1
|
Feng B, Liang G, Zetterberg C, Li S, Huang H, Williams J, Gao H, Morikawa Y, Kumar S. Utility of Chimeric Mice with Humanized Livers for Predicting Hepatic Organic Anion-Transporting Polypeptide 1B-Mediated Clinical Drug-Drug Interactions. Drug Metab Dispos 2024; 52:1073-1082. [PMID: 39103225 DOI: 10.1124/dmd.124.001792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024] Open
Abstract
The influence of transporters on the pharmacokinetics of drugs is being increasingly recognized, and drug-drug interactions (DDIs) via modulation of transporters could lead to clinical adverse events. Organic anion-transporting polypeptide 1B (OATP1B) is a liver-specific uptake transporter in humans that can transport a broad range of substrates, including statins. It is a challenge to predict OATP1B-mediated DDIs using preclinical animal models because of species differences in substrate specificity and abundance levels of transporters. PXB-mice are chimeric mice with humanized livers that are highly repopulated with human hepatocytes and have been widely used for drug metabolism and pharmacokinetics studies in drug discovery. In the present study, we measured the exposure increases [blood AUC (area under the blood/plasma concentration-time curve) and Cmax] of 10 OATP1B substrates in PXB-mice upon coadministration with rifampin, a potent OATP1B specific inhibitor. These data in PXB-mice were then compared with the observed DDIs between OATP1B substrates and single-dose rifampin in humans. Our findings suggest that the DDIs between OATP1B substrates and rifampin in PXB-mouse are comparable with the observed DDIs in the clinic. Since most OATP1B substrates are metabolized by cytochromes P450 (CYPs) and/or are substrates of P-glycoprotein (P-gp), we further validated the utility of PXB-mice to predict complex DDIs involving inhibition of OATP1B, CYPs, and P-gp using cyclosporin A (CsA) and gemfibrozil as perpetrators. Overall, the data support that the chimeric mice with humanized livers could be a useful tool for the prediction of hepatic OATP1B-mediated DDIs in humans. SIGNIFICANCE STATEMENT: The ability of PXB-mouse with humanized liver to predict organic anion-transporting polypeptide 1B (OATP1B)-mediated drug-drug interactions (DDIs) in humans was evaluated. The blood exposure increases of 10 OATP1B substrates with rifampin, an OATP1B inhibitor, in PXB-mice have a good correlation with those observed in humans. More importantly, PXB-mice can predict complex DDIs, including inhibition of OATP1B, cytochromes P450 (CYPs), and P-glycoprotein (P-gp) in humans. PXB-mice are a promising useful tool to assess OATP1B-mediated clinical DDIs.
Collapse
Affiliation(s)
- Bo Feng
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts (B.F., G.L., C.Z., S.L., H.H., J.W., H.G., S.K.) and PhoenixBio USA Corporation, New York, New York (Y.M.)
| | - Guiqing Liang
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts (B.F., G.L., C.Z., S.L., H.H., J.W., H.G., S.K.) and PhoenixBio USA Corporation, New York, New York (Y.M.)
| | - Craig Zetterberg
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts (B.F., G.L., C.Z., S.L., H.H., J.W., H.G., S.K.) and PhoenixBio USA Corporation, New York, New York (Y.M.)
| | - Shaolan Li
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts (B.F., G.L., C.Z., S.L., H.H., J.W., H.G., S.K.) and PhoenixBio USA Corporation, New York, New York (Y.M.)
| | - Hui Huang
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts (B.F., G.L., C.Z., S.L., H.H., J.W., H.G., S.K.) and PhoenixBio USA Corporation, New York, New York (Y.M.)
| | - John Williams
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts (B.F., G.L., C.Z., S.L., H.H., J.W., H.G., S.K.) and PhoenixBio USA Corporation, New York, New York (Y.M.)
| | - Hong Gao
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts (B.F., G.L., C.Z., S.L., H.H., J.W., H.G., S.K.) and PhoenixBio USA Corporation, New York, New York (Y.M.)
| | - Yoshio Morikawa
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts (B.F., G.L., C.Z., S.L., H.H., J.W., H.G., S.K.) and PhoenixBio USA Corporation, New York, New York (Y.M.)
| | - Sanjeev Kumar
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts (B.F., G.L., C.Z., S.L., H.H., J.W., H.G., S.K.) and PhoenixBio USA Corporation, New York, New York (Y.M.)
| |
Collapse
|
2
|
Bi G, Liang F, Wu T, Wang P, Jiang X, Hu S, Wu C, Zhou W, Guo J, Yang X, Fang JH, Chen W, Bi H. Pregnane X receptor activation induces liver enlargement and regeneration and simultaneously promotes the metabolic activity of CYP3A1/2 and CYP2C6/11 in rats. Basic Clin Pharmacol Toxicol 2024; 135:148-163. [PMID: 38887973 DOI: 10.1111/bcpt.14041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Human pregnane X receptor (PXR) is critical for regulating the expression of key drug-metabolizing enzymes such as CYP3A and CYP2C. Our recent study revealed that treatment with rodent-specific PXR agonist pregnenolone-16α-carbonitrile (PCN) significantly induced hepatomegaly and promoted liver regeneration after two-thirds partial hepatectomy (PHx) in mice. However, it remains unclear whether PXR activation induces hepatomegaly and liver regeneration and simultaneously promotes metabolic function of the liver. Here, we investigated the metabolism activity of CYP1A2, CYP3A1/2 and CYP2C6/11 during PXR activation-induced liver enlargement and regeneration in rats after cocktail dosing of CYP probe drugs. For PCN-induced hepatomegaly, a notable increase in the metabolic activity of CYP3A1/2 and CYP2C6/11, as evidenced by the plasma exposure of probe substrates and the AUC ratios of the characteristic metabolites to its corresponding probe substrates. The metabolic activity of CYP1A2, CYP3A1/2 and CYP2C6/11 decreased significantly after PHx. However, PCN treatment obviously enhanced the metabolic activity of CYP2C6/11 and CYP3A1/2 in PHx rats. Furthermore, the protein expression levels of CYP3A1/2 and CYP2C6/11 in liver were up-regulated. Taken together, this study demonstrates that PXR activation not only induces hepatomegaly and liver regeneration in rats, but also promotes the protein expression and metabolic activity of the PXR downstream metabolizing enzymes such as CYP3A1/2 and CYP2C6/11 in the body.
Collapse
Affiliation(s)
- Guofang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Fengting Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ting Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Peng Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaowen Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuang Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Chenghua Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wenhong Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiayin Guo
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jian-Hong Fang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wenying Chen
- Department of Pharmacy, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, China
| |
Collapse
|
3
|
Damoiseaux D, Schinkel AH, Beijnen JH, Huitema ADR, Dorlo TPC. Predictability of human exposure by human-CYP3A4-transgenic mouse models: A meta-analysis. Clin Transl Sci 2024; 17:e13668. [PMID: 38037826 PMCID: PMC10766057 DOI: 10.1111/cts.13668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 12/02/2023] Open
Abstract
First-in-human dose predictions are primarily based on no-observed-adverse-effect levels in animal studies. Predictions from these animal models are only as effective as their ability to predict human results. To narrow the gap between human and animals, researchers have, among other things, focused on the replacement of animal cytochrome P450 (CYP) enzymes with their human counterparts (called humanization), especially in mice. Whereas research in humanized mice is extensive, the emphasis has been particularly on qualitative rather than quantitative predictions. Because the CYP3A4 enzyme is most involved in the metabolism of clinically used drugs, most benefit was expected from CYP3A4 models. There are several applications of these mouse models regarding in vivo CYP3A4 functionality, one of which might be their capacity to help improve first-in-human (FIH) dose predictions for CYP3A4-metabolized drugs. To evaluate whether human-CYP3A4-transgenic mouse models are better predictors of human exposure compared to the wild-type mouse model, we performed a meta-analysis comparing both mouse models in their ability to accurately predict human exposure of small-molecule drugs metabolized by CYP3A4. Results showed that, in general, the human-CYP3A4-transgenic mouse model had similar accuracy in the prediction of human exposure compared to the wild-type mouse model, suggesting that there is limited added value in humanization of the mouse Cyp3a enzymes if the primary aim is to acquire more accurate FIH dose predictions. Despite the results of this meta-analysis, corrections for interspecies differences through extension of human-CYP3A4-transgenic mouse models with pharmacokinetic modeling approaches seems a promising contribution to more accurate quantitative predictions of human pharmacokinetics.
