1
|
Ailabouni AS, Singh DK, Thakur A, Boone EC, Gaedigk A, Paine MF, Prasad B. Quantitative Contributions of Hepatic and Renal Organic Cation Transporters to the Clinical Pharmacokinetic Cimetidine-Metformin Interaction. Clin Pharmacol Ther 2025. [PMID: 40098288 DOI: 10.1002/cpt.3639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
The widely prescribed oral anti-diabetic drug metformin is eliminated unchanged in the urine primarily through active tubular secretion. This process is mediated by organic cation transporter 2 (OCT2), an uptake transporter expressed on the basolateral membrane of renal proximal tubule cells. Metformin uptake into the liver, the site of action, is mediated by organic cation transporter 1 (OCT1), which is expressed on the sinusoidal membrane of hepatocytes. Sixteen healthy adults participated in a clinical pharmacokinetic drug-drug interaction study in which they were orally administered metformin (50 mg) as a dual OCT1/2 substrate alone (baseline) and with cimetidine (400 mg) as an OCT inhibitor. Relative to baseline, metformin systemic plasma exposure increased by 24% (p < 0.05) in the presence of cimetidine, which was accompanied by a disproportional decrease (8%) in metformin renal clearance (p = 0.005). Genetic variants of OCT1 and OCT2 moderately impacted the significance and magnitude of the interaction. Collectively, we hypothesized that the cimetidine-metformin interaction involves inhibition of hepatic OCT1 as well as renal OCT2. We tested this hypothesis by developing a physiologically based pharmacokinetic (PBPK) model and assessing potential OCT biomarkers in plasma and urine to gain mechanistic insight into the transporters involved in this interaction. The PBPK model predicted that cimetidine primarily inhibits hepatic OCT1 and, to a lesser extent, renal OCT2. The unchanged renal clearance of potential OCT2 biomarkers following cimetidine exposure supports a minimal role for renal OCT2 in this interaction.
Collapse
Affiliation(s)
- Anoud Sameer Ailabouni
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Dilip Kumar Singh
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Aarzoo Thakur
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Erin C Boone
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy Research Institute, Kansas City, Missouri, USA
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy Research Institute, Kansas City, Missouri, USA
- School of Medicine, Department of Pediatrics, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Mary F Paine
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
2
|
Ailabouni AS, Vijaywargi G, Subash S, Singh DK, Gaborik Z, Prasad B. Is N1-Methylnicotinamide a Good Organic Cation Transporter 2 (OCT2) Biomarker? Metabolites 2025; 15:80. [PMID: 39997705 PMCID: PMC11857448 DOI: 10.3390/metabo15020080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/15/2025] [Accepted: 01/25/2025] [Indexed: 02/26/2025] Open
Abstract
Background/Objectives: The impact of potential precipitant drugs on plasma or urinary exposure of endogenous biomarkers is emerging as an alternative approach to evaluating drug-drug interaction (DDI) liability. N1-Methylnicotinamide (NMN) has been proposed as a potential biomarker for renal organic cation transporter 2 (OCT2). NMN is synthesized in the liver from nicotinamide by nicotinamide N-methyltransferase (NNMT) and is subsequently metabolized by aldehyde oxidase (AO). Multiple clinical studies have shown a reduction in NMN plasma concentration following the administration of OCT inhibitors such as cimetidine, trimethoprim, and pyrimethamine, which contrasts with their inhibition of NMN renal clearance by OCT2. We hypothesized that OCT1-mediated NMN release from hepatocytes is inhibited by the administration of OCT inhibitors. Methods: Re-analysis of the reported NMN pharmacokinetics with and without OCT inhibitor exposure was performed. We assessed the effect of cimetidine on NMN uptake in OCT1-HEK293 cells and evaluated the potential confounding effects of cimetidine on enzymes involved in NMN formation and metabolism. Results: A re-analysis of previous NMN pharmacokinetic DDI data suggests that NMN plasma systemic exposure decreased by 17-41% during the first 4 h following different OCT inhibitor administration except dolutegravir. Our findings indicate that NMN uptake was significantly higher (by 2.5-fold) in OCT1-HEK293 cells compared to mock cells, suggesting that NMN is a substrate of OCT1. Additionally, our results revealed that cimetidine does not inhibit NNMT and AO activity. Conclusions: Our findings emphasize the limitations of using NMN as an OCT2 biomarker and reveal potential mechanisms behind the reduction in NMN plasma levels associated with OCT inhibitors. Instead, our data suggest that NMN could be tested further as a potential biomarker for OCT1 activity.
Collapse
Affiliation(s)
- Anoud Sameer Ailabouni
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (A.S.A.); (G.V.); (S.S.); (D.K.S.)
| | - Gautam Vijaywargi
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (A.S.A.); (G.V.); (S.S.); (D.K.S.)
| | - Sandhya Subash
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (A.S.A.); (G.V.); (S.S.); (D.K.S.)
| | - Dilip Kumar Singh
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (A.S.A.); (G.V.); (S.S.); (D.K.S.)
| | | | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (A.S.A.); (G.V.); (S.S.); (D.K.S.)
| |
Collapse
|
3
|
Karlsson IB, Ekdahl A, Etchingham-Coll H, Li XQ, Ericsson C, Ahlqvist M, Samuelsson K. Investigation of Biotransformation Pathways in a Chimeric Mouse with a Humanized Liver. Int J Mol Sci 2025; 26:1141. [PMID: 39940909 PMCID: PMC11818726 DOI: 10.3390/ijms26031141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Xenobiotics, including drugs, undergo metabolism to facilitate detoxification and excretion. Predicting a compound's metabolic fate before clinical trials is crucial for efficacy and safety. The existing methods rely on in vitro systems and in vivo animal testing. In vitro systems do not replicate the complexity of in vivo systems, and differences in biotransformation pathways between humans and nonclinical species may occur; thus, accurate predictions of human-specific drug metabolism are not always achieved. The aim of this study was to evaluate whether a chimeric mouse with a humanized liver, specifically the PXB-mouse, can mimic human metabolic profiles. PXB-mice have livers engrafted with up to 95% human hepatocytes. The biotransformation of 12 different small-molecule drugs were evaluated in PXB-mice (through analysis of blood and urine) and compared with the metabolism by hepatocytes from humans and mice and, when available, literature reports on human in vivo metabolism. The detected metabolites included major Phase I and II transitions, such as hydroxylation, and N- and O-dealkylation and glucuronidation. The metabolic patterns of the PXB-mice closely matched human in vivo data. It is also worth noting that the human hepatocytes formed most of the circulating metabolites, indicating that hepatocytes provide reliable predictions of human metabolic pathways. Thus, for drugs with human biotransformation pathways that are not observed in nonclinical species, the PXB-mouse model can be valuable in predicting human-specific metabolism.
Collapse
Affiliation(s)
- Isabella B. Karlsson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden; (A.E.); (H.E.-C.); (X.-Q.L.); (C.E.); (M.A.)
| | | | | | | | | | | | - Kristin Samuelsson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden; (A.E.); (H.E.-C.); (X.-Q.L.); (C.E.); (M.A.)
| |
Collapse
|
4
|
Rodrigues D, Wezalis S. Clinical Assessment of Drug Transporter Inhibition Using Biomarkers: Review of the Literature (2015-2024). J Clin Pharmacol 2025. [PMID: 39828904 DOI: 10.1002/jcph.6183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
As part of a narrative review of various publications describing the clinical use of urine- and plasma-based drug transporter biomarkers, it was determined that the utilization of coproporphyrin I, a hepatic organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 biomarker, has been reported for 28 different drug-drug interaction (DDI) perpetrator drugs. Similarly, biomarkers for liver organic cation transporter 1 (isobutyryl-l-carnitine, N = 7 inhibitors), renal organic cation transporter 2 and multidrug and toxin extrusion proteins (N1-methylnicotinamide, N = 13 inhibitors), renal organic anion transporter (OAT) 1 and 3 (pyridoxic acid, N = 7 inhibitors), and breast cancer resistance protein (riboflavin, N = 3 inhibitors) have also been described. Increased use of biomarkers has also been accompanied by modeling efforts to enable DDI predictions and development of multiplexed methods to facilitate their bioanalysis. Overall, there is consensus that exploratory biomarkers such as coproporphyrin I can be integrated into decision trees encompassing in vitro transporter inhibition data, DDI risk assessments, and follow-up Phase 1 studies. Therefore, sponsors can leverage biomarkers to evaluate dose-dependent inhibition of selected transporters, use them jointly with drug probes to deconvolute DDI mechanisms, and integrate in vitro data packages to establish calibrated (biomarker informed) DDI risk assessment cutoffs. Although transporter biomarker science has progressed, reflected by its inclusion in the recently issued International Council for Harmonisation DDI guidance document (M12), some biomarkers still require further validation. There is also a need for biomarkers that can differentiate specific transporters (e.g., OATP1B3 vs OATP1B1 and OAT1 vs OAT3).
