1
|
Li W, Picard F. Toxicokinetics in preclinical drug development of small-molecule new chemical entities. Biomed Chromatogr 2022:e5553. [PMID: 36415962 DOI: 10.1002/bmc.5553] [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: 10/24/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
Toxicokinetics (TK) is an integral part of nonclinical (preclinical) safety assessment of small-molecule new chemical entities in drug development. It is employed to describe the systemic exposure of a drug candidate and/or its important metabolite(s) achieved in study animals and elucidate the relationship (proportional, over-proportional, or under-proportional) between systemic exposure and dose administered and the associated differences/similarities between male and female animals along with the possible accumulation/induction. TK data and the derived parameters are employed to propose safe starting doses for clinical use of the new drug candidate through proper extrapolation of findings in study animals to humans. This review has attempted to highlight the health authority expectations on TK assessment in supporting preclinical safety profiling of new chemical entities. A robust TK assessment requires good understanding of absorption, distribution, metabolism, and elimination processes of drug candidate, adequate TK sampling (e.g., controls where relevant), implementation of fit-for-purpose bioanalytical methods (validated or scientifically qualified) along with necessary measures to prevent mis-dosing or ex vivo contamination, and establishment of stability of the drug candidate and/or its metabolite(s) in the intended species matrix to ensure the reliability of bioanalytical and TK data. The latter provides a vital link between animal experiments and human safety.
Collapse
Affiliation(s)
- Wenkui Li
- Pharmacokinetic Sciences-Drug Disposition, Novartis Institutes for BioMedical Research, East Hanover, New Jersey, USA
| | - Franck Picard
- Pharmacokinetic Sciences-Drug Disposition, Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
2
|
Loerracher AK, Braunbeck T. Inducibility of cytochrome P450-mediated 7-methoxycoumarin-O-demethylase activity in zebrafish (Danio rerio) embryos. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 225:105540. [PMID: 32569997 DOI: 10.1016/j.aquatox.2020.105540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/31/2020] [Accepted: 06/07/2020] [Indexed: 06/11/2023]
Abstract
The zebrafish (Danio rerio) embryo has increasingly been used as an alternative model in human and environmental toxicology. Since the cytochrome P450 (CYP) system is of fundamental importance for the understanding and correct interpretation of the outcome of toxicological studies, constitutive and xenobiotic-induced 7-methoxycoumarin-O-demethylase (MCOD), i.e. 'mammalian CYP2-like', activities were monitored in vivo in zebrafish embryos via confocal laser scanning microscopy. In order to elucidate molecular mechanisms underlying the MCOD induction, dose-dependent effects of the prototypical CYP inducers β-naphthoflavone (aryl hydrocarbon receptor (AhR) agonist), rifampicin (pregnane X receptor (PXR) agonist), carbamazepine and phenobarbital (constitutive androstane receptor (CAR) agonists) were analyzed in zebrafish embryos of varying age. Starting from 36 h of age, all embryonic stages of zebrafish could be shown to have constitutive MCOD activity, albeit with spatial variation and at distinct levels. Whereas carbamazepine, phenobarbital and rifampicin had no effect on in vivo MCOD activity in 96 h old zebrafish embryos, the model aryl hydrocarbon receptor agonist β-naphthoflavone significantly induced MCOD activity in 96 h old zebrafish embryos at 46-734 nM, however, without a clear concentration-effect relationship. Induction of MCOD activity by β-naphthoflavone gradually decreased with progression of embryonic development. By in vivo characterization of constitutive and xenobiotic-induced MCOD activity patterns in 36, 60, 84 and 108 h old zebrafish embryos, this decrease could primarily be attributed to an age-related decline in the induction of MCOD activity in the cardiovascular system. Results of this study provide novel insights into the mechanism and extent, by which specific CYP activities in early life-stages of zebrafish can be influenced by exposure to xenobiotics. The study thus lends further support to the view that zebrafish embryos- at least from an age of 36 h - have an elaborate and inducible biotransformation system.
Collapse
Affiliation(s)
- Ann-Kathrin Loerracher
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120, Heidelberg, Germany.