Collapse
Affiliation(s)
- David Damoiseaux
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Alfred H. Schinkel
- Division of PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jos H. Beijnen
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Utrecht Institute of Pharmaceutical Sciences, Utrecht UniversityUtrechtThe Netherlands
| | - Alwin D. R. Huitema
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of PharmacologyPrincess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Department of Clinical PharmacyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Thomas P. C. Dorlo
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of PharmacyUppsala UniversityUppsalaSweden
| |
Collapse
|
4
|
Mezler M, Jones RS, Sangaraju D, Goldman DC, Hoffmann M, Heikkinen AT, Mannila J, Chang JH, Foquet L, Pusalkar S, Chothe PP, Scheer N. Analysis of the Bile Acid Composition in a Fibroblast Growth Factor 19-Expressing Liver-Humanized Mouse Model and Its Use for CYP3A4-Mediated Drug-Drug Interaction Studies. Drug Metab Dispos 2023; 51:1391-1402. [PMID: 37524541 DOI: 10.1124/dmd.123.001398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/02/2023] Open
Abstract
Numerous biomedical applications have been described for liver-humanized mouse models, such as in drug metabolism or drug-drug interaction (DDI) studies. However, the strong enlargement of the bile acid (BA) pool due to lack of recognition of murine intestine-derived fibroblast growth factor-15 by human hepatocytes and a resulting upregulation in the rate-controlling enzyme for BA synthesis, cytochrome P450 (CYP) 7A1, may pose a challenge in interpreting the results obtained from such mice. To address this challenge, the human fibroblast growth factor-19 (FGF19) gene was inserted into the Fah-/- , Rag2-/- , Il2rg-/- NOD (FRGN) mouse model, allowing repopulation with human hepatocytes capable of responding to FGF19. While a decrease in CYP7A1 expression in human hepatocytes from humanized FRGN19 mice (huFRGN19) and a concomitant reduction in BA production was previously shown, a detailed analysis of the BA pool in these animals has not been elucidated. Furthermore, there are sparse data on the use of this model to assess potential clinical DDI. In the present work, the change in BA composition in huFRGN19 compared with huFRGN control animals was systematically evaluated, and the ability of the model to recapitulate a clinically described CYP3A4-mediated DDI was assessed. In addition to a massive reduction in the total amount of BA, FGF19 expression in huFRGN19 mice resulted in significant changes in the profile of various primary, secondary, and sulfated BAs in serum and feces. Moreover, as observed clinically, administration of the pregnane X receptor agonist rifampicin reduced the oral exposure of the CYP3A4 substrate triazolam. SIGNIFICANCE STATEMENT: Transgenic expression of FGF19 normalizes the unphysiologically high level of bile acids in a chimeric liver-humanized mouse model and leads to massive changes in bile acid composition. These adaptations could overcome one of the potential impediments in the use of these mouse models for drug-drug interaction studies.
Collapse
Affiliation(s)
- Mario Mezler
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Robert S Jones
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Dewakar Sangaraju
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Devorah C Goldman
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Matthew Hoffmann
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Aki T Heikkinen
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Janne Mannila
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Jae H Chang
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Lander Foquet
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Sandeepraj Pusalkar
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Paresh P Chothe
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| | - Nico Scheer
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (R.S.J., D.S., J.C.C.); Yecuris Corporation, Tualatin, Oregon (D.C.G., L.F.); Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey (M.H.); Symeres Finland Oy, Oulu, Finland, operating under Admescope brand (A.T.H., J.M.); Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. Cambridge, Massachusetts (S.P., P.P.C.); and FH Aachen University of Applied Sciences, Jülich, Germany (N.S.)
| |
Collapse
|
5
|
Asano D, Nakamura K, Nishiya Y, Shiozawa H, Takakusa H, Shibayama T, Inoue SI, Shinozuka T, Hamada T, Yahara C, Watanabe N, Yoshinari K. Physiologically Based Pharmacokinetic Modeling for Quantitative Prediction of Exposure to a Human Disproportionate Metabolite of the Selective Na V1.7 Inhibitor DS-1971a, a Mixed Substrate of Cytochrome P450 and Aldehyde Oxidase, Using Chimeric Mice With Humanized Liver. Drug Metab Dispos 2023; 51:67-80. [PMID: 36273823 DOI: 10.1124/dmd.122.001000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
In a previous study on the human mass balance of DS-1971a, a selective NaV1.7 inhibitor, its CYP2C8-dependent metabolite M1 was identified as a human disproportionate metabolite. The present study assessed the usefulness of pharmacokinetic evaluation in chimeric mice grafted with human hepatocytes (PXB-mice) and physiologically based pharmacokinetic (PBPK) simulation of M1. After oral administration of radiolabeled DS-1971a, the most abundant metabolite in the plasma, urine, and feces of PXB-mice was M1, while those of control SCID mice were aldehyde oxidase-related metabolites including M4, suggesting a drastic difference in the metabolism between these mouse strains. From a qualitative perspective, the metabolite profile observed in PXB-mice was remarkably similar to that in humans, but the quantitative evaluation indicated that the area under the plasma concentration-time curve (AUC) ratio of M1 to DS-1971a (M1/P ratio) was approximately only half of that in humans. A PXB-mouse-derived PBPK model was then constructed to achieve a more accurate prediction, giving an M1/P ratio (1.3) closer to that in humans (1.6) than the observed value in PXB-mice (0.69). In addition, simulated maximum plasma concentration and AUC values of M1 (3429 ng/ml and 17,116 ng·h/ml, respectively) were similar to those in humans (3180 ng/ml and 18,400 ng·h/ml, respectively). These results suggest that PBPK modeling incorporating pharmacokinetic parameters obtained with PXB-mice is useful for quantitatively predicting exposure to human disproportionate metabolites. SIGNIFICANCE STATEMENT: The quantitative prediction of human disproportionate metabolites remains challenging. This paper reports on a successful case study on the practical estimation of exposure (C max and AUC) to DS-1971a and its CYP2C8-dependent, human disproportionate metabolite M1, by PBPK simulation utilizing pharmacokinetic parameters obtained from PXB-mice and in vitro kinetics in human liver fractions. This work adds to the growing knowledge regarding metabolite exposure estimation by static and dynamic models.
Collapse
Affiliation(s)
- Daigo Asano
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Koichi Nakamura
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Yumi Nishiya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Hideyuki Shiozawa
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Hideo Takakusa
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Takahiro Shibayama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Shin-Ichi Inoue
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Tsuyoshi Shinozuka
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Takakazu Hamada
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Chizuko Yahara
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Nobuaki Watanabe
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Kouichi Yoshinari
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan (D.A., K.N., N.Y., H.S., H.T., T. Shibayama, S.-i.I., C.Y., N.W.), R&D Planning & Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T. Shinozuka), Research Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan (T.H.), Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| |
Collapse
|
6
|
Zerdoug A, Le Vée M, Uehara S, Lopez B, Chesné C, Suemizu H, Fardel O. Contribution of Humanized Liver Chimeric Mice to the Study of Human Hepatic Drug Transporters: State of the Art and Perspectives. Eur J Drug Metab Pharmacokinet 2022; 47:621-637. [DOI: 10.1007/s13318-022-00782-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/03/2022]
|
7
|
Fernandez-Checa JC, Bagnaninchi P, Ye H, Sancho-Bru P, Falcon-Perez JM, Royo F, Garcia-Ruiz C, Konu O, Miranda J, Lunov O, Dejneka A, Elfick A, McDonald A, Sullivan GJ, Aithal GP, Lucena MI, Andrade RJ, Fromenty B, Kranendonk M, Cubero FJ, Nelson LJ. Advanced preclinical models for evaluation of drug-induced liver injury - consensus statement by the European Drug-Induced Liver Injury Network [PRO-EURO-DILI-NET]. J Hepatol 2021; 75:935-959. [PMID: 34171436 DOI: 10.1016/j.jhep.2021.06.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023]
Abstract
Drug-induced liver injury (DILI) is a major cause of acute liver failure (ALF) and one of the leading indications for liver transplantation in Western societies. Given the wide use of both prescribed and over the counter drugs, DILI has become a major health issue for which there is a pressing need to find novel and effective therapies. Although significant progress has been made in understanding the molecular mechanisms underlying DILI, our incomplete knowledge of its pathogenesis and inability to predict DILI is largely due to both discordance between human and animal DILI in preclinical drug development and a lack of models that faithfully recapitulate complex pathophysiological features of human DILI. This is exemplified by the hepatotoxicity of acetaminophen (APAP) overdose, a major cause of ALF because of its extensive worldwide use as an analgesic. Despite intensive efforts utilising current animal and in vitro models, the mechanisms involved in the hepatotoxicity of APAP are still not fully understood. In this expert Consensus Statement, which is endorsed by the European Drug-Induced Liver Injury Network, we aim to facilitate and outline clinically impactful discoveries by detailing the requirements for more realistic human-based systems to assess hepatotoxicity and guide future drug safety testing. We present novel insights and discuss major players in APAP pathophysiology, and describe emerging in vitro and in vivo pre-clinical models, as well as advanced imaging and in silico technologies, which may improve prediction of clinical outcomes of DILI.