Collapse
Affiliation(s)
- David Rodrigues
- Drug Metabolism and Nonclinical Pharmacokinetics, Translational Medicine, Incyte, Wilmington, DE, USA
| | - Stephanie Wezalis
- Drug Metabolism and Nonclinical Pharmacokinetics, Translational Medicine, Incyte, Wilmington, DE, USA
| |
Collapse
|
5
|
Ailabouni AS, Singh DK, Thakur A, Paine MF, Boone EC, Gaedigk A, Prasad B. Quantitative contributions of hepatic and renal organic cation transporters to the clinical pharmacokinetic cimetidine-metformin interaction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.624371. [PMID: 39605472 PMCID: PMC11601659 DOI: 10.1101/2024.11.19.624371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The widely prescribed oral anti-diabetic drug metformin is eliminated unchanged in the urine primarily through active tubular secretion. This process is mediated by organic cation transporter 2 (OCT2), an uptake transporter expressed on the basolateral membrane of renal proximal tubule cells. Metformin uptake into the liver, the site of action, is mediated by OCT1, which is expressed on the sinusoidal membrane of hepatocytes. Sixteen healthy adults participated in a clinical pharmacokinetic drug-drug interaction study in which they were orally administered metformin (50 mg) as a dual OCT1/2 substrate alone (baseline) and with cimetidine (400 mg) as an OCT inhibitor. Relative to baseline, metformin systemic plasma exposure increased by 24% ( p <0.05) in the presence of cimetidine, which was accompanied by a disproportional decrease (8%) in metformin renal clearance ( p =0.005). Genetic variants of OCT1 and OCT2 moderately impacted the significance and magnitude of the interaction. Collectively, we hypothesized that the cimetidine-metformin interaction involves inhibition of hepatic OCT1 as well as renal OCT2. We tested this hypothesis by developing a physiologically based pharmacokinetic (PBPK) model and assessing potential OCT biomarkers in plasma and urine to gain mechanistic insight into the transporters involved in this interaction. The PBPK model predicted that cimetidine primarily inhibits hepatic OCT1 and, to a lesser extent, renal OCT2. The unchanged renal clearance of potential OCT2 biomarkers following cimetidine exposure supports a minimal role for renal OCT2 in this interaction.
Collapse
|
6
|
Isaiah S, Loots DT, van Reenen M, Solomons R, van Elsland S, Tutu van Furth AM, van der Kuip M, Mason S. Urinary metabolic characterization of advanced tuberculous meningitis cases in a South African paediatric population. Front Mol Biosci 2024; 11:1253983. [PMID: 38560518 PMCID: PMC10978807 DOI: 10.3389/fmolb.2024.1253983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Tuberculous meningitis (TBM) is a severe form of tuberculosis with high neuro-morbidity and mortality, especially among the paediatric population (aged ≤12 years). Little is known of the associated metabolic changes. This study aimed to identify characteristic metabolic markers that differentiate severe cases of paediatric TBM from controls, through non-invasive urine collection. Urine samples selected for this study were from two paediatric groups. Group 1: controls (n = 44): children without meningitis, no neurological symptoms and from the same geographical region as group 2. Group 2: TBM cases (n = 13): collected from paediatric patients that were admitted to Tygerberg Hospital in South Africa on the suspicion of TBM, mostly severely ill; with a later confirmation of TBM. Untargeted 1H NMR-based metabolomics data of urine were generated, followed by statistical analyses via MetaboAnalyst (v5.0), and the identification of important metabolites. Twenty nine urinary metabolites were identified as characteristic of advanced TBM and categorized in terms of six dysregulated metabolic pathways: 1) upregulated tryptophan catabolism linked to an altered vitamin B metabolism; 2) perturbation of amino acid metabolism; 3) increased energy production-metabolic burst; 4) disrupted gut microbiota metabolism; 5) ketoacidosis; 6) increased nitrogen excretion. We also provide original biological insights into this biosignature of urinary metabolites that can be used to characterize paediatric TBM patients in a South African cohort.
Collapse
Affiliation(s)
- Simon Isaiah
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Mari van Reenen
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Regan Solomons
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Sabine van Elsland
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- MRC Centre for Global Infectious Disease Analysis, Imperial College London, London, United Kingdom
| | - A. Marceline Tutu van Furth
- Vrije Universiteit, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Centers, Emma Children’s Hospital, Amsterdam, Netherlands
| | - Martijn van der Kuip
- Vrije Universiteit, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Centers, Emma Children’s Hospital, Amsterdam, Netherlands
| | - Shayne Mason
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
7
|
Dutta T, Kapoor N, Mathew M, Chakraborty SS, Ward NP, Prieto-Farigua N, Falzone A, DeLany JP, Smith SR, Coen PM, DeNicola GM, Gardell SJ. Source of nicotinamide governs its metabolic fate in cultured cells, mice, and humans. Cell Rep 2023; 42:112218. [PMID: 36897780 DOI: 10.1016/j.celrep.2023.112218] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/17/2023] [Accepted: 02/17/2023] [Indexed: 03/11/2023] Open
Abstract
Metabolic routing of nicotinamide (NAM) to NAD+ or 1-methylnicotinamide (MeNAM) has impacts on human health and aging. NAM is imported by cells or liberated from NAD+. The fate of 2H4-NAM in cultured cells, mice, and humans was determined by stable isotope tracing. 2H4-NAM is an NAD+ precursor via the salvage pathway in cultured A549 cells and human PBMCs and in A549 cell xenografts and PBMCs from 2H4-NAM-dosed mice and humans, respectively. 2H4-NAM is a MeNAM precursor in A549 cell cultures and xenografts, but not isolated PBMCs. NAM released from NAD+ is a poor MeNAM precursor. Additional A549 cell tracer studies yielded further mechanistic insight. NAMPT activators promote NAD+ synthesis and consumption. Surprisingly, NAM liberated from NAD+ in NAMPT activator-treated A549 cells is also routed toward MeNAM production. Metabolic fate mapping of the dual NAM sources across the translational spectrum (cells, mice, humans) illuminates a key regulatory node governing NAD+ and MeNAM synthesis.
Collapse
Affiliation(s)
- Tumpa Dutta
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, USA.
| | - Nidhi Kapoor
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, USA
| | - Meril Mathew
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, USA
| | - Suban S Chakraborty
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, USA
| | - Nathan P Ward
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Nicolas Prieto-Farigua
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Aimee Falzone
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - James P DeLany
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, USA
| | - Steven R Smith
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, USA
| | - Paul M Coen
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, USA
| | - Gina M DeNicola
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Stephen J Gardell
- Translational Research Institute, AdventHealth Orlando, Orlando, FL 32804, USA.
| |
Collapse
|
8
|
Rendić SP, Crouch RD, Guengerich FP. Roles of selected non-P450 human oxidoreductase enzymes in protective and toxic effects of chemicals: review and compilation of reactions. Arch Toxicol 2022; 96:2145-2246. [PMID: 35648190 PMCID: PMC9159052 DOI: 10.1007/s00204-022-03304-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/26/2022] [Indexed: 12/17/2022]
Abstract
This is an overview of the metabolic reactions of drugs, natural products, physiological compounds, and other (general) chemicals catalyzed by flavin monooxygenase (FMO), monoamine oxidase (MAO), NAD(P)H quinone oxidoreductase (NQO), and molybdenum hydroxylase enzymes (aldehyde oxidase (AOX) and xanthine oxidoreductase (XOR)), including roles as substrates, inducers, and inhibitors of the enzymes. The metabolism and bioactivation of selected examples of each group (i.e., drugs, "general chemicals," natural products, and physiological compounds) are discussed. We identified a higher fraction of bioactivation reactions for FMO enzymes compared to other enzymes, predominately involving drugs and general chemicals. With MAO enzymes, physiological compounds predominate as substrates, and some products lead to unwanted side effects or illness. AOX and XOR enzymes are molybdenum hydroxylases that catalyze the oxidation of various heteroaromatic rings and aldehydes and the reduction of a number of different functional groups. While neither of these two enzymes contributes substantially to the metabolism of currently marketed drugs, AOX has become a frequently encountered route of metabolism among drug discovery programs in the past 10-15 years. XOR has even less of a role in the metabolism of clinical drugs and preclinical drug candidates than AOX, likely due to narrower substrate specificity.