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120, Heidelberg, Germany
| |
Collapse
|
3
|
Aubets J, Jansat J, Salva M, Birks VM, Cole RJ, Lewis J, Pitcher A, Hall M. No evidence for interactions of dimethylfumarate (DMF) and its main metabolite monomethylfumarate (MMF) with human cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) drug transporter. Pharmacol Res Perspect 2019; 7:e00540. [PMID: 31832203 PMCID: PMC6887663 DOI: 10.1002/prp2.540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 01/28/2023] Open
Abstract
Dimethylfumarate (DMF) has long been used as part of a fixed combination of fumaric acid esters (FAE) in some European countries and is now available as an oral monotherapy for psoriasis. The present investigation determined whether DMF and its main metabolite monomethylfumarate (MMF) interact with hepatic cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) transporter, and was performed as part of DMF's regulatory commitments. Although referred to in the available product labels/summary of product characteristics, the actual data have not yet been made publicly available. In vitro inhibition experiments using CYP-selective substrates with human liver microsomes showed 50% inhibitory concentrations (IC50) of >666 µmol/L for DMF and >750 µmol/L for MMF. MMF (≤250 μmol/L; 72 hours) was not cytotoxic in cultured human hepatocyte experiments and mRNA expression data indicated no CYP induction by MMF (1-250 µmol/L). DMF (≤6.66 mmol/L) showed moderate-to-high absorption (apparent permeability [Papp] ≥2.3-29.7 x 10-6 cm/s) across a Caucasian colon adenocarcinoma (Caco-2) cell monolayer, while MMF (≤7.38 mmol/L) demonstrated low-to-moderate permeability (Papp 1.2-8.9 × 10-6 cm/s). DMF was not a substrate for P-gp (net efflux ratios ≤1.22) but was a weak inhibitor of P-gp at supratherapeutic concentrations (estimated IC50 relative to solvent control of 1.5 mmol/L; [3H]digoxin efflux in Caco-2 cells). This inhibition is unlikely to be clinically relevant. MMF was not a substrate or inhibitor of P-gp. Thus, DMF and MMF should not affect the absorption, distribution, metabolism or excretion of coadministered drugs that are CYP and P-gp substrates.
Collapse
Affiliation(s)
- Jordi Aubets
- Department of DMPK DevelopmentAlmirall S.A.BarcelonaSpain
| | | | - Miquel Salva
- Department of DMPK DevelopmentAlmirall S.A.BarcelonaSpain
| | | | | | | | | | | |
Collapse
|
4
|
Yoshioka M, Takenouchi T, Kitani H, Guruge KS, Yamanaka N. Synergistic induction of drug-metabolizing enzymes in co-cultures of bovine hepatocytic and sinusoidal cell lines. In Vitro Cell Dev Biol Anim 2019; 56:2-9. [PMID: 31722089 DOI: 10.1007/s11626-019-00408-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/25/2019] [Indexed: 11/26/2022]
Abstract
Hepatocyte-derived cell lines provide useful experimental systems for studying liver metabolism. Unlike human and rodents, few hepatocyte-derived cell lines have been generated from cattle. Here, we established two immortalized bovine hepatocyte-derived cell lines (BH4 and BH5) via transfection with a SV40 large T-antigen construct. Morphological and immunocytochemical analyses revealed that BH4 and BH5 originated from hepatocytes and biliary-epithelial cells, respectively. A potent carcinogen, 3-methylcholanthrene (3-MC), upregulated gene expression of cytochrome P450 (CYP)1A1, CYP1A2, and CYP1B1 in BH4 and BH5, but only to levels less than one-fifteenth of those in primary cultured bovine hepatocytes. Phenobarbital (PB) also increased expression levels of CYP2B6, CYP2C18, and CYP3A4 in BH4 and BH, but at a lower level than 3-MC. By contrast, when BH4 or BH5 was co-cultured with previously established bovine liver sinusoidal cell lines and treated with 3-MC, the gene expression levels of CYP1A1, CYP1A2, and CYP1B1 increased by 38~290%, compared with those in BH4 or BH5 cells cultured alone. PB-treated co-cultures of BH4 or BH5 cells and liver sinusoidal cell lines also showed synergistic increases in CYP2B6 and CYP2C18 expression. Together, the results suggest that these co-cultures could provide an in vitro model for investigations into pharmacological and toxicological properties of drugs in cattle liver.
