1
|
Rijmers J, Retmana IA, Bui V, Arguedas D, Lebre MC, Sparidans RW, Beijnen JH, Schinkel AH. ABCB1 attenuates brain exposure to the KRAS G12C inhibitor opnurasib whereas binding to mouse carboxylesterase 1c influences its plasma exposure. Biomed Pharmacother 2024; 175:116720. [PMID: 38733773 DOI: 10.1016/j.biopha.2024.116720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Opnurasib (JDQ443) is a newly developed oral KRASG12C inhibitor, with a binding mechanism distinct from the registered KRASG12C inhibitors sotorasib and adagrasib. Phase I and II clinical trials for opnurasib in NSCLC are ongoing. We evaluated the pharmacokinetic roles of the ABCB1 (P-gp/MDR1) and ABCG2 (BCRP) efflux and OATP1 influx transporters, and of the metabolizing enzymes CYP3A and CES1 in plasma and tissue disposition of oral opnurasib, using genetically modified cell lines and mouse models. In vitro, opnurasib was potently transported by human (h)ABCB1 and slightly by mouse (m)Abcg2. In Abcb1a/b- and Abcb1a/b;Abcg2-deficient mice, a significant ∼100-fold increase in brain-to-plasma ratios was observed. Brain penetration was unchanged in Abcg2-/- mice. ABCB1 activity in the blood-brain barrier may therefore potentially limit the efficacy of opnurasib against brain metastases. The Abcb1a/b transporter activity could be almost completely reversed by co-administration of elacridar, a dual ABCB1/ABCG2 inhibitor, increasing the brain penetration without any behavioral or postural signs of acute CNS-related toxicity. No significant pharmacokinetic roles of the OATP1 transporters were observed. Transgenic human CYP3A4 did not substantially affect the plasma exposure of opnurasib, indicating that opnurasib is likely not a sensitive CYP3A4 substrate. Interestingly, Ces1-/- mice showed a 4-fold lower opnurasib plasma exposure compared to wild-type mice, whereas no strong effect was seen on the tissue distribution. Plasma Ces1c therefore likely binds opnurasib, increasing its retention in plasma. The obtained pharmacokinetic insights may be useful for further optimization of the clinical efficacy and safety of opnurasib, and might reveal potential drug-drug interaction risks.
Collapse
Affiliation(s)
- Jamie Rijmers
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Irene A Retmana
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands; Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, the Netherlands
| | - Viët Bui
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Davinia Arguedas
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Maria C Lebre
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Rolf W Sparidans
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, the Netherlands
| | - Jos H Beijnen
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands; Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht, the Netherlands; The Netherlands Cancer Institute, Division of Pharmacy and Pharmacology, Amsterdam, the Netherlands
| | - Alfred H Schinkel
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Kobayashi K, Minegishi G, Kuriyama N, Miyajima A, Abe S, Kazuki K, Kazuki Y. Metabolic Disposition of Triazolam and Clobazam in Humanized CYP3A Mice with a Double-Knockout Background of Mouse Cyp2c and Cyp3a Genes. Drug Metab Dispos 2023; 51:174-182. [PMID: 36379710 DOI: 10.1124/dmd.122.001087] [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: 08/18/2022] [Revised: 10/16/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022] Open
Abstract
Knockout (KO) of mouse Cyp3a genes increases the expression of hepatic CYP2C enzymes, which can metabolize triazolam, a typical substrate of human CYP3A. There is still marked formation of 1'-hydroxytriazolam in Cyp3a-KO (3aKO) mice after triazolam dosing. Here, we generated a new model of humanized CYP3A (hCYP3A) mice with a double-KO background of Cyp3a and Cyp2c genes (2c3aKO), and we examined the metabolic profiles of triazolam in wild-type (WT), 2c3aKO, and hCYP3A/2c3aKO mice in vitro and in vivo In vitro studies using liver microsomes showed that the formation of 1'-hydroxytriazolam in 2c3aKO mice was less than 8% of that in WT mice. The formation rate of 1'-hydroxytriazolam in hCYP3A/2c3aKO mice was eightfold higher than that in 2c3aKO mice. In vivo studies showed that area under the curve (AUC) of 1'-hydroxytriazolam in 2c3aKO mice was less than 3% of that in WT mice. The AUC of 1'-hydroxytriazolam in hCYP3A/2c3aKO mice was sixfold higher than that in 2c3aKO mice. These results showed that formation of 1'-hydroxytriazolam was significantly decreased in 2c3aKO mice. Metabolic functions of human CYP3A enzymes were distinctly found in hCYP3A mice with the 2c3aKO background. Moreover, hCYP3A/2c3aKO mice treated with clobazam showed human CYP3A-mediated formation of desmethylclobazam and prolonged elimination of desmethylclobazam, which is found in poor metabolizers of CYP2C19. The novel hCYP3A mouse model without mouse Cyp2c and Cyp3a genes (hCYP3A/2c3aKO) is expected to be useful to evaluate human CYP3A-mediated metabolism in vivo SIGNIFICANT STATEMENT: Humanized CYP3A (hCYP3A/2c3aKO) mice with a background of double knockout (KO) for mouse Cyp2c and Cyp3a genes were generated. Although CYP2C enzymes played a compensatory role in the metabolism of triazolam to 1'-hydroxytriazolam in the previous hCYP3A/3aKO mice with Cyp2c genes, the novel hCYP3A/2c3aKO mice clearly showed functions of human CYP3A enzymes introduced by chromosome engineering technology.
Collapse
Affiliation(s)
- Kaoru Kobayashi
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Genki Minegishi
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Nina Kuriyama
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Atsushi Miyajima
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Satoshi Abe
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Kanako Kazuki
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Yasuhiro Kazuki
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| |
Collapse
|
3
|
Albadry M, Höpfl S, Ehteshamzad N, König M, Böttcher M, Neumann J, Lupp A, Dirsch O, Radde N, Christ B, Christ M, Schwen LO, Laue H, Klopfleisch R, Dahmen U. Periportal steatosis in mice affects distinct parameters of pericentral drug metabolism. Sci Rep 2022; 12:21825. [PMID: 36528753 PMCID: PMC9759570 DOI: 10.1038/s41598-022-26483-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Little is known about the impact of morphological disorders in distinct zones on metabolic zonation. It was described recently that periportal fibrosis did affect the expression of CYP proteins, a set of pericentrally located drug-metabolizing enzymes. Here, we investigated whether periportal steatosis might have a similar effect. Periportal steatosis was induced in C57BL6/J mice by feeding a high-fat diet with low methionine/choline content for either two or four weeks. Steatosis severity was quantified using image analysis. Triglycerides and CYP activity were quantified in photometric or fluorometric assay. The distribution of CYP3A4, CYP1A2, CYP2D6, and CYP2E1 was visualized by immunohistochemistry. Pharmacokinetic parameters of test drugs were determined after injecting a drug cocktail (caffeine, codeine, and midazolam). The dietary model resulted in moderate to severe mixed steatosis confined to periportal and midzonal areas. Periportal steatosis did not affect the zonal distribution of CYP expression but the activity of selected CYPs was associated with steatosis severity. Caffeine elimination was accelerated by microvesicular steatosis, whereas midazolam elimination was delayed in macrovesicular steatosis. In summary, periportal steatosis affected parameters of pericentrally located drug metabolism. This observation calls for further investigations of the highly complex interrelationship between steatosis and drug metabolism and underlying signaling mechanisms.
Collapse
Affiliation(s)
- Mohamed Albadry
- grid.275559.90000 0000 8517 6224Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany ,grid.411775.10000 0004 0621 4712Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkom, Menoufia, Egypt
| | - Sebastian Höpfl
- grid.5719.a0000 0004 1936 9713Institute for Systems Theory and Automatic Control, Faculty of Engineering Design, Production Engineering and Automotive Engineering, University of Stuttgart, Stuttgart, Germany
| | - Nadia Ehteshamzad
- grid.275559.90000 0000 8517 6224Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Matthias König
- grid.7468.d0000 0001 2248 7639Institute for Theoretical Biology, Institute of Biology, Humboldt-University, Berlin, Germany
| | - Michael Böttcher
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstraße 6, 06847 Dessau-Roßlau, Germany
| | - Jasna Neumann
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstraße 6, 06847 Dessau-Roßlau, Germany
| | - Amelie Lupp
- grid.275559.90000 0000 8517 6224Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Olaf Dirsch
- grid.459629.50000 0004 0389 4214Institute of Pathology, Klinikum Chemnitz, Chemnitz, Germany
| | - Nicole Radde
- grid.5719.a0000 0004 1936 9713Institute for Systems Theory and Automatic Control, Faculty of Engineering Design, Production Engineering and Automotive Engineering, University of Stuttgart, Stuttgart, Germany
| | - Bruno Christ
- grid.9647.c0000 0004 7669 9786Cell Transplantation/Molecular Hepatology Lab, Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig Medical Center, Leipzig, Germany
| | - Madlen Christ
- grid.9647.c0000 0004 7669 9786Cell Transplantation/Molecular Hepatology Lab, Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig Medical Center, Leipzig, Germany
| | - Lars Ole Schwen
- grid.428590.20000 0004 0496 8246Fraunhofer MEVIS, Max-Von-Laue-Str. 2, 28359 Bremen, Germany
| | - Hendrik Laue
- grid.428590.20000 0004 0496 8246Fraunhofer MEVIS, Max-Von-Laue-Str. 2, 28359 Bremen, Germany
| | - Robert Klopfleisch
- grid.14095.390000 0000 9116 4836Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Uta Dahmen
- grid.275559.90000 0000 8517 6224Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| |
Collapse
|
4
|
Wang J, Susam MM, Gan C, Sparidans RW, Lebre MC, Beijnen JH, Schinkel AH. P-Glycoprotein (MDR1/ABCB1) Restricts Brain Accumulation of the Novel EGFR Inhibitor EAI045 and Oral Elacridar Coadministration Enhances Its Brain Accumulation and Oral Exposure. Pharmaceuticals (Basel) 2022; 15:ph15091124. [PMID: 36145346 PMCID: PMC9505538 DOI: 10.3390/ph15091124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
EAI045 is a fourth-generation allosteric tyrosine kinase inhibitor (TKI) of the epidermal growth factor receptor (EGFR). It targets T790M and C797S EGFR mutants in the treatment of non-small cell lung cancer (NSCLC). EAI045 and cetuximab combined induce tumor regression in mouse models of EGFR-mutant lung cancer. We investigated the pharmacokinetic roles of the multidrug efflux and uptake transporters ABCB1 (P-gp), ABCG2 (BCRP), and OATP1A/1B, and of the drug-metabolizing enzyme CYP3A in plasma and tissue distribution of EAI045 and its metabolites, using genetically modified mouse models. In vitro, EAI045 was a good transport substrate of human ABCB1. In vivo, oral EAI045 (20 mg/kg) was rapidly absorbed. Relative to wild-type mice, EAI045 brain-to-plasma ratios were increased 3.9-fold in Abcb1a/1b-/- and 4.8-fold in Abcb1a/1b;Abcg2-/- mice. However, in single Abcg2-/- mice they were unchanged. EAI045 oral availability was not markedly altered. Oral coadministration of elacridar, an ABCB1/ABCG2 inhibitor, increased the plasma AUC0–30min and brain-to-plasma ratios of EAI045 by 4.0-fold and 5.4-fold, respectively, in wild-type mice. EAI045 glucuronide showed an increased plasma AUC0–30min and a markedly decreased accumulation and tissue-to-plasma ratio in the small intestinal content when Abcb1a/1b and Abcg2 were absent. A large fraction of oral EAI045 was converted to its hydrolyzed metabolite PIA, but Abcb1a/1b, Abcg2, and Oatp1a/1b had little impact on PIA pharmacokinetics. Mouse Cyp3a knockout or transgenic human CYP3A4 overexpression did not significantly affect oral EAI045 pharmacokinetics. Our results show that blood–brain barrier ABCB1 can markedly limit EAI045 brain accumulation. Moreover, elacridar coadministration can effectively reverse this process.
Collapse
Affiliation(s)
- Jing Wang
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - M. Merve Susam
- Division of Pharmacoepidemiology & Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Changpei Gan
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Rolf W. Sparidans
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Maria C. Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Jos H. Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Division of Pharmacoepidemiology & Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CS Utrecht, The Netherlands
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Slotervaart Hospital, 1066 CX Amsterdam, The Netherlands
| | - Alfred H. Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-20-5122046
| |
Collapse
|
5
|
Hannon SL, Ding X. Assessing cytochrome P450 function using genetically engineered mouse models. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:253-284. [PMID: 35953157 PMCID: PMC10544722 DOI: 10.1016/bs.apha.2022.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The ability to knock out and/or humanize different genes in experimental animals, globally or in cell- and tissue-specific patterns, has revolutionized scientific research in many areas. Genetically engineered mouse models, including knockout models, transgenic models, and humanized models, have played important roles in revealing the in vivo functions of various cytochrome P450 (CYP) enzymes. These functions are very diverse, ranging from the biotransformation of drugs and other xenobiotics, events that often dictate their pharmacokinetic or toxicokinetic properties and the associated therapeutic or adverse actions, to the metabolism of endogenous compounds, such as steroid hormones and other bioactive substances, that may determine susceptibility to many diseases, such as cancer and metabolic diseases. In this review, we provide a comprehensive list of Cyp-knockout, human CYP-transgenic, and CYP-humanized mouse models that target genes in the CYP1-4 gene families, and highlight their utility in assessing the in vivo metabolism, bioactivation, and toxicity of various xenobiotic compounds, including therapeutic agents and chemical carcinogens. We aim to showcase the advantages of utilizing these mouse models for in vivo drug metabolism and toxicology studies, and to encourage and facilitate greater utility of engineered mouse models to further improve our knowledge of the in vivo functions of various P450 enzymes, which is integral to our ability to develop safer and more effective therapeutics and to identify individuals predisposed to adverse drug reactions or environmental diseases.
