1
|
Buchl SC, Kim HN, Hur B, Simon WL, Langley MR, Sung J, Scarisbrick IA. Delayed atorvastatin delivery promotes recovery after experimental spinal cord injury. Neurotherapeutics 2025:e00517. [PMID: 39755500 DOI: 10.1016/j.neurot.2024.e00517] [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: 09/06/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025] Open
Abstract
Spinal cord injury (SCI) significantly alters gene expression, potentially impeding functional recovery. This study investigated the effects of atorvastatin, a widely prescribed cholesterol-lowering drug, on gene expression and functional recovery in a chronic murine SCI model. Female C57BL/6J mice underwent moderate 0.25 mm lateral compression SCI and received daily atorvastatin (10 mg/kg) or vehicle-only injections from two weeks post-injury for four weeks. Sensorimotor functions were assessed using the Basso Mouse Scale (BMS), its subscore, and the inclined plane test. RNA sequencing of spinal cord tissues identified robust transcriptomic changes from SCI and a smaller subset from atorvastatin treatment. Atorvastatin enhanced sensorimotor recovery within two weeks of treatment initiation, with effects persisting to the experimental endpoint. Pathway analysis showed atorvastatin enriched neural regeneration processes including Fatty Acid Transport, Axon Guidance, and the Endocannabinoid Developing Neuron Pathway; improved mitochondrial function via increased TCA Cycle II and reduced Mitochondrial Dysfunction; and decreased Inhibition of Matrix Metalloproteases. Key gene drivers included Fabp7, Unc5c, Rest, and Klf4. Together, these results indicate atorvastatin's potential in chronic SCI recovery, especially where already indicated for cardiovascular protection.
Collapse
Affiliation(s)
- Samuel C Buchl
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Ha Neui Kim
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Benjamin Hur
- Microbiomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA; Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Whitney L Simon
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Monica R Langley
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Jaeyun Sung
- Microbiomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA; Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Isobel A Scarisbrick
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Uno Y, Shimizu M, Yamazaki H. A variety of cytochrome P450 enzymes and flavin-containing monooxygenases in dogs and pigs commonly used as preclinical animal models. Biochem Pharmacol 2024; 228:116124. [PMID: 38490520 DOI: 10.1016/j.bcp.2024.116124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/29/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024]
Abstract
Drug oxygenation is mainly mediated by cytochromes P450 (P450s, CYPs) and flavin-containing monooxygenases (FMOs). Polymorphic variants of P450s and FMOs are known to influence drug metabolism. Species differences exist in terms of drug metabolism and can be important when determining the contributions of individual enzymes. The success of research into drug-metabolizing enzymes and their impacts on drug discovery and development has been remarkable. Dogs and pigs are often used as preclinical animal models. This research update provides information on P450 and FMO enzymes in dogs and pigs and makes comparisons with their human enzymes. Newly identified dog CYP3A98, a testosterone 6β- and estradiol 16α-hydroxylase, is abundantly expressed in small intestine and is likely the major CYP3A enzyme in small intestine, whereas dog CYP3A12 is the major CYP3A enzyme in liver. The roles of recently identified dog CYP2J2 and pig CYP2J33/34/35 were investigated. FMOs have been characterized in humans and several other species including dogs and pigs. P450 and FMO family members have been characterized also in cynomolgus macaques and common marmosets. P450s have industrial applications and have been the focus of attention of many pharmaceutical companies. The techniques used to investigate the roles of P450/FMO enzymes in drug oxidation and clinical treatments have not yet reached maturity and require further development. The findings summarized here provide a foundation for understanding individual pharmacokinetic and toxicological results in dogs and pigs as preclinical models and will help to further support understanding of the molecular mechanisms of human P450/FMO functionality.
Collapse
Affiliation(s)
- Yasuhiro Uno
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima 890-0065, Japan
| | - Makiko Shimizu
- Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Hiroshi Yamazaki
- Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan.
| |
Collapse
|
3
|
Yamazaki H, Shimizu M. Species Specificity and Selection of Models for Drug Oxidations Mediated by Polymorphic Human Enzymes. Drug Metab Dispos 2023; 51:123-129. [PMID: 35772770 DOI: 10.1124/dmd.121.000742] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/28/2022] [Accepted: 06/01/2022] [Indexed: 01/03/2023] Open
Abstract
Many drug oxygenations are mainly mediated by polymorphic cytochromes P450 (P450s) and also by flavin-containing monooxygenases (FMOs). More than 50 years of research on P450/FMO-mediated drug oxygenations have clarified their catalytic roles. The natural product coumarin causes hepatotoxicity in rats via the reactive coumarin 3,4-epoxide, a reaction catalyzed by P450 1A2; however, coumarin undergoes rapid 7-hydroxylation by polymorphic P450 2A6 in humans. The primary oxidation product of the teratogen thalidomide in rats is deactivated 5'-hydroxythalidomide plus sulfate and glucuronide conjugates; however, similar 5'-hydroxythalidomide and 5-hydroxythalidomide are formed in rabbits in vivo. Thalidomide causes human P450 3A enzyme induction in liver (and placenta) and is also activated in vitro and in vivo by P450 3A through the primary human metabolite 5-hydroxythalidomide (leading to conjugation with glutathione/nonspecific proteins). Species differences exist in terms of drug metabolism in rodents and humans, and such differences can be very important when determining the contributions of individual enzymes. The approaches used for investigating the roles of human P450 and FMO enzymes in understanding drug oxidations and clinical therapy have not yet reached maturity and still require further development. SIGNIFICANCE STATEMENT: Drug oxidations in animals and humans mediated by P450s and FMOs are important for understanding the pharmacological properties of drugs, such as the species-dependent teratogenicity of the reactive metabolites of thalidomide and the metabolism of food-derived odorous trimethylamine to non-odorous (but proatherogenic) trimethylamine N-oxide. Recognized differences exist in terms of drug metabolism between rodents, non-human primates, and humans, and such differences are important when determining individual liver enzyme contributions with substrates in in vitro and in vivo systems.
Collapse
Affiliation(s)
- Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan
| |
Collapse
|
4
|
Formation of Reactive Metabolites of Benzbromarone in Humanized-Liver Mice. Drug Metab Pharmacokinet 2022; 47:100467. [DOI: 10.1016/j.dmpk.2022.100467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/21/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022]
|
5
|
Guengerich FP. Roles of cytochrome P450 enzymes in pharmacology and toxicology: Past, present, and future. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:1-47. [PMID: 35953152 PMCID: PMC9869358 DOI: 10.1016/bs.apha.2021.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The development of the cytochrome P450 (P450) field has been remarkable in the areas of pharmacology and toxicology, particularly in drug development. Today it is possible to use the knowledge base and relatively straightforward assays to make intelligent predictions about drug disposition prior to human dosing. Much is known about the structures, regulation, chemistry of catalysis, and the substrate and inhibitor specificity of human P450s. Many aspects of drug-drug interactions and side effects can be understood in terms of P450s. This knowledge has also been useful in pharmacy practice, as well as in the pharmaceutical industry and medical practice. However, there are still basic and practical questions to address regarding P450s and their roles in pharmacology and toxicology. Another aspect is the discovery of drugs that inhibit P450 to treat diseases.