Collapse
Affiliation(s)
| | - Rachel D Crouch
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, 37204, USA
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, USA
| |
Collapse
|
9
|
Rodrigues AD. Reimagining the Framework Supporting the Static Analysis of Transporter Drug Interaction Risk; Integrated Use of Biomarkers to Generate
Pan‐Transporter
Inhibition Signatures. Clin Pharmacol Ther 2022; 113:986-1002. [PMID: 35869864 DOI: 10.1002/cpt.2713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/14/2022] [Indexed: 11/11/2022]
Abstract
Solute carrier (SLC) transporters present as the loci of important drug-drug interactions (DDIs). Therefore, sponsors generate in vitro half-maximal inhibitory concentration (IC50 ) data and apply regulatory agency-guided "static" methods to assess DDI risk and the need for a formal clinical DDI study. Because such methods are conservative and high false-positive rates are likely (e.g., DDI study triggered when liver SLC R value ≥ 1.04 and renal SLC maximal unbound plasma (Cmax,u )/IC50 ratio ≥ 0.02), investigators have attempted to deploy plasma- and urine-based SLC biomarkers in phase I studies to de-risk DDI and obviate the need for drug probe-based studies. In this regard, it was possible to generate in-house in vitro SLC IC50 data for various clinically (biomarker)-qualified perpetrator drugs, under standard assay conditions, and then estimate "% inhibition" for each SLC and relate it empirically to published clinical biomarker data (area under the plasma concentration vs. time curve (AUC) ratio (AUCR, AUCinhibitor /AUCreference ) and % decrease in renal clearance (ΔCLrenal )). After such a "calibration" exercise, it was determined that only compounds with high R values (> 1.5) and Cmax,u /IC50 ratios (> 0.5) are likely to significantly modulate liver (AUCR > 1.25) and renal (ΔCLrenal > 25%) biomarkers and evoke DDI risk. The % inhibition approach supports integration of liver and renal SLC data and allows one to generate pan-SLC inhibition signatures for different test perpetrators (e.g., SLC % inhibition ranking). In turn, such signatures can guide the selection of the most appropriate individual (or combinations of) biomarkers for testing in phase I studies.
Collapse
Affiliation(s)
- A. David Rodrigues
- Pharmacokinetics & Drug Metabolism, Medicine Design, Worldwide Research & Development, Pfizer Inc Groton CT USA
| |
Collapse
|
10
|
Sato K, Sanoh S, Ishida Y, Tateno C, Ohta S, Kotake Y. Assessment of metabolic activation of felbamate in chimeric mice with humanized liver in combination with in vitro metabolic assays. J Toxicol Sci 2022; 47:277-288. [PMID: 35786679 DOI: 10.2131/jts.47.277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Felbamate (FBM) is an antiepileptic drug that has minimal toxicity in preclinical toxicological species but has a serious idiosyncratic drug toxicity (IDT) in humans. The formation of reactive metabolites is common among most drugs associated with IDT, and 2-phenylpropenal (2-PP) is believed to be the cause of IDT by FBM. It is important to consider the species difference in susceptibility to IDT between experimental animals and humans. In the present study, we used an in vitro and in vivo model system to reveal species difference in IDT of FBM. Human cytochrome P450 (CYP) and carboxylesterase (CES) expressing microsomes were used to clarify the isozymes involved in the metabolism of FBM. The remaining amount of FBM was significantly reduced in incubation with microsomes expressing human CYP2C8, 2C9, 2E1, and CES1c isozymes. Chimeric mice with humanized liver are expected to predict IDT in humans. Therefore, metabolite profiles in chimeric mice with humanized liver were investigated after administration of FBM. Metabolites after glutathione (GSH) conjugation of 2-phenylpropenal (2-PP), which is the reactive metabolite responsible for FBM-induced IDT, were detected in chimeric mice plasma and liver homogenate. Mass spectrometry imaging (MSI) visualizes distribution of FBM and endogenous GSH, and GSH levels in human hepatocyte were decreased after administration of FBM. In this study, we identified CYP and CES isozymes involved in the metabolism of FBM and confirmed reactive metabolite formation and subsequent decrease in GSH using humanized animal model. These results would provide useful information for the susceptibility to IDT between experimental animals and humans.
Collapse
Affiliation(s)
- Koya Sato
- Graduate School of Biomedical and Health Sciences, Hiroshima University.,Non-Clinical Regulatory Science, Applied Research & Operations, Astellas Pharma Inc
| | - Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University.,School of Pharmaceutical Sciences, Wakayama Medical University
| | - Yuji Ishida
- R&D Dept., PhoenixBio, Co., Ltd.,Research Center for Hepatology and Gastroenterology, Hiroshima University
| | - Chise Tateno
- School of Pharmaceutical Sciences, Wakayama Medical University.,R&D Dept., PhoenixBio, Co., Ltd.,Research Center for Hepatology and Gastroenterology, Hiroshima University
| | - Shigeru Ohta
- Graduate School of Biomedical and Health Sciences, Hiroshima University.,School of Pharmaceutical Sciences, Wakayama Medical University
| | - Yaichiro Kotake
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| |
Collapse
|
11
|
Hayat F, Sonavane M, Makarov MV, Trammell SAJ, McPherson P, Gassman NR, Migaud ME. The Biochemical Pathways of Nicotinamide-Derived Pyridones. Int J Mol Sci 2021; 22:ijms22031145. [PMID: 33498933 PMCID: PMC7866226 DOI: 10.3390/ijms22031145] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
As catabolites of nicotinamide possess physiological relevance, pyridones are often included in metabolomics measurements and associated with pathological outcomes in acute kidney injury (AKI). Pyridones are oxidation products of nicotinamide, its methylated form, and its ribosylated form. While they are viewed as markers of over-oxidation, they are often wrongly reported or mislabeled. To address this, we provide a comprehensive characterization of these catabolites of vitamin B3, justify their nomenclature, and differentiate between the biochemical pathways that lead to their generation. Furthermore, we identify an enzymatic and a chemical process that accounts for the formation of the ribosylated form of these pyridones, known to be cytotoxic. Finally, we demonstrate that the ribosylated form of one of the pyridones, the 4-pyridone-3-carboxamide riboside (4PYR), causes HepG3 cells to die by autophagy; a process that occurs at concentrations that are comparable to physiological concentrations of this species in the plasma in AKI patients.
Collapse
Affiliation(s)
- Faisal Hayat
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (F.H.); (M.S.)
- Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA; (M.V.M.); (P.M.); (N.R.G.)
| | - Manoj Sonavane
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (F.H.); (M.S.)
- Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA; (M.V.M.); (P.M.); (N.R.G.)
- Department of Physiology & Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Mikhail V. Makarov
- Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA; (M.V.M.); (P.M.); (N.R.G.)
| | - Samuel A. J. Trammell
- Novo Nordisk Foundation, Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Pamela McPherson
- Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA; (M.V.M.); (P.M.); (N.R.G.)
| | - Natalie R. Gassman
- Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA; (M.V.M.); (P.M.); (N.R.G.)
- Department of Physiology & Cell Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Marie E. Migaud
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA; (F.H.); (M.S.)
- Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA; (M.V.M.); (P.M.); (N.R.G.)