Collapse
Affiliation(s)
- Sarrah L Hannon
- Department of Pharmacology and Toxicology, Ken R. Coit College of Pharmacy, The University of Arizona, Tucson, AZ, United States
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, Ken R. Coit College of Pharmacy, The University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
6
|
Wu Q, Hu Y, Wang C, Wei W, Gui L, Zeng W, Liu C, Jia W, Miao J, Lan K. Reevaluate In Vitro CYP3A Index Reactions of Benzodiazepines and Steroids between Humans and Dogs. Drug Metab Dispos 2022; 50:741-749. [PMID: 35351776 DOI: 10.1124/dmd.122.000864] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/15/2022] [Indexed: 02/13/2025] Open
Abstract
Cytochrome P450 3A (CYP3A), the most important class of drug-metabolizing enzymes, participates in the metabolism of half of clinically used drugs. The CYP3A index reactions of dogs, one of the most widely used preclinical nonrodent species, are still poorly understood. This work evaluated the activity and selectivity of 10 CYP3A index reactions, including midazolam (MDZ) 1'- and 4-hydroxylation, alprazolam (APZ) and triazolam (TRZ) α- and 4-hydroxylation, testosterone (T) 6β-hydroxylation, lithocholate (LCA) 6α-hydroxylation, deoxycholate (DCA) 1β- and 5β-hydroxylation, with quantitative reaction phenotyping and kinetic analysis in human and canine recombinant CYP enzymes (rCYPs). In human studies, all reactions are reconfirmed as mixed index reactions of CYP3A with minor contributions from non-CYP3A isoforms. In canine studies, all reactions are also primarily catalyzed by CYP3A12 with lower contributions from CYP3A26. However, the canine CYP2B11 appreciably contributes to the hydroxylation of benzodiazepines except for APZ 4-hydroxylation. The canine CYP3A isoforms have lower activity than human isoforms toward T 6β-hydroxylation and LCA 6α-hydroxylation and both substrates undergo non-CYP3A catalyzed side reactions. DCA 1β- and 5β-hydroxylation are validated as the CYP3A index reactions in both humans and dogs with limited non-CYP3A contributions and side reactions. In conclusion, this work provides a comprehensive overview for the selectivity and activity of in vitro CYP3A index reactions in humans and dogs. The validated CYP3A index reactions between humans and dogs may benefit future practices in drug metabolism and drug interaction studies. SIGNIFICANCE STATEMENT: Dogs are one of the most important nonrodent animals with limited studies of cytochrome P450 enzymes than humans. This work provides the most comprehensive quantitative data to date for the selectivity and activity of CYP3A index reactions in humans and dogs. The canine CYP2B11 was found to appreciably contribute to hydroxylation of midazolam, alprazolam and triazolam, the well-known probes for human CYP3A. Deoxycholate 1β- and 5β-hydroxylation are validated as the CYP3A index reactions in both humans and dogs.
Collapse
Affiliation(s)
- QingLiang Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| | - YiTing Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| | - CuiTong Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| | - Wei Wei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| | - LanLan Gui
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| | - WuShuang Zeng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| | - Changxiao Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| | - Wei Jia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| | - Jia Miao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| | - Ke Lan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy (Q.W., Y.H., C.W., W.W., K.L.), and Institute of Clinical Pharmacology, West China Hospital (J.M.), Sichuan University, Chengdu, China; Chengdu Health-Balance Medical Technology Co., Ltd., Chengdu, China (L.G., W.Z., K.L.); State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China (C.L.); and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China (W.J.)
| |
Collapse
|
7
|
F. Martins ML, Heydari P, Li W, Martínez-Chávez A, Venekamp N, Lebre MC, Lucas L, Beijnen JH, Schinkel AH. Drug Transporters ABCB1 (P-gp) and OATP, but not Drug-Metabolizing Enzyme CYP3A4, Affect the Pharmacokinetics of the Psychoactive Alkaloid Ibogaine and its Metabolites. Front Pharmacol 2022; 13:855000. [PMID: 35308219 PMCID: PMC8931498 DOI: 10.3389/fphar.2022.855000] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
The psychedelic alkaloid ibogaine is increasingly used as an oral treatment for substance use disorders, despite being unlicensed in most countries and having reported adverse events. Using wild-type and genetically modified mice, we investigated the impact of mouse (m)Abcb1a/1b and Abcg2 drug efflux transporters, human and mouse OATP drug uptake transporters, and the CYP3A drug-metabolizing complex on the pharmacokinetics of ibogaine and its main metabolites. Following oral ibogaine administration (10 mg/kg) to mice, we observed a rapid and extensive conversion of ibogaine to noribogaine (active metabolite) and noribogaine glucuronide. Mouse Abcb1a/1b, in combination with mAbcg2, modestly restricted the systemic exposure (plasma AUC) and peak plasma concentration (Cmax) of ibogaine. Accordingly, we found a ∼2-fold decrease in the relative recovery of ibogaine in the small intestine with fecal content in the absence of both transporters compared to the wild-type situation. Ibogaine presented good intrinsic brain penetration even in wild-type mice (brain-to-plasma ratio of 3.4). However, this was further increased by 1.5-fold in Abcb1a/1b;Abcg2−/− mice, but not in Abcg2−/− mice, revealing a stronger effect of mAbcb1a/1b in restricting ibogaine brain penetration. The studied human OATP transporters showed no major impact on ibogaine plasma and tissue disposition, but the mOatp1a/1b proteins modestly affected the plasma exposure of ibogaine metabolites and the tissue disposition of noribogaine glucuronide. No considerable role of mouse Cyp3a knockout or transgenic human CYP3A4 overexpression was observed in the pharmacokinetics of ibogaine and its metabolites. In summary, ABCB1, in combination with ABCG2, limits the oral availability of ibogaine, possibly by mediating its hepatobiliary and/or direct intestinal excretion. Moreover, ABCB1 restricts ibogaine brain penetration. Variation in ABCB1/ABCG2 activity due to genetic variation and/or pharmacologic inhibition might therefore affect ibogaine exposure in patients, but only to a limited extent. The insignificant impact of human CYP3A4 and OATP1B1/1B3 transporters may be clinically advantageous for ibogaine and noribogaine use, as it decreases the risks of undesirable drug interactions or interindividual variation related to CYP3A4 and/or OATP activity.
Collapse
Affiliation(s)
| | - Paniz Heydari
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Wenlong Li
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Alejandra Martínez-Chávez
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Nikkie Venekamp
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maria C. Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Luc Lucas
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jos H. Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Alfred H. Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- *Correspondence: Alfred H. Schinkel,
| |
Collapse
|
8
|
Wang Y, Sparidans RW, Potters S, Lebre MC, Beijnen JH, Schinkel AH. ABCB1 and ABCG2, but not CYP3A4 limit oral availability and brain accumulation of the RET inhibitor pralsetinib. Pharmacol Res 2021; 172:105850. [PMID: 34450308 DOI: 10.1016/j.phrs.2021.105850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/02/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE Pralsetinib is an FDA-approved oral small-molecule inhibitor for treatment of rearranged during transfection (RET) proto-oncogene fusion-positive non-small cell lung cancer. We investigated how the efflux transporters ABCB1 and ABCG2, the SLCO1A/1B uptake transporters and the drug-metabolizing enzyme CYP3A influence pralsetinib pharmacokinetics. EXPERIMENTAL APPROACH In vitro, transepithelial pralsetinib transport was assessed. In vivo, pralsetinib (10 mg/kg) was administered orally to relevant genetically modified mouse models. Pralsetinib concentrations in cell medium, plasma samples and organ homogenates were measured using liquid chromatography-tandem mass spectrometry. KEY RESULTS Pralsetinib was efficiently transported by human (h)ABCB1 and mouse (m)Abcg2, but not hACBG2. In vivo, mAbcb1a/1b markedly and mAbcg2 slightly limited pralsetinib brain penetration (6.3-and 1.8-fold, respectively). Testis distribution showed similar results. Abcb1a/1b;Abcg2-/- mice showed 1.5-fold higher plasma exposure, 23-fold increased brain penetration, and 4-fold reduced recovery of pralsetinib in the small intestinal content. mSlco1a/1b deficiency did not affect pralsetinib oral availability or tissue exposure. Oral coadministration of the ABCB1/ABCG2 inhibitor elacridar boosted pralsetinib plasma exposure (1.3-fold) and brain penetration (19.6-fold) in wild-type mice. Additionally, pralsetinib was a modest substrate of mCYP3A, but not of hCYP3A4, which did not noticeably restrict the oral availability or tissue distribution of pralsetinib. CONCLUSIONS AND IMPLICATIONS SLCO1A/1B and CYP3A4 are unlikely to affect the pharmacokinetics of pralsetinib, but ABCG2 and especially ABCB1 markedly limit its brain and testis penetration, as well as oral availability. These effects are mostly reversed by oral coadministration of the ABCB1/ABCG2 inhibitor elacridar. These insights may be useful in the further clinical development of pralsetinib.
Collapse
Affiliation(s)
- Yaogeng Wang
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Rolf W Sparidans
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Sander Potters
- Leiden university, Faculty of Science, Leiden Academic Centre for Drug Research (LACDR), Einsteinweg 55, 2300 RA Leiden, The Netherlands
| | - Maria C Lebre
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Jos H Beijnen
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; The Netherlands Cancer Institute, Department of Pharmacy & Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Alfred H Schinkel
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Li S, Li X, Yuan D, Wang B, Yang R, Zhang M, Li J, Zeng F. Effects of paeoniflorin on the activities and mRNA expression of rat CYP1A2, CYP2C11 and CYP3A1 enzymes in vivo. Xenobiotica 2021; 51:961-967. [PMID: 29160125 DOI: 10.1080/00498254.2017.1404659] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Paeoniflorin is the major constituent in extracts of the paeony root, the purpose of the present study was to assess the effects of paeoniflorin on the activities and mRNA expression of the rat hepatic drug-metabolizing enzymes cytochrome P450 (CYP1A2), CYP2C11 and CYP3A1 in vivo.Sprague-Dawley (SD) male rats were treated with paeoniflorin at the dosage of 25, 50 and 100 mg/kg or 0.9% sodium chloride solution by intragastric administration for 7 days, then were given probe drugs phenacetin (CYP1A2), tolbutamide (CYP2C11), or midazolam (CYP3A1) orally on the eighth day. Blood samples were collected at various times, and the plasma concentrations of the probe drugs were estimated with ultra-high-performance liquid chromatography. The mRNA expression levels of rat hepatic CYP1A2, CYP2C11 and CYP3A1 were analysed with real-time PCR.The pharmacokinetic results indicated that paeoniflorin inhibits the activities of CYP1A2, CYP2C11 and CYP3A1 in vivo. The effect was most pronounced on CYP3A1, according to the United States Food and Drug Administration classification of inhibitors of CYP3A, it reached the category of moderate inhibition. The mRNA expression levels of 3 CYP enzymes were also tended to be inhibited.We conclude that paeoniflorin can inhibit the activities of CYP1A2, CYP2C11 and CYP3A1 in vivo, which may affect the metabolism of drugs that are primarily dependent on these pathways.
Collapse
Affiliation(s)
- Sicong Li
- Sichuan Animal Science Academy, Chengdu, China
| | - Xuting Li
- Sichuan Animal Science Academy, Chengdu, China
| | | | - Bin Wang
- Sichuan Animal Science Academy, Chengdu, China
| | - Rui Yang
- Sichuan Animal Science Academy, Chengdu, China
| | - Min Zhang
- Sichuan Animal Science Academy, Chengdu, China
| | - Jinliang Li
- Sichuan Animal Science Academy, Chengdu, China
| | | |
Collapse
|
10
|
Deguchi S, Shintani T, Harada K, Okamoto T, Takemura A, Hirata K, Ito K, Takayama K, Mizuguchi H. In Vitro Model for a Drug Assessment of Cytochrome P450 Family 3 Subfamily A Member 4 Substrates Using Human Induced Pluripotent Stem Cells and Genome Editing Technology. Hepatol Commun 2021; 5:1385-1399. [PMID: 34430783 PMCID: PMC8369939 DOI: 10.1002/hep4.1729] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 02/23/2021] [Accepted: 03/19/2021] [Indexed: 02/04/2023] Open
Abstract
In drug development, a system for predicting drug metabolism and drug-induced toxicity is necessary to ensure drug safety. Cytochrome P450 family 3 subfamily A member 4 (CYP3A4) is an important drug-metabolizing enzyme expressed in the liver and small intestine, and predicting CYP3A4-mediated drug metabolism and drug-induced toxicity is essential. We previously developed procedures to differentiate human induced pluripotent stem (iPS) cells into hepatocyte-like cells (HLCs) or intestinal epithelial-like cells (IECs) with a fetal phenotype as well as a highly efficient genome editing technology that could enhance the homologous recombination efficiency at any locus, including CYP3A4. By using human iPS cells and our genome editing technology, we generated CYP3A4-knockout (KO) iPS cell-derived HLCs and IECs for the evaluation of CYP3A4-mediated drug metabolism and drug-induced toxicity. CYP3A4 deficiency did not affect pluripotency and hepatic and intestinal differentiation capacities, and CYP3A4 activity was entirely eradicated by CYP3A4 KO. Off-target effects (e.g., inhibition of bile acid excretion) were hardly observed in CYP3A4-KO cells but were observed in CYP3A4 inhibitor-treated (e.g., ketoconazole) cells. To evaluate whether drug-induced hepatotoxicity and enterotoxicity could be predicted using our model, we exposed CYP3A4-KO HLCs and IECs to acetaminophen, amiodarone, desipramine, leflunomide, tacrine, and tolcapone and confirmed that these cells could predict CYP3A4-mediated toxicity. Finally, we examined whether the therapeutic effects of an anti-hepatitis C virus (HCV) drug metabolized by CYP3A4 would be predicted using our model. CYP3A4-KO HLCs were treated with asunaprevir (antiviral drug metabolized by CYP3A4) after HCV infection, and the anti-viral effect was indeed strengthened by CYP3A4 KO. Conclusion: We succeeded in generating a novel evaluation system for prediction of CYP3A4-mediated drug metabolism and drug-induced toxicity.