Collapse
Affiliation(s)
- Miyako Yoshioka
- Pathology and Pathophysiology Research Division, National Institute of Animal Health, NARO, Kannondai 3-1-5, Tsukuba, Ibaraki, 305-0856, Japan.
| | - Takato Takenouchi
- Division of Animal Sciences, Institute of Agrobiological Sciences, NARO, 1-2 Ohwashi, Tsukuba, Ibaraki, 305-8634, Japan
| | - Hiroshi Kitani
- Division of Animal Sciences, Institute of Agrobiological Sciences, NARO, 1-2 Ohwashi, Tsukuba, Ibaraki, 305-8634, Japan
| | - Keerthi S Guruge
- Pathology and Pathophysiology Research Division, National Institute of Animal Health, NARO, Kannondai 3-1-5, Tsukuba, Ibaraki, 305-0856, Japan
| | - Noriko Yamanaka
- Pathology and Pathophysiology Research Division, National Institute of Animal Health, NARO, Kannondai 3-1-5, Tsukuba, Ibaraki, 305-0856, Japan
| |
Collapse
|
5
|
|
6
|
Ploemen JPHTM, Kramer H, Krajnc EI, Martin I. The Use of Toxicokinetic Data in Preclinical Safety Assessment: A Toxicologic Pathologist Perspective. Toxicol Pathol 2016; 35:836-9. [DOI: 10.1080/01926230701584247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Collection of toxicokinetic data has become a routine practice during the last 15 years in most general toxicity studies on pharma. It enables the correlation of pathological changes with the plasma concentration of drugs and/or their metabolites. This overview summarizes the use of the toxicokinetic data from the perspective of the toxicologic pathologist.
Collapse
Affiliation(s)
| | - Hester Kramer
- Clinical Pharmacology & Kinetics, Organon, 5340 BH Oss, The Netherlands
| | - Ernö I. Krajnc
- Department of Toxicology & Drug Disposition, Organon, 5340 BH Oss, The Netherlands
| | - Iain Martin
- Department of Pharmacology, Organon Research, Scotland
| |
Collapse
|
7
|
Gómez-Icazbalceta G, González-Sánchez I, Moreno J, Cerbón MA, Cervantes A. In vitro drug metabolism testing using blood-monocyte derivatives. Expert Opin Drug Metab Toxicol 2013; 9:1571-80. [PMID: 23984653 DOI: 10.1517/17425255.2013.831069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Monocytes and their cell derivatives can participate in drug metabolism. These cells express different Phase-I or -II drug metabolizing enzymes and can be differentiated into neo-hepatocytes (NeoHep) and represent a promising alternative strategy to test drug metabolism. This is particularly useful as primary human hepatocytes (PHH), are difficult to obtain and maintain in culture. AREAS COVERED The authors analyze the use of blood monocytes and their derivatives for the study of drug metabolism. They also compare them to the in vitro ability of cells from different sources including: PHH, immortalized hepatocytes, tumor cell lines and NeoHep. EXPERT OPINION The use of monocytes, macrophages, dendritic or Kupffer cells, to test drug metabolism, has serious limitations because these cells express lower levels of cytochrome P450 enzymes than PHH. The best available option, to replace PHH, have been tumor cell lines such as HepaRG, as well as immortalized hepatocytes from adult or fetal sources. Monocyte-derived NeoHep cells are novel and easily accessible cells, which express many drug metabolizing enzymes at levels comparable to PHH. These cells allow drug evaluation under a diverse genetic background. While these cells are in the early stages of evaluation and do need to be examined more thoroughly, they constitute a promising new tool for in vitro drug testing.
