed-Boussema, I., J.M. Pascussi, P. Maurel, et al., Effect of aflatoxin B1 on nuclear receptors PXR, CAR, and AhR and their target cytochromes P450 mRNA expression in primary cultures of human hepatocytes. Int J Toxicol, 2012. 31: p. 86-93 Lemaire, G., W. Mnif, J.M. Pascussi, et al., Identification of new human pregnane X receptor ligands among pesticides using a stable reporter cell system. Toxicol Sci, 2006. 91: p. 501-9 Vrba, J., E. Vrublova, M. Modriansky, et al., Protopine and allocryptopine increase mRNA levels of cytochromes P450 1A in human hepatocytes and HepG2 cells independently of AhR. Toxicol Lett, 2011. 203: p. 135-41 Yang, K., K.H. Koh, and H. Jeong, Induction of CYP2B6 and CYP3A4 expression by 1aminobenzotriazole (ABT) in human hepatocytes. Drug Metab Lett, 2010. 4: p. 129-33 Rencurel, F., A. Stenhouse, S.A. Hawley, et al., AMP-activated protein kinase mediates phenobarbital induction of CYP2B gene expression in hepatocytes and a newly derived human hepatoma cell line. J Biol Chem, 2005. 280: p. 4367-73 Wahlang, B., K.C. Falkner, H.B. Clair, et al., Human receptor activation by aroclor 1260, a polychlorinated biphenyl mixture. Toxicol Sci, 2014. 140: p. 283-97

22

12.

13. 14.

15.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

16.

17. 18.

19.

20.

21. 22.

23. 24.

25. 26. 27.

28.

29.

30.

Vernhet, L., N. Allain, M. Le Vee, et al., Blockage of multidrug resistance-associated proteins potentiates the inhibitory effects of arsenic trioxide on CYP1A1 induction by polycyclic aromatic hydrocarbons. J Pharmacol Exp Ther, 2003. 304: p. 145-55 Noreault, T.L., V.E. Kostrubsky, S.G. Wood, et al., Arsenite decreases CYP3A4 and RXRalpha in primary human hepatocytes. Drug Metab Dispos, 2005. 33: p. 993-1003 Noreault-Conti, T.L., A. Fellows, J.M. Jacobs, et al., Arsenic decreases RXRalpha-dependent transcription of CYP3A and suppresses immune regulators in hepatocytes. Int Immunopharmacol, 2012. 12: p. 651-6 Burk, O., K.A. Arnold, A.K. Nussler, et al., Antimalarial artemisinin drugs induce cytochrome P450 and MDR1 expression by activation of xenosensors pregnane X receptor and constitutive androstane receptor. Mol Pharmacol, 2005. 67: p. 1954-65 Burk, O., R. Piedade, L. Ghebreghiorghis, et al., Differential effects of clinically used derivatives and metabolites of artemisinin in the activation of constitutive androstane receptor isoforms. Br J Pharmacol, 2012. 167: p. 666-81 Kistler, A., H. Liechti, L. Pichard, et al., Metabolism and CYP-inducer properties of astaxanthin in man and primary human hepatocytes. Arch Toxicol, 2002. 75: p. 665-75 Kocarek, T.A., M.S. Dahn, H. Cai, et al., Regulation of CYP2B6 and CYP3A expression by hydroxymethylglutaryl coenzyme A inhibitors in primary cultured human hepatocytes. Drug Metab Dispos, 2002. 30: p. 1400-5 Hoffart, E., L. Ghebreghiorghis, A.K. Nussler, et al., Effects of atorvastatin metabolites on induction of drug-metabolizing enzymes and membrane transporters through human pregnane X receptor. Br J Pharmacol, 2012. 165: p. 1595-608 Monostory, K., J.M. Pascussi, P. Szabo, et al., Drug interaction potential of 2-((3,4dichlorophenethyl)(propyl)amino)-1-(pyridin-3-yl)ethanol (LK-935), the novel nonstatin-type cholesterol-lowering agent. Drug Metab Dispos, 2009. 37: p. 375-85 Schroder, A., J. Wollnik, C. Wrzodek, et al., Inferring statin-induced gene regulatory relationships in primary human hepatocytes. Bioinformatics, 2011. 27: p. 2473-7 Sahi, J., M.A. Milad, X. Zheng, et al., Avasimibe induces CYP3A4 and multiple drug resistance protein 1 gene expression through activation of the pregnane X receptor. J Pharmacol Exp Ther, 2003. 306: p. 1027-34 Stapleton, H.M., S.M. Kelly, R. Pei, et al., Metabolism of polybrominated diphenyl ethers (PBDEs) by human hepatocytes in vitro. Environ Health Perspect, 2009. 117: p. 197-202 Wen, Y.H., J. Sahi, E. Urda, et al., Effects of bergamottin on human and monkey drugmetabolizing enzymes in primary cultured hepatocytes. Drug Metab Dispos, 2002. 30: p. 97784 Liu, N., Q.Y. Zhang, D. Vakharia, et al., Induction of CYP1A by benzo[k]fluoranthene in human hepatocytes: CYP1A1 or CYP1A2? Arch Biochem Biophys, 2001. 389: p. 130-4 Benod, C., G. Subra, V. Nahoum, et al., N-1H-benzimidazol-5-ylbenzenesulfonamide derivatives as potent hPXR agonists. Bioorg Med Chem, 2008. 16: p. 3537-49 Moon, Y.J., S. Zhang, D.A. Brazeau, et al., Effects of the flavonoid biochanin A on gene expression in primary human hepatocytes and human intestinal cells. Mol Nutr Food Res, 2007. 51: p. 317-23 Vrzal, R., O. Zenata, A. Doricakova, et al., Environmental pollutants parathion, paraquat and bisphenol A show distinct effects towards nuclear receptors-mediated induction of xenobiotics-metabolizing cytochromes P450 in human hepatocytes. Toxicol Lett, 2015. 238: p. 43-53 Price, R.J., M.P. Scott, A.M. Giddings, et al., Effect of butylated hydroxytoluene, curcumin, propyl gallate and thiabendazole on cytochrome P450 forms in cultured human hepatocytes. Xenobiotica, 2008. 38: p. 574-86 Yang, J., X. Luan, H. Gui, et al., Byakangelicin induces cytochrome P450 3A4 expression via transactivation of pregnane X receptors in human hepatocytes. Br J Pharmacol, 2011. 162: p. 441-51 23

31.