Collapse
Affiliation(s)
- Sayaka Deguchi
- Laboratory of Biochemistry and Molecular BiologyGraduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan
| | - Tomohiro Shintani
- Laboratory of Biochemistry and Molecular BiologyGraduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan
| | - Kazuo Harada
- Laboratory of Applied Environmental BiologyGraduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan
| | - Toru Okamoto
- Institute for Advanced Co-creation Studies, Research Institute for Microbial DiseasesOsaka UniversityOsakaJapan
| | - Akinori Takemura
- Laboratory of BiopharmaceuticsGraduate School of Pharmaceutical SciencesChiba UniversityChibaJapan
| | - Kazumasa Hirata
- Laboratory of Applied Environmental BiologyGraduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan
| | - Kousei Ito
- Laboratory of BiopharmaceuticsGraduate School of Pharmaceutical SciencesChiba UniversityChibaJapan
| | - Kazuo Takayama
- Laboratory of Biochemistry and Molecular BiologyGraduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan.,Center for Induced Pluripotent Stem Cell Research and ApplicationKyoto UniversityKyotoJapan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular BiologyGraduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan.,Laboratory of Hepatocyte RegulationNational Institutes of Biomedical Innovation, Health and NutritionOsakaJapan.,Global Center for Medical Engineering and InformaticsOsaka UniversityOsakaJapan.,Integrated Frontier Research for Medical Science Division of the Institute for Open and Transdisciplinary Research InitiativesOsaka UniversityOsakaJapan
| |
Collapse
|
11
|
Ly JQ, Wong S, Liu L, Li R, Messick K, Chang JH. Investigating the Utility of Humanized Pregnane X Receptor-Constitutive Androstane Receptor-CYP3A4/7 Mouse Model to Assess CYP3A-Mediated Induction. Drug Metab Dispos 2021; 49:540-547. [PMID: 33863817 DOI: 10.1124/dmd.121.000439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/02/2021] [Indexed: 11/22/2022] Open
Abstract
Clinical induction liability is assessed with human hepatocytes. However, underpredictions in the magnitude of clinical induction have been reported. Unfortunately, in vivo studies in animals do not provide additional insight because of species differences in drug metabolizing enzymes and their regulatory pathways. To circumvent this limitation, transgenic animals expressing human orthologs were developed. The aim of this work was to investigate the utility of mouse models expressing human orthologs of pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 (Tg-Composite) in evaluating clinical induction. Rifampin, efavirenz, and pioglitazone, which were employed to represent strong, moderate, and weak inducers, were administered at multiple doses to Tg-Composite animals. In vivo CYP3A activity was monitored by measuring changes in the exposure of the CYP3A probe substrate triazolam. After the in vivo studies, microsomes were prepared from their livers to measure changes of in vitro CYP3A4 activity. In both in vivo and in vitro, distinction of clinic induction was recapitulated as rifampin yielded the greatest inductive effect followed by efavirenz and pioglitazone. Interestingly, with rifampin, in vivo CYP3A activity was approximately 4-fold higher than in vitro activity. Conversely, there was no difference between in vivo and in vitro CYP3A activity with efavirenz. These findings are consistent with the report that, although rifampin exhibits differential inductive effects between the intestines and liver, efavirenz does not. These data highlight the promise of transgenic models, such as Tg-Composite, to complement human hepatocytes to enhance the translatability of clinical induction as well as become a powerful tool to further study mechanisms of drug disposition. SIGNIFICANCE STATEMENT: Underprediction of the magnitude of clinical induction when using human hepatocytes has been reported, and transgenic models may improve clinical translatability. The work presented here showcases the human orthologs of pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 model, which was able to recapitulate the magnitude of clinical induction and to differentiate tissue-dependent induction observed with rifampin but not with efavirenz. These results not only foreshadow the potential application of such transgenic models in assessing clinical induction but also in further investigation of the mechanism of drug disposition.
Collapse
Affiliation(s)
- Justin Q Ly
- Genentech, Inc., South San Francisco, California (J.Q.L., S.W., L.L., R.L., K.M.), and ORIC Pharmaceuticals, South San Francisco, California (J.H.C.)
| | - Susan Wong
- Genentech, Inc., South San Francisco, California (J.Q.L., S.W., L.L., R.L., K.M.), and ORIC Pharmaceuticals, South San Francisco, California (J.H.C.)
| | - Liling Liu
- Genentech, Inc., South San Francisco, California (J.Q.L., S.W., L.L., R.L., K.M.), and ORIC Pharmaceuticals, South San Francisco, California (J.H.C.)
| | - Ruina Li
- Genentech, Inc., South San Francisco, California (J.Q.L., S.W., L.L., R.L., K.M.), and ORIC Pharmaceuticals, South San Francisco, California (J.H.C.)
| | - Kirsten Messick
- Genentech, Inc., South San Francisco, California (J.Q.L., S.W., L.L., R.L., K.M.), and ORIC Pharmaceuticals, South San Francisco, California (J.H.C.)
| | - Jae H Chang
- Genentech, Inc., South San Francisco, California (J.Q.L., S.W., L.L., R.L., K.M.), and ORIC Pharmaceuticals, South San Francisco, California (J.H.C.)
| |
Collapse
|
12
|
Howell L, Jenkins RE, Lynch S, Duckworth C, Kevin Park B, Goldring C. Proteomic profiling of murine biliary-derived hepatic organoids and their capacity for drug disposition, bioactivation and detoxification. Arch Toxicol 2021; 95:2413-2430. [PMID: 34050779 PMCID: PMC8241807 DOI: 10.1007/s00204-021-03075-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 05/06/2021] [Indexed: 11/26/2022]
Abstract
Hepatic organoids are a recent innovation in in vitro modeling. Initial studies suggest that organoids better recapitulate the liver phenotype in vitro compared to pre-existing proliferative cell models. However, their potential for drug metabolism and detoxification remains poorly characterized, and their global proteome has yet to be compared to their tissue of origin. This analysis is urgently needed to determine what gain-of-function this new model may represent for modeling the physiological and toxicological response of the liver to xenobiotics. Global proteomic profiling of undifferentiated and differentiated hepatic murine organoids and donor-matched livers was, therefore, performed to assess both their similarity to liver tissue, and the expression of drug-metabolizing enzymes and transporters. This analysis quantified 4405 proteins across all sample types. Data are available via ProteomeXchange (PXD017986). Differentiation of organoids significantly increased the expression of multiple cytochrome P450, phase II enzymes, liver biomarkers and hepatic transporters. While the final phenotype of differentiated organoids is distinct from liver tissue, the organoids contain multiple drug metabolizing and transporter proteins necessary for liver function and drug metabolism, such as cytochrome P450 3A, glutathione-S-transferase alpha and multidrug resistance protein 1A. Indeed, the differentiated organoids were shown to exhibit increased sensitivity to midazolam (10–1000 µM) and irinotecan (1–100 µM), when compared to the undifferentiated organoids. The predicted reduced activity of HNF4A and a resulting dysregulation of RNA polymerase II may explain the partial differentiation of the organoids. Although further experimentation, optimization and characterization is needed relative to pre-existing models to fully contextualize their use as an in vitro model of drug-induced liver injury, hepatic organoids represent an attractive novel model of the response of the liver to xenobiotics. The current study also highlights the utility of global proteomic analyses for rapid and accurate evaluation of organoid-based test systems.
Collapse
Affiliation(s)
- Lawrence Howell
- Department of Pharmacology and Therapeutics, MRC Centre of Drug Safety Science, University of Liverpool, The Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Rosalind E Jenkins
- Department of Pharmacology and Therapeutics, MRC Centre of Drug Safety Science, University of Liverpool, The Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Stephen Lynch
- Department of Pharmacology and Therapeutics, MRC Centre of Drug Safety Science, University of Liverpool, The Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Carrie Duckworth
- Department of Pharmacology and Therapeutics, MRC Centre of Drug Safety Science, University of Liverpool, The Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - B Kevin Park
- Department of Pharmacology and Therapeutics, MRC Centre of Drug Safety Science, University of Liverpool, The Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Christopher Goldring
- Department of Pharmacology and Therapeutics, MRC Centre of Drug Safety Science, University of Liverpool, The Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK.
| |
Collapse
|
13
|
Rapid quantification of vincristine in mouse plasma using ESI-LC-MS/MS: Application to pharmacokinetic studies. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1168:122591. [PMID: 33684722 DOI: 10.1016/j.jchromb.2021.122591] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/30/2021] [Accepted: 02/09/2021] [Indexed: 11/20/2022]
Abstract
A simple, rapid, and sensitive LC-MS/MS method for determining concentrations of the anticancer alkaloid vincristine in micro volumes of mouse plasma was developed and validated in positive ion mode. Separation of vincristine and the internal standard [2H3]-vincristine was achieved on an Accucore aQ column with a gradient mobile phase delivered at a flow rate of 0.4 mL/min and a run time of 2.2 min. Calibration curves were linear (r2 > 0.99, n = 8) up to 250 ng/mL, with a lower limit of quantitation of 2.5 ng/mL. The matrix effect and extraction recovery for vincristine were ranging 108-110% and 88.4-107%, respectively. The intra-day and inter-day precision of quality controls tested at 3 different concentrations were always less than 15%, and accuracy ranged from 91.7 to 107%. The method was successfully applied to evaluate the pharmacokinetic profile of vincristine in wild-type and CYP3A-deficient mice in support of a project to provide mechanistic insight into drug-drug interactions and to identify sources of inter-individual pharmacokinetic variability associated with vincristine-induced peripheral neuropathy.
Collapse
|
14
|
Pang X, Tang C, Kong F, Chen M, Chen X. CYP2C and CYP2B Mediated Metabolic Activation of Retrorsine in Cyp3a Knockout Mice. Curr Drug Metab 2020; 21:1040-1051. [DOI: 10.2174/1389200221666201202101715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/04/2020] [Accepted: 10/01/2020] [Indexed: 11/22/2022]
Abstract
Background:
Retrorsine is one of the hepatotoxic pyrrolizidine alkaloids, which could be converted
into a highly reactive metabolite, dehydroretrorsine, by CYP3A, and to a lesser extent by CYP2C and CYP2B.
Objective:
We employed Cyp3a knockout (3AKO) mice to investigate whether the absence of CYP3A could attenuate
dehydroretrorsine formation and the role of CYP2C and CYP2B in the formation.
Methods:
Blood and liver samples were collected after intragastrical administration of 35 mg/kg retrorsine or
saline for seven days in wild-type (WT) and 3AKO mice. Blood pyrrole-protein adducts were semi quantified
by high-performance liquid chromatography/quadrupole time-of-flight mass spectrometry. The formations of
glutathionyl-6,7-dihydro-1-hydroxymethyl-5H-pyrrolizine (GSH-DHP) and the activities of CYP3A, CYP2B
and CYP2C were evaluated in the liver microsomes of WT and 3AKO mice before and after treatment. The
metabolic phenotype of retrorsine was determined in human liver microsomes. The gene and protein expression
of retrorsine metabolism-related CYP450s in the liver was measured by quantitative real-time PCR method and
western blotting method. The serum cytokine level was detected by the ELISA method to reveal the potential
mechanism of Cyp3a, Cyp2b and Cyp2c downregulation.
Results:
After an oral administration of 35 mg/kg retrorsine for seven days, the blood exposures of DHP
adducts between WT and 3AKO mice were similar, consistent with the comparable formation of GSH-DHP in
their liver microsomes. The chemical inhibitor experiment in liver microsomes indicated the predominant role
of CYP3A and CYP2C in GSH-DHP formation in WT and 3AKO mice, respectively. Real-time qPCR analysis
showed that the expressions of Cyp2b10 and Cyp2cs increased 2.3-161-fold in 3AKO mice, which was consistent
with protein changes. The increased CYP2B activity in 3AKO mice supported the potential role of CYP2B
in GSH-DHP formation. After a seven-day treatment of retrorsine, the yields of GSH-DHP were lower than the
untreated ones in both alleles, accompanied by the decreased mRNA of Cyp3a, Cyp2b and Cyp2c. The increased
serum IL6 might mediate the retrorsine-induced downregulation of Cyp450s.
Conclusion:
These data demonstrated the increased transcription of Cyp2c and Cyp2b caused by Cyp3a ablation,
which played a vital role in the metabolic activation of retrorsine, and long-term exposure of retrorsine can
reduce the CYP450 activities.