Collapse
Affiliation(s)
- F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, United States.
| |
Collapse
|
6
|
Kuwagata M, Hasegawa T, Takashima H, Shimizu M, Kitajima S, Yamazaki H. Pharmacokinetics of primary metabolites 5-hydroxythalidomide and 5'-hydroxythalidomide formed after oral administration of thalidomide in the rabbit, a thalidomide-sensitive species. J Toxicol Sci 2021; 46:553-560. [PMID: 34853241 DOI: 10.2131/jts.46.553] [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
The teratogenicity of the chemotherapeutic drug thalidomide is species-specific and affects humans, non-human primates, and rabbits. The primary oxidation of thalidomide in previously investigated rodents predominantly resulted in the formation of deactivated 5'-hydroxythalidomide. In the current study, similar in vivo biotransformations to 5-hydroxythalidomide and 5'-hydroxythalidomide were confirmed by the analysis of blood plasma from male rabbits, a thalidomide-sensitive species, after oral administration of thalidomide (2.0 mg/kg). Similar levels of thalidomide in seminal plasma and in blood plasma were detected using liquid chromatography-tandem mass spectrometry at 4 hr and 7 hr after oral doses in male rabbits. Seminal plasma concentrations of 5-hydroxythalidomide and 5'-hydroxythalidomide were also seen in male rabbits in a roughly similar time-dependent manner to those in the blood plasma after oral doses of thalidomide (2.0 mg/kg). Furthermore, the values generated by a simplified physiologically based pharmacokinetic rabbit model were in agreement with the measured in vivo blood plasma data under metabolic ratios of 0.01 for the hepatic intrinsic clearance of thalidomide to both unconjugated 5-hydroxythalidomide and 5'-hydroxythalidomide. These results suggest that metabolic activation of thalidomide may be dependent on rabbit liver enzymes just it was for cytochrome P450 enzymes in humanized-liver mice; in contrast, rodent livers predominantly mediate biotransformation of thalidomide to 5'-hydroxythalidomide. A developmental toxicity test system with experimental animals that involves intravaginal exposures to the chemotherapeutic drug thalidomide via semen should be considered in the future.
Collapse
Affiliation(s)
- Makiko Kuwagata
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences
| | | | | | | | - Satoshi Kitajima
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences
| | | |
Collapse
|
7
|
Miura T, Kamiya Y, Hina S, Kobayashi Y, Murayama N, Shimizu M, Yamazaki H. Metabolic profiles of coumarin in human plasma extrapolated from a rat data set with a simplified physiologically based pharmacokinetic model. J Toxicol Sci 2020; 45:695-700. [PMID: 33132243 DOI: 10.2131/jts.45.695] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Coumarin is a dietary-derived substance that is extensively metabolized by human liver to excretable 7-hydroxycoumarin. Although coumarin under daily dietary consumption is generally regarded as nontoxic, the substance is of toxicological and clinical interest because of its potential association with hepatotoxicity, which is especially evident in rats. In this study, the pharmacokinetics of coumarin were modeled after virtual oral administration in humans. The adjusted monitoring equivalents of coumarin, along with the biotransformation of coumarin to o-hydroxyphenylacetic acid (via 3,4-epoxidation) based on reported plasma concentrations from rat studies, were scaled to human coumarin equivalents using known species allometric scaling factors. Using rat and human liver preparations, data on the rapid in vitro metabolic clearance for humans (~50-fold faster than in rats) were obtained for in vitro-in vivo extrapolation. For human physiologically based pharmacokinetic (PBPK) modeling, the metabolic ratios to o-hydroxyphenylacetic acid and 7-hydroxycoumarin were set at minor (0.1) and major (0.9) levels for the total disappearance of coumarin. The resulting modeled plasma concentration curves in humans generated by simple PBPK models were consistent with reported simulated coumarin maximum concentrations. These results provide basic information to simulate plasma levels of coumarin and its primary metabolite 7-hydroxycoumarin or its secondary activated metabolite o-hydroxyphenylacetic acid (via 3,4-epoxidation) resulting from dietary foodstuff consumption. Under the current assumptions, little toxicological impact of coumarin was evident in humans, thereby indicating the usefulness of forward dosimetry using PBPK modeling for human risk assessment.
Collapse
|
8
|
Toda A, Shimizu M, Uehara S, Sasaki T, Miura T, Mogi M, Utoh M, Suemizu H, Yamazaki H. Plasma and hepatic concentrations of acetaminophen and its primary conjugates after oral administrations determined in experimental animals and humans and extrapolated by pharmacokinetic modeling. Xenobiotica 2020; 51:316-323. [PMID: 33179995 DOI: 10.1080/00498254.2020.1849872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Plasma concentrations of acetaminophen, its glucuronide and sulfate conjugates, and cysteinyl acetaminophen were experimentally determined after oral administrations of 10 mg/kg in humanised-liver mice, control mice, rats, common marmosets, cynomolgus monkeys, and minipigs; the results were compared with reported human pharmacokinetic data. Among the animals tested, only rats predominantly converted acetaminophen to sulfate conjugates, rather than glucuronide conjugates. In contrast, the values of area under the plasma concentration curves of acetaminophen, its glucuronide and sulfate conjugates, and cysteinyl acetaminophen after oral administration of acetaminophen in marmosets and minipigs were consistent with those reported in humans under the present conditions. Physiologically based pharmacokinetic (PBPK) models (consisting of the gut, liver, and central compartments) for acetaminophen and its primary metabolite could reproduce and estimate, respectively, the plasma and hepatic concentrations of acetaminophen in experimental animals and humans after single virtual oral doses. The values of area under the curves of hepatic concentrations of acetaminophen estimated using PBPK models were correlated with the measured levels of cysteinyl acetaminophen (a deactivated metabolite) in plasma fractions in these species. Consequently, using simple PBPK models and plasma data to predict hepatic chemical concentrations after oral doses could be helpful as an indicator of in vivo possible hepatotoxicity of chemicals such as acetaminophen.