32. 33.

34.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

35. 36.

37.

38.

39.

40.

41. 42.

43.

44.

45.

46.

47.

Chen, Y., Y. Tang, G.T. Robbins, et al., Camptothecin attenuates cytochrome P450 3A4 induction by blocking the activation of human pregnane X receptor. J Pharmacol Exp Ther, 2010. 334: p. 999-1008 Faucette, S.R., H. Wang, G.A. Hamilton, et al., Regulation of CYP2B6 in primary human hepatocytes by prototypical inducers. Drug Metab Dispos, 2004. 32: p. 348-58 Piedade, R., S. Traub, A. Bitter, et al., Carboxymefloquine, the major metabolite of the antimalarial drug mefloquine, induces drug-metabolizing enzyme and transporter expression by activation of pregnane X receptor. Antimicrob Agents Chemother, 2015. 59: p. 96-104 Dickmann, L.J., B.M. VandenBrink, and Y.S. Lin, In vitro hepatotoxicity and cytochrome P450 induction and inhibition characteristics of carnosic acid, a dietary supplement with antiadipogenic properties. Drug Metab Dispos, 2012. 40: p. 1263-7 Aninat, C., P. Seguin, P.N. Descheemaeker, et al., Catecholamines induce an inflammatory response in human hepatocytes. Crit Care Med, 2008. 36: p. 848-54 Das, P.C., Y. Cao, R.L. Rose, et al., Enzyme induction and cytotoxicity in human hepatocytes by chlorpyrifos and N,N-diethyl-m-toluamide (DEET). Drug Metabol Drug Interact, 2008. 23: p. 237-60 Maglich, J.M., D.J. Parks, L.B. Moore, et al., Identification of a novel human constitutive androstane receptor (CAR) agonist and its use in the identification of CAR target genes. J Biol Chem, 2003. 278: p. 17277-83 Ferguson, S.S., Y. Chen, E.L. LeCluyse, et al., Human CYP2C8 is transcriptionally regulated by the nuclear receptors constitutive androstane receptor, pregnane X receptor, glucocorticoid receptor, and hepatic nuclear factor 4alpha. Mol Pharmacol, 2005. 68: p. 747-57 Zhang, J.G., S.S. Dehal, T. Ho, et al., Human cytochrome p450 induction and inhibition potential of clevidipine and its primary metabolite h152/81. Drug Metab Dispos, 2006. 34: p. 734-7 Yasuda, K., A. Ranade, R. Venkataramanan, et al., A comprehensive in vitro and in silico analysis of antibiotics that activate pregnane X receptor and induce CYP3A4 in liver and intestine. Drug Metab Dispos, 2008. 36: p. 1689-97 Prueksaritanont, T., K.M. Richards, Y. Qiu, et al., Comparative effects of fibrates on drug metabolizing enzymes in human hepatocytes. Pharm Res, 2005. 22: p. 71-8 Dvorak, Z., J. Ulrichova, L. Pichard-Garcia, et al., Comparative effect of colchicine and colchiceine on cytotoxicity and CYP gene expression in primary human hepatocytes. Toxicol In Vitro, 2002. 16: p. 219-27 Dvorak, Z., M. Modriansky, L. Pichard-Garcia, et al., Colchicine down-regulates cytochrome P450 2B6, 2C8, 2C9, and 3A4 in human hepatocytes by affecting their glucocorticoid receptor-mediated regulation. Mol Pharmacol, 2003. 64: p. 160-9 Vrzal, R., M. Daujat-Chavanieu, J.M. Pascussi, et al., Microtubules-interfering agents restrict aryl hydrocarbon receptor-mediated CYP1A2 induction in primary cultures of human hepatocytes via c-jun-N-terminal kinase and glucocorticoid receptor. Eur J Pharmacol, 2008. 581: p. 244-54 Papageorgiou, I., S. Grepper, and J.D. Unadkat, Induction of hepatic CYP3A enzymes by pregnancy-related hormones: studies in human hepatocytes and hepatic cell lines. Drug Metab Dispos, 2013. 41: p. 281-90 Mao, J., T.R. Johnson, Z. Shen, et al., Prediction of crizotinib-midazolam interaction using the Simcyp population-based simulator: comparison of CYP3A time-dependent inhibition between human liver microsomes versus hepatocytes. Drug Metab Dispos, 2013. 41: p. 34352 Kamenickova, A., E. Anzenbacherova, P. Pavek, et al., Pelargonidin activates the AhR and induces CYP1A1 in primary human hepatocytes and human cancer cell lines HepG2 and LS174T. Toxicol Lett, 2013. 218: p. 253-9

24

48.

49.

50.

51.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

52.

53.

54.

55.

56.

57. 58.

59.

60.

61.

62.

63. 64.

65.