- Correspondence:
| |
Collapse
|
12
|
Paragas EM, Choughule K, Jones JP, Barr JT. Enzyme Kinetics, Pharmacokinetics, and Inhibition of Aldehyde Oxidase. Methods Mol Biol 2021; 2342:257-284. [PMID: 34272698 DOI: 10.1007/978-1-0716-1554-6_10] [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] [Indexed: 06/13/2023]
Abstract
Aldehyde oxidase (AO) has emerged as an important drug metabolizing enzyme over the last decade. Several compounds have failed in the clinic because the clearance or toxicity was underestimated by preclinical species. Human AO is much more active than rodent AO, and dogs do not have functional AO. Metabolic products from AO-catalyzed oxidation are generally nonreactive and often they have much lower solubility. AO metabolism is not limited to oxidation as AO can also catalyze reduction of oxygen and nitrite. Reduction of oxygen leads to the reactive oxygen species (ROS) superoxide radical anion and hydrogen peroxide. Reduction of nitrite leads to the formation of nitric oxide with potential pharmacological implications. AO is also reported to catalyze the reductive metabolism of nitro-compounds, N-oxides, sulfoxides, isoxazoles, isothiazoles, nitrite, and hydroxamic acids. These reductive transformations may cause toxicity due to the formation of reactive metabolites. Moreover, the inhibition kinetics are complex, and multiple probe substrates should be used when assessing the potential for DDIs. Finally, AO appears to be amenable to computational predictions of both regioselectivity and rates of reaction, which holds promise for virtual screening.
Collapse
Affiliation(s)
- Erickson M Paragas
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Kanika Choughule
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck, Boston, MA, USA
| | - Jeffrey P Jones
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | - John T Barr
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck, South San Francisco, CA, USA.
| |
Collapse
|
13
|
Miyake T, Kimoto E, Luo L, Mathialagan S, Horlbogen LM, Ramanathan R, Wood LS, Johnson JG, Le VH, Vourvahis M, Rodrigues AD, Muto C, Furihata K, Sugiyama Y, Kusuhara H. Identification of Appropriate Endogenous Biomarker for Risk Assessment of Multidrug and Toxin Extrusion Protein-Mediated Drug-Drug Interactions in Healthy Volunteers. Clin Pharmacol Ther 2020; 109:507-516. [PMID: 32866300 PMCID: PMC7891601 DOI: 10.1002/cpt.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/26/2020] [Indexed: 12/27/2022]
Abstract
Endogenous biomarkers are emerging to advance clinical drug‐drug interaction (DDI) risk assessment in drug development. Twelve healthy subjects received a multidrug and toxin exclusion protein (MATE) inhibitor (pyrimethamine, 10, 25, and 75 mg) in a crossover fashion to identify an appropriate endogenous biomarker to assess MATE1/2‐K‐mediated DDI in the kidneys. Metformin (500 mg) was also given as reference probe drug for MATE1/2‐K. In addition to the previously reported endogenous biomarker candidates (creatinine and N1‐methylnicotinamide (1‐NMN)), N1‐methyladenosine (m1A) was included as novel biomarkers. 1‐NMN and m1A presented as superior MATE1/2‐K biomarkers since changes in their renal clearance (CLr) along with pyrimethamine dose were well‐correlated with metformin CLr changes. The CLr of creatinine was reduced by pyrimethamine, however, its changes poorly correlated with metformin CLr changes. Nonlinear regression analysis (CLr vs. mean total concentration of pyrimethamine in plasma) yielded an estimate of the inhibition constant (Ki) of pyrimethamine and the fraction of the clearance pathway sensitive to pyrimethamine. The in vivoKi value thus obtained was further converted to unbound Ki using plasma unbound fraction of pyrimethamine, which was comparable to the in vitroKi for MATE1 (1‐NMN) and MATE2‐K (1‐NMN and m1A). It is concluded that 1‐NMN and m1A CLr can be leveraged as quantitative MATE1/2‐K biomarkers for DDI risk assessment in healthy volunteers.
Collapse
Affiliation(s)
- Takeshi Miyake
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Emi Kimoto
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Lina Luo
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | | | | | - Ragu Ramanathan
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Linda S Wood
- Clinical Pharmacogenomics Laboratory, Early Clinical Development, Pfizer Inc, Groton, Connecticut, USA
| | - Jillian G Johnson
- Clinical Pharmacogenomics Laboratory, Early Clinical Development, Pfizer Inc, Groton, Connecticut, USA
| | - Vu H Le
- Biostatics, Pfizer Inc., Collegeville, Pennsylvania, USA
| | | | - A David Rodrigues
- ADME Sciences, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Chieko Muto
- Clinical Pharmacology, Pfizer R&D Japan, Tokyo, Japan
| | | | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama, Kanagawa, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
14
|
Manevski N, King L, Pitt WR, Lecomte F, Toselli F. Metabolism by Aldehyde Oxidase: Drug Design and Complementary Approaches to Challenges in Drug Discovery. J Med Chem 2019; 62:10955-10994. [PMID: 31385704 DOI: 10.1021/acs.jmedchem.9b00875] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aldehyde oxidase (AO) catalyzes oxidations of azaheterocycles and aldehydes, amide hydrolysis, and diverse reductions. AO substrates are rare among marketed drugs, and many candidates failed due to poor pharmacokinetics, interspecies differences, and adverse effects. As most issues arise from complex and poorly understood AO biology, an effective solution is to stop or decrease AO metabolism. This perspective focuses on rational drug design approaches to modulate AO-mediated metabolism in drug discovery. AO biological aspects are also covered, as they are complementary to chemical design and important when selecting the experimental system for risk assessment. The authors' recommendation is an early consideration of AO-mediated metabolism supported by computational and in vitro experimental methods but not an automatic avoidance of AO structural flags, many of which are versatile and valuable building blocks. Preferably, consideration of AO-mediated metabolism should be part of the multiparametric drug optimization process, with the goal to improve overall drug-like properties.
Collapse
Affiliation(s)
- Nenad Manevski
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - Lloyd King
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - William R Pitt
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - Fabien Lecomte
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - Francesca Toselli
- UCB BioPharma , Chemin du Foriest 1 , 1420 Braine-l'Alleud , Belgium
| |
Collapse
|
15
|
Cheshmazar N, Dastmalchi S, Terao M, Garattini E, Hamzeh-Mivehroud M. Aldehyde oxidase at the crossroad of metabolism and preclinical screening. Drug Metab Rev 2019; 51:428-452. [DOI: 10.1080/03602532.2019.1667379] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Narges Cheshmazar
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mineko Terao
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Enrico Garattini
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Dalvie D, Di L. Aldehyde oxidase and its role as a drug metabolizing enzyme. Pharmacol Ther 2019; 201:137-180. [PMID: 31128989 DOI: 10.1016/j.pharmthera.2019.05.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/27/2019] [Indexed: 11/29/2022]
Abstract
Aldehyde oxidase (AO) is a cytosolic enzyme that belongs to the family of structurally related molybdoflavoproteins like xanthine oxidase (XO). The enzyme is characterized by broad substrate specificity and marked species differences. It catalyzes the oxidation of aromatic and aliphatic aldehydes and various heteroaromatic rings as well as reduction of several functional groups. The references to AO and its role in metabolism date back to the 1950s, but the importance of this enzyme in the metabolism of drugs has emerged in the past fifteen years. Several reviews on the role of AO in drug metabolism have been published in the past decade indicative of the growing interest in the enzyme and its influence in drug metabolism. Here, we present a comprehensive monograph of AO as a drug metabolizing enzyme with emphasis on marketed drugs as well as other xenobiotics, as substrates and inhibitors. Although the number of drugs that are primarily metabolized by AO are few, the impact of AO on drug development has been extensive. We also discuss the effect of AO on the systemic exposure and clearance these clinical candidates. The review provides a comprehensive analysis of drug discovery compounds involving AO with the focus on developmental candidates that were reported in the past five years with regards to pharmacokinetics and toxicity. While there is only one known report of AO-mediated clinically relevant drug-drug interaction (DDI), a detailed description of inhibitors and inducers of AO known to date has been presented here and the potential risks associated with DDI. The increasing recognition of the importance of AO has led to significant progress in predicting the site of AO-mediated metabolism using computational methods. Additionally, marked species difference in expression of AO makes it is difficult to predict human clearance with high confidence. The progress made towards developing in vivo, in vitro and in silico approaches for predicting AO metabolism and estimating human clearance of compounds that are metabolized by AO have also been discussed.