Collapse
Affiliation(s)
- Akiko Toda
- Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Wakayama , Japan
| | - Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , Tokyo , Japan
| | - Shotaro Uehara
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , Tokyo , Japan.,Laboratory Animal Research Department, Central Institute for Experimental Animals , Kawasaki , Japan
| | - Tatsuro Sasaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , Tokyo , Japan
| | - Tomonori Miura
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , Tokyo , Japan
| | - Masayuki Mogi
- Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Wakayama , Japan.,Drug Safety Research Laboratories, Shin Nippon Biomedical Laboratories, Ltd., Kagoshima , Japan
| | - Masahiro Utoh
- Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Wakayama , Japan.,Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , Tokyo , Japan.,Scientific Affairs Division, Shin Nippon Biomedical Laboratories, Ltd., Tokyo , Japan
| | - Hiroshi Suemizu
- Laboratory Animal Research Department, Central Institute for Experimental Animals , Kawasaki , Japan
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , Tokyo , Japan
| |
Collapse
|
9
|
Ogawa SI, Shimizu M, Kamiya Y, Uehara S, Suemizu H, Yamazaki H. Increased plasma concentrations of an antidyslipidemic drug pemafibrate co-administered with rifampicin or cyclosporine A in cynomolgus monkeys genotyped for the organic anion transporting polypeptide 1B1. Drug Metab Pharmacokinet 2020; 35:354-360. [DOI: 10.1016/j.dmpk.2020.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/02/2020] [Accepted: 03/24/2020] [Indexed: 12/21/2022]
|
10
|
Kamiya Y, Otsuka S, Miura T, Yoshizawa M, Nakano A, Iwasaki M, Kobayashi Y, Shimizu M, Kitajima M, Shono F, Funatsu K, Yamazaki H. Physiologically Based Pharmacokinetic Models Predicting Renal and Hepatic Concentrations of Industrial Chemicals after Virtual Oral Doses in Rats. Chem Res Toxicol 2020; 33:1736-1751. [PMID: 32500706 DOI: 10.1021/acs.chemrestox.0c00009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recently developed high-throughput in vitro assays in combination with computational models could provide alternatives to animal testing. The purpose of the present study was to model the plasma, hepatic, and renal pharmacokinetics of approximately 150 structurally varied types of drugs, food components, and industrial chemicals after virtual external oral dosing in rats and to determine the relationship between the simulated internal concentrations in tissue/plasma and their lowest-observed-effect levels. The model parameters were based on rat plasma data from the literature and empirically determined pharmacokinetics measured after oral administrations to rats carried out to evaluate hepatotoxic or nephrotic potentials. To ensure that the analyzed substances exhibited a broad diversity of chemical structures, their structure-based location in the chemical space underwent projection onto a two-dimensional plane, as reported previously, using generative topographic mapping. A high-throughput in silico one-compartment model and a physiologically based pharmacokinetic (PBPK) model consisting of chemical receptor (gut), metabolizing (liver), central (main), and excreting (kidney) compartments were developed in parallel. For 159 disparate chemicals, the maximum plasma concentrations and the areas under the concentration-time curves obtained by one-compartment models and modified simple PBPK models were closely correlated. However, there were differences between the PBPK modeled and empirically obtained hepatic/renal concentrations and plasma maximal concentrations/areas under the concentration-time curves of the 159 chemicals. For a few compounds, the lowest-observed-effect levels were available for hepatotoxicity and nephrotoxicity in the Hazard Evaluation Support System Integrated Platform in Japan. The areas under the renal or hepatic concentration-time curves estimated using PBPK modeling were inversely associated with these lowest-observed-effect levels. Using PBPK forward dosimetry could provide the plasma/tissue concentrations of drugs and chemicals after oral dosing, thereby facilitating estimates of nephrotoxic or hepatotoxic potential as a part of the risk assessment.
Collapse
Affiliation(s)
- Yusuke Kamiya
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| | - Shohei Otsuka
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| | - Tomonori Miura
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| | - Manae Yoshizawa
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| | - Ayane Nakano
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| | - Miyu Iwasaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| | - Yui Kobayashi
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| | - Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| | - Masato Kitajima
- Fujitsu Kyusyu Systems, Higashi-hie, Hakata-ku, Fukuoka 812-0007, Japan
| | - Fumiaki Shono
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kimito Funatsu
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| |
Collapse
|
11
|
Yamazaki H, Kamiya Y. Extrapolation of Hepatic Concentrations of Industrial Chemicals Using Pharmacokinetic Models to Predict Hepatotoxicity. Toxicol Res 2019; 35:295-301. [PMID: 31636840 PMCID: PMC6791659 DOI: 10.5487/tr.2019.35.4.295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/12/2019] [Accepted: 08/26/2019] [Indexed: 11/20/2022] Open
Abstract
In this review, we describe the absorption rates (Caco-2 cell permeability) and hepatic/plasma pharmacokinetics of 53 diverse chemicals estimated by modeling virtual oral administration in rats. To ensure that a broad range of chemical structures is present among the selected substances, the properties described by 196 chemical descriptors in a chemoinformatics tool were calculated for 50,000 randomly selected molecules in the original chemical space. To allow visualization, the resulting chemical space was projected onto a two-dimensional plane using generative topographic mapping. The calculated absorbance rates of the chemicals based on cell permeability studies were found to be inversely correlated to the no-observed-effect levels for hepatoxicity after oral administration, as obtained from the Hazard Evaluation Support System Integrated Platform in Japan (r = -0.88, p < 0.01, n = 27). The maximum plasma concentrations and the areas under the concentration-time curves (AUC) of a varied selection of chemicals were estimated using two different methods: simple one-compartment models (i.e., high-throughput toxicokinetic models) and simplified physiologically based pharmacokinetic (PBPK) modeling consisting of chemical receptor (gut), metabolizing (liver), and central (main) compartments. The results obtained from the two methods were consistent. Although the maximum concentrations and AUC values of the 53 chemicals roughly correlated in the liver and plasma, inconsistencies were apparent between empirically measured concentrations and the PBPK-modeled levels. The lowest-observed-effect levels and the virtual hepatic AUC values obtained using PBPK models were inversely correlated (r = -0.78, p < 0.05, n = 7). The present simplified PBPK models could estimate the relationships between hepatic/plasma concentrations and oral doses of general chemicals using both forward and reverse dosimetry. These methods are therefore valuable for estimating hepatotoxicity.
Collapse
Affiliation(s)
- Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Tokyo, Japan
| | - Yusuke Kamiya
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
12
|
Notsu Y, Shimizu M, Sasaki T, Nakano A, Ota M, Yoshida S, Yamazaki H. Simple pharmacokinetic models accounting for drug monitoring results of atomoxetine and its 4-hydroxylated metabolites in Japanese pediatric patients genotyped for cytochrome P450 2D6. Drug Metab Pharmacokinet 2019; 35:191-200. [PMID: 32184039 DOI: 10.1016/j.dmpk.2019.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/30/2019] [Accepted: 08/19/2019] [Indexed: 01/15/2023]
Abstract
Atomoxetine is an approved medicine for attention-deficit/hyperactivity disorder and a cytochrome P450 2D6 (CYP2D6) probe substrate. Simple physiologically based pharmacokinetic (PBPK) models and compartment models were set up to account for drug monitoring results of 33 Japanese patients (6-15 years of age) to help establish the correct dosage for the evaluation of clinical outcomes. The steady-state one-point drug monitoring data for the most participants indicated the extensive biotransformation of atomoxetine to 4-hydroxyatomoxetine under individually prescribed doses of atomoxetine. However, 5 participants (with impaired CYP2D6 activity scores based on the CYP2D6 genotypes) showed high plasma concentrations of atomoxetine (0.53-1.5 μM) compared with those of total 4-hydroxyatomoxetine (0.49-1.4 μM). Results from full PBPK models using the in-built Japanese pediatric system of software Simcyp, one-compartment models, and new simple PBPK models (using parameters that reflected the subjects' small body size and normal/reduced CYP2D6-dependent clearance) could overlay one-point measured drug/metabolite plasma concentrations from almost common 28 participants within threefold ranges. Validated one-compartment or simple PBPK models can be used to predict steady-state plasma concentrations of atomoxetine and/or its primary metabolites in Japanese pediatric patients (>6 years) who took a variety of individualized doses in a clinical setting.