Lindley, C., G. Hamilton, J.S. McCune, et al., The effect of cyclophosphamide with and without dexamethasone on cytochrome P450 3A4 and 2B6 in human hepatocytes. Drug Metab Dispos, 2002. 30: p. 814-22 Wang, D., L. Li, J. Fuhrman, et al., The role of constitutive androstane receptor in oxazaphosphorine-mediated induction of drug-metabolizing enzymes in human hepatocytes. Pharm Res, 2011. 28: p. 2034-44 Muntane-Relat, J., J.C. Ourlin, J. Domergue, et al., Differential effects of cytokines on the inducible expression of CYP1A1, CYP1A2, and CYP3A4 in human hepatocytes in primary culture. Hepatology, 1995. 22: p. 1143-53 Abdel-Razzak, Z., L. Corcos, A. Fautrel, et al., Transforming growth factor-beta 1 downregulates basal and polycyclic aromatic hydrocarbon-induced cytochromes P-450 1A1 and 1A2 in adult human hepatocytes in primary culture. Mol Pharmacol, 1994. 46: p. 1100-10 Donato, M.T., M.J. Gomez-Lechon, R. Jover, et al., Human hepatocyte growth factor downregulates the expression of cytochrome P450 isozymes in human hepatocytes in primary culture. J Pharmacol Exp Ther, 1998. 284: p. 760-7 Pascussi, J.M., S. Gerbal-Chaloin, L. Pichard-Garcia, et al., Interleukin-6 negatively regulates the expression of pregnane X receptor and constitutively activated receptor in primary human hepatocytes. Biochem Biophys Res Commun, 2000. 274: p. 707-13 Assenat, E., S. Gerbal-Chaloin, D. Larrey, et al., Interleukin 1beta inhibits CAR-induced expression of hepatic genes involved in drug and bilirubin clearance. Hepatology, 2004. 40: p. 951-60 Thasler, W.E., R. Dayoub, M. Muhlbauer, et al., Repression of cytochrome P450 activity in human hepatocytes in vitro by a novel hepatotrophic factor, augmenter of liver regeneration. J Pharmacol Exp Ther, 2006. 316: p. 822-9 Mao, Z., X. Luan, G. Cao, et al., DEC1 binding to the proximal promoter of CYP3A4 ascribes to the downregulation of CYP3A4 expression by IL-6 in primary human hepatocytes. Biochem Pharmacol, 2012. 84: p. 701-11 Lee, J.K., H.J. Chung, L. Fischer, et al., Human placental lactogen induces CYP2E1 expression via PI 3-kinase pathway in female human hepatocytes. Drug Metab Dispos, 2014. 42: p. 492-9 Rana, R., Y. Chen, S.S. Ferguson, et al., Hepatocyte nuclear factor 4{alpha} regulates rifampicin-mediated induction of CYP2C genes in primary cultures of human hepatocytes. Drug Metab Dispos, 2010. 38: p. 591-9 Dickmann, L.J., S.K. Patel, D.A. Rock, et al., Effects of interleukin-6 (IL-6) and an anti-IL-6 monoclonal antibody on drug-metabolizing enzymes in human hepatocyte culture. Drug Metab Dispos, 2011. 39: p. 1415-22 Lawrence, S.K., D. Nguyen, C. Bowen, et al., The metabolic drug-drug interaction profile of Dabrafenib: in vitro investigations and quantitative extrapolation of the P450-mediated DDI risk. Drug Metab Dispos, 2014. 42: p. 1180-90 Schuetz, J.D., E.G. Schuetz, J.V. Thottassery, et al., Identification of a novel dexamethasone responsive enhancer in the human CYP3A5 gene and its activation in human and rat liver cells. Mol Pharmacol, 1996. 49: p. 63-72 Pascussi, J.M., L. Drocourt, J.M. Fabre, et al., Dexamethasone induces pregnane X receptor and retinoid X receptor-alpha expression in human hepatocytes: synergistic increase of CYP3A4 induction by pregnane X receptor activators. Mol Pharmacol, 2000. 58: p. 361-72 McCune, J.S., R.L. Hawke, E.L. LeCluyse, et al., In vivo and in vitro induction of human cytochrome P4503A4 by dexamethasone. Clin Pharmacol Ther, 2000. 68: p. 356-66 Pascussi, J.M., S. Gerbal-Chaloin, J.M. Fabre, et al., Dexamethasone enhances constitutive androstane receptor expression in human hepatocytes: consequences on cytochrome P450 gene regulation. Mol Pharmacol, 2000. 58: p. 1441-50 Gerbal-Chaloin, S., M. Daujat, J.M. Pascussi, et al., Transcriptional regulation of CYP2C9 gene. Role of glucocorticoid receptor and constitutive androstane receptor. J Biol Chem, 2002. 277: p. 209-17 25

66.

67. 68.

69.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

70.

71.

72.

73. 74. 75.

76.

77.

78.

79.

80.

81. 82.

83.

84.

Pascussi, J.M., L. Drocourt, S. Gerbal-Chaloin, et al., Dual effect of dexamethasone on CYP3A4 gene expression in human hepatocytes. Sequential role of glucocorticoid receptor and pregnane X receptor. Eur J Biochem, 2001. 268: p. 6346-58 Raucy, J.L., L. Mueller, K. Duan, et al., Expression and induction of CYP2C P450 enzymes in primary cultures of human hepatocytes. J Pharmacol Exp Ther, 2002. 302: p. 475-82 Wang, H., S.R. Faucette, D. Gilbert, et al., Glucocorticoid receptor enhancement of pregnane X receptor-mediated CYP2B6 regulation in primary human hepatocytes. Drug Metab Dispos, 2003. 31: p. 620-30 Chen, Y., S.S. Ferguson, M. Negishi, et al., Identification of constitutive androstane receptor and glucocorticoid receptor binding sites in the CYP2C19 promoter. Mol Pharmacol, 2003. 64: p. 316-24 Monostory, K., K. Kohalmy, R.A. Prough, et al., The effect of synthetic glucocorticoid, dexamethasone on CYP1A1 inducibility in adult rat and human hepatocytes. FEBS Lett, 2005. 579: p. 229-35 Onica, T., K. Nichols, M. Larin, et al., Dexamethasone-mediated up-regulation of human CYP2A6 involves the glucocorticoid receptor and increased binding of hepatic nuclear factor 4 alpha to the proximal promoter. Mol Pharmacol, 2008. 73: p. 451-60 Belic, A., M. Temesvari, K. Kohalmy, et al., Investigation of the CYP2C9 induction profile in human hepatocytes by combining experimental and modelling approaches. Curr Drug Metab, 2009. 10: p. 1066-74 Kohalmy, K., V. Tamasi, L. Kobori, et al., Dehydroepiandrosterone induces human CYP2B6 through the constitutive androstane receptor. Drug Metab Dispos, 2007. 35: p. 1495-501 Belic, A., K. Toth, R. Vrzal, et al., Dehydroepiandrosterone post-transcriptionally modifies CYP1A2 induction involving androgen receptor. Chem Biol Interact, 2013. 203: p. 597-603 Pondugula, S.R., P.C. Flannery, K.L. Abbott, et al., Diindolylmethane, a naturally occurring compound, induces CYP3A4 and MDR1 gene expression by activating human PXR. Toxicol Lett, 2015. 232: p. 580-9 Gross-Steinmeyer, K., P.L. Stapleton, F. Liu, et al., Phytochemical-induced changes in gene expression of carcinogen-metabolizing enzymes in cultured human primary hepatocytes. Xenobiotica, 2004. 34: p. 619-32 Gross-Steinmeyer, K., P.L. Stapleton, J.H. Tracy, et al., Modulation of aflatoxin B1-mediated genotoxicity in primary cultures of human hepatocytes by diindolylmethane, curcumin, and xanthohumols. Toxicol Sci, 2009. 112: p. 303-10 Richert, L., G. Tuschl, C. Viollon-Abadie, et al., Species differences in the response of liver drug-metabolizing enzymes to (S)-4-O-tolylsulfanyl-2-(4-trifluormethyl-phenoxy)-butyric acid (EMD 392949) in vivo and in vitro. Drug Metab Dispos, 2008. 36: p. 702-14 Hariparsad, N., S.C. Nallani, R.S. Sane, et al., Induction of CYP3A4 by efavirenz in primary human hepatocytes: comparison with rifampin and phenobarbital. J Clin Pharmacol, 2004. 44: p. 1273-81 Williams, S.N., H. Shih, D.K. Guenette, et al., Comparative studies on the effects of green tea extracts and individual tea catechins on human CYP1A gene expression. Chem Biol Interact, 2000. 128: p. 211-29 Casabar, R.C., P.C. Das, G.K. Dekrey, et al., Endosulfan induces CYP2B6 and CYP3A4 by activating the pregnane X receptor. Toxicol Appl Pharmacol, 2010. 245: p. 335-43 Sahi, J., M.W. Sinz, S. Campbell, et al., Metabolism and transporter-mediated drug-drug interactions of the endothelin-A receptor antagonist CI-1034. Chem Biol Interact, 2006. 159: p. 156-68 Li, Y., J.S. Ross-Viola, N.F. Shay, et al., Human CYP3A4 and murine Cyp3A11 are regulated by equol and genistein via the pregnane X receptor in a species-specific manner. J Nutr, 2009. 139: p. 898-904 Higashi, E., T. Fukami, M. Itoh, et al., Human CYP2A6 is induced by estrogen via estrogen receptor. Drug Metab Dispos, 2007. 35: p. 1935-41 26