Collapse
Affiliation(s)
- Jose C Fernandez-Checa
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Consejo Superior Investigaciones Científicas (CSIC), Spain; Liver Unit, Hospital Clínic, Barcelona, Spain; Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; USC Research Center for ALPD, Keck School of Medicine, Los Angeles, United States, CA 90033.
| | - Pierre Bagnaninchi
- Center for Regenerative Medicine, Institute for Regenerative and Repair, The University of Edinburgh, Edinburgh, UK, EH16 4UU; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Faraday Building, Colin Maclaurin Road, EH9 3 DW, Scotland, UK
| | - Hui Ye
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Pau Sancho-Bru
- Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Juan M Falcon-Perez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, 48160, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia, 48015, Spain
| | - Felix Royo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, 48160, Spain
| | - Carmen Garcia-Ruiz
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Consejo Superior Investigaciones Científicas (CSIC), Spain; Liver Unit, Hospital Clínic, Barcelona, Spain; Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; USC Research Center for ALPD, Keck School of Medicine, Los Angeles, United States, CA 90033
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey; Interdisciplinary Neuroscience Program, Bilkent University, Ankara, Turkey; UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Joana Miranda
- Research Institute for iMedicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alistair Elfick
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh EH8 3DW, UK
| | - Alison McDonald
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh EH8 3DW, UK
| | - Gareth J Sullivan
- University of Oslo and the Oslo University Hospital, Oslo, Norway; Hybrid Technology Hub-Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Pediatric Research, Oslo University Hosptial, Oslo, Norway
| | - Guruprasad P Aithal
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospital NHS Trust and University of Nottingham, Nottingham, UK
| | - M Isabel Lucena
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, UICEC SCReN, Universidad de Málaga, Málaga, Spain
| | - Raul J Andrade
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Unidad de Gestión Clínica de Enfermedades Digestivas, Instituto de Investigación, Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Malaga, Spain
| | - Bernard Fromenty
- INSERM, Univ Rennes, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, F-35000 Rennes, France
| | - Michel Kranendonk
- Center for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculty of Medical Sciences, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Francisco Javier Cubero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Leonard J Nelson
- Center for Regenerative Medicine, Institute for Regenerative and Repair, The University of Edinburgh, Edinburgh, UK, EH16 4UU; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Faraday Building, Colin Maclaurin Road, EH9 3 DW, Scotland, UK; Institute of Biological Chemistry, Biophysics and Bioengineering (IB3), School of Engineering and Physical Sciences (EPS), Heriot-Watt University, Edinburgh EH12 2AS, Scotland, UK.
| |
Collapse
|
8
|
An improved TK-NOG mouse as a novel platform for humanized liver that overcomes limitations in both male and female animals. Drug Metab Pharmacokinet 2021; 42:100410. [PMID: 34839181 DOI: 10.1016/j.dmpk.2021.100410] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/10/2021] [Accepted: 06/07/2021] [Indexed: 11/21/2022]
Abstract
We developed a novel immunodeficient NOG mouse expressing HSVtk mutant clone 30 cDNA under the control of mouse transthyretin gene enhancer/promoter (NOG-TKm30) to acquire fertility in males and high inducibility of liver injury in females. Maximum human albumin levels (approx. 15 mg/mL plasma) in both male and female NOG-TKm30 mice engrafted with human hepatocytes (humanized liver mice) were observed 8-12 weeks after transplantation. Immunohistochemical analyses revealed abundant expression of major human cytochrome P450 (CYP) enzymes (CYP1A2, CYP2C9, CYP2D6, CYP2E1, and CYP3A4) in reconstituted liver with original zonal distribution. In vivo drug-drug interactions were observed in humanized liver mice as decreased area under the curve of midazolam (CYP3A4/5 substrate) and omeprazole (CYP3A4/5 and CYP2C19 substrate) after oral administration of rifampicin. Furthermore, we developed a pregnant model for evaluating prenatal exposure to drugs. The detection of thalidomide metabolites in the fetuses of pregnant humanized liver mice indicates that the novel TK model can be used for developmental toxicity studies requiring the assessment of human drug metabolism. These results suggest that the limitations of traditional TK-NOG mice can be addressed using NOG-TKm30 mice, which constitute a novel platform for humanized liver for both in vivo and in vitro studies.
Collapse
|
9
|
Kato S, Shah A, Plesescu M, Miyata Y, Bolleddula J, Chowdhury S, Zhu X. Prediction of Human Disproportionate and Biliary Excreted Metabolites Using Chimeric Mice with Humanized Liver. Drug Metab Dispos 2020; 48:934-943. [PMID: 32665417 DOI: 10.1124/dmd.120.000128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/07/2020] [Indexed: 11/22/2022] Open
Abstract
The PXB-mouse is potentially a useful in vivo model to predict human hepatic metabolism and clearance. Four model compounds, [14C]desloratadine, [3H]mianserin, cyproheptadine, and [3H]carbazeran, all reported with disproportionate human metabolites, were orally administered to PXB- or control SCID mice to elucidate the biotransformation of each of them. For [14C]desloratadine in PXB-mice, O-glucuronide of 3-hydroxydesloratadine was observed as the predominant metabolite in both the plasma and urine. Both 3-hydroxydesloratadine and its O-glucuronide were detected as major drug-related materials in the bile, whereas only 3-hydroxydesloratadine was detected in the feces, suggesting that a fraction of 3-hydroxydesloratadine in feces was derived from deconjugation of its O-glucuronide by gut microflora. This information can help understand the biliary clearance mechanism of a drug and may fill the gap in a human absorption, distribution, metabolism, and excretion study, in which the bile samples are typically not available. The metabolic profiles in PXB-mice were qualitatively similar to those reported in humans in a clinical study in which 3-hydroxydesloratadine and its O-glucuronide were major and disproportionate metabolites compared with rat, mouse, and monkey. In the control SCID mice, neither of the metabolites was detected in any matrix. Similarly, for the other three compounds, all human specific or disproportionate metabolites were detected at a high level in PXB-mice, but they were either minimally observed or not observed in the control mice. Data from these four compounds indicate that studies in PXB-mice can help predict the potential for the presence of human disproportionate metabolites (relative to preclinical species) prior to conducting clinical studies and understand the biliary clearance mechanism of a drug. SIGNIFICANCE STATEMENT: Studies in PXB-mice have successfully predicted the human major and disproportionate metabolites compared with preclinical safety species for desloratadine, mianserin, cyproheptadine, and carbazeran. In addition, biliary excretion data from PXB-mice can help illustrate the human biliary clearance mechanism of a drug.