Collapse
Affiliation(s)
- Yuki Notsu
- Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan
| | - Makiko Shimizu
- Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan
| | - Tatsuro Sasaki
- Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan
| | - Ayane Nakano
- Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan
| | - Miki Ota
- Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan
| | - Sayaka Yoshida
- Nara Prefecture General Medical Center, Nara, 630-8581, Japan
| | - Hiroshi Yamazaki
- Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan.
| |
Collapse
|
13
|
Chen M, Chen Y, Wang X, Zhou Y. Quantitative determination of talatisamine and its pharmacokinetics and bioavailability in mouse plasma by UPLC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1124:180-187. [PMID: 31207562 DOI: 10.1016/j.jchromb.2019.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/17/2019] [Accepted: 06/07/2019] [Indexed: 11/17/2022]
Abstract
Talatisamine, as the efficacy ingredient of Aconitum, was known as a novel specific blocker for the delayed rectifier K+ channels in rat hippocampal neurons. In this study, a rapid, selective and reproducible UPLC-MS/MS separation method was established and fully validated for the quantitative determination of talatisamine levels in ICR (Institute of Cancer Research) mouse blood. A total of 24 healthy male ICR mice were divided into four groups that was administered talatisamine via intravenous at a dose of 1 mg/kg and oral administration of three doses (2, 4, 8 mg/kg). All blood samples were protein precipitate by using acetonitrile with an internal standard (IS) deltaline. The effective chromatographic separation was carried out through an UPLC BEH C18 analytical column (2.1 mm × 50 mm, 1.7 μm) with an initial mobile phase that consisted of acetonitrile and 10 mmol/L ammonium acetate aqueous solution (containing 0.1% formic acid) with a gradient elution pumped at a flow rate of 0.4 mL/min. Also, an electrospray ionization (ESI) was applied to quantify the talatisamine in the positive ions mode. The method validation demonstrated good linearity over the range of 1-1000 ng/mL (r2 ≥ 0.9993) for talatisamine in mouse blood with a lower limit of quantification (LLOQ) at 1 ng/mL. The accuracy values of the method were within 89.4% to 113.3%, and the matrix effects were between 103.2% and 106.3%. The mean extraction recoveries for talatisamine obtained from four concentrations of QC blood samples were exceeded 71.7%, and the relative standard deviation (RSD) both of intra- and inter-day precision values for replicate quality control samples did not exceed 15% respectively for all analytes during the assay validation. This method was successfully applied to the evaluation of the pharmacokinetic of talatisamine, regardless of intragastric or intravenous administration in mice. Based on the pharmacokinetics data, the bioavailability of talatisamine in mice was >65.0% after oral administration, exhibiting an excellent oral absorption.
Collapse
Affiliation(s)
- Mengchun Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Yijie Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Xianqin Wang
- Analytical and Testing Centre, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yunfang Zhou
- The Laboratory of Clinical Pharmacy, The People's Hospital of Lishui, Lishui 323000, China.
| |
Collapse
|
14
|
Miura T, Uehara S, Mizuno S, Yoshizawa M, Murayama N, Kamiya Y, Shimizu M, Suemizu H, Yamazaki H. Steady-State Human Pharmacokinetics of Monobutyl Phthalate Predicted by Physiologically Based Pharmacokinetic Modeling Using Single-Dose Data from Humanized-Liver Mice Orally Administered with Dibutyl Phthalate. Chem Res Toxicol 2019; 32:333-340. [DOI: 10.1021/acs.chemrestox.8b00361] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Tomonori Miura
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Shotaro Uehara
- Laboratory Animal Research Department, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Sawa Mizuno
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Manae Yoshizawa
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Norie Murayama
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Yusuke Kamiya
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Hiroshi Suemizu
- Laboratory Animal Research Department, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| |
Collapse
|
15
|
Miura T, Uehara S, Nakazato M, Kusama T, Toda A, Kamiya Y, Murayama N, Shimizu M, Suemizu H, Yamazaki H. Human plasma and liver concentrations of styrene estimated by combining a simple physiologically based pharmacokinetic model with rodent data. J Toxicol Sci 2019; 44:543-548. [DOI: 10.2131/jts.44.543] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
| | | | | | | | - Akiko Toda
- Shin Nippon Biomedical Laboratories, Ltd
| | | | | | | | | | | |
Collapse
|
16
|
Iwata H, Goto M, Sakai N, Suemizu H, Yamazaki H. Predictability of human pharmacokinetics of diisononyl phthalate (DINP) using chimeric mice with humanized liver. Xenobiotica 2018; 49:1311-1322. [PMID: 30589368 DOI: 10.1080/00498254.2018.1564087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
1. In order to investigate the pharmacokinetics of diisononyl phthalate (DINP) in humans, we administered [phenyl-U-14C]DINP at a dose of 50.0 mg/kg orally to chimeric mice (humanized-liver mice) in which the liver of TK-NOG mice (control mice) was replaced with human hepatocytes. 2. The plasma radioactivity concentrations peaked (18.0 and 59.9 µg equivalent of DINP/mL, respectively) at 2 h after administration in control and humanized-liver mice. Concentrations rose again at 8 h in controls, but not in humanized-liver mice. 3. The cumulative excretion rates in urine and feces, respectively, were 58.1% and 37.3% of the doses in controls up to 48 h, but were 86.0% and 7.7% in humanized-liver mice. 4. The main circulating metabolites in control and humanized-liver mice were monoisononyl phthalate (MINP) and the glucuronide of oxidized MINP, respectively. The urinary excretion ratio of the glucuronide of oxidized MINP in control mice was one-third of that in humanized-liver mice. 5. The present results suggested that the oxidation rates of the primary metabolite of DINP and their excretion routes to urine/feces were different for control and humanized-liver mice. Species differences in liver activities could be a determinant factor in the in vivo metabolism and disposition of diallyl phthalates such as DINP.