Collapse
Affiliation(s)
- Deepak Dalvie
- Drug Metabolism and Pharmacokinetics, Celgene Corporation, 10300, Campus Point Drive, San Diego, CA 92121, USA.
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, UK
| |
Collapse
|
17
|
Affiliation(s)
- Christine Beedham
- Honorary Senior Lecturer, Faculty of Life Sciences, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
18
|
Takaoka N, Sanoh S, Okuda K, Kotake Y, Sugahara G, Yanagi A, Ishida Y, Tateno C, Tayama Y, Sugihara K, Kitamura S, Kurosaki M, Terao M, Garattini E, Ohta S. Inhibitory effects of drugs on the metabolic activity of mouse and human aldehyde oxidases and influence on drug–drug interactions. Biochem Pharmacol 2018; 154:28-38. [DOI: 10.1016/j.bcp.2018.04.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
|
19
|
Amano T, Fukami T, Ogiso T, Hirose D, Jones JP, Taniguchi T, Nakajima M. Identification of enzymes responsible for dantrolene metabolism in the human liver: A clue to uncover the cause of liver injury. Biochem Pharmacol 2018. [DOI: 10.1016/j.bcp.2018.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
20
|
A pharmaceutical industry perspective on transporter and CYP-mediated drug–drug interactions: kidney transporter biomarkers. Bioanalysis 2018; 10:625-631. [DOI: 10.4155/bio-2017-0265] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
21
|
Müller F, Sharma A, König J, Fromm MF. Biomarkers for In Vivo Assessment of Transporter Function. Pharmacol Rev 2018; 70:246-277. [PMID: 29487084 DOI: 10.1124/pr.116.013326] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Drug-drug interactions are a major concern not only during clinical practice, but also in drug development. Due to limitations of in vitro-in vivo predictions of transporter-mediated drug-drug interactions, multiple clinical Phase I drug-drug interaction studies may become necessary for a new molecular entity to assess potential drug interaction liabilities. This is a resource-intensive process and exposes study participants, who frequently are healthy volunteers without benefit from study treatment, to the potential risks of a new drug in development. Therefore, there is currently a major interest in new approaches for better prediction of transporter-mediated drug-drug interactions. In particular, researchers in the field attempt to identify endogenous compounds as biomarkers for transporter function, such as hexadecanedioate, tetradecanedioate, coproporphyrins I and III, or glycochenodeoxycholate sulfate for hepatic uptake via organic anion transporting polypeptide 1B or N1-methylnicotinamide for multidrug and toxin extrusion protein-mediated renal secretion. We summarize in this review the currently proposed biomarkers and potential limitations of the substances identified to date. Moreover, we suggest criteria based on current experiences, which may be used to assess the suitability of a biomarker for transporter function. Finally, further alternatives and supplemental approaches to classic drug-drug interaction studies are discussed.
Collapse
Affiliation(s)
- Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| | - Ashish Sharma
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| |
Collapse
|
22
|
Sanoh S, Ohta S. [Contribution of chimeric mice with a humanized liver to the evaluation of pharmacology, toxicity, and pharmacokinetics in drug discovery and development]. Nihon Yakurigaku Zasshi 2018; 151:213-220. [PMID: 29760366 DOI: 10.1254/fpj.151.213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
To develop new drugs with high efficacy and safety, it is important to predict the pharmacological, toxicological, and pharmacokinetic profiles of drug candidates in humans. Chimeric mice with a humanized liver are mice in which human hepatocytes have been transplanted, such that mouse liver cells are replaced with human hepatocytes; these mice have been used as prediction models. Studies performed thus far indicate that chimeric mice with a humanized liver can be used for the prediction of human-specific metabolite formation and plasma concentration-time curves for several drugs. Furthermore, studies advocate the utility of chimeric mice with a humanized liver for modelling drug-induced hepatotoxicity and disease such as hepatitis virus infection in safety and pharmacological evaluations respectively. Taken together, these findings indicate that chimeric mice with a humanized liver can be used to evaluate the relationship between pharmacokinetics, toxicity, and efficacy; the contribution by active metabolites may also be assessed. In recent years, new and improved animal models have been developed to overcome the disadvantages of chimeric mice with a humanized liver. It is expected that their usefulness for optimization of drug candidates and translational research in drug discovery and development will further increase.
Collapse
Affiliation(s)
- Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Shigeru Ohta
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| |
Collapse
|
23
|
Kücükgöze G, Terao M, Garattini E, Leimkühler S. Direct Comparison of the Enzymatic Characteristics and Superoxide Production of the Four Aldehyde Oxidase Enzymes Present in Mouse. Drug Metab Dispos 2017; 45:947-955. [PMID: 28526768 DOI: 10.1124/dmd.117.075937] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/15/2017] [Indexed: 02/13/2025] Open
Abstract
Aldehyde oxidases (AOXs) are molybdoflavoenzymes with an important role in the metabolism and detoxification of heterocyclic compounds and aliphatic as well as aromatic aldehydes. The enzymes use oxygen as the terminal electron acceptor and produce reduced oxygen species during turnover. Four different enzymes, mAOX1, mAOX3, mAOX4, and mAOX2, which are the products of distinct genes, are present in the mouse. A direct and simultaneous comparison of the enzymatic properties and characteristics of the four enzymes has never been performed. In this report, the four catalytically active mAOX enzymes were purified after heterologous expression in Escherichia coli The kinetic parameters of the four mouse AOX enzymes were determined and compared with the use of six predicted substrates of physiologic and toxicological interest, i.e., retinaldehyde, N1-methylnicotinamide, pyridoxal, vanillin, 4-(dimethylamino)cinnamaldehyde (p-DMAC), and salicylaldehyde. While retinaldehyde, vanillin, p-DMAC, and salycilaldehyde are efficient substrates for the four mouse AOX enzymes, N1-methylnicotinamide is not a substrate of mAOX1 or mAOX4, and pyridoxal is not metabolized by any of the purified enzymes. Overall, mAOX1, mAOX2, mAOX3, and mAOX4 are characterized by significantly different KM and kcat values for the active substrates. The four mouse AOXs are also characterized by quantitative differences in their ability to produce superoxide radicals. With respect to this last point, mAOX2 is the enzyme generating the largest rate of superoxide radicals of around 40% in relation to moles of substrate converted, and mAOX1, the homolog to the human enzyme, produces a rate of approximately 30% of superoxide radicals with the same substrate.
Collapse
Affiliation(s)
- Gökhan Kücükgöze
- Institut für Biochemie and Biologie, Universität Potsdam, Potsdam, Germany (G.K., S.L.); Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy (M.T., E.G.)
| | - Mineko Terao
- Institut für Biochemie and Biologie, Universität Potsdam, Potsdam, Germany (G.K., S.L.); Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy (M.T., E.G.)
| | - Enrico Garattini
- Institut für Biochemie and Biologie, Universität Potsdam, Potsdam, Germany (G.K., S.L.); Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy (M.T., E.G.)
| | - Silke Leimkühler
- Institut für Biochemie and Biologie, Universität Potsdam, Potsdam, Germany (G.K., S.L.); Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy (M.T., E.G.)