Collapse
Affiliation(s)
- Guillermo Gómez-Icazbalceta
- National Autonomous University of Mexico, Faculty of Chemistry, Department of Biology , Mexico City, D.F. 04510 , Mexico +52 55 5622 3820 ; +52 55 5616 2010 ;
| | | | | | | | | |
Collapse
|
8
|
Bjornsson TD, Callaghan JT, Einolf HJ, Fischer V, Gan L, Grimm S, Kao J, King SP, Miwa G, Ni L, Kumar G, McLeod J, Obach SR, Roberts S, Roe A, Shah A, Snikeris F, Sullivan JT, Tweedie D, Vega JM, Walsh J, Wrighton SA. The Conduct of In Vitro and In Vivo Drug-Drug Interaction Studies: A PhRMA Perspective. J Clin Pharmacol 2013. [DOI: 10.1177/0091270003252519] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
9
|
Sinz MW. Evaluation of pregnane X receptor (PXR)-mediated CYP3A4 drug-drug interactions in drug development. Drug Metab Rev 2013; 45:3-14. [DOI: 10.3109/03602532.2012.743560] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
10
|
Ettlin RA, Kuroda J, Plassmann S, Prentice DE. Successful drug development despite adverse preclinical findings part 1: processes to address issues and most important findings. J Toxicol Pathol 2010; 23:189-211. [PMID: 22272031 PMCID: PMC3234634 DOI: 10.1293/tox.23.189] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 09/06/2010] [Indexed: 01/08/2023] Open
Abstract
Unexpected adverse preclinical findings (APFs) are not infrequently encountered during drug development. Such APFs can be functional disturbances such as QT prolongation, morphological toxicity or carcinogenicity. The latter is of particular concern in conjunction with equivocal genotoxicity results. The toxicologic pathologist plays an important role in recognizing these effects, in helping to characterize them, to evaluate their risk for man, and in proposing measures to mitigate the risk particularly in early clinical trials. A careful scientific evaluation is crucial while termination of the development of a potentially useful drug must be avoided. This first part of the review discusses processes to address unexpected APFs and provides an overview over typical APFs in particular classes of drugs. If the mode of action (MoA) by which a drug candidate produces an APF is known, this supports evaluation of its relevance for humans. Tailor-made mechanistic studies, when needed, must be planned carefully to test one or several hypotheses regarding the potential MoA and to provide further data for risk evaluation. Safety considerations are based on exposure at no-observed-adverse-effect levels (NOAEL) of the most sensitive and relevant animal species and guide dose escalation in clinical trials. The availability of early markers of toxicity for monitoring of humans adds further safety to clinical studies. Risk evaluation is concluded by a weight of evidence analysis (WoE) with an array of parameters including drug use, medical need and alternatives on the market. In the second part of this review relevant examples of APFs will be discussed in more detail.
Collapse
Affiliation(s)
- Robert A. Ettlin
- Ettlin Consulting Ltd., 14 Mittelweg, 4142 Muenchenstein,
Switzerland
| | - Junji Kuroda
- KISSEI Pharmaceutical Co., Ltd., 2320–1 Maki, Hotaka, Azumino,
Nagano 399-8305, Japan
| | - Stephanie Plassmann
- PreClinical Safety (PCS) Consultants Ltd., 7 Gartenstrasse, 4132
Muttenz, Switzerland
| | - David E. Prentice
- PreClinical Safety (PCS) Consultants Ltd., 7 Gartenstrasse, 4132
Muttenz, Switzerland
| |
Collapse
|
11
|
Sato W, Suzuki H, Sasaki T, Kumagai T, Sakaguchi S, Mizugaki M, Miyairi S, Yamazoe Y, Nagata K. Construction of a system that simultaneously evaluates CYP1A1 and CYP1A2 induction in a stable human-derived cell line using a dual reporter plasmid. Drug Metab Pharmacokinet 2010; 25:180-9. [PMID: 20460824 DOI: 10.2133/dmpk.25.180] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human CYP1A1 and CYP1A2 genes are in a head-to-head orientation on chromosome 15 and are separated by a 23-kb intergenic space. To our knowledge, this is the first report on a stable cell line that contains the 23-kb full-length regulatory region and is able to simultaneously assess the transcriptional activation of CYP1A1 and CYP1A2 genes. The stable cell line that constitutively expresses the reporter activities was constructed by inserting the dual reporter plasmid containing the 23-kb region between the CYP1A1 and CYP1A2 genes into the chromosome. Transcriptional activation of the CYP1A1 and CYP1A2 genes was measured simultaneously using luciferase (Luc) and secreted alkaline phosphatase (SEAP) activities, respectively. To demonstrate the utility of the stable cell line, CYP1A1/1A2 induction by the majority of compounds previously identified as CYP1A1/1A2 inducers was measured. The results clearly show that all compounds caused induction of reporter activities. In addition to assessing transcriptional activation of the CYP1A1 and CYP1A2 genes by measuring reporter activities, we determined the intrinsic CYP1A1 and CYP1A2 mRNA levels by treating them with the same compounds. The results suggest that this stable cell line may be used to rapidly and accurately predict CYP1A1/1A2 induction.