Collapse
Affiliation(s)
| | | | | | - Hiroshi Suemizu
- b Central Institute for Experimental Animals , Kawasaki , Japan
| | - Hiroshi Yamazaki
- c Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , Machida , Japan
| |
Collapse
|
17
|
Kamiya Y, Otsuka S, Miura T, Takaku H, Yamada R, Nakazato M, Nakamura H, Mizuno S, Shono F, Funatsu K, Yamazaki H. Plasma and Hepatic Concentrations of Chemicals after Virtual Oral Administrations Extrapolated Using Rat Plasma Data and Simple Physiologically Based Pharmacokinetic Models. Chem Res Toxicol 2018; 32:211-218. [PMID: 30511563 DOI: 10.1021/acs.chemrestox.8b00307] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Only a small fraction of chemicals possesses adequate in vivo toxicokinetic data for assessing potential hazards. The aim of the present study was to model the plasma and hepatic pharmacokinetics of more than 50 disparate types of chemicals and drugs after virtual oral administrations in rats. The models were based on reported pharmacokinetics determined after oral administration to rats. An inverse relationship was observed between no-observed-effect levels after oral administration and chemical absorbance rates evaluated for cell permeability ( r = -0.98, p < 0.001, n = 17). For a varied selection of more than 30 chemicals, the plasma concentration curves and the maximum concentrations obtained using a simple one-compartment model (recently recommended as a high-throughput toxicokinetic model) and a simple physiologically based pharmacokinetic (PBPK) model (consisting of chemical receptor, metabolizing, and central compartments) were highly consistent. The hepatic and plasma concentrations and the hepatic and plasma areas under the concentration-time curves of more than 50 chemicals were roughly correlated; however, differences were evident between the PBPK-modeled values in livers and empirically obtained values in plasma. Of the compounds selected for analysis, only seven had the lowest observed effect level (LOEL) values for hepatoxicity listed in the Hazard Evaluation Support System Integrated Platform in Japan. For these seven compounds, the LOEL values and the areas under the hepatic concentration-time curves estimated using PBPK modeling were inversely correlated ( r = -0.78, p < 0.05, n = 7). This study provides important information to help simulate the high hepatic levels of potent hepatotoxic compounds. Using suitable PBPK parameters, the present models could estimate the plasma/hepatic concentrations of chemicals and drugs after oral doses using both PBPK forward and reverse dosimetry, thereby indicating the potential value of this modeling approach in predicting hepatic toxicity as a part of risk assessments of chemicals absorbed in the human body.
Collapse
Affiliation(s)
- Yusuke Kamiya
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen , Machida, Tokyo 194-8543 , Japan
| | - Shohei Otsuka
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen , Machida, Tokyo 194-8543 , Japan
| | - Tomonori Miura
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen , Machida, Tokyo 194-8543 , Japan
| | - Hiroka Takaku
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen , Machida, Tokyo 194-8543 , Japan
| | - Rio Yamada
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen , Machida, Tokyo 194-8543 , Japan
| | - Mayuko Nakazato
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen , Machida, Tokyo 194-8543 , Japan
| | - Hitomi Nakamura
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen , Machida, Tokyo 194-8543 , Japan
| | - Sawa Mizuno
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen , Machida, Tokyo 194-8543 , Japan
| | - Fumiaki Shono
- Department of Chemical System Engineering, School of Engineering , The University of Tokyo , Bunkyo-ku, Tokyo 113-8656 , Japan
| | - Kimito Funatsu
- Department of Chemical System Engineering, School of Engineering , The University of Tokyo , Bunkyo-ku, Tokyo 113-8656 , Japan
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen , Machida, Tokyo 194-8543 , Japan
| |
Collapse
|
18
|
Murayama N, Suemizu H, Uehara S, Kusama T, Mitsui M, Kamiya Y, Shimizu M, Guengerich FP, Yamazaki H. Association of pharmacokinetic profiles of lenalidomide in human plasma simulated using pharmacokinetic data in humanized-liver mice with liver toxicity detected by human serum albumin RNA. J Toxicol Sci 2018; 43:369-375. [PMID: 29877213 DOI: 10.2131/jts.43.369] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Lenalidomide has been shown to be potentially teratogenic in thalidomide-sensitive animal species. Screening for thalidomide analogs devoid of teratogenicity/toxicity-attributable to drug metabolism and disposition, but having immunomodulatory properties-is a strategic pathway towards development of new anticancer drugs. Plasma concentrations of lenalidomide were investigated in immunodeficient control and humanized-liver mice following oral administration of lenalidomide (50 mg/kg). Plasma concentrations of lenalidomide (1-2 hr after administration) were slightly but significantly higher in humanized-liver mice than in control mice (p < 0.05). Human albumin mRNA, a liver-specific toxicity marker, was found in the blood of humanized-liver mice 24 hr after lenalidomide administration. Simulations of human plasma concentrations of lenalidomide were achieved with simplified physiologically-based pharmacokinetic models in control and humanized-liver mice or by the direct fitting analysis of reported human data, in accordance with reported lenalidomide concentrations after low dose administration in humans. The results indicate that pharmacokinetic profiles of lenalidomide, a compound resulting from introducing one aromatic amino group into thalidomide and removing one keto group, resulted in less species variation in in vivo pharmacokinetics in control and humanized-liver mice and that immunodeficient humanized-liver mice can serve as experimental model animals for human liver injury in drug development at high doses, with human albumin RNA analysis in plasma.
Collapse
Affiliation(s)
- Norie Murayama
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | | | | | - Takashi Kusama
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | - Marina Mitsui
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | - Yusuke Kamiya
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | - Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, USA
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| |
Collapse
|
19
|
Shimizu M, Suemizu H, Mizuno S, Kusama T, Miura T, Uehara S, Yamazaki H. Human plasma concentrations of trimethylamine N-oxide extrapolated using pharmacokinetic modeling based on metabolic profiles of deuterium-labeled trimethylamine in humanized-liver mice. J Toxicol Sci 2018; 43:387-393. [PMID: 29877215 DOI: 10.2131/jts.43.387] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Medicinal carnitine-derived and dietary-derived malodorous trimethylamine and its non-malodorous metabolite trimethylamine N-oxide were historically regarded as nontoxic. Clinical and toxicological interest has recently arisen because of their potential association with atherosclerosis. We previously reported a human physiologically based pharmacokinetic (PBPK) model for trimethylamine and its primary metabolite, trimethylamine N-oxide, based on reported rat trimethylamine pharmacokinetics. However, rats are poor metabolizers with respect to trimethylamine N-oxygenation, and this species difference was investigated in vitro using substrate depletion rates in rat and human liver microsomes. The current study investigated the pharmacokinetics of deuterium-labeled trimethylamine orally administered to immunodeficient humanized-liver mice transplanted with commercially available human hepatocytes. Trimethylamine N-oxide was extensively formed in vivo in humanized-liver mice, but not in control mice. The experimental pharmacokinetic data of deuterium-labeled trimethylamine and its N-oxide in humanized-liver mice were scaled up for application to a human PBPK model. The human plasma concentration curves generated by the resulting simple PBPK model were consistent with concentrations in humans reported in the literature. The model can also simulate human plasma levels of trimethylamine and trimethylamine N-oxide during treatment with the prescription medicine L-carnitine and in trimethylamine loading tests. The predicted plasma levels were in the ranges that occur under the consumption of daily dietary foodstuff; such levels are associated with few toxicological impacts. The present PBPK model for trimethylamine and trimethylamine N-oxide could estimate daily doses by both forward and reverse dosimetry and could facilitate risk assessment in humans.