| |
Collapse
|
24
|
Szafarz M, Kus K, Walczak M, Zakrzewska A, Niemczak M, Pernak J, Chlopicki S. Pharmacokinetic Profile of 1-Methylnicotinamide Nitrate in Rats. J Pharm Sci 2017; 106:1412-1418. [DOI: 10.1016/j.xphs.2017.01.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/17/2022]
|
25
|
Jensen KG, Jacobsen AM, Bundgaard C, Nilausen DØ, Thale Z, Chandrasena G, Jørgensen M. Lack of Exposure in a First-in-Man Study Due to Aldehyde Oxidase Metabolism: Investigated by Use of 14C-microdose, Humanized Mice, Monkey Pharmacokinetics, and In Vitro Methods. Drug Metab Dispos 2017; 45:68-75. [PMID: 27737930 DOI: 10.1124/dmd.116.072793] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/12/2016] [Indexed: 11/22/2022] Open
Abstract
Inclusion of a microdose of 14C-labeled drug in the first-in-man study of new investigational drugs and subsequent analysis by accelerator mass spectrometry has become an integrated part of drug development at Lundbeck. It has been found to be highly informative with regard to investigations of the routes and rates of excretion of the drug and the human metabolite profiles according to metabolites in safety testing guidance and also when additional metabolism-related issues needed to be addressed. In the first-in-man study with the NCE Lu AF09535, contrary to anticipated, surprisingly low exposure was observed when measuring the parent compound using conventional bioanalysis. Parallel accelerator mass spectrometry analysis revealed that the low exposure was almost exclusively attributable to extensive metabolism. The metabolism observed in humans was mediated via a human specific metabolic pathway, whereas an equivalent extent of metabolism was not observed in preclinical species. In vitro, incubation studies in human liver cytosol revealed involvement of aldehyde oxidase (AO) in the biotransformation of Lu AF09535. In vivo, substantially lower plasma exposure of Lu AF09535 was observed in chimeric mice with humanized livers compared with control animals. In addition, Lu AF09535 exhibited very low oral bioavailability in monkeys despite relatively low clearance after intravenous administration in contrast to the pharmacokinetics in rats and dogs, both showing low clearance and high bioavailability. The in vitro and in vivo methods applied were proved useful for identifying and evaluating AO-dependent metabolism. Different strategies to integrate these methods for prediction of in vivo human clearance of AO substrates were evaluated.
Collapse
Affiliation(s)
- Klaus Gjervig Jensen
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Anne-Marie Jacobsen
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Christoffer Bundgaard
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Dorrit Østergaard Nilausen
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Zia Thale
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Gamini Chandrasena
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| | - Martin Jørgensen
- Drug ADME Research (K.G.J., M.J.), Department of Drug Metabolism (A.J.), Discovery DMPK (C.B.), Clinical Pharmacology (D.Ø.N.), and Department of Bioanalysis (Z.T.), H. Lundbeck A/S, Valby Denmark; and Discovery Chemistry & DMPK, Lundbeck Research, New Jersey (G.C.)
| |
Collapse
|
26
|
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
|
27
|
Sanoh S, Naritomi Y, Fujimoto M, Sato K, Kawamura A, Horiguchi A, Sugihara K, Kotake Y, Ohshita H, Tateno C, Horie T, Kitamura S, Ohta S. Predictability of plasma concentration–time curves in humans using single-species allometric scaling of chimeric mice with humanized liver. Xenobiotica 2015; 45:605-14. [DOI: 10.3109/00498254.2015.1007112] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
28
|
Liu M, Wang H, Zeng H, Li CJ. Silver(I) as a widely applicable, homogeneous catalyst for aerobic oxidation of aldehydes toward carboxylic acids in water-"silver mirror": From stoichiometric to catalytic. SCIENCE ADVANCES 2015; 1:e1500020. [PMID: 26601150 PMCID: PMC4643818 DOI: 10.1126/sciadv.1500020] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/02/2015] [Indexed: 05/30/2023]
Abstract
The first example of a homogeneous silver(I)-catalyzed aerobic oxidation of aldehydes in water is reported. More than 50 examples of different aliphatic and aromatic aldehydes, including natural products, were tested, and all of them successfully underwent aerobic oxidation to give the corresponding carboxylic acids in extremely high yields. The reaction conditions are very mild and greener, requiring only a very low silver(I) catalyst loading, using atmospheric oxygen as the oxidant and water as the solvent, and allowing gram-scale oxidation with only 2 mg of our catalyst. Chromatography is completely unnecessary for purification in most cases.
Collapse
|
29
|
Sanoh S, Tayama Y, Sugihara K, Kitamura S, Ohta S. Significance of aldehyde oxidase during drug development: Effects on drug metabolism, pharmacokinetics, toxicity, and efficacy. Drug Metab Pharmacokinet 2015; 30:52-63. [DOI: 10.1016/j.dmpk.2014.10.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/03/2014] [Accepted: 10/03/2014] [Indexed: 12/28/2022]
|
30
|
Müller F, Pontones CA, Renner B, Mieth M, Hoier E, Auge D, Maas R, Zolk O, Fromm MF. N(1)-methylnicotinamide as an endogenous probe for drug interactions by renal cation transporters: studies on the metformin-trimethoprim interaction. Eur J Clin Pharmacol 2015; 71:85-94. [PMID: 25552403 DOI: 10.1007/s00228-014-1770-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE N(1)-methylnicotinamide (NMN) was proposed as an in vivo probe for drug interactions involving renal cation transporters, which, for example, transport the oral antidiabetic drug metformin, based on a study with the inhibitor pyrimethamine. The role of NMN for predicting other interactions with involvement of renal cation transporters (organic cation transporter 2, OCT2; multidrug and toxin extrusion proteins 1 and 2-K, MATE1 and MATE2-K) is unclear. METHODS We determined inhibition of metformin or NMN transport by trimethoprim using cell lines expressing OCT2, MATE1, or MATE2-K. Moreover, a randomized, open-label, two-phase crossover study was performed in 12 healthy volunteers. In each phase, 850 mg metformin hydrochloride was administered p.o. in the evening of day 4 and in the morning of day 5. In phase B, 200 mg trimethoprim was administered additionally p.o. twice daily for 5 days. Metformin pharmacokinetics and effects (measured by OGTT) and NMN pharmacokinetics were determined. RESULTS Trimethoprim inhibited metformin transport with K i values of 27.2, 6.3, and 28.9 μM and NMN transport with IC50 values of 133.9, 29.1, and 0.61 μM for OCT2, MATE1, and MATE2-K, respectively. In the clinical study, trimethoprim increased metformin area under the plasma concentration-time curve (AUC) by 29.5 % and decreased metformin and NMN renal clearances by 26.4 and 19.9 %, respectively (p ≤ 0.01). Moreover, decreases of NMN and metformin renal clearances due to trimethoprim correlated significantly (r S=0.727, p=0.010). CONCLUSIONS These data on the metformin-trimethoprim interaction support the potential utility of N(1)-methylnicotinamide as an endogenous probe for renal drug-drug interactions with involvement of renal cation transporters.
Collapse
Affiliation(s)
- Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstr. 17, 91054, Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Samuelsson K, Pickup K, Sarda S, Foster JR, Randall K, Abrahamsson A, Jacobsen M, Weidolf L, Wilson I. Troglitazone metabolism and transporter effects in chimeric mice: a comparison between chimeric humanized and chimeric murinized FRG mice. Xenobiotica 2014; 44:186-95. [PMID: 24417752 DOI: 10.3109/00498254.2013.879237] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
1. The biotransformation, hepatic transporter and blood chemistry effects of troglitazone were investigated following 7 days of dosing at 600 mg/kg/day to chimeric murinized or humanized FRG mice, Mo-FRG and Hu-FRG mice, respectively. 2. Clinical chemistry and histopathology analysis revealed a significant drop in humanization over the time course of the study for the Hu-FRG mice but no significant changes associated with troglitazone treatment in either the Mo-FRG or the Hu-FRG models. No changes in transporter expression in livers of these mice were observed. Oxidative and conjugative metabolic pathways were identified with a 15- to 18-fold increase in formation of troglitazone sulfate in the Hu-FRG mice compared with the Mo-FRG mice in blood and bile, respectively. This resembles the troglitazone metabolism in human and these data are comparable with the formation of this metabolite in the chimeric uPA(+/+)/SCID mice. 3. However, larger amounts of troglitazone glucuronide were also observed in the Hu-FRG mouse compared with the Mo-FRG mouse which may be an effect of the drop in humanization of the Hu-FRG mouse during the study. 4. Highly humanized mice have a considerable potential in providing a useful first insight into circulating human metabolites of candidate drugs metabolized in the liver.