Collapse
Affiliation(s)
- Xiaoyan Pang
- Centre for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Chongzhuang Tang
- Centre for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Fandi Kong
- Centre for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Meixia Chen
- Centre for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Xiaoyan Chen
- Centre for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| |
Collapse
|
15
|
Ji JZ, Li YF, Jiang LP, Tai T, Ge PX, Mi QY, Zhu T, Xie HG. P-glycoprotein deficiency enhances metabolic activation of and platelet response to clopidogrel through marked up-regulation of Cyp3a11 in mice: Direct evidence for the interplay between P-glycoprotein and Cyp3a. Biochem Pharmacol 2020; 183:114313. [PMID: 33137324 DOI: 10.1016/j.bcp.2020.114313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Variability in P-glycoprotein (P-gp) efflux transporting activity was supposed to be involved in altered intestinal absorption and bioavailability of clopidogrel in patients; however, reliable evidence is still lacking. In this study, we sought to determine whether P-gp could play an important role in the metabolic activation of and platelet response to clopidogrel in mice. Abcb1a/1b knock-out (KO) and wild-type (WT) mice were used to evaluate differences in the intracellular accumulation of clopidogrel in the intestine, liver, and brain tissues and in systemic exposure of clopidogrel and its main metabolites as well as the mechanisms involved. Results indicated that, compared with WT mice, KO mice exhibited an 84% increase in systemic exposure of clopidogrel active thiol metabolite H4 and a 14.5% rise of suppression of ADP-induced platelet integrin αIIbβ3 activation, paralleled by a 41% decrease in systemic exposure of clopidogrel due to enhanced systemic clearance. Furthermore, KO mice displayed a 45% increase in Cyp3a11 but a 23% decrease in Ces1 at their protein levels compared with WT mice. Concurrently, intracellular clopidogrel concentrations in the tissues examined did not differ significantly between KO and WT mice. We conclude that although P-gp does not transport clopidogrel and its major metabolites in mice, P-gp-deficient mice exhibit elevated formation of the active metabolite H4 and enhanced antiplatelet effect of clopidogrel through up-regulation of Cyp3a11 and down-regulation of Ces1, suggesting that P-gp activity may correlate inversely with the formation of H4 and antiplatelet efficacy of clopidogrel in clinical settings due to P-gp and CYP3A4 interplay.
Collapse
Affiliation(s)
- Jin-Zi Ji
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yi-Fei Li
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Li-Ping Jiang
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Ting Tai
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Peng-Xin Ge
- Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qiong-Yu Mi
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Ting Zhu
- Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hong-Guang Xie
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Department of Clinical Pharmacy, Nanjing Medical University School of Pharmacy, Nanjing 211166, China.
| |
Collapse
|
16
|
Gabás‐Rivera C, Jurado‐Ruiz E, Sánchez‐Ortiz A, Romanos E, Martínez‐Beamonte R, Navarro MA, Surra JC, Arnal C, Rodríguez‐Yoldi MJ, Andrés‐Lacueva C, Osada J. Dietary Squalene Induces CytochromesCyp2b10andCyp2c55Independently of Sex, Dose, and Diet in Several Mouse Models. Mol Nutr Food Res 2020; 64:e2000354. [DOI: 10.1002/mnfr.202000354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Clara Gabás‐Rivera
- Departamento Bioquímica y Biología Molecular y Celular Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | | | | | - Eduardo Romanos
- Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50009 Spain
| | - Roberto Martínez‐Beamonte
- Departamento Bioquímica y Biología Molecular y Celular Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - María A. Navarro
- Departamento Bioquímica y Biología Molecular y Celular Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Joaquín C. Surra
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- Departamento de Producción Animal Instituto de Investigación Sanitaria de Aragón (IISA) Escuela Politécnica Superior de Huesca Huesca 22071 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- Departamento de Patología Animal Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - María J. Rodríguez‐Yoldi
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- Departamento de Farmacología y Fisiología Instituto de Investigación Sanitaria de Aragón (IISA) Facultad de Veterinaria Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Cristina Andrés‐Lacueva
- Biomarkers and Nutrimetabolomics Laboratory Department of Nutrition Food Sciences and Gastronomy Faculty of Pharmacy and Food Sciences University of Barcelona Barcelona 08028 Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Jesús Osada
- Departamento Bioquímica y Biología Molecular y Celular Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| |
Collapse
|
17
|
Togao M, Kawakami K, Otsuka J, Wagai G, Ohta-Takada Y, Kado S. Effects of gut microbiota on in vivo metabolism and tissue accumulation of cytochrome P450 3A metabolized drug: Midazolam. Biopharm Drug Dispos 2020; 41:275-282. [PMID: 32562497 PMCID: PMC7497050 DOI: 10.1002/bdd.2244] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/21/2020] [Accepted: 06/03/2020] [Indexed: 12/04/2022]
Abstract
The link between drug‐metabolizing enzymes and gut microbiota is well established. In particular, hepatic cytochrome P450 (CYP) 3A activities are presumed to be affected by gut microbiota. However, there is no direct evidence that the gut microbiota affects CYP3A metabolism or the clearance of clinically relevant drugs in vivo. Our purpose was to evaluate the effects of gut microbiota on in vitro and in vivo drug metabolism and on the clearance of midazolam, which is a standard CYP3A metabolized drug. Hepatic Cyp3a activity and in vitro midazolam hydroxylase activity were compared using specific pathogen‐free (SPF) and germ‐free (GF) mice. In a pharmacokinetics (PK) study, SPF and GF mice were intraperitoneally injected with 60 mg/kg of midazolam, and plasma and tissue concentrations were measured. Hepatic Cyp3a activity and midazolam hydroxylase activity were significantly lower in GF mice than in SPF mice. Notably, in the PK study, the area under the plasma concentration–time curve from time zero to infinity and the elimination half‐life were approximately four‐fold higher in GF mice compared with SPF mice. Furthermore, the concentration of midazolam in the brain 180 min after administration was about 14‐fold higher in GF mice compared with SPF mice. Together, our results demonstrated that the gut microbiota altered the metabolic ability of Cyp3a and the tissue accumulation of midazolam.
Collapse
Affiliation(s)
- Masao Togao
- Safety Research Department, Yakult Central Institute, Kunitachi-shi, Tokyo, Japan
| | - Koji Kawakami
- Safety Research Department, Yakult Central Institute, Kunitachi-shi, Tokyo, Japan
| | - Jun Otsuka
- Safety Research Department, Yakult Central Institute, Kunitachi-shi, Tokyo, Japan
| | - Gaku Wagai
- Safety Research Department, Yakult Central Institute, Kunitachi-shi, Tokyo, Japan
| | - Yuki Ohta-Takada
- Safety Research Department, Yakult Central Institute, Kunitachi-shi, Tokyo, Japan
| | - Shoichi Kado
- Safety Research Department, Yakult Central Institute, Kunitachi-shi, Tokyo, Japan
| |
Collapse
|
18
|
Li F, MacKenzie KR, Jain P, Santini C, Young DW, Matzuk MM. Metabolism of JQ1, an inhibitor of bromodomain and extra terminal bromodomain proteins, in human and mouse liver microsomes†. Biol Reprod 2020; 103:427-436. [PMID: 32285106 PMCID: PMC7401416 DOI: 10.1093/biolre/ioaa043] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/30/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022] Open
Abstract
JQ1 is a small-molecule inhibitor of the bromodomain and extra terminal (BET) protein family that potently inhibits the bromodomain testis-specific protein (BRDT), which is essential for spermatogenesis. JQ1 treatment produces a reversible contraceptive effect by targeting the activity of BRDT in mouse male germ cells, validating BRDT as a male contraceptive target. Although JQ1 possesses favourable physical properties, it exhibits a short half-life. Because the details of xenobiotic metabolism play important roles in the optimization of drug candidates and in determining the role of metabolism in drug efficacy, we investigated the metabolism of JQ1 in human and mouse liver microsomes. We present the first comprehensive view of JQ1 metabolism in liver microsomes, distinguishing nine JQ1 metabolites, including three monohydroxylated, one de-tert-butylated, two dihydroxylated, one monohydroxylated/dehydrogenated, one monohydroxylated-de-tert-butylated and one dihydroxylated/dehydrogenated variant of JQ1. The dominant metabolite (M1) in both human and mouse liver microsomes is monohydroxylated on the fused three-ring core. Using recombinant cytochrome P450 (CYP) enzymes, chemical inhibitors and the liver S9 fraction of Cyp3a-null mice, we identify enzymes that contribute to the formation of these metabolites. Cytochrome P450 family 3 subfamily A member 4 (CYP3A4) is the main contributor to the production of JQ1 metabolites in vitro, and the CYP3A4/5 inhibitor ketoconazole strongly inhibits JQ1 metabolism in both human and mouse liver microsomes. Our findings suggest that JQ1 half-life and efficacy might be improved in vivo by co-administration of a selective CYP inhibitor, thereby impacting the use of JQ1 as a probe for BRDT activity in spermatogenesis and as a probe or therapeutic in other systems.
Collapse
Affiliation(s)
- Feng Li
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA.,NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine Houston, TX, USA
| | - Kevin R MacKenzie
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA.,NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine Houston, TX, USA
| | - Prashi Jain
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Conrad Santini
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Damian W Young
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| | - Martin M Matzuk
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
19
|
Brain accumulation of tivozanib is restricted by ABCB1 (P-glycoprotein) and ABCG2 (breast cancer resistance protein) in mice. Int J Pharm 2020; 581:119277. [PMID: 32234426 DOI: 10.1016/j.ijpharm.2020.119277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/18/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
Tivozanib is a potent and selective inhibitor of VEGFR1-3, recently approved by the EMA for first-line treatment of renal cell carcinoma. We used wild-type, knockout, and transgenic mouse strains to study the effects of the drug transporters ABCB1, ABCG2, and OATP1A/1B, and of the CYP3A enzymes on the oral availability and tissue distribution of tivozanib. Tivozanib was transported by human ABCB1 and mouse Abcg2 in polarized MDCK-II cells. Upon oral administration, tivozanib showed rapid absorption and the plasma concentration-time curves showed secondary peaks in all mouse strains, suggesting enterohepatic recirculation. The brain-to-plasma ratios were significantly increased in Abcb1a/1b-/- (2.2-fold) and Abcb1a/1b;Abcg2-/- (2.6-fold) mice compared to wild-type mice, indicating a modest protective role of these transporters in the blood-brain barrier. Slco1a/1b-/- mice showed a 1.2-fold lower liver-to-plasma ratio than wild-type mice, suggesting a minor role of mOatp1a/1b in tivozanib liver distribution. Oral plasma pharmacokinetics of tivozanib was not significantly altered in these mouse strains, nor in Cyp3a knockout and CYP3A4-humanized mice. The modest effect of ABC transporters on tivozanib brain accumulation, if also true in humans, might mean that this drug is not strongly limited in its therapeutic efficacy against malignant lesions situated partly or completely behind the blood-brain barrier.
Collapse
|
20
|
Park E, Kim HK, Jee J, Hahn S, Jeong S, Yoo J. Development of organoid-based drug metabolism model. Toxicol Appl Pharmacol 2019; 385:114790. [DOI: 10.1016/j.taap.2019.114790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/27/2019] [Accepted: 10/22/2019] [Indexed: 12/23/2022]
|
21
|
Li Y, Meng Q, Yang M, Liu D, Hou X, Tang L, Wang X, Lyu Y, Chen X, Liu K, Yu AM, Zuo Z, Bi H. Current trends in drug metabolism and pharmacokinetics. Acta Pharm Sin B 2019; 9:1113-1144. [PMID: 31867160 PMCID: PMC6900561 DOI: 10.1016/j.apsb.2019.10.001] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/23/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
Pharmacokinetics (PK) is the study of the absorption, distribution, metabolism, and excretion (ADME) processes of a drug. Understanding PK properties is essential for drug development and precision medication. In this review we provided an overview of recent research on PK with focus on the following aspects: (1) an update on drug-metabolizing enzymes and transporters in the determination of PK, as well as advances in xenobiotic receptors and noncoding RNAs (ncRNAs) in the modulation of PK, providing new understanding of the transcriptional and posttranscriptional regulatory mechanisms that result in inter-individual variations in pharmacotherapy; (2) current status and trends in assessing drug-drug interactions, especially interactions between drugs and herbs, between drugs and therapeutic biologics, and microbiota-mediated interactions; (3) advances in understanding the effects of diseases on PK, particularly changes in metabolizing enzymes and transporters with disease progression; (4) trends in mathematical modeling including physiologically-based PK modeling and novel animal models such as CRISPR/Cas9-based animal models for DMPK studies; (5) emerging non-classical xenobiotic metabolic pathways and the involvement of novel metabolic enzymes, especially non-P450s. Existing challenges and perspectives on future directions are discussed, and may stimulate the development of new research models, technologies, and strategies towards the development of better drugs and improved clinical practice.
Collapse
Affiliation(s)
- Yuhua Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
- The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qiang Meng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Mengbi Yang
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Xiangyu Hou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lan Tang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xin Wang
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuanfeng Lyu
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ai-Ming Yu
- UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Zhong Zuo
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Huichang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
22
|
Lin Y, Wang S, Zhou Z, Guo L, Yu F, Wu B. Bmal1 regulates circadian expression of cytochrome P450 3a11 and drug metabolism in mice. Commun Biol 2019; 2:378. [PMID: 31633069 PMCID: PMC6795895 DOI: 10.1038/s42003-019-0607-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 09/10/2019] [Indexed: 12/17/2022] Open
Abstract
Metabolism is a major defense mechanism of the body against xenobiotic threats. Here we unravel a critical role of Bmal1 for circadian clock-controlled Cyp3a11 expression and xenobiotic metabolism. Bmal1 deficiency decreases the mRNA, protein and microsomal activity of Cyp3a11, and blunts their circadian rhythms in mice. A screen for Cyp3a11 regulators identifies two circadian genes Dbp and Hnf4α as potential regulatory mediators. Cell-based experiments confirm that Dbp and Hnf4α activate Cyp3a11 transcription by their binding to a D-box and a DR1 element in the Cyp3a11 promoter, respectively. Bmal1 binds to the P1 distal promoter to regulate Hnf4α transcriptionally. Cellular regulation of Cyp3a11 by Bmal1 is Dbp- and Hnf4α-dependent. Bmal1 deficiency sensitizes mice to toxicities of drugs such as aconitine and triptolide (and blunts circadian toxicity rhythmicities) due to elevated drug exposure. In summary, Bmal1 connects circadian clock and Cyp3a11 metabolism, thereby impacting drug detoxification as a function of daily time.