Collapse
Affiliation(s)
- Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | | | - Sawa Mizuno
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | - Takashi Kusama
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | - Tomonori Miura
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | | | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| |
Collapse
|
20
|
Chen L, Zhang B, Liu J, Fan Z, Weng Z, Geng P, Wang X, Lin G. Pharmacokinetics and Bioavailability Study of Monocrotaline in Mouse Blood by Ultra-Performance Liquid Chromatography-Tandem Mass Spectrometry. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1578643. [PMID: 30186850 PMCID: PMC6110008 DOI: 10.1155/2018/1578643] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/05/2018] [Accepted: 07/29/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS The present study aimed to develop a simple and sensitive method for quantitative determination of monocrotaline (MCT) in mouse blood employing ultra-performance liquid chromatography-electrospray ionization tandem mass spectrometry (UPLC-ESI/MS/MS) using rhynchophylline as an internal standard. METHODS Proteins present in the blood samples were precipitated using acetonitrile. MCT was separated using a 1.7-μm ethylene bridged hybrid (BEH) C18 column (2.1 mm × 50 mm) with a gradient elution program and a constant flow rate of 0.4 mL/min. The LC mobile phase consisted of 10 mmol/L ammonium acetate (containing 0.1% formic acid) and acetonitrile. The total elution time was 4.0 min. The analytes were detected on a UPLC-ESI mass spectrometer in multiple reaction monitoring (MRM) mode and quantified. RESULTS The new method for the determination of MCT has a satisfactory linear detection range of 1-2000 ng/mL and excellent linearity (r = 0.9971). The lower limit of quantification (LLOQ) of MCT is 1.0 ng/mL. Intra- and interassay precisions of MCT were ≤13% with an accuracy from 96.2% to 106.6%. The average recovery of the new method was >75.0%, and matrix effects were between 89.0% and 94.3%. Based on the pharmacokinetics data, the bioavailability of MCT in mice was 88.3% after oral administration. CONCLUSIONS The results suggest that the newly standardized method for quantitative determination of MCT in whole blood is fast, reliable, specific, sensitive, and suitable for pharmacokinetic studies of MCT after intravenous or intragastric administration.
Collapse
Affiliation(s)
- Lianguo Chen
- The Third Clinical Institute Affiliated with Wenzhou Medical University & Wenzhou People's Hospital, Wenzhou 325000, China
| | - Bin Zhang
- Analytical and Testing Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jinlai Liu
- The Third Clinical Institute Affiliated with Wenzhou Medical University & Wenzhou People's Hospital, Wenzhou 325000, China
| | - Zhehua Fan
- Analytical and Testing Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Ziwei Weng
- Analytical and Testing Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Peiwu Geng
- Laboratory of Clinical Pharmacy, The People's Hospital of Lishui, Lishui 323000, China
| | - Xianqin Wang
- Analytical and Testing Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Guanyang Lin
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
21
|
Bissig KD, Han W, Barzi M, Kovalchuk N, Ding L, Fan X, Pankowicz FP, Zhang QY, Ding X. P450-Humanized and Human Liver Chimeric Mouse Models for Studying Xenobiotic Metabolism and Toxicity. Drug Metab Dispos 2018; 46:1734-1744. [PMID: 30093418 DOI: 10.1124/dmd.118.083303] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023] Open
Abstract
Preclinical evaluation of drug candidates in experimental animal models is an essential step in drug development. Humanized mouse models have emerged as a promising alternative to traditional animal models. The purpose of this mini-review is to provide a brief survey of currently available mouse models for studying human xenobiotic metabolism. Here, we describe both genetic humanization and human liver chimeric mouse models, focusing on the advantages and limitations while outlining their key features and applications. Although this field of biomedical science is relatively young, these humanized mouse models have the potential to transform preclinical drug testing and eventually lead to a more cost-effective and rapid development of new therapies.
Collapse
Affiliation(s)
- Karl-Dimiter Bissig
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Weiguo Han
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Mercedes Barzi
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Nataliia Kovalchuk
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Liang Ding
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Xiaoyu Fan
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Francis P Pankowicz
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Qing-Yu Zhang
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Xinxin Ding
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| |
Collapse
|
22
|
Chen L, Weng Q, Li F, Liu J, Zhang X, Zhou Y. Pharmacokinetics and Bioavailability Study of Tubeimoside I in ICR Mice by UPLC-MS/MS. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2018; 2018:9074893. [PMID: 30116651 PMCID: PMC6079592 DOI: 10.1155/2018/9074893] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/19/2018] [Indexed: 06/08/2023]
Abstract
The aim of this study is to establish and validate a rapid, selective, and sensitive ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method to determine tubeimoside I (TBMS-I) in ICR (Institute of Cancer Research) mouse whole blood and its application in the pharmacokinetics and bioavailability study. The blood samples were precipitated by acetonitrile to extract the analytes. Chromatographic separation was performed on a UPLC BEH C18 column (2.1 mm × 50 mm, 1.7 μm). The mobile phase consisted of water with 0.1% formic acid and methanol (1 : 1, v/v) at a flow rate of 0.4 mL/min. The total eluting time was 4 min. The TBMS-I and ardisiacrispin A (internal standard (IS)) were quantitatively detected by a tandem mass spectrometry equipped with an electrospray ionization (ESI) in a positive mode by multiple reaction monitoring (MRM). A validation of this method was in accordance with the US Food and Drug Administration (FDA) guidelines. The lower limit of quantification (LLOQ) of TBMS-I was 2 ng/mL, and the calibration curve was linearly ranged from 2 to 2000 ng/mL (r2 ≥ 0.995). The relative standard deviation (RSD) of interday precision and intraday precision was both lower than 15%, and the accuracy was between 91.7% and 108.0%. The average recovery was >66.9%, and the matrix effects were from 104.8% to 111.0%. In this assay, a fast, highly sensitive, and reproducible quantitative method was developed and validated in mouse blood for the first time. The absolute availability of TBMS-I in the mouse was only 1%, exhibiting a poor oral absorption.