85. 86.

87.

88.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

89. 90.

91.

92. 93.

94.

95. 96. 97. 98.

99.

100.

101.

102. 103. 104.

Choi, S.Y., K.H. Koh, and H. Jeong, Isoform-specific regulation of cytochromes P450 expression by estradiol and progesterone. Drug Metab Dispos, 2013. 41: p. 263-9 Koh, K.H., S. Jurkovic, K. Yang, et al., Estradiol induces cytochrome P450 2B6 expression at high concentrations: implication in estrogen-mediated gene regulation in pregnancy. Biochem Pharmacol, 2012. 84: p. 93-103 Kostrubsky, V.E., S.C. Strom, S.G. Wood, et al., Ethanol and isopentanol increase CYP3A and CYP2E in primary cultures of human hepatocytes. Arch Biochem Biophys, 1995. 322: p. 51620 Yanakakis, L.J. and N.N. Bumpus, Biotransformation of the antiretroviral drug etravirine: metabolite identification, reaction phenotyping, and characterization of autoinduction of cytochrome P450-dependent metabolism. Drug Metab Dispos, 2012. 40: p. 803-14 Das, P.C., Y. Cao, N. Cherrington, et al., Fipronil induces CYP isoforms and cytotoxicity in human hepatocytes. Chem Biol Interact, 2006. 164: p. 200-14 Huwyler, J., M.B. Wright, H. Gutmann, et al., Induction of cytochrome P450 3A4 and Pglycoprotein by the isoxazolyl-penicillin antibiotic flucloxacillin. Curr Drug Metab, 2006. 7: p. 119-26 Deng, Y., L. Wang, Y. Yang, et al., In vitro inhibition and induction of human liver cytochrome P450 enzymes by gentiopicroside: potent effect on CYP2A6. Drug Metab Pharmacokinet, 2013. 28: p. 339-44 He, N., H.B. Cai, H.G. Xie, et al., Induction of cyp3a in primary cultures of human hepatocytes by ginkgolides a and B. Clin Exp Pharmacol Physiol, 2007. 34: p. 632-5 Brobst, D.E., X. Ding, K.L. Creech, et al., Guggulsterone activates multiple nuclear receptors and induces CYP3A gene expression through the pregnane X receptor. J Pharmacol Exp Ther, 2004. 310: p. 528-35 Watanabe, K., K. Sakurai, Y. Tsuchiya, et al., Dual roles of nuclear receptor liver X receptor alpha (LXRalpha) in the CYP3A4 expression in human hepatocytes as a positive and negative regulator. Biochem Pharmacol, 2013. 86: p. 428-36 Cho, Y.Y., H.U. Jeong, J.H. Kim, et al., Effect of honokiol on the induction of drug-metabolizing enzymes in human hepatocytes. Drug Des Devel Ther, 2014. 8: p. 2137-45 Moore, L.B., B. Goodwin, S.A. Jones, et al., St. John's wort induces hepatic drug metabolism through activation of the pregnane X receptor. Proc Natl Acad Sci U S A, 2000. 97: p. 7500-2 Goodwin, B., L.B. Moore, C.M. Stoltz, et al., Regulation of the human CYP2B6 gene by the nuclear pregnane X receptor. Mol Pharmacol, 2001. 60: p. 427-31 Chen, Y., S.S. Ferguson, M. Negishi, et al., Induction of human CYP2C9 by rifampicin, hyperforin, and phenobarbital is mediated by the pregnane X receptor. J Pharmacol Exp Ther, 2004. 308: p. 495-501 Nishimura, M., T. Imai, Y. Morioka, et al., Effects of NO-1886 (Ibrolipim), a lipoprotein lipasepromoting agent, on gene induction of cytochrome P450s, carboxylesterases, and sulfotransferases in primary cultures of human hepatocytes. Drug Metab Pharmacokinet, 2004. 19: p. 422-9 Morioka, Y., M. Nishimura, T. Imai, et al., Assessment of induction of cytochrome P450 by NO-1886 (ibrolipim), a lipoprotein lipase-promoting agent, in primary cultures of human hepatocytes and in female rat liver. Drug Metab Pharmacokinet, 2006. 21: p. 19-28 Drocourt, L., J.M. Pascussi, E. Assenat, et al., Calcium channel modulators of the dihydropyridine family are human pregnane X receptor activators and inducers of CYP3A, CYP2B, and CYP2C in human hepatocytes. Drug Metab Dispos, 2001. 29: p. 1325-31 Novotna, A., K. Krasulova, I. Bartonkova, et al., Dual effects of ketoconazole cis-enantiomers on CYP3A4 in human hepatocytes and HepG2 Cells. PLoS One, 2014. 9: p. e111286 Novotna, A., M. Korhonova, I. Bartonkova, et al., Enantiospecific effects of ketoconazole on aryl hydrocarbon receptor. PLoS One, 2014. 9: p. e101832 Vrzal, R., K. Frauenstein, P. Proksch, et al., Khellin and visnagin differentially modulate AHR signaling and downstream CYP1A activity in human liver cells. PLoS One, 2013. 8: p. e74917 27

105.