Collapse
Affiliation(s)
- Yanke Lin
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632 Guangzhou, China
| | - Shuai Wang
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632 Guangzhou, China
- Integrated Chinese and Western Medicine Postdoctoral research station, Jinan University, 601 Huangpu Avenue West, Guangzhou, China
| | - Ziyue Zhou
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632 Guangzhou, China
| | - Lianxia Guo
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632 Guangzhou, China
| | - Fangjun Yu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632 Guangzhou, China
| | - Baojian Wu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632 Guangzhou, China
| |
Collapse
|
23
|
Rixen S, Havemeyer A, Tyl-Bielicka A, Pysniak K, Gajewska M, Kulecka M, Ostrowski J, Mikula M, Clement B. Mitochondrial amidoxime-reducing component 2 (MARC2) has a significant role in N-reductive activity and energy metabolism. J Biol Chem 2019; 294:17593-17602. [PMID: 31554661 DOI: 10.1074/jbc.ra119.007606] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 09/19/2019] [Indexed: 01/29/2023] Open
Abstract
The mitochondrial amidoxime-reducing component (MARC) is a mammalian molybdenum-containing enzyme. All annotated mammalian genomes harbor two MARC genes, MARC1 and MARC2, which share a high degree of sequence similarity. Both molybdoenzymes reduce a variety of N-hydroxylated compounds. Besides their role in N-reductive drug metabolism, only little is known about their physiological functions. In this study, we characterized an existing KO mouse model lacking the functional MARC2 gene and fed a high-fat diet and also performed in vivo and in vitro experiments to characterize reductase activity toward known MARC substrates. MARC2 KO significantly decreased reductase activity toward several N-oxygenated substrates, and for typical MARC substrates, only small residual reductive activity was still detectable in MARC2 KO mice. The residual detected reductase activity in MARC2 KO mice could be explained by MARC1 expression that was hardly unaffected by KO, and we found no evidence of significant activity of other reductase enzymes. These results clearly indicate that MARC2 is mainly responsible for N-reductive biotransformation in mice. Striking phenotypical features of MARC2 KO mice were lower body weight, increased body temperature, decreased levels of total cholesterol, and increased glucose levels, supporting previous findings that MARC2 affects energy pathways. Of note, the MARC2 KO mice were resistant to high-fat diet-induced obesity. We propose that the MARC2 KO mouse model could be a powerful tool for predicting MARC-mediated drug metabolism and further investigating MARC's roles in energy homeostasis.
Collapse
Affiliation(s)
- Sophia Rixen
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Christian Albrechts University, 24118 Kiel, Germany
| | - Antje Havemeyer
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Christian Albrechts University, 24118 Kiel, Germany
| | - Anita Tyl-Bielicka
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland
| | - Kazimiera Pysniak
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland
| | - Marta Gajewska
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland
| | - Maria Kulecka
- Department of Gastroenterology, Hepatology, and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
| | - Jerzy Ostrowski
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland.,Department of Gastroenterology, Hepatology, and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
| | - Michal Mikula
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland
| | - Bernd Clement
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Christian Albrechts University, 24118 Kiel, Germany
| |
Collapse
|
24
|
Minegishi G, Kazuki Y, Yamasaki Y, Okuya F, Akita H, Oshimura M, Kobayashi K. Comparison of the hepatic metabolism of triazolam in wild-type andCyp3a-knockout mice for understanding CYP3A-mediated metabolism inCYP3A-humanised mice in vivo. Xenobiotica 2019; 49:1303-1310. [DOI: 10.1080/00498254.2018.1560516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Genki Minegishi
- Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Tottori, Japan
| | - Yuki Yamasaki
- Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Fuka Okuya
- Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hidetaka Akita
- Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
| | - Kaoru Kobayashi
- Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
25
|
Henderson CJ, Kapelyukh Y, Scheer N, Rode A, McLaren AW, MacLeod AK, Lin D, Wright J, Stanley LA, Wolf CR. An Extensively Humanized Mouse Model to Predict Pathways of Drug Disposition and Drug/Drug Interactions, and to Facilitate Design of Clinical Trials. Drug Metab Dispos 2019; 47:601-615. [PMID: 30910785 PMCID: PMC6505380 DOI: 10.1124/dmd.119.086397] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
Species differences in drug metabolism and disposition can confound the extrapolation of in vivo PK data to man and also profoundly compromise drug efficacy studies owing to differences in pharmacokinetics, in metabolites produced (which are often pharmacologically active), and in differential activation of the transcription factors constitutive androstane receptor (CAR) and pregnane X receptor (PXR), which regulate the expression of such enzymes as P450s and drug transporters. These differences have gained additional importance as a consequence of the use of genetically modified mouse models for drug-efficacy testing and also patient-derived xenografts to predict individual patient responses to anticancer drugs. A number of humanized mouse models for cytochrome P450s, CAR, and PXR have been reported. However, the utility of these models has been compromised by the redundancy in P450 reactions across gene families, whereby the remaining murine P450s can metabolize the compounds being tested. To remove this confounding factor and create a mouse model that more closely reflects human pathways of drug disposition, we substituted 33 murine P450s from the major gene families involved in drug disposition, together with Car and Pxr, for human CAR, PXR, CYP1A1, CYP1A2, CYP2C9, CYP2D6, CYP3A4, and CYP3A7. We also created a mouse line in which 34 P450s were deleted from the mouse genome. Using model compounds and anticancer drugs, we demonstrated how these mouse lines can be applied to predict drug-drug interactions in patients and discuss here their potential application in the more informed design of clinical trials and the personalized treatment of cancer.
Collapse
Affiliation(s)
- C J Henderson
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - Y Kapelyukh
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - N Scheer
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - A Rode
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - A W McLaren
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - A K MacLeod
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - D Lin
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - J Wright
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - L A Stanley
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - C R Wolf
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| |
Collapse
|
26
|
The transcription factor E4bp4 regulates the expression and activity of Cyp3a11 in mice. Biochem Pharmacol 2019; 163:215-224. [DOI: 10.1016/j.bcp.2019.02.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/19/2019] [Indexed: 11/17/2022]
|
27
|
Ye H, Sui D, Liu W, Yuan Y, Ouyang Z, Wei Y. Effects of CYP2C11 gene knockout on the pharmacokinetics and pharmacodynamics of warfarin in rats. Xenobiotica 2019; 49:1478-1484. [PMID: 30724651 DOI: 10.1080/00498254.2019.1579006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
1. CYP2C11 is the most abundant isoform of cytochrome P450s (CYPs) in male rats and is considered the main enzyme for warfarin metabolism. 2. To further access the in vivo function of CYP2C11 in warfarin metabolism and efficacy, a CYP2C11-null rat model was used to study warfarin metabolism with both in vitro and in vivo approaches. Prothrombin time (PT) of warfarin was also determined. 3. The maximum rate of metabolism (Vmax) and intrinsic clearance (CLint) of liver microsomes from CYP2C11-null males were reduced by 37 and 64%, respectively, compared to those in Sprague Dawley (S-D) rats. The Km of liver microsomes from CYP2C11-null males was increased by 73% compared to that of S-D rats. The time to reach the maximum plasma concentration (Tmax) of warfarin in CYP2C11-null males was significantly delayed compared to that in S-D males, and the CL rate was also reduced. The PT of CYP2C11-null rats was moderately longer than that of S-D rats. 4. In conclusion, the clearance rate of warfarin was mildly decreased and its anticoagulant effect was moderately increased in male rats following CYP2C11 gene knockout. CYP2C11 played a certain role in the clearance and efficacy of warfarin, while it did not seem to be essential.
Collapse
Affiliation(s)
- Huanying Ye
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Danjuan Sui
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Wei Liu
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Yuannan Yuan
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Zhen Ouyang
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Yuan Wei
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| |
Collapse
|
28
|
Wang J, Gan C, Retmana IA, Sparidans RW, Li W, Lebre MC, Beijnen JH, Schinkel AH. P-glycoprotein (MDR1/ABCB1) and Breast Cancer Resistance Protein (BCRP/ABCG2) limit brain accumulation of the FLT3 inhibitor quizartinib in mice. Int J Pharm 2018; 556:172-180. [PMID: 30553002 DOI: 10.1016/j.ijpharm.2018.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 12/31/2022]
Abstract
Quizartinib, a second-generation FLT3 inhibitor, is in clinical development for the treatment of acute myeloid leukemia. We studied its pharmacokinetic interactions with the multidrug efflux transporters ABCB1 and ABCG2 and the multidrug metabolizing enzyme CYP3A, using in vitro transport assays and knockout and transgenic mouse models. Quizartinib was transported by human ABCB1 in vitro, and by mouse (m)Abcb1 and mAbcg2 in vivo. Upon oral administration, the brain accumulation of quizartinib was 6-fold decreased by mAbcb1 and 2-fold by mAbcg2 (together: 12-fold). Unexpectedly, the absence of mAbcb1 resulted in a ∼2-fold lower plasma exposure in Abcb1a/1b-/- and Abcb1a/1b;Abcg2-/- mice, suggesting that loss of mAbcb1 causes compensatory alterations in alternative quizartinib elimination or uptake systems. mAbcb1 and mAbcg2 themselves did not appear to restrict quizartinib oral availability. Oral and intravenous pharmacokinetics of quizartinib were not substantially altered between wild-type, Cyp3a knockout and CYP3A4-humanized mice. All three strains showed relatively high (33-51%) oral bioavailability. If this also applies in humans, this would suggest a limited risk of CYP3A-related inter-individual variation in exposure for this drug. Our results provide a possible rationale for using pharmacological ABCB1/ABCG2 inhibitors together with quizartinib when treating malignant lesions situated in part or in whole behind the blood-brain barrier.
Collapse
Affiliation(s)
- Jing Wang
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Changpei Gan
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Irene A Retmana
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Rolf W Sparidans
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Wenlong Li
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Maria C Lebre
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Jos H Beijnen
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; The Netherlands Cancer Institute/Slotervaart Hospital, Department of Pharmacy & Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Alfred H Schinkel
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
29
|
Millecam J, De Clerck L, Govaert E, Devreese M, Gasthuys E, Schelstraete W, Deforce D, De Bock L, Van Bocxlaer J, Sys S, Croubels S. The Ontogeny of Cytochrome P450 Enzyme Activity and Protein Abundance in Conventional Pigs in Support of Preclinical Pediatric Drug Research. Front Pharmacol 2018; 9:470. [PMID: 29867477 PMCID: PMC5960725 DOI: 10.3389/fphar.2018.00470] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/23/2018] [Indexed: 01/08/2023] Open
Abstract
Since the implementation of several legislations to improve pediatric drug research, more pediatric clinical trials are being performed. In order to optimize these pediatric trials, adequate preclinical data are necessary, which are usually obtained by juvenile animal models. The growing piglet has been increasingly suggested as a potential animal model due to a high degree of anatomical and physiological similarities with humans. However, physiological data in pigs on the ontogeny of major organs involved in absorption, distribution, metabolism, and excretion of drugs are largely lacking. The aim of this study was to unravel the ontogeny of porcine hepatic drug metabolizing cytochrome P450 enzyme (CYP450) activities as well as protein abundances. Liver microsomes from 16 conventional pigs (8 males and 8 females) per age group: 2 days, 4 weeks, 8 weeks, and 6-7 months were prepared. Activity measurements were performed with substrates of major human CYP450 enzymes: midazolam (CYP3A), tolbutamide (CYP2C), and chlorzoxazone (CYP2E). Next, the hepatic scaling factor, microsomal protein per gram liver (MPPGL), was determined to correct for enzyme losses during the fractionation process. Finally, protein abundance was determined using proteomics and correlated with enzyme activity. No significant sex differences within each age category were observed in enzyme activity or MPPGL. The biotransformation rate of all three substrates increased with age, comparable with human maturation of CYP450 enzymes. The MPPGL decreased from birth till 8 weeks of age followed by an increase till 6-7 months of age. Significant sex differences in protein abundance were observed for CYP1A2, CYP2A19, CYP3A22, CYP4V2, CYP2C36, CYP2E_1, and CYP2E_2. Midazolam and tolbutamide are considered good substrates to evaluate porcine CYP3A/2C enzymes, respectively. However, chlorzoxazone is not advised to evaluate porcine CYP2E enzyme activity. The increase in biotransformation rate with age can be attributed to an increase in absolute amount of CYP450 proteins. Finally, developmental changes were observed regarding the involvement of specific CYP450 enzymes in the biotransformation of the different substrates.
Collapse
Affiliation(s)
- Joske Millecam
- Laboratory of Pharmacology and Toxicology, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Laura De Clerck
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Elisabeth Govaert
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Mathias Devreese
- Laboratory of Pharmacology and Toxicology, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Elke Gasthuys
- Laboratory of Pharmacology and Toxicology, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wim Schelstraete
- Laboratory of Pharmacology and Toxicology, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Lies De Bock
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Jan Van Bocxlaer
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Stanislas Sys
- Department of Internal Medicine and Clinical Biology of Large Animals, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Siska Croubels
- Laboratory of Pharmacology and Toxicology, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
30
|
P-glycoprotein (MDR1/ABCB1) and Breast Cancer Resistance Protein (BCRP/ABCG2) affect brain accumulation and intestinal disposition of encorafenib in mice. Pharmacol Res 2017; 129:414-423. [PMID: 29155017 DOI: 10.1016/j.phrs.2017.11.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/08/2017] [Indexed: 12/29/2022]
Abstract
Encorafenib (LGX818) is a promising BRAFV600E inhibitor that has efficacy against metastatic melanoma. To better understand its pharmacokinetics, we studied its interactions with the multidrug efflux transporters ABCB1 and ABCG2 and the multidrug metabolizing enzyme CYP3A. In polarized MDCK-II cells, encorafenib was efficiently transported by canine and human ABCB1 and ABCG2 and by mouse Abcg2. Upon oral administration to wild-type, Abcb1a/1b-/-, Abcg2-/-, and Abcb1a/1b;Abcg2-/- mice, encorafenib was absorbed very quickly and to very high plasma levels, but without clear changes in oral availability between the strains. Upon oral or intravenous administration, encorafenib brain accumulation was markedly increased in Abcb1a/1b;Abcg2-/- mice and to a lesser extent in Abcb1a/1b-/- mice. However, absolute brain concentrations and brain-to-plasma ratios remained very low in all strains, indicating intrinsically poor brain penetration of encorafenib. Upon intravenous administration, Abcb1a/1b;Abcg2-/- mice showed somewhat reduced plasma elimination of encorafenib compared to wild-type mice, and lower accumulation of the drug in the intestinal tract, suggesting a limited role for these transporters in intestinal elimination of the drug. In Cyp3a-/- mice plasma levels of encorafenib were not markedly increased, suggesting a limited impact of Cyp3a on encorafenib oral availability. The low brain penetration of encorafenib might limit its efficacy against malignancies positioned behind a functional blood-brain barrier, but its oral bioavailability and distribution to other tested organs (liver, kidney, spleen, testis) was high.