Collapse
Affiliation(s)
- Suguru Kato
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (S.K., A.S., M.P., J.B., S.C., X.Z.) and Research Planning and Business Development, PhoenixBio USA Corporation, New York City, New York (Y.M.)
| | - Abhi Shah
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (S.K., A.S., M.P., J.B., S.C., X.Z.) and Research Planning and Business Development, PhoenixBio USA Corporation, New York City, New York (Y.M.)
| | - Mihaela Plesescu
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (S.K., A.S., M.P., J.B., S.C., X.Z.) and Research Planning and Business Development, PhoenixBio USA Corporation, New York City, New York (Y.M.)
| | - Yoshinari Miyata
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (S.K., A.S., M.P., J.B., S.C., X.Z.) and Research Planning and Business Development, PhoenixBio USA Corporation, New York City, New York (Y.M.)
| | - Jayaprakasam Bolleddula
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (S.K., A.S., M.P., J.B., S.C., X.Z.) and Research Planning and Business Development, PhoenixBio USA Corporation, New York City, New York (Y.M.)
| | - Swapan Chowdhury
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (S.K., A.S., M.P., J.B., S.C., X.Z.) and Research Planning and Business Development, PhoenixBio USA Corporation, New York City, New York (Y.M.)
| | - Xiaochun Zhu
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (S.K., A.S., M.P., J.B., S.C., X.Z.) and Research Planning and Business Development, PhoenixBio USA Corporation, New York City, New York (Y.M.)
| |
Collapse
|
10
|
Yamasaki C, Ishida Y, Yanagi A, Yoshizane Y, Kojima Y, Ogawa Y, Kageyama Y, Iwasaki Y, Ishida S, Chayama K, Tateno C. Culture density contributes to hepatic functions of fresh human hepatocytes isolated from chimeric mice with humanized livers: Novel, long-term, functional two-dimensional in vitro tool for developing new drugs. PLoS One 2020; 15:e0237809. [PMID: 32915792 PMCID: PMC7485858 DOI: 10.1371/journal.pone.0237809] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/03/2020] [Indexed: 01/25/2023] Open
Abstract
Chimeric mice with humanized livers are considered a useful animal model for predicting human (h-) drug metabolism and toxicity. In this study, the characteristics of fresh h-hepatocytes (cFHHs, PXB-cells®) isolated from chimeric mice (PXB-mice®) were evaluated in vitro to confirm their utility for drug development. cFHHs cultured at high density (2.13 × 105 cells/cm2) displayed stable production of h-albumin and cytochrome P450 (CYP) 3A activities for at least 21 days. The mRNA expression levels of 10 of 13 CYP, UDP-glucuronosyltransferase (UGT), and transporters were maintained at >10% of the levels of freshly isolated cFHHs after 21 days. From 1 week, many bile canaliculi were observed between cFHHs, and the accumulation of the multidrug resistance-associated protein and bile salt export pump substrates in these bile canaliculi was clearly inhibited by cyclosporin A. Microarray analysis of cFHHs cultured at high density and at low density (0.53 × 105 cells/cm2) revealed that high density culture maintained high expressions of some transcription factors (HNF4α, PXR, and FXR) perhaps involved in the high CYP, UGT and transporter gene expressions of cFHHs. These results strongly suggest that cFHHs could be a novel in vitro tool for drug development studies.
Collapse
Affiliation(s)
| | - Yuji Ishida
- PhoenixBio Co., Ltd., Higashi-Hiroshima, Hiroshima, Japan
- Research Center for Hepatology and Gastroenterology, Hiroshima University, Hiroshima, Japan
| | - Ami Yanagi
- PhoenixBio Co., Ltd., Higashi-Hiroshima, Hiroshima, Japan
| | | | - Yuha Kojima
- PhoenixBio Co., Ltd., Higashi-Hiroshima, Hiroshima, Japan
| | - Yuko Ogawa
- PhoenixBio Co., Ltd., Higashi-Hiroshima, Hiroshima, Japan
| | | | - Yumiko Iwasaki
- PhoenixBio Co., Ltd., Higashi-Hiroshima, Hiroshima, Japan
| | - Seiichi Ishida
- Department of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Higashihiroshima, Hiroshima, Japan
| | - Chise Tateno
- PhoenixBio Co., Ltd., Higashi-Hiroshima, Hiroshima, Japan
- Research Center for Hepatology and Gastroenterology, Hiroshima University, Hiroshima, Japan
- * E-mail:
| |
Collapse
|
11
|
Miyamoto M, Kosugi Y, Iwasaki S, Chisaki I, Nakagawa S, Amano N, Hirabayashi H. Characterization of plasma protein binding in two mouse models of humanized liver, PXB mouse and humanized TK-NOG mouse. Xenobiotica 2020; 51:51-60. [PMID: 32779988 DOI: 10.1080/00498254.2020.1808735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The unbound fractions in plasma (f up) in two mouse models of humanized liver mice, PXB and humanized TK-NOG mice, were compared with human f up values using equilibrium dialysis method. A good relationship between f up values obtained from PXB mice and humans was observed; the f up of 34/39 compounds (87.2%) in PXB mice were within 3-fold of human f up. In contrast, a weak correlation was observed between human and humanized TK-NOG mouse f up values; the f up of 15/24 compounds (62.5%) in humanized TK-NOG mice were within 3-fold of human f up. As different profiles of plasma protein binding (PPB) profiles were observed between PXB and humanized TK-NOG mice, f up evaluation is necessary in each mouse model to utilize these humanized liver mice for pharmacological, drug-drug interaction (DDI), and toxicity studies. The unbound fraction in the mixed plasma of human and SCID mouse plasma (85:15) was well correlated with f up in PXB mice (38/39 compounds within a 3-fold). Thus, this artificial PXB mouse plasma could be used to evaluate PPB.
Collapse
Affiliation(s)
- Maki Miyamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Yohei Kosugi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Shinji Iwasaki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Ikumi Chisaki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Sayaka Nakagawa
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Nobuyuki Amano
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| |
Collapse
|
12
|
Lu C, Di L. In vitro
and
in vivo
methods to assess pharmacokinetic drug– drug interactions in drug discovery and development. Biopharm Drug Dispos 2020; 41:3-31. [DOI: 10.1002/bdd.2212] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/27/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Chuang Lu
- Department of DMPKSanofi Company Waltham MA 02451
| | - Li Di
- Pharmacokinetics, Dynamics and MetabolismPfizer Worldwide Research & Development Groton CT 06340
| |
Collapse
|
13
|
Shehu AI, Ma X. Pregnane X receptor in drug-induced liver injury: Friend or foe? LIVER RESEARCH 2018. [DOI: 10.1016/j.livres.2018.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
14
|
Bissig KD, Han W, Barzi M, Kovalchuk N, Ding L, Fan X, Pankowicz FP, Zhang QY, Ding X. P450-Humanized and Human Liver Chimeric Mouse Models for Studying Xenobiotic Metabolism and Toxicity. Drug Metab Dispos 2018; 46:1734-1744. [PMID: 30093418 PMCID: PMC6199624 DOI: 10.1124/dmd.118.083303] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023] Open
Abstract
Preclinical evaluation of drug candidates in experimental animal models is an essential step in drug development. Humanized mouse models have emerged as a promising alternative to traditional animal models. The purpose of this mini-review is to provide a brief survey of currently available mouse models for studying human xenobiotic metabolism. Here, we describe both genetic humanization and human liver chimeric mouse models, focusing on the advantages and limitations while outlining their key features and applications. Although this field of biomedical science is relatively young, these humanized mouse models have the potential to transform preclinical drug testing and eventually lead to a more cost-effective and rapid development of new therapies.