Collapse
|
32
|
Kitamura S, Sugihara K. Current status of prediction of drug disposition and toxicity in humans using chimeric mice with humanized liver. Xenobiotica 2013; 44:123-34. [PMID: 24329499 DOI: 10.3109/00498254.2013.868062] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
1. Human-chimeric mice with humanized liver have been constructed by transplantation of human hepatocytes into several types of mice having genetic modifications that injure endogenous liver cells. Here, we focus on liver urokinase-type plasminogen activator-transgenic severe combined immunodeficiency (uPA/SCID) mice, which are the most widely used human-chimeric mice. Studies so far indicate that drug metabolism, drug transport, pharmacological effects and toxicological action in these mice are broadly similar to those in humans. 2. Expression of various drug-metabolizing enzymes is known to be different between humans and rodents. However, the expression pattern of cytochrome P450, aldehyde oxidase and phase II enzymes in the liver of human-chimeric mice resembles that in humans, not that in the host mice. 3. Metabolism of various drugs, including S-warfarin, zaleplon, ibuprofen, naproxen, coumarin, troglitazone and midazolam, in human-chimeric mice is mediated by human drug-metabolizing enzymes, not by host mouse enzymes, and thus resembles that in humans. 4. Pharmacological and toxicological effects of various drugs in human-chimeric mice are also similar to those in humans. 5. The current consensus is that chimeric mice with humanized liver are useful to predict drug metabolism catalyzed by cytochrome P450, aldehyde oxidase and phase II enzymes in humans in vivo and in vitro. Some remaining issues are discussed in this review.
Collapse
Affiliation(s)
- Shigeyuki Kitamura
- Department of Environmental Science, Nihon Pharmaceutical University , Saitama , Japan and
| | | |
Collapse
|
33
|
Igawa Y, Fujitani T, Shah B, Oo C, Kanai Y. In vitroandin vivometabolism of a novel chymase inhibitor, SUN13834, and the predictability of human metabolism using mice with humanized liver. Xenobiotica 2013; 44:154-63. [DOI: 10.3109/00498254.2013.865857] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Sanoh S, Ohta S. Chimeric mice transplanted with human hepatocytes as a model for prediction of human drug metabolism and pharmacokinetics. Biopharm Drug Dispos 2013; 35:71-86. [DOI: 10.1002/bdd.1864] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 09/09/2013] [Accepted: 09/21/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Seigo Sanoh
- Graduate School of Biomedical and Health Sciences; Hiroshima University; Hiroshima Japan
| | - Shigeru Ohta
- Graduate School of Biomedical and Health Sciences; Hiroshima University; Hiroshima Japan
| |
Collapse
|
35
|
Tanoue C, Sugihara K, Uramaru N, Tayama Y, Watanabe Y, Horie T, Ohta S, Kitamura S. Prediction of human metabolism of the sedative-hypnotic zaleplon using chimeric mice transplanted with human hepatocytes. Xenobiotica 2013; 43:956-62. [DOI: 10.3109/00498254.2013.788232] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
36
|
Patel M, Vasaya MM, Asker D, Parsons RB. HPLC–UV method for measuring nicotinamide N-methyltransferase activity in biological samples: Evidence for substrate inhibition kinetics. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 921-922:87-95. [DOI: 10.1016/j.jchromb.2013.01.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 12/20/2012] [Accepted: 01/09/2013] [Indexed: 12/01/2022]
|
37
|
Sanoh S, Horiguchi A, Sugihara K, Kotake Y, Tayama Y, Uramaru N, Ohshita H, Tateno C, Horie T, Kitamura S, Ohta S. Predictability of metabolism of ibuprofen and naproxen using chimeric mice with human hepatocytes. Drug Metab Dispos 2012; 40:2267-72. [PMID: 22936315 DOI: 10.1124/dmd.112.047555] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Prediction of human drug metabolism is important for drug development. Recently, the number of new drug candidates metabolized by not only cytochrome P450 (P450) but also non-P450 has been increasing. It is necessary to consider species differences in drug metabolism between humans and experimental animals. We examined species differences of drug metabolism, especially between humans and rats, for ibuprofen and (S)-naproxen as nonsteroidal anti-inflammatory drugs, which are metabolized by P450 and UDP-glucuronosyltransferase, sulfotransferase, and amino acid N-acyltransferase for taurine conjugation in liver, using human chimeric mice (h-PXB mice) repopulated with human hepatocytes and rat chimeric mice (r-PXB mice) transplanted with rat hepatocytes. We performed the direct comparison of excretory metabolites in urine between h-PXB mice and reported data for humans as well as between r-PXB mice and rats after administration of ibuprofen and (S)-naproxen. Good agreement for urinary metabolites (percentage of dose) was observed not only between humans and h-PXB mice but also between rats and r-PXB mice. Therefore, the metabolic profiles in humans and rats reflected those in h-PXB mice and r-PXB mice. Our results indicated that h-PXB mice should be helpful for predicting the quantitative metabolic profiles of drugs mediated by P450 and non-P450 in liver, and r-PXB mice should be helpful for evaluation of species differences in these metabolic enzymes.
Collapse
Affiliation(s)
- Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Samuelsson K, Pickup K, Sarda S, Swales JG, Morikawa Y, Schulz-Utermoehl T, Hutchison M, Wilson ID. Pharmacokinetics and metabolism of midazolam in chimeric mice with humanised livers. Xenobiotica 2012; 42:1128-37. [DOI: 10.3109/00498254.2012.689888] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Sanoh S, Horiguchi A, Sugihara K, Kotake Y, Tayama Y, Ohshita H, Tateno C, Horie T, Kitamura S, Ohta S. Prediction of in vivo hepatic clearance and half-life of drug candidates in human using chimeric mice with humanized liver. Drug Metab Dispos 2012; 40:322-8. [PMID: 22048522 DOI: 10.1124/dmd.111.040923] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Accurate prediction of pharmacokinetics (PK) parameters in humans from animal data is difficult for various reasons, including species differences. However, chimeric mice with humanized liver (PXB mice; urokinase-type plasminogen activator/severe combined immunodeficiency mice repopulated with approximately 80% human hepatocytes) have been developed. The expression levels and metabolic activities of cytochrome P450 (P450) and non-P450 enzymes in the livers of PXB mice are similar to those in humans. In this study, we examined the predictability for human PK parameters from data obtained in PXB mice. Elimination of selected drugs involves multiple metabolic pathways mediated not only by P450 but also by non-P450 enzymes, such as UDP-glucuronosyltransferase, sulfotransferase, and aldehyde oxidase in liver. Direct comparison between in vitro intrinsic clearance (CL(int,in vitro)) in PXB mice hepatocytes and in vivo intrinsic clearance (CL(int,in vivo)) in humans, calculated based on a well stirred model, showed a moderate correlation (r² = 0.475, p = 0.009). However, when CL(int,in vivo) values in humans and PXB mice were compared similarly, there was a good correlation (r² = 0.754, p = 1.174 × 10⁻⁴). Elimination half-life (t(1/2)) after intravenous administration also showed a good correlation (r² = 0.886, p = 1.506 × 10⁻⁴) between humans and PXB mice. The rank order of CL and t(1/2) in human could be predicted at least, although it may not be possible to predict absolute values due to rather large prediction errors. Our results indicate that in vitro and in vivo experiments with PXB mice should be useful at least for semiquantitative prediction of the PK characteristics of candidate drugs in humans.
Collapse
Affiliation(s)
- Seigo Sanoh
- Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553 Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sanoh S, Nozaki K, Murai H, Terashita S, Teramura T, Ohta S. Prediction of human metabolism of FK3453 by aldehyde oxidase using chimeric mice transplanted with human or rat hepatocytes. Drug Metab Dispos 2012; 40:76-82. [PMID: 21984595 DOI: 10.1124/dmd.111.041954] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
During drug development, it is important to predict the activities of multiple metabolic enzymes, not only cytochrome P450 (P450) but also non-P450 enzymes, such as conjugative enzymes and aldehyde oxidase (AO). In this study, we focused on prediction of AO-mediated human metabolism and pharmacokinetics (PK) of 6-(2-amino-4-phenylpyrimidine-5-yl)-2-isopropylpyridazin-3(2H)-one (FK3453) (Astellas Pharma Inc.), the development of which was suspended due to extremely low exposure in human, despite good oral bioavailability in rat and dog. We examined species difference in oxidative metabolism of the aminopyrimidine moiety of FK3453, catalyzed by AO, using human-chimeric mice with humanized liver (h-PXB mice) and rat-chimeric mice (r-PXB mice) transplanted with rat hepatocytes. AO activity of h-PXB mouse hepatocytes was higher than that of r-PXB mouse hepatocytes. Moreover, higher concentrations of human-specific AO-generated FK3453 metabolite A-M were detected in urine and feces after administration of FK3453 to h-PXB mice versus r-PXB mice. The total clearance of h-PXB mice was 2-fold higher than that of r-PXB mice. These results agreed reasonably well with the metabolism and PK profiles of FK3453 in human and rat. Our results indicated that h-PXB mice should be helpful for predicting the metabolic profile of drugs in humans, and the use of both h-PXB and r-PXB mice should be helpful for evaluation of species differences of AO metabolic activity.