Collapse
|
31
|
Kort A, van Hoppe S, Sparidans RW, Wagenaar E, Beijnen JH, Schinkel AH. Brain Accumulation of Ponatinib and Its Active Metabolite, N-Desmethyl Ponatinib, Is Limited by P-Glycoprotein (P-GP/ABCB1) and Breast Cancer Resistance Protein (BCRP/ABCG2). Mol Pharm 2017; 14:3258-3268. [DOI: 10.1021/acs.molpharmaceut.7b00257] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Anita Kort
- Division
of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan
121, 1066 CX Amsterdam, The Netherlands
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Stéphanie van Hoppe
- Division
of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan
121, 1066 CX Amsterdam, The Netherlands
| | - Rolf W. Sparidans
- Division of Pharmacoepidemiology & Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Els Wagenaar
- Division
of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan
121, 1066 CX Amsterdam, The Netherlands
| | - Jos H. Beijnen
- Division of Pharmacoepidemiology & Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Department
of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan
121, 1066 CX Amsterdam, The Netherlands
| | - Alfred H. Schinkel
- Division
of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan
121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
32
|
Lu W, Rettenmeier E, Paszek M, Yueh MF, Tukey RH, Trottier J, Barbier O, Chen S. Crypt Organoid Culture as an in Vitro Model in Drug Metabolism and Cytotoxicity Studies. Drug Metab Dispos 2017; 45:748-754. [PMID: 28468837 PMCID: PMC5478905 DOI: 10.1124/dmd.117.075945] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/27/2017] [Indexed: 12/14/2022] Open
Abstract
The gastrointestinal tract is enriched with xenobiotic processing proteins that play important roles in xenobiotic bioactivation, metabolism, and detoxification. The application of genetically modified mouse models has been instrumental in characterizing the function of xenobiotic processing genes (XPG) and their proteins in drug metabolism. Here, we report the utilization of three-dimensional crypt organoid cultures from these animal models to study intestinal drug metabolism and toxicity. With the successful culturing of crypt organoids, we profiled the abundance of Phase I and Phase II XPG expression, drug transporter gene expression, and xenobiotic nuclear receptor (XNR) gene expression. Functions of XNRs were examined by treating crypt cells with XNR prototypical agonists. Real-time quantitative polymerase chain reaction demonstrated that the representative downstream target genes were induced. These findings were validated from cultures developed from XNR-null mice. In crypt cultures isolated from Pxr-/- mice, pregnenolone 16α-carbonitrile failed to induce Cyp3a11 gene expression; similarly, WY14643 failed to induce Cyp4a10 in the Pparα-/- crypts. Crypt cultures from control (Ugt1F/F ) and intestinal epithelial cell (IEC) specific Ugt1 null mice (Ugt1ΔIEC ) were treated with camptothecin-11, an anticancer prodrug with severe intestinal toxicity that originates from insufficient UGT1A1-dependent glucuronidation of its active metabolite SN-38. In the absence of Ugt1 gene expression, Ugt1ΔIEC crypt cultures exhibit very limited production of SN-38 glucuronide, concordant with increased apoptosis in comparison with Ugt1F/F crypt cultures. This study suggests crypt organoid cultures as an effective in vitro model for studying intestinal drug metabolism and toxicity.
Collapse
Affiliation(s)
- Wenqi Lu
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (W.L., E.R., M.P., M-F.Y., R.H.T., S.C.); and Laboratory of Molecular Pharmacology, CHU de Quebec Research Centre and Faculty of Pharmacy, Laval University, Québec (Québec), Canada (J.T., O.B.)
| | - Eva Rettenmeier
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (W.L., E.R., M.P., M-F.Y., R.H.T., S.C.); and Laboratory of Molecular Pharmacology, CHU de Quebec Research Centre and Faculty of Pharmacy, Laval University, Québec (Québec), Canada (J.T., O.B.)
| | - Miles Paszek
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (W.L., E.R., M.P., M-F.Y., R.H.T., S.C.); and Laboratory of Molecular Pharmacology, CHU de Quebec Research Centre and Faculty of Pharmacy, Laval University, Québec (Québec), Canada (J.T., O.B.)
| | - Mei-Fei Yueh
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (W.L., E.R., M.P., M-F.Y., R.H.T., S.C.); and Laboratory of Molecular Pharmacology, CHU de Quebec Research Centre and Faculty of Pharmacy, Laval University, Québec (Québec), Canada (J.T., O.B.)
| | - Robert H Tukey
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (W.L., E.R., M.P., M-F.Y., R.H.T., S.C.); and Laboratory of Molecular Pharmacology, CHU de Quebec Research Centre and Faculty of Pharmacy, Laval University, Québec (Québec), Canada (J.T., O.B.)
| | - Jocelyn Trottier
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (W.L., E.R., M.P., M-F.Y., R.H.T., S.C.); and Laboratory of Molecular Pharmacology, CHU de Quebec Research Centre and Faculty of Pharmacy, Laval University, Québec (Québec), Canada (J.T., O.B.)
| | - Olivier Barbier
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (W.L., E.R., M.P., M-F.Y., R.H.T., S.C.); and Laboratory of Molecular Pharmacology, CHU de Quebec Research Centre and Faculty of Pharmacy, Laval University, Québec (Québec), Canada (J.T., O.B.)
| | - Shujuan Chen
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (W.L., E.R., M.P., M-F.Y., R.H.T., S.C.); and Laboratory of Molecular Pharmacology, CHU de Quebec Research Centre and Faculty of Pharmacy, Laval University, Québec (Québec), Canada (J.T., O.B.)
| |
Collapse
|
33
|
Lipiski M, Arras M, Jirkof P, Cesarovic N. Premedication with fentanyl-midazolam improves sevoflurane anesthesia for surgical intervention in laboratory mice. Exp Biol Med (Maywood) 2017; 242:1287-1298. [PMID: 28474988 PMCID: PMC5476341 DOI: 10.1177/1535370217707730] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 04/11/2017] [Indexed: 01/04/2023] Open
Abstract
Balanced anesthesia allows for a reduced dosage of each component, while inducing general anesthesia of sufficient depth with potentially fewer side effects. Here, we compare two anesthetic protocols combining sevoflurane anesthesia with pre-medication (ketamine [K] or fentanyl-midazolam [FM]) to a sevoflurane monoanesthesia (S) concerning their ability to provide reliable anesthesia suitable for moderate surgery in laboratory mice. Twenty-one female C57BL/6J mice assigned randomly to one of three protocols underwent a 50-min anesthesia and a sham embryo transfer. Heart rate and core body temperature were continuously recorded by telemetry intra-operatively and for three days pre- and three days post-surgery. Intra-operative respiratory rate was determined by counting thorax movements. Body weight, food, and water intake were measured daily for three days pre- and three days post-surgery. The heart rate in the KS group remained at baseline level throughout the 50-min of anesthesia and surgery. FMS caused a lower heart rate and S alone caused a higher heart rate compared to baseline values. Intra-operative body temperature was at baseline levels in all groups. A decreased respiratory rate was observed in all groups compared to baseline values obtained from resting mice of the same strain, sex and age-distribution. Surgical stimuli induced no significant changes in heart rate and respiratory rate in the KS or FMS group but significant respiratory alteration in the S group compared to baseline values obtained 10 s before applying the stimulus. Post-operative heart rate was above baseline values in all groups; with a significant deviation in the S group. There were no changes in body weight, food, and water intake. In summary, FMS was superior to KS and S for moderate surgery in laboratory mice resulting in less inter-individual variability in response to painful stimuli. Fentanyl and midazolam reduced the depressant effect of sevoflurane on the respiratory rate and the negative post-anesthetic effects on the heart rate. Impact statement With approximately 65 million animals used per year mice are still the most prevalent laboratory mammal species worldwide. In course of biomedical research projects approximately 40% of mice will undergo one or more short or long-term anesthesia. Sufficient anesthetic depth, cardiovascular stability, adequate analgesia, and short recovery times are essential requirements of anesthetic protocols to meet animal welfare. Anesthesia in mice and rats are only to be performed by personnel with appropriate basic training and experience. However, more and more adapted and advanced anesthetic protocols, required to answer very specific scientific questions, often exceed the skills acquired through basic training and present a major challenge to researchers. It is therefore of great importance to further develop and evaluate safe and reliable anesthetic protocols as presented in this study to provide new perspectives on this challenging problem.
Collapse
Affiliation(s)
- Miriam Lipiski
- Division of Surgical Research, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Margarete Arras
- Division of Surgical Research, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| | - Paulin Jirkof
- Division of Surgical Research, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| | - Nikola Cesarovic
- Division of Surgical Research, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| |
Collapse
|
34
|
Ly JQ, Messick K, Qin A, Takahashi RH, Choo EF. Utility of CYP3A4 and PXR-CAR-CYP3A4/3A7 Transgenic Mouse Models To Assess the Magnitude of CYP3A4 Mediated Drug–Drug Interactions. Mol Pharm 2017; 14:1754-1759. [DOI: 10.1021/acs.molpharmaceut.7b00006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Justin Q. Ly
- Genentech Inc., South San Francisco, California 94080, United States
| | - Kirsten Messick
- Genentech Inc., South San Francisco, California 94080, United States
| | - Ann Qin
- Genentech Inc., South San Francisco, California 94080, United States
| | - Ryan H. Takahashi
- Genentech Inc., South San Francisco, California 94080, United States
| | - Edna F. Choo
- Genentech Inc., South San Francisco, California 94080, United States
| |
Collapse
|
35
|
Kumar R, Mota LC, Litoff EJ, Rooney JP, Boswell WT, Courter E, Henderson CM, Hernandez JP, Corton JC, Moore DD, Baldwin WS. Compensatory changes in CYP expression in three different toxicology mouse models: CAR-null, Cyp3a-null, and Cyp2b9/10/13-null mice. PLoS One 2017; 12:e0174355. [PMID: 28350814 PMCID: PMC5370058 DOI: 10.1371/journal.pone.0174355] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/07/2017] [Indexed: 12/12/2022] Open
Abstract
Targeted mutant models are common in mechanistic toxicology experiments investigating the absorption, metabolism, distribution, or elimination (ADME) of chemicals from individuals. Key models include those for xenosensing transcription factors and cytochrome P450s (CYP). Here we investigated changes in transcript levels, protein expression, and steroid hydroxylation of several xenobiotic detoxifying CYPs in constitutive androstane receptor (CAR)-null and two CYP-null mouse models that have subfamily members regulated by CAR; the Cyp3a-null and a newly described Cyp2b9/10/13-null mouse model. Compensatory changes in CYP expression that occur in these models may also occur in polymorphic humans, or may complicate interpretation of ADME studies performed using these models. The loss of CAR causes significant changes in several CYPs probably due to loss of CAR-mediated constitutive regulation of these CYPs. Expression and activity changes include significant repression of Cyp2a and Cyp2b members with corresponding drops in 6α- and 16β-testosterone hydroxylase activity. Further, the ratio of 6α-/15α-hydroxylase activity, a biomarker of sexual dimorphism in the liver, indicates masculinization of female CAR-null mice, suggesting a role for CAR in the regulation of sexually dimorphic liver CYP profiles. The loss of Cyp3a causes fewer changes than CAR. Nevertheless, there are compensatory changes including gender-specific increases in Cyp2a and Cyp2b. Cyp2a and Cyp2b were down-regulated in CAR-null mice, suggesting activation of CAR and potentially PXR following loss of the Cyp3a members. However, the loss of Cyp2b causes few changes in hepatic CYP transcript levels and almost no significant compensatory changes in protein expression or activity with the possible exception of 6α-hydroxylase activity. This lack of a compensatory response in the Cyp2b9/10/13-null mice is probably due to low CYP2B hepatic expression, especially in male mice. Overall, compensatory and regulatory CYP changes followed the order CAR-null > Cyp3a-null > Cyp2b-null mice.