Collapse
Affiliation(s)
- Karl-Dimiter Bissig
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Weiguo Han
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Mercedes Barzi
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Nataliia Kovalchuk
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Liang Ding
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Xiaoyu Fan
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Francis P Pankowicz
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Qing-Yu Zhang
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Xinxin Ding
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| |
Collapse
|
15
|
Characterization of CYP2C Induction in Cryopreserved Human Hepatocytes and Its Application in the Prediction of the Clinical Consequences of the Induction. J Pharm Sci 2018; 107:2479-2488. [DOI: 10.1016/j.xphs.2018.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/08/2018] [Accepted: 05/16/2018] [Indexed: 12/19/2022]
|
16
|
Garimella T, Tao X, Sims K, Chang YT, Rana J, Myers E, Wind-Rotolo M, Bhatnagar R, Eley T, LaCreta F, AbuTarif M. Effects of a Fixed-Dose Co-Formulation of Daclatasvir, Asunaprevir, and Beclabuvir on the Pharmacokinetics of a Cocktail of Cytochrome P450 and Drug Transporter Substrates in Healthy Subjects. Drugs R D 2018; 18:55-65. [PMID: 29255971 PMCID: PMC5833906 DOI: 10.1007/s40268-017-0222-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background A fixed-dose combination of daclatasvir (DCV; hepatitis C virus NS5A inhibitor), asunaprevir (ASV; non-structural protein 3 inhibitor), and beclabuvir (BCV; non-structural protein 5B inhibitor) is approved in Japan for hepatitis C virus genotype 1. Objective The objective of this study was to assess the combination’s drug–drug interaction potential in vivo using a validated cocktail of eight cytochrome P450 (CYP) and transporter probes. Methods We conducted an open-label single-sequence study in healthy adults (n = 20) given single-dose caffeine (CYP1A2 substrate), metoprolol (CYP2D6), flurbiprofen (CYP2C9), montelukast (CYP2C8), omeprazole (CYP2C19), midazolam (CYP3A4), digoxin (P-glycoprotein), and pravastatin (organic anion-transporting polypeptide), alone or with steady-state twice-daily DCV/ASV/BCV 30/200/75 mg (with or without additional BCV 75 mg to adjust for higher exposure in hepatitis C virus infection). Results Daclatasvir/asunaprevir/beclabuvir did not affect CYP1A2, CYP2C8, or CYP2C9; the probe maximum observed concentration and area under the concentration–time curve extrapolated to infinite time geometric mean ratios and 90% confidence intervals were all within the 0.8–1.25 bioequivalence range. Beclabuvir showed moderate dose-dependent CYP2C19 induction; omeprazole maximum observed concentration and area under the concentration–time curve from 0 to the last quantifiable concentration were lower with additional BCV [geometric mean ratio 0.36 (90% confidence interval 0.23–0.55) and 0.34 (0.25–0.46), respectively] than without [0.57 (0.42–0.78), 0.48 (0.39–0.59)]. Weak-to-moderate CYP3A4 induction was observed, plus weak CYP2D6, P-glycoprotein, and organic anion-transporting polypeptide inhibition [maximum observed concentration and area under the concentration–time curve extrapolated to infinite time without additional BCV: midazolam 0.57 (0.50–0.65), 0.53 (0.47–0.60); metoprolol 1.40 (1.20–1.64), 1.71 (1.49–1.97); digoxin 1.23 (1.12–1.35), 1.23 (1.17–1.29); pravastatin 2.01 (1.63–2.47), 1.68 (1.43–1.97)]. Conclusions No dose adjustments with DCV/ASV/BCV are indicated for CYP1A2, CYP2C8, CYP2C9, or P-glycoprotein substrates. CYP3A4, CYP2D6, and OATP substrates should be co-administered with caution. Co-administration with agents solely metabolized by CYP2C19 is not recommended. Electronic supplementary material The online version of this article (10.1007/s40268-017-0222-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tushar Garimella
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA.
| | - Xiaolu Tao
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Karen Sims
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Yi-Ting Chang
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Jignasa Rana
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Elsa Myers
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | | | | | - Timothy Eley
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Frank LaCreta
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Malaz AbuTarif
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| |
Collapse
|
17
|
Sterol 27-hydroxylase gene dosage and the antiatherosclerotic effect of Rifampicin in mice. Biosci Rep 2018; 38:BSR20171162. [PMID: 29191818 PMCID: PMC5784176 DOI: 10.1042/bsr20171162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/03/2017] [Accepted: 11/29/2017] [Indexed: 11/17/2022] Open
Abstract
Sterol 27-hydroxylase (CYP27A1) catalyzes the hydroxylation of cholesterol to 27-hydroxycholesterol (27-OHC) and regulates cholesterol homeostasis. In Cyp27a1/ Apolipoprotein E (ApoE) double knockout (KO) mice fed with Western diet (WD), the atherosclerotic phenotype found in ApoE KO mice was reversed. As protective mechanism, up-regulation of Cyp3a11 and Cyp7a1 was proposed. Cyp27a1 heterozygote/ApoE KO (het) mice, with reduced Cyp27a1 expression and normal levels of Cyp7a1 and Cyp3a11, developed more severe lesions than ApoE KO mice. To analyze the contribution of Cyp3a11 to the protection of atherosclerosis development, Cyp3a11 was induced by Rifampicin (RIF) in ApoE KO and het mice. Males were fed with WD and treated daily with RIF (10 mg/kg ip) or vehicle for 4 weeks. Atherosclerosis was quantified in the aortic valve. Plasma lipids and 27-hydroxycholesterol (27-OHC), expression of cytochromes P450 and genes involved in cholesterol transport and bile acids (BAs) signaling in liver and intestine, and intestinal cholesterol absorption were analyzed. RIF increased expression of hepatic but not intestinal Cyp3a11 4-fold in both genotypes. In ApoE KO mice treated with RIF, we found a 2-fold decrease in plasma cholesterol, and a 2-fold increase in high-density lipoprotein/low-density lipoprotein ratio and CY27A1 activity. Intestinal cholesterol absorption remained unchanged and atherosclerotic lesions decreased approximately 3-fold. In het mice, RIF had no effect on plasma lipids composition, CYP27A1 activity, and atherosclerotic plaque development, despite a reduction in cholesterol absorption. In conclusion, the antiatherogenic effect of Cyp3a11 induction by RIF was also dependent on Cyp27a1 expression.
Collapse
|
18
|
Chimeric mice with humanized liver: Application in drug metabolism and pharmacokinetics studies for drug discovery. Drug Metab Pharmacokinet 2018; 33:31-39. [DOI: 10.1016/j.dmpk.2017.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/23/2017] [Accepted: 11/01/2017] [Indexed: 11/21/2022]
|
19
|
Uchida M, Tajima Y, Kakuni M, Kageyama Y, Okada T, Sakurada E, Tateno C, Hayashi R. Organic Anion-Transporting Polypeptide (OATP)-Mediated Drug-Drug Interaction Study between Rosuvastatin and Cyclosporine A in Chimeric Mice with Humanized Liver. Drug Metab Dispos 2018; 46:11-19. [PMID: 29051147 DOI: 10.1124/dmd.117.075994] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 10/13/2017] [Indexed: 02/13/2025] Open
Abstract
The influence of transporters on the pharmacokinetics of drugs is being increasingly recognized, and DDIs via transporters may be a risk factor for adverse events. Cyclosporine A, a strong OATP inhibitor, has been reported to increase the systemic exposure of rosuvastatin, an OATP substrate, by 7.1-fold in clinical studies. PXB mice are chimeric mice with humanized livers that are highly repopulated with human hepatocytes and have been widely used for drug discovery in drug metabolism and pharmacokinetics studies. In the present study, we examined in vivo and in vitro DDIs between rosuvastatin and cyclosporine A in PXB mice and fresh human hepatocytes (PXB cells) obtained from PXB mice. We initially investigated the active transport of rosuvastatin into PXB cells, and found concentration-dependent uptake with a Michaelis-Menten constant value of 4.0 μmol/l and a Vmax value of 4.63 pmol/min per 106 cells. Cyclosporine A inhibited the uptake of rosuvastatin with an IC50 value of 0.21 μmol/l. We then examined in vivo DDIs, and the exposure of orally administered rosuvastatin increased by 3.3-fold and 11-fold in PXB mice pretreated with 10 and 50 mg/kg cyclosporine A, whereas it increased by 2.5-fold and 6.2-fold when rosuvastatin was administered intravenously, in studies that were conducted for considering gastrointestinal DDIs. The liver-to-blood concentration ratio of rosuvastatin was dose-dependently decreased by pretreatment with cyclosporine A in PXB mice and SCID mice. Observed DDIs in vivo were considered to be reasonable based on the estimated concentrations of cyclosporine A at the inlet to the liver and in the liver tissues of both mice. In conclusion, our results indicate that PXB mice might be a useful tool for predicting human OATP-mediated DDIs in drug discovery, and its limitation due to the differences of gastrointestinal condition from human should also be considered.