Collapse
Affiliation(s)
- Lianguo Chen
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Qinghua Weng
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Feifei Li
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Jinlai Liu
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Xueliang Zhang
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Yunfang Zhou
- Laboratory of Clinical Pharmacy, The People's Hospital of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| |
Collapse
|
23
|
Miura T, Suemizu H, Goto M, Sakai N, Iwata H, Shimizu M, Yamazaki H. Human urinary concentrations of monoisononyl phthalate estimated using physiologically based pharmacokinetic modeling and experimental pharmacokinetics in humanized-liver mice orally administered with diisononyl phthalate. Xenobiotica 2018; 49:513-520. [DOI: 10.1080/00498254.2018.1471753] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
| | - Hiroshi Suemizu
- Central Institute for Experimental Animals, Kawasaki-ku, Japan
| | | | | | | | | | | |
Collapse
|
24
|
Nakayama K, Ito S, Suzuki M, Takubo H, Yamazaki H, Nomura Y. Prediction of human pharmacokinetics of typical compounds by a physiologically based method using chimeric mice with humanized liver. Xenobiotica 2018; 49:404-414. [DOI: 10.1080/00498254.2018.1460516] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Keigo Nakayama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| | - Soichiro Ito
- Drug Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| | - Masahiro Suzuki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| | - Hiroaki Takubo
- Drug Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Tokyo, Japan
| | - Yukihiro Nomura
- Drug Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| |
Collapse
|
25
|
Dihydrocodeine Overdoses in a Neonate and in a 14-year-old Girl Who Were Both Genotyped as Cytochrome P450 2D6*1/*10-*36: Comparing Developmental Ages and Drug Monitoring Data With the Results of Pharmacokinetic Modeling. Ther Drug Monit 2018; 40:162-165. [DOI: 10.1097/ftd.0000000000000482] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
Kusama T, Toda A, Shimizu M, Uehara S, Inoue T, Uno Y, Utoh M, Sasaki E, Yamazaki H. Association with polymorphic marmoset cytochrome P450 2C19 of in vivo hepatic clearances of chirally separated R-omeprazole and S-warfarin using individual marmoset physiologically based pharmacokinetic models. Xenobiotica 2017; 48:1072-1077. [DOI: 10.1080/00498254.2017.1393121] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
| | - Akiko Toda
- Shin Nippon Biomedical Laboratories Ltd, Kainan, Wakayama, Japan,
| | | | | | - Takashi Inoue
- Department of Marmoset Research, Central Institute for Experimental Animals, Kawasaki, Japan, and
| | - Yasuhiro Uno
- Shin Nippon Biomedical Laboratories Ltd, Kainan, Wakayama, Japan,
| | - Masahiro Utoh
- Showa Pharmaceutical University, Machida, Tokyo, Japan,
- Shin Nippon Biomedical Laboratories Ltd, Kainan, Wakayama, Japan,
| | - Erika Sasaki
- Department of Marmoset Research, Central Institute for Experimental Animals, Kawasaki, Japan, and
- Keio Advanced Research Center, Keio University, Minato-ku, Tokyo, Japan
| | | |
Collapse
|
27
|
Shimizu M, Yamazaki H. Human plasma and urinary metabolic profiles of trimethylamine and trimethylamine N-oxide extrapolated using a simple physiologically based pharmacokinetic model. J Toxicol Sci 2017; 42:485-490. [PMID: 28717107 DOI: 10.2131/jts.42.485] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Trimethylamine, a dietary- and medicinal carnitine-derived amine, is extensively metabolized by liver to non-malodorous trimethylamine N-oxide. Although trimethylamine and trimethylamine N-oxide under daily dietary consumption or carnitine treatment are generally regarded as nontoxic, they have been, and remain, of toxicological and clinical interest because of their potential association with atherosclerosis. The aim of the current study was to model the pharmacokinetics of trimethylamine after oral administration of trimethylamine in humans and compare the results with reported measured values. Adjusted biomonitoring equivalents from rat studies based on reported plasma concentrations were scaled to human equivalents using known species allometric scaling factors. In vitro metabolic clearance data were obtained using rat and human liver microsomal preparations. Renal clearances in humans for trimethylamine and trimethylamine N-oxide were calculated with a clearance concept approach using reported 24-hr urinary excretion rates and assumed areas under plasma concentration curves. The resulting modeled plasma and urinary concentration curves by simple physiologically based pharmacokinetic models (or semi-physiological pharmacokinetic models) were consistent with reported concentrations. This study provides important information to help simulate human plasma levels of trimethylamine and trimethylamine N-oxide in trimethylamine loading tests and during treatment with prescribed medicinal l-carnitine, showing the similar range as that resulting from daily dietary foodstuff consumption along with little toxicological impacts. The present models could estimate relationship between plasma and urine concentrations of trimethylamine or trimethylamine N-oxide and the daily oral doses by both forward and reverse dosimetry from viewpoint of human risk assessment.
Collapse
Affiliation(s)
- Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| |
Collapse
|
28
|
Toda A, Uehara S, Inoue T, Utoh M, Kusama T, Shimizu M, Uno Y, Mogi M, Sasaki E, Yamazaki H. Effects of aging and rifampicin pretreatment on the pharmacokinetics of human cytochrome P450 probes caffeine, warfarin, omeprazole, metoprolol and midazolam in common marmosets genotyped for cytochrome P450 2C19. Xenobiotica 2017; 48:720-726. [PMID: 28686070 DOI: 10.1080/00498254.2017.1353716] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
1. The pharmacokinetics were investigated for human cytochrome P450 probes after single intravenous and oral administrations of 0.20 and 1.0 mg/kg, respectively, of caffeine, warfarin, omeprazole, metoprolol and midazolam to aged (10-14 years old, n = 4) or rifampicin-treated/young (3 years old, n = 3) male common marmosets all genotyped as heterozygous for a cytochrome P450 2C19 variant. 2. Slopes of the plasma concentration-time curves after intravenous administration of warfarin and midazolam were slightly, but significantly (two-way analysis of variance), decreased in aged marmosets compared with young marmosets. The mean hepatic clearances determined by in silico fitting for individual pharmacokinetic models of warfarin and midazolam in the aged group were, respectively, 23% and 56% smaller than those for the young group. 3. Significantly enhanced plasma clearances of caffeine, warfarin, omeprazole and midazolam were evident in young marmosets pretreated with rifampicin (25 mg/kg daily for 4 days). Two- to three-fold increases in hepatic intrinsic clearance values were observed in the individual pharmacokinetic models. 4. The in vivo dispositions of multiple simultaneously administered drugs in old, young and P450-enzyme-induced marmosets were elucidated. The results suggest that common marmosets could be experimental models for aged, induced or polymorphic P450 enzymes in P450-dependent drug metabolism studies.