Collapse
Affiliation(s)
- Ramiya Kumar
- Biological Sciences, Clemson University, Clemson, SC, United States of America
| | - Linda C. Mota
- Environmental Toxicology, Clemson University, Pendleton, SC, United States of America
| | - Elizabeth J. Litoff
- Biological Sciences, Clemson University, Clemson, SC, United States of America
| | - John P. Rooney
- NHEERL, US-EPA, Research Triangle Park, NC, United States of America
| | - W. Tyler Boswell
- Biological Sciences, Clemson University, Clemson, SC, United States of America
| | - Elliott Courter
- Biological Sciences, Clemson University, Clemson, SC, United States of America
| | | | - Juan P. Hernandez
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | | | - David D. Moore
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - William S. Baldwin
- Biological Sciences, Clemson University, Clemson, SC, United States of America
- Environmental Toxicology, Clemson University, Pendleton, SC, United States of America
- * E-mail:
| |
Collapse
|
36
|
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
|
37
|
Chang JH, Chen J, Liu L, Messick K, Ly J. Rifampin-Mediated Induction of Tamoxifen Metabolism in a Humanized PXR-CAR-CYP3A4/3A7-CYP2D6 Mouse Model. Drug Metab Dispos 2016; 44:1736-1741. [PMID: 27538915 DOI: 10.1124/dmd.116.072132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/17/2016] [Indexed: 01/14/2023] Open
Abstract
Animals are not commonly used to assess drug-drug interactions due to poor clinical translatability arising from species differences that may exist in drug-metabolizing enzymes and transporters, and their regulation pathways. In this study, a transgenic mouse model expressing human pregnane X receptor (PXR), constitutive androstane receptor (CAR), CYP3A4/CYP3A7, and CYP2D6 (Tg-composite) was used to investigate the effect of induction mediated by rifampin on the pharmacokinetics of tamoxifen and its metabolites. In humans, tamoxifen is metabolized primarily by CYP3A4 and CYP2D6, and multiple-day treatment with rifampin decreased tamoxifen exposure by 6.2-fold. Interestingly, exposure of tamoxifen metabolites 4-hydroxytamoxifen (4OHT), N-desmethyltamoxifen (NDM), and endoxifen also decreased. In the Tg-composite model, pretreatment with rifampin decreased tamoxifen area under the time-concentration curve between 0 and 8 hours (AUC0-8) from 0.82 to 0.20 µM*h, whereas AUC0-8 of 4OHT, NDM, and endoxifen decreased by 3.4-, 4.7-, and 1.3-fold, respectively, mirroring the clinic observations. In the humanized PXR-CAR (hPXR-CAR) model, rifampin decreased AUC0-8 of tamoxifen and its metabolites by approximately 2-fold. In contrast, no significant modulation by rifampin was observed in the nonhumanized C57BL/6 (wild-type) animals. In vitro kinetics determined in microsomes prepared from livers of the Tg-composite animals showed that, although Km values were not different between vehicle- and rifampin-treated groups, rifampin increased the Vmax for the CYP3A4-mediated pathways. These data demonstrate that, although the hPXR-CAR model is responsive to rifampin, the extent of the clinical rifampin-tamoxifen interaction is better represented by the Tg-composite model. Consequently, the Tg-composite model may be a suitable tool to examine the extent of rifampin-mediated induction for other compounds whose metabolism is mediated by CYP3A4 and/or CYP2D6.
Collapse
Affiliation(s)
- Jae H Chang
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| | - John Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| | - Liling Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| | - Kirsten Messick
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| | - Justin Ly
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California
| |
Collapse
|
38
|
Xie F, Ding X, Zhang QY. An update on the role of intestinal cytochrome P450 enzymes in drug disposition. Acta Pharm Sin B 2016; 6:374-383. [PMID: 27709006 PMCID: PMC5045550 DOI: 10.1016/j.apsb.2016.07.012] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/05/2022] Open
Abstract
Oral administration is the most commonly used route for drug treatment. Intestinal cytochrome P450 (CYP)-mediated metabolism can eliminate a large proportion of some orally administered drugs before they reach systemic circulation, while leaving the passage of other drugs unimpeded. A better understanding of the ability of intestinal P450 enzymes to metabolize various clinical drugs in both humans and preclinical animal species, including the identification of the CYP enzymes expressed, their regulation, and the relative importance of intestinal metabolism compared to hepatic metabolism, is important for improving bioavailability of current drugs and new drugs in development. Here, we briefly review the expression of drug-metabolizing P450 enzymes in the small intestine of humans and several preclinical animal species, and provide an update of the various factors or events that regulate intestinal P450 expression, including a cross talk between the liver and the intestine. We further compare various clinical and preclinical approaches for assessing the impact of intestinal drug metabolism on bioavailability, and discuss the utility of the intestinal epithelium–specific NADPH-cytochrome P450 reductase-null (IECN) mouse as a useful model for studying in vivo roles of intestinal P450 in the disposition of orally administered drugs.
Collapse
|
39
|
de Vries EM, Lammers LA, Achterbergh R, Klümpen HJ, Mathot RAA, Boelen A, Romijn JA. Fasting-Induced Changes in Hepatic P450 Mediated Drug Metabolism Are Largely Independent of the Constitutive Androstane Receptor CAR. PLoS One 2016; 11:e0159552. [PMID: 27434302 PMCID: PMC4951123 DOI: 10.1371/journal.pone.0159552] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 07/04/2016] [Indexed: 12/25/2022] Open
Abstract
Introduction Hepatic drug metabolism by cytochrome P450 enzymes is altered by the nutritional status of patients. The expression of P450 enzymes is partly regulated by the constitutive androstane receptor (CAR). Fasting regulates the expression of both P450 enzymes and CAR and affects hepatic drug clearance. We hypothesized that the fasting-induced alterations in P450 mediated drug clearance are mediated by CAR. Methods To investigate this we used a drug cocktail validated in humans consisting of five widely prescribed drugs as probes for specific P450 enzymes: caffeine (CYP1A2), metoprolol (CYP2D6), omeprazole (CYP2C19), midazolam (CYP3A4) and s-warfarin (CYP2C9). This cocktail was administered to wild type (WT, C57Bl/6) mice or mice deficient for CAR (CAR-/-) that were either fed ad libitum or fasted for 24 hours. Blood was sampled at predefined intervals and drug concentrations were measured as well as hepatic mRNA expression of homologous/orthologous P450 enzymes (Cyp1a2, Cyp2d22, Cyp3a11, Cyp2c37, Cyp2c38 and Cyp2c65). Results Fasting decreased Cyp1a2 and Cyp2d22 expression and increased Cyp3a11 and Cyp2c38 expression in both WT and CAR-/- mice. The decrease in Cyp1a2 was diminished in CAR-/- in comparison with WT mice. Basal Cyp2c37 expression was lower in CAR-/- compared to WT mice. Fasting decreased the clearance of all drugs tested in both WT and CAR-/- mice. The absence of CAR was associated with an decrease in the clearance of omeprazole, metoprolol and midazolam in fed mice. The fasting-induced reduction in clearance of s-warfarin was greater in WT than in CAR-/-. The changes in drug clearance correlated with the expression pattern of the specific P450 enzymes in case of Cyp1a2-caffeine and Cyp2c37-omeprazole. Conclusion We conclude that CAR is important for hepatic clearance of several widely prescribed drugs metabolized by P450 enzymes. However the fasting-induced alterations in P450 mediated drug clearance are largely independent of CAR.
Collapse
Affiliation(s)
- E. M. de Vries
- Department of Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- * E-mail:
| | - L. A. Lammers
- Department of Hospital Pharmacy, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - R. Achterbergh
- Department of Medical Oncology, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - H-J Klümpen
- Department of Medical Oncology, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - R. A. A. Mathot
- Department of Hospital Pharmacy, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - A. Boelen
- Department of Endocrinology and Metabolism, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - J. A. Romijn
- Department of Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| |
Collapse
|
40
|
Peters SA, Jones CR, Ungell AL, Hatley OJD. Predicting Drug Extraction in the Human Gut Wall: Assessing Contributions from Drug Metabolizing Enzymes and Transporter Proteins using Preclinical Models. Clin Pharmacokinet 2016; 55:673-96. [PMID: 26895020 PMCID: PMC4875961 DOI: 10.1007/s40262-015-0351-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Intestinal metabolism can limit oral bioavailability of drugs and increase the risk of drug interactions. It is therefore important to be able to predict and quantify it in drug discovery and early development. In recent years, a plethora of models-in vivo, in situ and in vitro-have been discussed in the literature. The primary objective of this review is to summarize the current knowledge in the quantitative prediction of gut-wall metabolism. As well as discussing the successes of current models for intestinal metabolism, the challenges in the establishment of good preclinical models are highlighted, including species differences in the isoforms; regional abundances and activities of drug metabolizing enzymes; the interplay of enzyme-transporter proteins; and lack of knowledge on enzyme abundances and availability of empirical scaling factors. Due to its broad specificity and high abundance in the intestine, CYP3A is the enzyme that is frequently implicated in human gut metabolism and is therefore the major focus of this review. A strategy to assess the impact of gut wall metabolism on oral bioavailability during drug discovery and early development phases is presented. Current gaps in the mechanistic understanding and the prediction of gut metabolism are highlighted, with suggestions on how they can be overcome in the future.
Collapse
Affiliation(s)
- Sheila Annie Peters
- Translational Quantitative Pharmacology, BioPharma, R&D Global Early Development, Merck KGaA, Frankfurter Str. 250, F130/005, 64293, Darmstadt, Germany.
| | | | - Anna-Lena Ungell
- Investigative ADME, Non-Clinical Development, UCB New Medicines, BioPharma SPRL, Braine l'Alleud, Belgium
| | - Oliver J D Hatley
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, Sheffield, UK
| |
Collapse
|
41
|
Wang X, Tang Y, Lu J, Shao Y, Qin X, Li Y, Wang L, Li D, Liu M. Characterization of novel cytochrome P450 2E1 knockout rat model generated by CRISPR/Cas9. Biochem Pharmacol 2016; 105:80-90. [DOI: 10.1016/j.bcp.2016.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/01/2016] [Indexed: 12/26/2022]
|
42
|
Yin Q, Tai T, Ji JZ, Mi QY, Zhang MR, Huang WJ, Cao CC, Xie HG. Interleukin-10 does not modulate clopidogrel platelet response in mice. J Thromb Haemost 2016; 14:596-605. [PMID: 26712119 DOI: 10.1111/jth.13238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/06/2015] [Indexed: 02/04/2023]
Abstract
UNLABELLED ESSENTIALS: It is unclear whether interleukin-10 (IL-10) could affect clopidogrel metabolism and response. The bioactivation of and response to clopidogrel were determined between mice with or without IL-10. Maximum clopidogrel active metabolite levels were the major driver of platelet response to clopidogrel. IL-10 did not modulate maximum levels of clopidogrel active metabolite and its antiplatelet effects. SUMMARY BACKGROUND Elevated plasma interleukin-10 (IL-10) levels were observed in patients who responded less to clopidogrel (a prodrug that is required for further metabolic bioactivation in the liver). However, no data are currently available suggesting whether there is such an association. OBJECTIVE To systematically explore possible differences in the formation of and response to clopidogrel active metabolite (CAM) in mice with or without IL-10 gene expression. METHODS A single oral dose of clopidogrel (10 mg kg(-1)) was given to IL-10 knockout (KO) mice and wild-type (WT) control mice, respectively, and pharmacokinetic parameters of clopidogrel and CAM were calculated. Moreover, adenosine diphosphate-induced whole-blood platelet aggregation was measured in mice receiving 0, 5, 10, or 20 mg kg(-1) of clopidogrel, respectively. RESULTS Compared with IL-10 KO mice, WT mice had significantly lower area under the plasma concentration-time curve (AUC) of CAM as a result of a shorter mean elimination half-life but had significantly higher AUC of clopidogrel due to slower systemic clearance and smaller volume of distribution. Although AUC of CAM was significantly lower in WT mice than in KO mice, antiplatelet effects of clopidogrel did not differ significantly between the two mouse groups, as their maximum plasma concentrations (Cmax ) of CAM were not significantly different. CONCLUSIONS IL-10 expression level affects AUC rather than Cmax of CAM, but the Cmax of CAM is the major driver of antiplatelet effects of clopidogrel in mice.
Collapse
Affiliation(s)
- Q Yin
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - T Tai
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - J-Z Ji
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Q-Y Mi
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - M-R Zhang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - W-J Huang
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - C-C Cao
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - H-G Xie
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pharmacology, Nanjing Medical University School of Pharmacy, Nanjing, Jiangsu, China
| |
Collapse
|
43
|
Scheer N, Kapelyukh Y, Rode A, Oswald S, Busch D, McLaughlin LA, Lin D, Henderson CJ, Wolf CR. Defining Human Pathways of Drug Metabolism In Vivo through the Development of a Multiple Humanized Mouse Model. Drug Metab Dispos 2015; 43:1679-90. [PMID: 26265742 DOI: 10.1124/dmd.115.065656] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/10/2015] [Indexed: 11/22/2022] Open
Abstract
Variability in drug pharmacokinetics is a major factor in defining drug efficacy and side effects. There remains an urgent need, particularly with the growing use of polypharmacy, to obtain more informative experimental data predicting clinical outcomes. Major species differences in multiplicity, substrate specificity, and regulation of enzymes from the cytochrome P450-dependent mono-oxygenase system play a critical role in drug metabolism. To develop an in vivo model for predicting human responses to drugs, we generated a mouse, where 31 P450 genes from the Cyp2c, Cyp2d, and Cyp3a gene families were exchanged for their relevant human counterparts. The model has been improved through additional humanization for the nuclear receptors constitutive androgen receptor and pregnane X receptor that control the expression of key drug metabolizing enzymes and transporters. In this most complex humanized mouse model reported to date, the cytochromes P450 function as predicted and we illustrate how these mice can be applied to predict drug-drug interactions in humans.
Collapse
Affiliation(s)
- Nico Scheer
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Yury Kapelyukh
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Anja Rode
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Stefan Oswald
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Diana Busch
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Lesley A McLaughlin
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - De Lin
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Colin J Henderson
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - C Roland Wolf
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| |
Collapse
|
44
|
Abstract
Most of what we know about a drug prior to human clinical studies is derived from animal testing. Because animals and humans have substantial differences in their physiology and in their drug metabolism pathways, we do not know very much about the pharmacokinetic and pharmacodynamic behavior of a drug in humans until after it is administered to many people. Hence, drug-induced liver injury has become a significant public health problem, and we have a very inefficient drug development process with a high failure rate. Because the human liver is at the heart of these problems, chimeric mice with humanized livers could be used to address these issues. We examine recent evidence indicating that drug testing in chimeric mice could provide better information about a drug's metabolism, disposition, and toxicity (i.e., its "behavior") in humans and could aid in developing personalized medicine strategies, which would improve drug efficacy and safety.