Collapse
Affiliation(s)
- Masashi Uchida
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Yoriko Tajima
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Masakazu Kakuni
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Yutaka Kageyama
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Taro Okada
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Eri Sakurada
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Chise Tateno
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Ryoji Hayashi
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| |
Collapse
|
20
|
CRISPR knockout rat cytochrome P450 3A1/2 model for advancing drug metabolism and pharmacokinetics research. Sci Rep 2017; 7:42922. [PMID: 28218310 PMCID: PMC5317174 DOI: 10.1038/srep42922] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/16/2017] [Indexed: 12/16/2022] Open
Abstract
Cytochrome P450 (CYP) 3A accounts for nearly 30% of the total CYP enzymes in the human liver and participates in the metabolism of over 50% of clinical drugs. Moreover, CYP3A plays an important role in chemical metabolism, toxicity, and carcinogenicity. New animal models are needed to investigate CYP3A functions, especially for drug metabolism. In this report, Cyp3a1/2 double knockout (KO) rats were generated by CRISPR-Cas9 technology, and then were characterized for viability and physiological status. The Cyp3a1/2 double KO rats were viable and fertile, and had no obvious physiological abnormities. Compared with the wild-type (WT) rat, Cyp3a1/2 expression was completely absent in the liver of the KO rat. In vitro and in vivo metabolic studies of the CYP3A1/2 substrates indicated that CYP3A1/2 was functionally inactive in double KO rats. The Cyp3a1/2 double KO rat model was successfully generated and characterized. The Cyp3a1/2 KO rats are a novel rodent animal model that will be a powerful tool for the study of the physiological and pharmacological roles of CYP3A, especially in drug and chemical metabolism in vivo.
Collapse
|
21
|
Clinical Exposure Boost Predictions by Integrating Cytochrome P450 3A4-Humanized Mouse Studies With PBPK Modeling. J Pharm Sci 2016; 105:1398-404. [PMID: 27019957 DOI: 10.1016/j.xphs.2016.01.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 01/01/2023]
Abstract
NVS123 is a poorly water-soluble protease 56 inhibitor in clinical development. Data from in vitro hepatocyte studies suggested that NVS123 is mainly metabolized by CYP3A4. As a consequence of limited solubility, NVS123 therapeutic plasma exposures could not be achieved even with high doses and optimized formulations. One approach to overcome NVS123 developability issues was to increase plasma exposure by coadministrating it with an inhibitor of CYP3A4 such as ritonavir. A clinical boost effect was predicted by using physiologically based pharmacokinetic (PBPK) modeling. However, initial boost predictions lacked sufficient confidence because a key parameter, fraction of drug metabolized by CYP3A4 (fmCYP3A4), could not be estimated with accuracy on account of disconnects between in vitro and in vivo preclinical data. To accurately estimate fmCYP3A4 in human, an in vivo boost effect study was conducted using CYP3A4-humanized mouse model which showed a 33- to 56-fold exposure boost effect. Using a top-down approach, human fmCYP3A4 for NVS123 was estimated to be very high and included in the human PBPK modeling to support subsequent clinical study design. The combined use of the in vivo boost study in CYP3A4-humanized mouse model mice along with PBPK modeling accurately predicted the clinical outcome and identified a significant NVS123 exposure boost (∼42-fold increase) with ritonavir.
Collapse
|
22
|
Up-Regulation of CYP2C19 Expression by BuChang NaoXinTong via PXR Activation in HepG2 Cells. PLoS One 2016; 11:e0160285. [PMID: 27467078 PMCID: PMC4965156 DOI: 10.1371/journal.pone.0160285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 07/15/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cytochrome P450 2C19 (CYP2C19) is an important drug-metabolizing enzyme (DME), which is responsible for the biotransformation of several kinds of drugs such as proton pump inhibitors, platelet aggregation inhibitors and antidepressants. Previous studies showed that Buchang NaoXinTong capsules (NXT) increased the CYP2C19 metabolic activity in vitro and enhanced the antiplatelet effect of clopidogrel in vivo. However, the underlying molecular mechanism remained unclear. In the present study, we examined whether Pregnane X receptor (PXR) plays a role in NXT-mediated regulation of CYP2C19 expression. METHODS We applied luciferase assays, real-time quantitative PCR (qPCR), Western blotting and cell-based analysis of metabolic activity experiments to investigate the NXT regulatory effects on the CYP2C19 promoter activity, the mRNA/ protein expression and the metabolic activity. RESULTS Our results demonstrated that NXT significantly increased the CYP2C19 promoter activity when co-transfected with PXR in HepG2 cells. Mutations in PXR responsive element abolished the NXT inductive effects on the CYP2C19 promoter transcription. Additionally, NXT incubation (150 and 250μg/mL) also markedly up-regulated endogenous CYP2C19 mRNA and protein levels in PXR-transfected HepG2 cells. Correspondingly, NXT leaded to a significant enhancement of the CYP2C19 catalytic activity in PXR-transfected HepG2 cells. CONCLUSION In summary, this is the first study to suggest that NXT could induce CYP2C19 expression via PXR activation.
Collapse
|
23
|
Kamimura H, Ito S. Assessment of chimeric mice with humanized livers in new drug development: generation of pharmacokinetics, metabolism and toxicity data for selecting the final candidate compound. Xenobiotica 2015; 46:557-69. [DOI: 10.3109/00498254.2015.1091113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
24
|
Scheer N, Wilson ID. A comparison between genetically humanized and chimeric liver humanized mouse models for studies in drug metabolism and toxicity. Drug Discov Today 2015; 21:250-63. [PMID: 26360054 DOI: 10.1016/j.drudis.2015.09.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/07/2015] [Accepted: 09/01/2015] [Indexed: 12/12/2022]
Abstract
Mice that have been genetically humanized for proteins involved in drug metabolism and toxicity and mice engrafted with human hepatocytes are emerging and promising in vivo models for an improved prediction of the pharmacokinetic, drug-drug interaction and safety characteristics of compounds in humans. The specific advantages and disadvantages of these models should be carefully considered when using them for studies in drug discovery and development. Here, an overview on the corresponding genetically humanized and chimeric liver humanized mouse models described to date is provided and illustrated with examples of their utility in drug metabolism and toxicity studies. We compare the strength and weaknesses of the two different approaches, give guidance for the selection of the appropriate model for various applications and discuss future trends and perspectives.
Collapse
Affiliation(s)
| | - Ian D Wilson
- Imperial College London, South Kensington, London SW7 2AZ, UK.
| |
Collapse
|
25
|
Watanabe A, Watari R, Ogawa K, Shimizu R, Tanaka Y, Takai N, Nezasa KI, Yamaguchi Y. Using improved serial blood sampling method of mice to study pharmacokinetics and drug-drug interaction. J Pharm Sci 2014; 104:955-961. [PMID: 25452230 DOI: 10.1002/jps.24236] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 09/22/2014] [Accepted: 10/08/2014] [Indexed: 11/06/2022]
Abstract
In pharmacokinetic evaluation of mice, using serial sampling methods rather than a terminal blood sampling method could reduce the number of animals needed and lead to more reliable data by excluding individual differences. In addition, using serial sampling methods can be valuable for evaluation of the drug-drug interaction (DDI) potential of drug candidates. In this study, we established an improved method for serially sampling the blood from one mouse by only one incision of the lateral tail vein, and investigated whether our method could be adapted to pharmacokinetic and DDI studies. After intravenous and oral administration of ibuprofen and fexofenadine (BCS class II and III), the plasma concentration and pharmacokinetic parameters were evaluated by our method and a terminal blood sampling method, with the result that both methods gave comparable results (ibuprofen: 63.8 ± 4.0% and 64.4%, fexofenadine: 6.5 ± 0.7% and 7.9%, respectively, in bioavailability). In addition, our method could be adapted to DDI study for cytochrome P450 and organic anion transporting polypeptide inhibition. These results demonstrate that our method can be useful for pharmacokinetic evaluation from the perspective of reliable data acquisition as well as easy handling and low stress to mice and improve the quality of pharmacokinetic and DDI studies.
Collapse
Affiliation(s)
- Ayahisa Watanabe
- Drug Metabolism and Pharmacokinetics, Research Laboratory for Development, Shionogi & Co., Ltd. Toyonaka, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Jaiswal S, Sharma A, Shukla M, Vaghasiya K, Rangaraj N, Lal J. Novel pre-clinical methodologies for pharmacokinetic drug-drug interaction studies: spotlight on "humanized" animal models. Drug Metab Rev 2014; 46:475-93. [PMID: 25270219 DOI: 10.3109/03602532.2014.967866] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Poly-therapy is common due to co-occurrence of several ailments in patients, leading to the elevated possibility of drug-drug interactions (DDI). Pharmacokinetic DDI often accounts for severe adverse drug reactions in patients resulting in withdrawal of drug from the market. Hence, the prediction of DDI is necessary at pre-clinical stage of drug development. Several human tissue and cell line-based in vitro systems are routinely used for screening metabolic and transporter pathways of investigational drugs and for predicting their clinical DDI potentials. However, ample constraints are associated with the in vitro systems and sometimes in vitro-in vivo extrapolation (IVIVE) fail to assess the risk of DDI in clinic. In vitro-in vivo correlation model in animals combined with human in vitro studies may be helpful in better prediction of clinical outcome. Native animal models vary remarkably from humans in drug metabolizing enzymes and transporters, hence, the interpretation of results from animal DDI studies is difficult. With the advent of modern molecular biology and engineering tools, novel pre-clinical animal models, namely, knockout rat/mouse, transgenic rat/mouse with humanized drug metabolizing enzymes and/or transporters and chimeric rat/mouse with humanized liver are developed. These models nearly simulate human-like drug metabolism and help to validate the in vivo relevance of the in vitro human DDI data. This review briefly discusses the application of such novel pre-clinical models for screening various type of DDI along with their advantages and limitations.