Collapse
Affiliation(s)
- Seigo Sanoh
- Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553 Japan.
| | | | | | | | | | | |
Collapse
|
41
|
Schulz-Utermoehl T, Sarda S, Foster JR, Jacobsen M, Kenna JG, Morikawa Y, Salmu J, Gross G, Wilson ID. Evaluation of the pharmacokinetics, biotransformation and hepatic transporter effects of troglitazone in mice with humanized livers. Xenobiotica 2011; 42:503-17. [DOI: 10.3109/00498254.2011.640716] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
42
|
Ivers DJ, Veum TL. Effect of graded levels of niacin supplementation of a semipurified diet on energy and nitrogen balance, growth performance, diarrhea occurrence, and niacin metabolite excretion by growing swine. J Anim Sci 2011; 90:282-8. [PMID: 21873540 DOI: 10.2527/jas.2011-4035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Thirty-six crossbred barrows with an average initial age of 42 d and BW of 13.8 kg were placed in individual metabolism crates in a 35-d experiment to evaluate the supplementation of a semipurified diet with graded levels of crystalline niacin. Response criteria were energy and N balance, growth performance, occurrence of niacin deficiency diarrhea, and urinary excretion of the niacin metabolite N(1)-methyl-2-pyridone-5-carboxylamide (PYR). The basal diet met the true ileal Trp requirement of growing swine, and supplementation with 6, 10, 14, 18, 22, or 44 mg of niacin/kg made 6 treatments. Pigs were observed for scours twice daily, and pig BW and feed consumption were determined weekly. Total urine collections and fecal grab samples were made twice daily from each pig from d 28 to 35. Pigs fed the diet containing 14 mg of niacin/kg absorbed and retained more (P < 0.05) grams of N/d, had a greater N digestibility (%, P < 0.05), a greater ADFI and ADG (P < 0.10), and no diarrhea (P < 0.05) compared with pigs fed the diet containing 6 mg of niacin/kg, and pigs fed the diet containing 10 mg of niacin/kg were intermediate in ADG. There were no additional improvements in the response criteria with niacin supplementation greater than 14 mg/kg. Urinary PYR criteria (mg/L and mg/d) were greater (P < 0.001) for pigs fed the diet containing 44 mg of niacin/kg than for pigs fed the diets containing 6 to 22 mg of niacin/kg. However, urinary PYR criteria for pigs fed the diets containing 6 to 22 mg of niacin/kg did not differ from each other, indicating that PYR was not a sensitive indicator of niacin status for growing swine. Niacin treatment did not affect the percentages of N retained/N absorbed, N retained/N intake, DE, or ME. In conclusion, 14 mg of crystalline niacin/kg of semipurified diet adequate in Trp was the minimum concentration of niacin that maximized N utilization and growth performance, and prevented niacin deficiency diarrhea of growing swine in the current experiment. Because practical feed ingredients may be sources of available endogenous niacin, supplementation of practical diets with 100% of the current NRC requirement for niacin should provide adequate niacin for growing swine.
Collapse
Affiliation(s)
- D J Ivers
- Agricultural Experiment Station and Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | | |
Collapse
|
43
|
Garattini E, Terao M. Increasing recognition of the importance of aldehyde oxidase in drug development and discovery. Drug Metab Rev 2011; 43:374-86. [DOI: 10.3109/03602532.2011.560606] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
44
|
Zhou SS, Li D, Sun WP, Guo M, Lun YZ, Zhou YM, Xiao FC, Jing LX, Sun SX, Zhang LB, Luo N, Bian FN, Zou W, Dong LB, Zhao ZG, Li SF, Gong XJ, Yu ZG, Sun CB, Zheng CL, Jiang DJ, Li ZN. Nicotinamide overload may play a role in the development of type 2 diabetes. World J Gastroenterol 2009; 15:5674-84. [PMID: 19960564 PMCID: PMC2789220 DOI: 10.3748/wjg.15.5674] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether nicotinamide overload plays a role in type 2 diabetes.
METHODS: Nicotinamide metabolic patterns of 14 diabetic and 14 non-diabetic subjects were compared using HPLC. Cumulative effects of nicotinamide and N1-methylnicotinamide on glucose metabolism, plasma H2O2 levels and tissue nicotinamide adenine dinucleotide (NAD) contents of adult Sprague-Dawley rats were observed. The role of human sweat glands and rat skin in nicotinamide metabolism was investigated using sauna and burn injury, respectively.
RESULTS: Diabetic subjects had significantly higher plasma N1-methylnicotinamide levels 5 h after a 100-mg nicotinamide load than the non-diabetic subjects (0.89 ± 0.13 μmol/L vs 0.6 ± 0.13 μmol/L, P < 0.001). Cumulative doses of nicotinamide (2 g/kg) significantly increased rat plasma N1-methylnicotinamide concentrations associated with severe insulin resistance, which was mimicked by N1-methylnicotinamide. Moreover, cumulative exposure to N1-methylnicotinamide (2 g/kg) markedly reduced rat muscle and liver NAD contents and erythrocyte NAD/NADH ratio, and increased plasma H2O2 levels. Decrease in NAD/NADH ratio and increase in H2O2 generation were also observed in human erythrocytes after exposure to N1-methylnicotinamide in vitro. Sweating eliminated excessive nicotinamide (5.3-fold increase in sweat nicotinamide concentration 1 h after a 100-mg nicotinamide load). Skin damage or aldehyde oxidase inhibition with tamoxifen or olanzapine, both being notorious for impairing glucose tolerance, delayed N1-methylnicotinamide clearance.
CONCLUSION: These findings suggest that nicotinamide overload, which induced an increase in plasma N1-methylnicotinamide, associated with oxidative stress and insulin resistance, plays a role in type 2 diabetes.
Collapse
|
45
|
|
46
|
Inoue T, Sugihara K, Ohshita H, Horie T, Kitamura S, Ohta S. Prediction of Human Disposition toward S-3H-Warfarin using Chimeric Mice with Humanized Liver. Drug Metab Pharmacokinet 2009; 24:153-60. [DOI: 10.2133/dmpk.24.153] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Inoue T, Nitta K, Sugihara K, Horie T, Kitamura S, Ohta S. CYP2C9-catalyzed metabolism of S-warfarin to 7-hydroxywarfarin in vivo and in vitro in chimeric mice with humanized liver. Drug Metab Dispos 2008; 36:2429-33. [PMID: 18784266 DOI: 10.1124/dmd.108.022830] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Chimeric mice having humanized livers were constructed by transplantation of human hepatocytes. In this study, we investigated whether these mice have a capacity for drug metabolism similar to that of humans by examining hydroxylation of S-warfarin, which is predominantly metabolized to S-7-hydroxywarfarin, catalyzed by CYP2C9, in humans but not mice. The 7-hydroxylating activity of chimeric mouse liver microsomes toward S-warfarin was approximately 10-fold higher than that of control (urokinase-type plasminogen activator-transgenic severe combined immunodeficient) mice. The 7-hydroxylase activity of chimeric mouse liver microsomes was markedly inhibited by sulfaphenazole, as was that of human liver microsomes, whereas the activity of control mice was unaffected. The CYP2C isoform in chimeric mouse liver was also confirmed to be the human isoform, CYP2C9, by immunoblot analysis. In the present in vivo study, the level of S-7-hydroxywarfarin in plasma of chimeric mice was approximately 7-fold higher than that in control mice, in agreement with the in vitro data. Thus, the CYP2C isoform in chimeric mice functions in vivo and in vitro as a human isoform, CYP2C9. These results suggest that chimeric mice with humanized liver could be useful for predicting drug metabolism in humans, at least regarding CYP2C9-dependent metabolism.
Collapse
Affiliation(s)
- Tae Inoue
- Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan
| | | | | | | | | | | |
Collapse
|