106.

107.

108.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

109.

110.

111.

112. 113.

114.

115.

116.

117. 118. 119.

120.

121. 122.

DiNatale, B.C., I.A. Murray, J.C. Schroeder, et al., Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that synergistically induces interleukin-6 in the presence of inflammatory signaling. Toxicol Sci, 2010. 115: p. 89-97 Curi-Pedrosa, R., M. Daujat, L. Pichard, et al., Omeprazole and lansoprazole are mixed inducers of CYP1A and CYP3A in human hepatocytes in primary culture. J Pharmacol Exp Ther, 1994. 269: p. 384-92 Masubuchi, N., A.P. Li, and O. Okazaki, An evaluation of the cytochrome P450 induction potential of pantoprazole in primary human hepatocytes. Chem Biol Interact, 1998. 114: p. 113 Krusekopf, S., I. Roots, A.G. Hildebrandt, et al., Time-dependent transcriptional induction of CYP1A1, CYP1A2 and CYP1B1 mRNAs by H+/K+ -ATPase inhibitors and other xenobiotics. Xenobiotica, 2003. 33: p. 107-18 Novotna, A. and Z. Dvorak, Omeprazole and lansoprazole enantiomers induce CYP3A4 in human hepatocytes and cell lines via glucocorticoid receptor and pregnane X receptor axis. PLoS One, 2014. 9: p. e105580 Dhuria, S., H. Einolf, J. Mangold, et al., Time-dependent inhibition and induction of human cytochrome P4503A4/5 by an oral IAP antagonist, LCL161, in vitro and in vivo in healthy subjects. J Clin Pharmacol, 2013. 53: p. 642-53 Zamek-Gliszczynski, M.J., M.A. Mohutsky, J.L. Rehmel, et al., Investigational small-molecule drug selectively suppresses constitutive CYP2B6 activity at the gene transcription level: physiologically based pharmacokinetic model assessment of clinical drug interaction risk. Drug Metab Dispos, 2014. 42: p. 1008-15 Gong, X., Y. Gao, G. Guo, et al., Effect of matrine on primary human hepatocytes in vitro. Cytotechnology, 2015. 67: p. 255-65 Vrzal, R., K. Kubesova, P. Pavek, et al., Benzodiazepines medazepam and midazolam are activators of pregnane X receptor and weak inducers of CYP3A4: investigation in primary cultures of human hepatocytes and hepatocarcinoma cell lines. Toxicol Lett, 2010. 193: p. 183-8 Kim, Y.H., Y.J. Bae, H.S. Kim, et al., Measurement of Human Cytochrome P450 Enzyme Induction Based on Mesalazine and Mosapride Citrate Treatments Using a Luminescent Assay. Biomol Ther (Seoul), 2015. 23: p. 486-92 Rencurel, F., M. Foretz, M.R. Kaufmann, et al., Stimulation of AMP-activated protein kinase is essential for the induction of drug metabolizing enzymes by phenobarbital in human and mouse liver. Mol Pharmacol, 2006. 70: p. 1925-34 Yang, H., B. Garzel, S. Heyward, et al., Metformin represses drug-induced expression of CYP2B6 by modulating the constitutive androstane receptor signaling. Mol Pharmacol, 2014. 85: p. 249-60 Krausova, L., L. Stejskalova, H. Wang, et al., Metformin suppresses pregnane X receptor (PXR)-regulated transactivation of CYP3A4 gene. Biochem Pharmacol, 2011. 82: p. 1771-80 Campbell, S.D., A. Crafford, B.L. Williamson, et al., Mechanism of autoinduction of methadone N-demethylation in human hepatocytes. Anesth Analg, 2013. 117: p. 52-60 Tolson, A.H., H. Li, N.D. Eddington, et al., Methadone induces the expression of hepatic drugmetabolizing enzymes through the activation of pregnane X receptor and constitutive androstane receptor. Drug Metab Dispos, 2009. 37: p. 1887-94 Lemaire, G., C. Benod, V. Nahoum, et al., Discovery of a highly active ligand of human pregnane x receptor: a case study from pharmacophore modeling and virtual screening to "in vivo" biological activity. Mol Pharmacol, 2007. 72: p. 572-81 Mnif, W., J.M. Pascussi, A. Pillon, et al., Estrogens and antiestrogens activate hPXR. Toxicol Lett, 2007. 170: p. 19-29 Banerjee, M. and T. Chen, Thiazide-like diuretic drug metolazone activates human pregnane X receptor to induce cytochrome 3A4 and multidrug-resistance protein 1. Biochem Pharmacol, 2014. 92: p. 389-402 28

123.

124.

125.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

126.

127.

128.

129. 130.

131.

132.

133. 134. 135. 136. 137.

138.

139.

140. 141.