Collapse
Affiliation(s)
- Dan Xu
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California 94305;
| | - Gary Peltz
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California 94305;
| |
Collapse
|
45
|
Hendrikx JJMA, Lagas JS, Song JY, Rosing H, Schellens JHM, Beijnen JH, Rottenberg S, Schinkel AH. Ritonavir inhibits intratumoral docetaxel metabolism and enhances docetaxel antitumor activity in an immunocompetent mouse breast cancer model. Int J Cancer 2015; 138:758-69. [PMID: 26297509 DOI: 10.1002/ijc.29812] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 08/03/2015] [Indexed: 12/27/2022]
Abstract
Docetaxel (Taxotere(®)) is currently used intravenously as an anticancer agent and is primarily metabolized by Cytochrome P450 3A (CYP3A). The HIV protease inhibitor ritonavir, a strong CYP3A4 inhibitor, decreased first-pass metabolism of orally administered docetaxel. Anticancer effects of ritonavir itself have also been described. We here aimed to test whether ritonavir co-administration could decrease intratumoral metabolism of intravenously administered docetaxel and thus increase the antitumor activity of docetaxel in an orthotopic, immunocompetent mouse model for breast cancer. Spontaneously arising K14cre;Brca1(F/F) ;p53(F/F) mouse mammary tumors were orthotopically implanted in syngeneic mice lacking Cyp3a (Cyp3a(-/-)) to limit ritonavir effects on systemic docetaxel clearance. Over 3 weeks, docetaxel (20 mg/kg) was administered intravenously once weekly, with or without ritonavir (12.5 mg/kg) administered orally for 5 days per week. Untreated mice were used as control for tumor growth. Ritonavir treatment alone did not significantly affect the median time of survival (14 vs. 10 days). Median time of survival in docetaxel-treated mice was 54 days. Ritonavir co-treatment significantly increased this to 66 days, and substantially reduced relative average tumor size, without altering tumor histology. Concentrations of the major docetaxel metabolite M2 in tumor tissue were reduced by ritonavir co-administration, whereas tumor RNA expression of Cyp3a was unaltered. In this breast cancer model, we observed no direct antitumor effect of ritonavir alone, but we found enhanced efficacy of docetaxel treatment when combined with ritonavir. Our data, therefore, suggest that decreased docetaxel metabolism inside the tumor as a result of Cyp3a inhibition contributes to increased antitumor activity.
Collapse
Affiliation(s)
- Jeroen J M A Hendrikx
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jurjen S Lagas
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Department of Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Sven Rottenberg
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alfred H Schinkel
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
46
|
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
|
47
|
Kato K, Ohbuchi M, Hamamura S, Ohshita H, Kazuki Y, Oshimura M, Sato K, Nakada N, Kawamura A, Usui T, Kamimura H, Tateno C. Development of Murine Cyp3a Knockout Chimeric Mice with Humanized Liver. Drug Metab Dispos 2015; 43:1208-17. [PMID: 25979261 DOI: 10.1124/dmd.115.063479] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/15/2015] [Indexed: 02/13/2025] Open
Abstract
We developed murine CYP3A knockout ko chimeric mice with humanized liver expressing human P450S similar to those in humans and whose livers and small intestines do not express murine CYP3A this: approach may overcome effects of residual mouse metabolic enzymes like Cyp3a in conventional chimeric mice with humanized liver, such as PXB-mice [urokinase plasminogen activator/severe combined immunodeficiency (uPA/SCID) mice repopulated with over 70% human hepatocytes] to improve the prediction of drug metabolism and pharmacokinetics in humans. After human hepatocytes were transplanted into Cyp3a KO/uPA/SCID host mice, human albumin levels logarithmically increased until approximately 60 days after transplantation, findings similar to those in PXB-mice. Quantitative real-time-polymerase chain reaction analyses showed that hepatic human P450s, UGTs, SULTs, and transporters mRNA expression levels in Cyp3a KO chimeric mice were also similar to those in PXB-mice and confirmed the absence of Cyp3a11 mRNA expression in mouse liver and intestine. Findings for midazolam and triazolam metabolic activities in liver microsomes were comparable between Cyp3a KO chimeric mice and PXB-mice. In contrast, these activities in the intestine of Cyp3a KO chimeric mice were attenuated compared with PXB-mice. Owing to the knockout of murine Cyp3a, hepatic Cyp2b10 and 2c55 mRNA levels in Cyp3a KO/uPA/SCID mice (without hepatocyte transplants) were 8.4- and 61-fold upregulated compared with PXB-mice, respectively. However, human hepatocyte transplantation successfully restored Cyp2b10 level nearly fully and Cyp2c55 level partly (still 13-fold upregulated) compared with those in PXB-mice. Intestinal Cyp2b10 and 2c55 were also repressed by human hepatocyte transplantation in Cyp3a KO chimeric mice.
Collapse
Affiliation(s)
- Kota Kato
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Masato Ohbuchi
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Satoko Hamamura
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Hiroki Ohshita
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Yasuhiro Kazuki
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Mitsuo Oshimura
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Koya Sato
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Naoyuki Nakada
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Akio Kawamura
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Takashi Usui
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Hidetaka Kamimura
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| | - Chise Tateno
- Drug Metabolism Research Laboratories, Astellas Pharma Inc., Osaka, Japan (K.K., Ma.O., K.S., N.N., A.K., T.U.); PhoenixBio Co., Ltd., Hiroshima, Japan (S.H., H.O., C.T.); Liver Research Project Center, Hiroshima University, Hiroshima, Japan (C.T.); Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science (Y.K., Mi.O.), Chromosome Engineering Research Center (Y.K., Mi.O.), Tottori University, Tottori, Japan; ADME & Tox Research Institute, Sekisui Medical Co., Ltd., Tokyo, Japan (H.K.)
| |
Collapse
|
48
|
Olsen LR, Gabel-Jensen C, Wubshet SG, Kongstad KT, Janfelt C, Badolo L, Hansen SH. Characterization of midazolam metabolism in locusts: the role of a CYP3A4-like enzyme in the formation of 1'-OH and 4-OH midazolam. Xenobiotica 2015. [PMID: 26207435 DOI: 10.3109/00498254.2015.1051604] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
1. The metabolism of midazolam was investigated in vivo in locusts in order to evaluate the presence of an enzyme with functionality similar to human CYP3A4/5. 2. Hydroxylated metabolites of midazolam identical to human metabolites were detected in locusts and the apparent affinities (Km values) were in the same range as reported in humans (in locusts: 7-23 and 33-85 µM for the formation of the 1'-OH and 4-OH metabolites, respectively). 3. The formation of hydroxylated metabolites could successfully be inhibited by co-administration of ketoconazole, a known CYP3A4/5 inhibitor. 4. Besides phase I metabolites, a number of conjugated metabolites were detected using high-resolution mass spectrometry. The most abundant metabolites detected were structurally identified by (1)H NMR as two N-glucosides. NMR analysis strongly suggested that the glycosylation occurred at the two nitrogens (either one in each case) of the imidazole ring. 5. Distribution of midazolam and the glucose conjugates were successfully measured using desorption electrospray mass spectrometry imaging revealing time-dependent changes in distribution over time. 6. In conclusion, it appears that an enzyme with functionality similar to human CYP3A4/5 is present in locusts. However, it appears that conjugation with glucose is the main detoxification pathway of midazolam in locusts.
Collapse
Affiliation(s)
| | | | - Sileshi Gizachew Wubshet
- b Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark , and
| | - Kenneth Thermann Kongstad
- b Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark , and
| | | | - Lassina Badolo
- c Department of Discovery ADME , H. Lundbeck A/S , Valby , Denmark
| | | |
Collapse
|
49
|
Deol P, Evans JR, Dhahbi J, Chellappa K, Han DS, Spindler S, Sladek FM. Soybean Oil Is More Obesogenic and Diabetogenic than Coconut Oil and Fructose in Mouse: Potential Role for the Liver. PLoS One 2015. [PMID: 26200659 PMCID: PMC4511588 DOI: 10.1371/journal.pone.0132672] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The obesity epidemic in the U.S. has led to extensive research into potential contributing dietary factors, especially fat and fructose. Recently, increased consumption of soybean oil, which is rich in polyunsaturated fatty acids (PUFAs), has been proposed to play a causal role in the epidemic. Here, we designed a series of four isocaloric diets (HFD, SO-HFD, F-HFD, F-SO-HFD) to investigate the effects of saturated versus unsaturated fat, as well as fructose, on obesity and diabetes. C57/BL6 male mice fed a diet moderately high in fat from coconut oil and soybean oil (SO-HFD, 40% kcal total fat) showed statistically significant increases in weight gain, adiposity, diabetes, glucose intolerance and insulin resistance compared to mice on a diet consisting primarily of coconut oil (HFD). They also had fatty livers with hepatocyte ballooning and very large lipid droplets as well as shorter colonic crypt length. While the high fructose diet (F-HFD) did not cause as much obesity or diabetes as SO-HFD, it did cause rectal prolapse and a very fatty liver, but no balloon injury. The coconut oil diet (with or without fructose) increased spleen weight while fructose in the presence of soybean oil increased kidney weight. Metabolomics analysis of the liver showed an increased accumulation of PUFAs and their metabolites as well as γ-tocopherol, but a decrease in cholesterol in SO-HFD. Liver transcriptomics analysis revealed a global dysregulation of cytochrome P450 (Cyp) genes in SO-HFD versus HFD livers, most notably in the Cyp3a and Cyp2c families. Other genes involved in obesity (e.g., Cidec, Cd36), diabetes (Igfbp1), inflammation (Cd63), mitochondrial function (Pdk4) and cancer (H19) were also upregulated by the soybean oil diet. Taken together, our results indicate that in mice a diet high in soybean oil is more detrimental to metabolic health than a diet high in fructose or coconut oil.
Collapse
Affiliation(s)
- Poonamjot Deol
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, California, United States of America
| | - Jane R. Evans
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, California, United States of America
| | - Joseph Dhahbi
- Department of Biochemistry, University of California, Riverside, Riverside, California, United States of America
| | - Karthikeyani Chellappa
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, California, United States of America
| | - Diana S. Han
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, California, United States of America
| | - Stephen Spindler
- Department of Biochemistry, University of California, Riverside, Riverside, California, United States of America
| | - Frances M. Sladek
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, California, United States of America
- * E-mail:
| |
Collapse
|
50
|
Choo EF, Woolsey S, DeMent K, Ly J, Messick K, Qin A, Takahashi R. Use of transgenic mouse models to understand the oral disposition and drug-drug interaction potential of cobimetinib, a MEK inhibitor. Drug Metab Dispos 2015; 43:864-9. [PMID: 25813936 DOI: 10.1124/dmd.115.063743] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 03/26/2015] [Indexed: 02/13/2025] Open
Abstract
Data from the clinical absolute bioavailability (F) study with cobimetinib suggested that F was lower than predicted based on its low hepatic extraction and good absorption. The CYP3A4 transgenic (Tg) mouse model with differential expression of CYP3A4 in the liver (Cyp3a(-/-)Tg-3A4Hep) or intestine (Cyp3a(-/-)Tg-3A4Int) and both liver and intestine (Cyp3a(-/-)Tg-3A4Hep/Int) were used to study the contribution of intestinal metabolism to the F of cobimetinib. In addition, the effect of CYP3A4 inhibition and induction on cobimetinib exposures was tested in the Cyp3a(-/-)Tg-3A4Hep/Int and PXR-CAR-CYP3A4/CYP3A7 mouse models, respectively. After i.v. administration of 1 mg/kg cobimetinib to wild-type [(WT) FVB], Cyp3a(-/-)Tg-3A4Hep, Cyp3a(-/-)Tg-3A4Int, or Cyp3a(-/-)Tg-3A4Hep/Int mice, clearance (CL) (26-35 ml/min/kg) was similar in the CYP3A4 transgenic and WT mice. After oral administration of 5 mg/kg cobimetinib, the area under the curve (AUC) values of cobimetinib in WT, Cyp3a(-/-)Tg-3A4Hep, Cyp3a(-/-)Tg-3A4Int, or Cyp3a(-/-)Tg-3A4Hep/Int mice were 1.35, 3.39, 1.04, and 0.701 μM⋅h, respectively. The approximately 80% lower AUC of cobimetinib in transgenic mice when intestinal CYP3A4 was present suggested that the intestinal first pass contributed to the oral CL of cobimetinib. Oxidative metabolites observed in human circulation were also observed in the transgenic mice. In drug-drug interaction (DDI) studies using Cyp3a(-/-)Tg-3A4Hep/Int mice, 8- and 4-fold increases in oral and i.v. cobimetinib exposure, respectively, were observed with itraconazole co-administration. In PXR-CAR-CYP3A4/CYP3A7 mice, rifampin induction decreased cobimetinib oral exposure by approximately 80%. Collectively, these data support the conclusion that CYP3A4 intestinal metabolism contributes to the oral disposition of cobimetinib and suggest that under certain circumstances the transgenic model may be useful in predicting clinical DDIs.
Collapse
Affiliation(s)
- Edna F Choo
- Genentech Inc., South San Francisco, California
| | | | | | - Justin Ly
- Genentech Inc., South San Francisco, California
| | | | - Ann Qin
- Genentech Inc., South San Francisco, California
| | | |
Collapse
|