Collapse
Affiliation(s)
- Akiko Toda
- a Shin Nippon Biomedical Laboratories Ltd. , Kainan , Japan
| | - Shotaro Uehara
- b Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Tokyo , Japan
| | - Takashi Inoue
- c Marmoset Research Department, Central Institute for Experimental Animals , Kawasaki , Japan
| | - Masahiro Utoh
- a Shin Nippon Biomedical Laboratories Ltd. , Kainan , Japan.,b Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Tokyo , Japan
| | - Takashi Kusama
- b Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Tokyo , Japan
| | - Makiko Shimizu
- b Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Tokyo , Japan
| | - Yasuhiro Uno
- a Shin Nippon Biomedical Laboratories Ltd. , Kainan , Japan
| | - Masayuki Mogi
- a Shin Nippon Biomedical Laboratories Ltd. , Kainan , Japan
| | - Erika Sasaki
- d Center of Applied Developmental Biology, Central Institute for Experimental Animals , Kawasaki , Japan , and.,e Keio Advanced Research Center, Keio University , Tokyo , Japan
| | - Hiroshi Yamazaki
- b Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Tokyo , Japan
| |
Collapse
|
29
|
Yamazaki-Nishioka M, Shimizu M, Suemizu H, Nishiwaki M, Mitsui M, Yamazaki H. Human plasma metabolic profiles of benzydamine, a flavin-containing monooxygenase probe substrate, simulated with pharmacokinetic data from control and humanized-liver mice. Xenobiotica 2017; 48:117-123. [DOI: 10.1080/00498254.2017.1288280] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Miho Yamazaki-Nishioka
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan,
| | - Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan,
| | - Hiroshi Suemizu
- Laboratory Animal Research Department, Central Institute for Experimental Animals, Kawasaki-ku, Japan, and
| | - Megumi Nishiwaki
- Laboratory Animal Research Department, Central Institute for Experimental Animals, Kawasaki-ku, Japan, and
- Technical Service Department, CLEA Japan, Inc, Fujinomiya, Japan
| | - Marina Mitsui
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan,
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan,
| |
Collapse
|
30
|
Utoh M, Kusama T, Miura T, Mitsui M, Kawano M, Hirano T, Shimizu M, Uno Y, Yamazaki H. R-warfarin clearances from plasma associated with polymorphic cytochrome P450 2C19 and simulated by individual physiologically based pharmacokinetic models for 11 cynomolgus monkeys. Xenobiotica 2017; 48:206-210. [DOI: 10.1080/00498254.2017.1288945] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Masahiro Utoh
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan, and
- Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories Ltd, Kainan, Wakayama, Japan
| | - Takashi Kusama
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan, and
| | - Tomonori Miura
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan, and
| | - Marina Mitsui
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan, and
| | - Mirai Kawano
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan, and
| | - Takahiro Hirano
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan, and
- Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories Ltd, Kainan, Wakayama, Japan
| | - Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan, and
| | - Yasuhiro Uno
- Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories Ltd, Kainan, Wakayama, Japan
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan, and
| |
Collapse
|
31
|
Isin EM, Turesky RJ. Introduction: Mass Spectrometry and Emerging Technologies for Biomarker Discovery in the Assessment of Human Health and Disease. Chem Res Toxicol 2016; 29:1901-1902. [PMID: 27989143 DOI: 10.1021/acs.chemrestox.6b00429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Emre M Isin
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal SE-431 83, Sweden
| | - Robert J Turesky
- Masonic Cancer Center and Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota , Minneapolis, Minnesota 55455, United States
| |
Collapse
|
32
|
Shimizu M, Suemizu H, Mitsui M, Shibata N, Guengerich FP, Yamazaki H. Metabolic profiles of pomalidomide in human plasma simulated with pharmacokinetic data in control and humanized-liver mice. Xenobiotica 2016; 47:844-848. [PMID: 27852146 DOI: 10.1080/00498254.2016.1247218] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
1. Pomalidomide has been shown to be potentially teratogenic in thalidomide-sensitive animal species such as rabbits. Screening for thalidomide analogs devoid of teratogenicity/toxicity - attributable to metabolites formed by cytochrome P450 enzymes - but having immunomodulatory properties is a strategic pathway towards development of new anticancer drugs. 2. In this study, plasma concentrations of pomalidomide, its primary 5-hydroxylated metabolite, and its glucuronide conjugate(s) were investigated in control and humanized-liver mice. Following oral administration of pomalidomide (100 mg/kg), plasma concentrations of 7-hydroxypomalidomide and 5-hydroxypomalidomide glucuronide were slightly higher in humanized-liver mice than in control mice. 3. Simulations of human plasma concentrations of pomalidomide were achieved with simplified physiologically-based pharmacokinetic models in both groups of mice in accordance with reported pomalidomide concentrations after low dose administration in humans. 4. The results indicate that pharmacokinetic profiles of pomalidomide were roughly similar between control mice and humanized-liver mice and that control and humanized-liver mice mediated pomalidomide 5-hydroxylation in vivo. Introducing one aromatic amino group into thalidomide resulted in less species differences in in vivo pharmacokinetics in control and humanized-liver mice.
Collapse
Affiliation(s)
- Makiko Shimizu
- a Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Tokyo , Japan
| | - Hiroshi Suemizu
- b Central Institute for Experimental Animals , Kawasaki , Japan
| | - Marina Mitsui
- a Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Tokyo , Japan
| | - Norio Shibata
- c Graduate School of Engineering, Nagoya Institute of Technology , Nagoya , Japan , and
| | - F Peter Guengerich
- d Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , United States
| | - Hiroshi Yamazaki
- a Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Tokyo , Japan
| |
Collapse
|
33
|
Yamazaki H. Differences in Toxicological and Pharmacological Responses Mediated by Polymorphic Cytochromes P450 and Related Drug-Metabolizing Enzymes. Chem Res Toxicol 2016; 30:53-60. [PMID: 27750412 DOI: 10.1021/acs.chemrestox.6b00286] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Research over the past 30 years has elucidated the roles of polymorphic human liver cytochrome P450 (P450) enzymes associated with toxicological and/or pharmacological actions. Thalidomide exerts its various pharmacological and toxic actions in primates through multiple mechanisms, including nonspecific modification of many protein networks after bioactivation by autoinduced human P450 enzymes. To overcome species differences between rodents, currently, nonhuman primates and/or mouse models with transplanted human hepatocytes are used. Interindividual variability of P450-dependent drug clearances in cynomolgus monkeys and common marmosets is partly accounted for by polymorphic P450 variants and/or aging, just as it is in humans with increased prevalence of polypharmacy. Genotyping of P450 genes in nonhuman primates would be beneficial before and/or after drug metabolism and toxicity testing and evaluation as well in humans. Genome-wide association studies in humans have been rapidly advanced; however, unique whole-gene deletion of P450 2A6 was subsequently developed to cover nicotine-related lung cancer risk study. Regarding polypharmacy, toxicological research should generally be aimed at identifying the risk of adverse drug events following specific potential drug exposures by examining single or multiple metabolic pathways involving single or multiple drug-metabolizing enzymes. Current and next-generation research of drug metabolism and disposition resulting in drug toxicity would be addressed under advanced knowledge of polymorphic and age-related intra- and/or interspecies differences of drug-metabolizing enzymes. In the near future, humanized animal models combining transplanted hepatocytes and a humanized immune system may be available to study human immune reactions caused by human-type drug metabolites. Such sophisticated models should provide preclinical predictions of human drug metabolism and potential toxicity.
Collapse
Affiliation(s)
- Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , 3-3165 Higashi-tamagawa Gakuen, Machida, Tokyo 194-8543, Japan
| |
Collapse
|