Harvey, J.L., A.J. Paine, P. Maurel, et al., Effect of the adrenal 11-beta-hydroxylase inhibitor metyrapone on human hepatic cytochrome P-450 expression: induction of cytochrome P-450 3A4. Drug Metab Dispos, 2000. 28: p. 96-101 Svecova, L., R. Vrzal, L. Burysek, et al., Azole antimycotics differentially affect rifampicininduced pregnane X receptor-mediated CYP3A4 gene expression. Drug Metab Dispos, 2008. 36: p. 339-48 Jang, G.R. and L.Z. Benet, Antiprogestin pharmacodynamics, pharmacokinetics, and metabolism: implications for their long-term use. J Pharmacokinet Biopharm, 1997. 25: p. 647-72 Shimizu, T., K. Akimoto, T. Yoshimura, et al., Autoinduction of MKC-963 [(R)-1-(1cyclohexylethylamino)-4-phenylphthalazine] metabolism in healthy volunteers and its retrospective evaluation using primary human hepatocytes and cDNA-expressed enzymes. Drug Metab Dispos, 2006. 34: p. 950-4 Dixit, V., N. Hariparsad, F. Li, et al., Cytochrome P450 enzymes and transporters induced by anti-human immunodeficiency virus protease inhibitors in human hepatocytes: implications for predicting clinical drug interactions. Drug Metab Dispos, 2007. 35: p. 1853-9 Kirby, B.J., A.C. Collier, E.D. Kharasch, et al., Complex drug interactions of HIV protease inhibitors 2: in vivo induction and in vitro to in vivo correlation of induction of cytochrome P450 1A2, 2B6, and 2C9 by ritonavir or nelfinavir. Drug Metab Dispos, 2011. 39: p. 2329-37 Hernandez, J.P., W. Huang, L.M. Chapman, et al., The environmental estrogen, nonylphenol, activates the constitutive androstane receptor. Toxicol Sci, 2007. 98: p. 416-26 Doricakova, A. and R. Vrzal, A food contaminant ochratoxin A suppresses pregnane X receptor (PXR)-mediated CYP3A4 induction in primary cultures of human hepatocytes. Toxicology, 2015. 337: p. 72-8 Ayed-Boussema, I., J.M. Pascussi, C. Zaied, et al., Ochratoxin A induces CYP3A4, 2B6, 3A5, 2C9, 1A1, and CYP1A2 gene expression in primary cultured human hepatocytes: a possible activation of nuclear receptors. Drug Chem Toxicol, 2012. 35: p. 71-80 Piton, A., C. Rauch, S. Langouet, et al., Involvement of pregnane X receptor in the regulation of CYP2B6 gene expression by oltipraz in human hepatocytes. Toxicol In Vitro, 2010. 24: p. 452-9 Daujat, M., B. Peryt, P. Lesca, et al., Omeprazole, an inducer of human CYP1A1 and 1A2, is not a ligand for the Ah receptor. Biochem Biophys Res Commun, 1992. 188: p. 820-5 Raucy, J.L., Regulation of CYP3A4 expression in human hepatocytes by pharmaceuticals and natural products. Drug Metab Dispos, 2003. 31: p. 533-9 Gerbal-Chaloin, S., L. Pichard-Garcia, J.M. Fabre, et al., Role of CYP3A4 in the regulation of the aryl hydrocarbon receptor by omeprazole sulphide. Cell Signal, 2006. 18: p. 740-50 Guillen, M.I., M.T. Donato, R. Jover, et al., Oncostatin M down-regulates basal and induced cytochromes P450 in human hepatocytes. J Pharmacol Exp Ther, 1998. 285: p. 127-34 Novotna, A., A. Doricakova, R. Vrzal, et al., Investigation of Orlistat effects on PXR activation and CYP3A4 expression in primary human hepatocytes and human intestinal LS174T cells. Eur J Pharm Sci, 2010. 41: p. 276-80 Budinsky, R.A., E.L. LeCluyse, S.S. Ferguson, et al., Human and rat primary hepatocyte CYP1A1 and 1A2 induction with 2,3,7,8-tetrachlorodibenzo-p-dioxin, 2,3,7,8-tetrachlorodibenzofuran, and 2,3,4,7,8-pentachlorodibenzofuran. Toxicol Sci, 2010. 118: p. 224-35 Wojcikowski, J., P. Maurel, and W.A. Daniel, Autoinduction of the metabolism of phenothiazine neuroleptics in a primary culture of human hepatocytes. Pharmacol Rep, 2012. 64: p. 1578-83 Gervot, L., B. Rochat, J.C. Gautier, et al., Human CYP2B6: expression, inducibility and catalytic activities. Pharmacogenetics, 1999. 9: p. 295-306 Gerbal-Chaloin, S., J.M. Pascussi, L. Pichard-Garcia, et al., Induction of CYP2C genes in human hepatocytes in primary culture. Drug Metabolism and Disposition, 2001. 29: p. 242-51 29

142.

143.

144.

145.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

146.

147.

148. 149.

150.

151.

152.

153. 154.

155. 156.

157.

158.

159.