Collapse
Affiliation(s)
- Swati Jaiswal
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute , Lucknow , India
| | | | | | | | | | | |
Collapse
|
27
|
Ohtsuki S, Kawakami H, Inoue T, Nakamura K, Tateno C, Katsukura Y, Obuchi W, Uchida Y, Kamiie J, Horie T, Terasaki T. Validation of uPA/SCID mouse with humanized liver as a human liver model: protein quantification of transporters, cytochromes P450, and UDP-glucuronosyltransferases by LC-MS/MS. Drug Metab Dispos 2014; 42:1039-43. [PMID: 24711249 DOI: 10.1124/dmd.114.057646] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Chimeric mice with humanized liver (PXB mice) have been generated by transplantation of urokinase-type plasminogen activator/severe combined immunodeficiency mice with human hepatocytes. The purpose of the present study was to clarify the protein expression levels of metabolizing enzymes and transporters in humanized liver of PXB mice transplanted with hepatocytes from three different donors, and to compare their protein expressions with those of human livers to validate this human liver model. The protein expression levels of metabolizing enzymes and transporters were quantified in microsomal fraction and plasma membrane fraction, respectively, by means of liquid chromatography-tandem mass spectrometry. Protein expression levels of 12 human P450 enzymes, two human UDP-glucuronosyltransferases, eight human ATP binding cassette (ABC) transporters, and eight human solute carrier transporters were determined. The variances of protein expression levels among samples from mice humanized with hepatocytes from all donors were significantly greater than those from samples obtained from mice derived from each individual donor. Compared with the protein expression levels in human livers, all of the quantified metabolizing enzymes and transporters were within a range of 4-fold difference, except for CYP2A6, CYP4A11, bile salt export pump (BSEP), and multidrug resistance protein 3 (MDR3), which showed 4- to 5-fold differences between PXB mouse and human livers. The present study indicates that humanized liver of PXB mice is a useful model of human liver from the viewpoint of protein expression of metabolizing enzymes and transporters, but the results are influenced by the characteristics of the human hepatocyte donor.
Collapse
Affiliation(s)
- Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O., K.N.); Laboratory of Veterinary Pathology, School of Veterinary Medicine, Azabu University, Kanagawa, Japan (H.K., J.K.); Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (T.I, Y.K., W.O., Y.U., T.T.); PhoenixBio, Co., Ltd., Hiroshima, Japan (C.T.); DeThree Research Laboratory Inc., Ibaraki, Japan (T.H.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Comparative study of the effects of antituberculosis drugs and antiretroviral drugs on cytochrome P450 3A4 and P-glycoprotein. Antimicrob Agents Chemother 2014; 58:3168-76. [PMID: 24663015 DOI: 10.1128/aac.02278-13] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Predicting drug-drug interactions (DDIs) related to cytochrome P450 (CYP), such as CYP3A4 and one of the major drug transporters, P-glycoprotein (P-gp), is crucial in the development of future chemotherapeutic regimens to treat tuberculosis (TB) and TB/AIDS coinfection cases. We evaluated the effects of 30 anti-TB drugs, novel candidates, macrolides, and representative antiretroviral drugs on human CYP3A4 activity using a commercially available screening kit for CYP3A4 inhibitors and a human hepatocyte, HepaRG. Moreover, in order to estimate the interactions of these drugs with human P-gp, screening for substrates was performed. For some substrates, P-gp inhibition tests were carried out using P-gp-expressing MDCK cells. As a result, almost all the compounds showed the expected effects on human CYP3A4 both in the in vitro screening and in HepaRG cells. Importantly, the unproven mechanisms of DDIs caused by WHO group 5 drugs, thioamides, and p-aminosalicylic acid were elucidated. Intriguingly, clofazimine (CFZ) exhibited weak inductive effects on CYP3A4 at >0.25 μM in HepaRG cells, while an inhibitory effect was observed at 1.69 μM in the in vitro screening, suggesting that CFZ autoinduces CYP3A4 in the human liver. Our method, based on one of the pharmacokinetics parameters in humans, provides more practical information associated with not only DDIs but also with drug metabolism.
Collapse
|
29
|
Rathod V, Jain S, Nandekar P, Sangamwar AT. Human pregnane X receptor: a novel target for anticancer drug development. Drug Discov Today 2014; 19:63-70. [DOI: 10.1016/j.drudis.2013.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/02/2013] [Accepted: 08/15/2013] [Indexed: 02/07/2023]
|
30
|
Kakuni M, Yamasaki C, Tachibana A, Yoshizane Y, Ishida Y, Tateno C. Chimeric mice with humanized livers: a unique tool for in vivo and in vitro enzyme induction studies. Int J Mol Sci 2013; 15:58-74. [PMID: 24362577 PMCID: PMC3907798 DOI: 10.3390/ijms15010058] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/05/2013] [Accepted: 12/06/2013] [Indexed: 12/04/2022] Open
Abstract
We performed in vivo and in vitro studies to determine the induction of human cytochrome P450 (CYP) using chimeric mice with humanized liver (PXB-mice®) and human hepatocytes isolated from the PXB-mice (PXB-cells), which were derived from the same donor. For the in vivo study, PXB-mice were injected with 3-methylcholanthrene (3-MC, 2 or 20 mg/kg) or rifampicin (0.1 or 10 mg/kg) for four days. For the in vitro study, PXB-cells were incubated with 3-MC (10, 50, or 250 ng/mL) or with rifampicin (5 or 25 μg/mL). The CYP1A1 and 1A2, and CYP3A4 mRNA expression levels increased significantly in the PXB-mouse livers with 20 mg/kg of 3-MC (Cmax, 12.2 ng/mL), and 10 mg/kg rifampicin (Cmax, 6.9 μg/mL), respectively. The CYP1A1 mRNA expression level increased significantly in PXB-cells with 250 ng/mL of 3-MC, indicating lower sensitivity than in vivo. The CYP1A2 and CYP3A4 mRNA expression levels increased significantly with 50 ng/mL of 3-MC, and 5 μg/mL of rifampicin, respectively, which indicated that the sensitivities were similar between in vivo and in vitro studies. In conclusion, PXB-mice and PXB-cells provide a robust model as an intermediate between in vivo and in vitro human metabolic enzyme induction studies.
Collapse
Affiliation(s)
- Masakazu Kakuni
- PhoenixBio Co., Ltd., 3-4-1, Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan; E-Mails: (M.K.); (C.Y.); (A.T.); (Y.Y.); (Y.I.)
| | - Chihiro Yamasaki
- PhoenixBio Co., Ltd., 3-4-1, Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan; E-Mails: (M.K.); (C.Y.); (A.T.); (Y.Y.); (Y.I.)
| | - Asato Tachibana
- PhoenixBio Co., Ltd., 3-4-1, Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan; E-Mails: (M.K.); (C.Y.); (A.T.); (Y.Y.); (Y.I.)
| | - Yasumi Yoshizane
- PhoenixBio Co., Ltd., 3-4-1, Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan; E-Mails: (M.K.); (C.Y.); (A.T.); (Y.Y.); (Y.I.)
| | - Yuji Ishida
- PhoenixBio Co., Ltd., 3-4-1, Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan; E-Mails: (M.K.); (C.Y.); (A.T.); (Y.Y.); (Y.I.)
- Liver Research Project Center, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, Hiroshima 734-8551, Japan
| | - Chise Tateno
- PhoenixBio Co., Ltd., 3-4-1, Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan; E-Mails: (M.K.); (C.Y.); (A.T.); (Y.Y.); (Y.I.)