Courtois, A., L. Payen, E. Le Ferrec, et al., Differential regulation of multidrug resistanceassociated protein 2 (MRP2) and cytochromes P450 2B1/2 and 3A1/2 in phenobarbitaltreated hepatocytes. Biochem Pharmacol, 2002. 63: p. 333-41 Wang, H., S. Faucette, T. Sueyoshi, et al., A novel distal enhancer module regulated by pregnane X receptor/constitutive androstane receptor is essential for the maximal induction of CYP2B6 gene expression. J Biol Chem, 2003. 278: p. 14146-52 Itoh, M., M. Nakajima, E. Higashi, et al., Induction of human CYP2A6 is mediated by the pregnane X receptor with peroxisome proliferator-activated receptor-gamma coactivator 1alpha. J Pharmacol Exp Ther, 2006. 319: p. 693-702 Wang, H., S. Faucette, R. Moore, et al., Human constitutive androstane receptor mediates induction of CYP2B6 gene expression by phenytoin. J Biol Chem, 2004. 279: p. 29295-301 Chaudhry, A.S., T.J. Urban, J.K. Lamba, et al., CYP2C9*1B promoter polymorphisms, in linkage with CYP2C19*2, affect phenytoin autoinduction of clearance and maintenance dose. J Pharmacol Exp Ther, 2010. 332: p. 599-611 Arora, V., M.L. Cate, C. Ghosh, et al., Phosphorodiamidate morpholino antisense oligomers inhibit expression of human cytochrome P450 3A4 and alter selected drug metabolism. Drug Metab Dispos, 2002. 30: p. 757-62 Cooper, B.W., T.M. Cho, P.M. Thompson, et al., Phthalate induction of CYP3A4 is dependent on glucocorticoid regulation of PXR expression. Toxicol Sci, 2008. 103: p. 268-77 DeKeyser, J.G., E.M. Laurenzana, E.C. Peterson, et al., Selective phthalate activation of naturally occurring human constitutive androstane receptor splice variants and the pregnane X receptor. Toxicol Sci, 2011. 120: p. 381-91 Wang, Y.M., W. Lin, S.C. Chai, et al., Piperine activates human pregnane X receptor to induce the expression of cytochrome P450 3A4 and multidrug resistance protein 1. Toxicol Appl Pharmacol, 2013. 272: p. 96-107 Fontaine, F., C. Delescluse, G. de Sousa, et al., Cytochrome 1A1 induction by primaquine in human hepatocytes and HepG2 cells: absence of binding to the aryl hydrocarbon receptor. Biochem Pharmacol, 1999. 57: p. 255-62 Donato, M.T., P. Viitala, C. Rodriguez-Antona, et al., CYP2A5/CYP2A6 expression in mouse and human hepatocytes treated with various in vivo inducers. Drug Metab Dispos, 2000. 28: p. 1321-6 Price, R.J., A.M. Giddings, M.P. Scott, et al., Effect of Pyrethrins on cytochrome P450 forms in cultured rat and human hepatocytes. Toxicology, 2008. 243: p. 84-95 Yang, D., X. Wang, Y.T. Chen, et al., Pyrethroid insecticides: isoform-dependent hydrolysis, induction of cytochrome P450 3A4 and evidence on the involvement of the pregnane X receptor. Toxicol Appl Pharmacol, 2009. 237: p. 49-58 Das, P.C., T.M. Streit, Y. Cao, et al., Pyrethroids: cytotoxicity and induction of CYP isoforms in human hepatocytes. Drug Metabol Drug Interact, 2008. 23: p. 211-36 Hirose, Y., H. Nagahori, T. Yamada, et al., Comparison of the effects of the synthetic pyrethroid Metofluthrin and phenobarbital on CYP2B form induction and replicative DNA synthesis in cultured rat and human hepatocytes. Toxicology, 2009. 258: p. 64-9 Qian, L., R. Zolfaghari, and A.C. Ross, Liver-specific cytochrome P450 CYP2C22 is a direct target of retinoic acid and a retinoic acid-metabolizing enzyme in rat liver. J Lipid Res, 2010. 51: p. 1781-92 Li, A.P., M.K. Reith, A. Rasmussen, et al., Primary human hepatocytes as a tool for the evaluation of structure-activity relationship in cytochrome P450 induction potential of xenobiotics: evaluation of rifampin, rifapentine and rifabutin. Chem Biol Interact, 1997. 107: p. 17-30 Reinach, B., G. de Sousa, P. Dostert, et al., Comparative effects of rifabutin and rifampicin on cytochromes P450 and UDP-glucuronosyl-transferases expression in fresh and cryopreserved human hepatocytes. Chem Biol Interact, 1999. 121: p. 37-48 30

160.

161.

162.

163.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

164.

165.

166. 167.

168.

169. 170. 171.

172. 173.

174.

175. 176.

177.

Williamson, B., K.E. Dooley, Y. Zhang, et al., Induction of influx and efflux transporters and cytochrome P450 3A4 in primary human hepatocytes by rifampin, rifabutin, and rifapentine. Antimicrob Agents Chemother, 2013. 57: p. 6366-9 Greuet, J., L. Pichard, C. Bonfils, et al., The fetal specific gene CYP3A7 is inducible by rifampicin in adult human hepatocytes in primary culture. Biochem Biophys Res Commun, 1996. 225: p. 689-94 Rae, J.M., M.D. Johnson, M.E. Lippman, et al., Rifampin is a selective, pleiotropic inducer of drug metabolism genes in human hepatocytes: studies with cDNA and oligonucleotide expression arrays. J Pharmacol Exp Ther, 2001. 299: p. 849-57 Duniec-Dmuchowski, Z., H.L. Fang, S.C. Strom, et al., Human pregnane X receptor activation and CYP3A4/CYP2B6 induction by 2,3-oxidosqualene:lanosterol cyclase inhibition. Drug Metab Dispos, 2009. 37: p. 900-8 Pichard-Garcia, L., R.J. Weaver, N. Eckett, et al., The olivacine derivative s 16020 (9-hydroxy5,6-dimethyl-N-[2-(dimethylamino)ethyl)-6H-pyrido(4,3-B)-carbazole-1-c arboxamide) induces CYP1A and its own metabolism in human hepatocytes in primary culture. Drug Metab Dispos, 2004. 32: p. 80-8 Ayan-Oshodi, M.A., B.A. Willis, W.F. Annes, et al., The effects on metabolic clearance when administering a potent CYP3A autoinducer with the prototypic CYP3A inhibitor, ketoconazole. Drug Metab Dispos, 2012. 40: p. 1945-52 Fan, G., Y. Cao, and J. Yang, In vitro inhibition and induction of human cytochrome P450 enzymes by SIPI5357, a potential antidepressant. Biopharm Drug Dispos, 2015 Dvorak, Z., R. Vrzal, P. Henklova, et al., JNK inhibitor SP600125 is a partial agonist of human aryl hydrocarbon receptor and induces CYP1A1 and CYP1A2 genes in primary human hepatocytes. Biochem Pharmacol, 2008. 75: p. 580-8 Gross-Steinmeyer, K., P.L. Stapleton, J.H. Tracy, et al., Sulforaphane- and phenethyl isothiocyanate-induced inhibition of aflatoxin B1-mediated genotoxicity in human hepatocytes: role of GSTM1 genotype and CYP3A4 gene expression. Toxicol Sci, 2010. 116: p. 422-32 Yokota, S., E. Higashi, T. Fukami, et al., Human CYP2A6 is regulated by nuclear factorerythroid 2 related factor 2. Biochem Pharmacol, 2011. 81: p. 289-94 Duniec-Dmuchowski, Z., E. Ellis, S.C. Strom, et al., Regulation of CYP3A4 and CYP2B6 expression by liver X receptor agonists. Biochem Pharmacol, 2007. 74: p. 1535-40 Tippin, T.K., G. Hamilton, L. Moore, et al., CYP3A induction by N-hydroxyformamide tumor necrosis factor-alpha converting enzyme/matrix metalloproteinase inhibitors use of a pregname X receptor activation assay and primary hepatocyte culture for assessing induction potential in humans. Drug Metab Dispos, 2003. 31: p. 870-7 Ring, B.J., B.E. Patterson, M.I. Mitchell, et al., Effect of tadalafil on cytochrome P450 3A4mediated clearance: studies in vitro and in vivo. Clin Pharmacol Ther, 2005. 77: p. 63-75 Desai, P.B., S.C. Nallani, R.S. Sane, et al., Induction of cytochrome P450 3A4 in primary human hepatocytes and activation of the human pregnane X receptor by tamoxifen and 4hydroxytamoxifen. Drug Metab Dispos, 2002. 30: p. 608-12 Sane, R.S., D.J. Buckley, A.R. Buckley, et al., Role of human pregnane X receptor in tamoxifenand 4-hydroxytamoxifen-mediated CYP3A4 induction in primary human hepatocytes and LS174T cells. Drug Metab Dispos, 2008. 36: p. 946-54 Kostrubsky, V.E., L.D. Lewis, S.C. Strom, et al., Induction of cytochrome P4503A by taxol in primary cultures of human hepatocytes. Arch Biochem Biophys, 1998. 355: p. 131-6 Nallani, S.C., B. Goodwin, A.R. Buckley, et al., Differences in the induction of cytochrome P450 3A4 by taxane anticancer drugs, docetaxel and paclitaxel, assessed employing primary human hepatocytes. Cancer Chemother Pharmacol, 2004. 54: p. 219-29 Schrenk, D., T. Stuven, G. Gohl, et al., Induction of CYP1A and glutathione S-transferase activities by 2,3,7,8-tetrachlorodibenzo-p-dioxin in human hepatocyte cultures. Carcinogenesis, 1995. 16: p. 943-6 31

178.

179.

180.

181.

Downloaded by [University of California, San Diego] at 00:32 04 December 2015

182.

183.

184.

185.

186.

187. 188.

189.

190.

191.

192.

193.

194.

Xu, L., A.P. Li, D.L. Kaminski, et al., 2,3,7,8 Tetrachlorodibenzo-p-dioxin induction of cytochrome P4501A in cultured rat and human hepatocytes. Chem Biol Interact, 2000. 124: p. 173-89 Zhang, Z.Y., R.D. Pelletier, Y.N. Wong, et al., Preferential inducibility of CYP1A1 and CYP1A2 by TCDD: differential regulation in primary human hepatocytes versus transformed human cells. Biochem Biophys Res Commun, 2006. 341: p. 399-407 Le Vee, M., E. Jouan, and O. Fardel, Involvement of aryl hydrocarbon receptor in basal and 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced expression of target genes in primary human hepatocytes. Toxicol In Vitro, 2010. 24: p. 1775-81 Murayama, N., R. van Beuningen, H. Suemizu, et al., Thalidomide increases human hepatic cytochrome P450 3A enzymes by direct activation of the pregnane X receptor. Chem Res Toxicol, 2014. 27: p. 304-8 Gonzalez, R., J.A. Collado, S. Nell, et al., Cytoprotective properties of alpha-tocopherol are related to gene regulation in cultured D-galactosamine-treated human hepatocytes. Free Radic Biol Med, 2007. 43: p. 1439-52 Nallani, S.C., T.A. Glauser, N. Hariparsad, et al., Dose-dependent induction of cytochrome P450 (CYP) 3A4 and activation of pregnane X receptor by topiramate. Epilepsia, 2003. 44: p. 1521-8 Kim, J., C. Peraire, J. Sola, et al., Drug interaction potential of toremifene and Ndesmethyltoremifene with multiple cytochrome P450 isoforms. Xenobiotica, 2011. 41: p. 851-62 Ledirac, N., G. de Sousa, F. Fontaine, et al., Effects of macrolide antibiotics on CYP3A expression in human and rat hepatocytes: interspecies differences in response to troleandomycin. Drug Metab Dispos, 2000. 28: p. 1391-3 Ramachandran, V., V.E. Kostrubsky, B.J. Komoroski, et al., Troglitazone increases cytochrome P-450 3A protein and activity in primary cultures of human hepatocytes. Drug Metab Dispos, 1999. 27: p. 1194-9 Sahi, J., G. Hamilton, M. Sinz, et al., Effect of troglitazone on cytochrome P450 enzymes in primary cultures of human and rat hepatocytes. Xenobiotica, 2000. 30: p. 273-84 Kitamura, R., H. Asanoma, S. Nagayama, et al., Identification of human liver cytochrome P450 isoforms involved in autoinduced metabolism of the antiangiogenic agent (Z)-5-[(1,2-dihydro2-oxo-3H-indol-3-ylidene)methyl]-2,4-dimethyl-1H-pyrrole-3-pr opanoic acid (TSU-68). Drug Metab Dispos, 2008. 36: p. 1003-9 Smutny, T., M. Bitman, M. Urban, et al., U0126, a mitogen-activated protein kinase kinase 1 and 2 (MEK1 and 2) inhibitor, selectively up-regulates main isoforms of CYP3A subfamily via a pregnane X receptor (PXR) in HepG2 cells. Arch Toxicol, 2014. 88: p. 2243-59 Cerveny, L., L. Svecova, E. Anzenbacherova, et al., Valproic acid induces CYP3A4 and MDR1 gene expression by activation of constitutive androstane receptor and pregnane X receptor pathways. Drug Metab Dispos, 2007. 35: p. 1032-41 Rulcova, A., I. Prokopova, L. Krausova, et al., Stereoselective interactions of warfarin enantiomers with the pregnane X nuclear receptor in gene regulation of major drugmetabolizing cytochrome P450 enzymes. J Thromb Haemost, 2010. 8: p. 2708-17 Thomas, M., O. Burk, B. Klumpp, et al., Direct transcriptional regulation of human hepatic cytochrome P450 3A4 (CYP3A4) by peroxisome proliferator-activated receptor alpha (PPARalpha). Mol Pharmacol, 2013. 83: p. 709-18 Rakhshandehroo, M., G. Hooiveld, M. Muller, et al., Comparative analysis of gene regulation by the transcription factor PPARalpha between mouse and human. PLoS One, 2009. 4: p. e6796 Umehara, K.I., Y. Susaki, R.H. Van Teylingen, et al., Evaluation of the inhibitory and induction potential of YM758, a novel If channel inhibitor, for human P450-mediated metabolism. Eur J Drug Metab Pharmacokinet, 2008. 33: p. 211-23 32

Opportunities and challenges in using human hepatocytes in cytochromes P450 induction assays.

Identification of inducers of xenobiotic-metabolizing cytochromes P450 (CYP) is of topical interest. The issue mainly concerns three sectors: (i) prec...
565B Sizes 0 Downloads 12 Views