- Liver Research Project Center, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, Hiroshima 734-8551, Japan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-82-431-0016; Fax: +81-82-431-0017
| |
Collapse
|
31
|
Kitamura S, Sugihara K. Current status of prediction of drug disposition and toxicity in humans using chimeric mice with humanized liver. Xenobiotica 2013; 44:123-34. [PMID: 24329499 DOI: 10.3109/00498254.2013.868062] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
1. Human-chimeric mice with humanized liver have been constructed by transplantation of human hepatocytes into several types of mice having genetic modifications that injure endogenous liver cells. Here, we focus on liver urokinase-type plasminogen activator-transgenic severe combined immunodeficiency (uPA/SCID) mice, which are the most widely used human-chimeric mice. Studies so far indicate that drug metabolism, drug transport, pharmacological effects and toxicological action in these mice are broadly similar to those in humans. 2. Expression of various drug-metabolizing enzymes is known to be different between humans and rodents. However, the expression pattern of cytochrome P450, aldehyde oxidase and phase II enzymes in the liver of human-chimeric mice resembles that in humans, not that in the host mice. 3. Metabolism of various drugs, including S-warfarin, zaleplon, ibuprofen, naproxen, coumarin, troglitazone and midazolam, in human-chimeric mice is mediated by human drug-metabolizing enzymes, not by host mouse enzymes, and thus resembles that in humans. 4. Pharmacological and toxicological effects of various drugs in human-chimeric mice are also similar to those in humans. 5. The current consensus is that chimeric mice with humanized liver are useful to predict drug metabolism catalyzed by cytochrome P450, aldehyde oxidase and phase II enzymes in humans in vivo and in vitro. Some remaining issues are discussed in this review.
Collapse
Affiliation(s)
- Shigeyuki Kitamura
- Department of Environmental Science, Nihon Pharmaceutical University , Saitama , Japan and
| | | |
Collapse
|
32
|
Foster JR, Lund G, Sapelnikova S, Tyrrell DL, Kneteman NM. Chimeric rodents with humanized liver: bridging the preclinical/clinical trial gap in ADME/toxicity studies. Xenobiotica 2013; 44:109-22. [DOI: 10.3109/00498254.2013.867553] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
33
|
Grompe M, Strom S. Mice with human livers. Gastroenterology 2013; 145:1209-14. [PMID: 24042096 DOI: 10.1053/j.gastro.2013.09.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/04/2013] [Accepted: 09/04/2013] [Indexed: 12/28/2022]
Abstract
Animal models are used to study many aspects of human disease and to test therapeutic interventions. However, some very important features of human biology cannot be replicated in animals, even in nonhuman primates or transgenic rodents engineered with human genes. Most human microbial pathogens do not infect animals and the metabolism of many xenobiotics is different between human beings and animals. The advent of transgenic immune-deficient mice has made it possible to generate chimeric animals harboring human tissues and cells, including hepatocytes. The liver plays a central role in many human-specific biological processes and mice with humanized livers can be used to model human metabolism, liver injury, gene regulation, drug toxicity, and hepatotropic infections.
Collapse
Affiliation(s)
- Markus Grompe
- Oregon Stem Cell Center, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon.
| | | |
Collapse
|
34
|
Sanoh S, Ohta S. Chimeric mice transplanted with human hepatocytes as a model for prediction of human drug metabolism and pharmacokinetics. Biopharm Drug Dispos 2013; 35:71-86. [DOI: 10.1002/bdd.1864] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 09/09/2013] [Accepted: 09/21/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Seigo Sanoh
- Graduate School of Biomedical and Health Sciences; Hiroshima University; Hiroshima Japan
| | - Shigeru Ohta
- Graduate School of Biomedical and Health Sciences; Hiroshima University; Hiroshima Japan
| |
Collapse
|
35
|
Huang L, Huang M, Li YH, Li RM, Zeng Y, Kuang SY, Zhang L, Wang YT, Bi HC. Up-regulatation of CYP3A expression through pregnent X receptor by praeruptorin D isolated from Peucedanum praeruptorum Dunn. JOURNAL OF ETHNOPHARMACOLOGY 2013; 148:596-602. [PMID: 23702042 DOI: 10.1016/j.jep.2013.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 03/10/2013] [Accepted: 05/01/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qianhu, the dried roots of Peucedanum praeruptorum DUNN (Umbelliferae), is a well-known traditional Chinese medicinal herb which was officially listed in the Chinese Pharmacopoeia. Praeruptorin D (PD) is one of the major active constituents of Peucedanum praeruptorum Dunn (Qianhu). The Pregnane X receptor (PXR) is an orphan nuclear receptor and plays a pivotal role in the activation of human cytochrome P450 3A4 (CYP3A4) gene. AIM OF THE STUDY The purpose of this study was to investigate the effect of PD on the PXR-mediated transactivation of CYP3A4, and thus to predict potential herb-drug interactions between PD, Qianhu, and the other co-administered drugs that metabolized by CYP3A4. MATERIALS AND METHODS The effect of PD on the Cyp3a11, mPXR mRNA expression in mice primary hepatocytes was measured using real-time PCR. The gene expression, protein expression, and catalytic activity of CYP3A4 in the LS174T cells after transfected with PXR expression plasmids were determined by real-time PCR, Western blot analysis, and LC-MS/MS based CYP3A4 substrate assay. RESULTS The results revealed that the level of Cyp3a11 gene expression in mice primary hepatocytes was significantly increased by PD, but PD cannot induce the mPXR gene expression. On the other hand, CYP3A4 mRNA, protein expression and functional activity in PXR-over-expression LS174T cells were significantly increased by PD through PXR-mediated pathway; conversely, no significant change was found in the untransfected cells. CONCLUSIONS These findings suggest that PD can significantly up-regulate CYP3A4 expression and activity via the PXR-mediated pathway and this should be taken into consideration to predict any potential herb-drug interactions when PD and Peucedanum praeruptorum Dunn are co-administered with other drugs.
Collapse
Affiliation(s)
- Ling Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132# Waihuan Dong Road, University City, Guangzhou 510006, China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
PXR polymorphisms and their impact on pharmacokinetics/pharmacodynamics of repaglinide in healthy Chinese volunteers. Eur J Clin Pharmacol 2013; 69:1917-25. [PMID: 23807564 DOI: 10.1007/s00228-013-1552-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/10/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE CYP3A4 is the main isoform of cytochrome P450 oxidases involved in the metabolism of approximately 60 % drugs, and its expression level is highly variable in human subjects. CYP3A4 is regulated by many transcription factors, among which the pregnane X receptor/steroid and xenobiotic receptor (PXR/SXR, NR1I2) have been identified as the most critical. Genetic polymorphisms (such as SNPs) in PXR may affect the expression level of CYP3A4. Although numerous SNPs have been identified in PXR and have appeared to affect PXR function, their impact on the expression of CYP3A4 in human subjects has not been well studied. Thus, a clinical study in healthy Chinese subjects was conducted to investigate the impact of PXR polymorphisms on repaglinide (an endogenous marker for CYP3A4 activity) pharmacokinetics used alone or in combination with a PXR inducer, flucloxacillin. METHOD Two SNPs, -298A>G and 11193T>C, were identified as the tag SNPs to represent the overall genetic polymorphic profile of PXR. To evaluate the potential functional change of these two SNPs, 24 healthy subjects were recruited in a pharmacokinetics/pharmacodynamics study of repaglinide with or without flucloxacillin. RESULTS The pharmacokinetic parameters including AUC and T1/2 were significantly different among the PXR genotype groups. The SNPs of -298G/G and 11193C/C were found to be associated with a lower PXR activity resulting in reduction of CYP3A4 activity in vivo. After administration of flucloxacillin, a significant drug-drug interaction was observed. The clearance of repagnilide was significantly increased by concomitant flucloxacillin in a genotype dependent manner. The subjects with SNPs of -298G/G and 11193C/C appeared to be less sensitive to flucloxacillin. CONCLUSION Our study results demonstrated for the first time the impact of genetic polymorphisms of PXR on the PK and PD of repaglinide, and showed that subjects with genotype of -298G/G and 11193C/C in PXR has a decreased elimination rate of 3A4/2C8. Furthermore, flucloxacillin was able to induce 3A4/2C8 expression mediated by PXR in a genotype dependent manner.
Collapse
|