Collapse
Affiliation(s)
- Wataru Sato
- Department of Environmental and Health Science, Tohoku Pharmaceutical University, Sendai, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Tang C, Prueksaritanont T. Use of in vivo animal models to assess pharmacokinetic drug-drug interactions. Pharm Res 2010; 27:1772-87. [PMID: 20428930 DOI: 10.1007/s11095-010-0157-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 04/08/2010] [Indexed: 12/31/2022]
Abstract
Animal models are used commonly in various stages of drug discovery and development to aid in the prospective assessment of drug-drug interaction (DDI) potential and the understanding of the underlying mechanism for DDI of a drug candidate. In vivo assessments in an appropriate animal model can be very valuable, when used in combination with in vitro systems, to help verify in vivo relevance of the in vitro animal-based results, and thus substantiate the extrapolation of in vitro human data to clinical outcomes. From a pharmacokinetic standpoint, a key consideration for rational selection of an animal model is based on broad similarities to humans in important physiological and biochemical parameters governing drug absorption, distribution, metabolism or excretion (ADME) processes in question for both the perpetrator and victim drugs. Equally critical are specific in vitro and/or in vivo experiments to demonstrate those similarities, usually both qualitative and quantitative, in the ADME properties/processes under investigation. In this review, theoretical basis and specific examples are presented to illustrate the utility of the animal models in assessing the potential and understanding the mechanisms of DDIs.
Collapse
Affiliation(s)
- Cuyue Tang
- Department of Drug Metabolism and Pharmacokinetics, Merck Research Laboratories, Merck & Co., Inc., WP75A-203, West Point, Pennsylvania 19486, USA
| | | |
Collapse
|
13
|
TANIGUCHI A, WADA KI, OHNO M. Development of Novel Culture System Using Nano-biotechnology. YAKUGAKU ZASSHI 2010; 130:529-35. [DOI: 10.1248/yakushi.130.529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Akiyoshi TANIGUCHI
- Advanced Medical Materials Group, Biomaterials center, National Institute for Materials Science
| | - Ken-ichi WADA
- Advanced Medical Materials Group, Biomaterials center, National Institute for Materials Science
| | - Maki OHNO
- Advanced Medical Materials Group, Biomaterials center, National Institute for Materials Science
| |
Collapse
|
14
|
Krämer S, Testa B. The Biochemistry of Drug Metabolism - An Introduction. Chem Biodivers 2009; 6:1477-660, table of contents. [DOI: 10.1002/cbdv.200900233] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
15
|
Ohno M, Motojima K, Okano T, Taniguchi A. Induction of Drug-Metabolizing Enzymes by Phenobarbital in Layered Co-culture of a Human Liver Cell Line and Endothelial Cells. Biol Pharm Bull 2009; 32:813-7. [DOI: 10.1248/bpb.32.813] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Maki Ohno
- Advanced Medical Materials Group, Biomaterials Center, National Institute for Materials Science, MANA
- Department of Biochemistry, Meiji Pharmaceutical University
| | | | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University
| | - Akiyoshi Taniguchi
- Advanced Medical Materials Group, Biomaterials Center, National Institute for Materials Science, MANA
| |
Collapse
|
16
|
Pelkonen O, Turpeinen M. In vitro–in vivoextrapolation of hepatic clearance: Biological tools, scaling factors, model assumptions and correct concentrations. Xenobiotica 2008; 37:1066-89. [DOI: 10.1080/00498250701620726] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Meneses-Lorente G, Pattison C, Guyomard C, Chesné C, Heavens R, Watt AP, Sohal B. Utility of long-term cultured human hepatocytes as an in vitro model for cytochrome p450 induction. Drug Metab Dispos 2006; 35:215-20. [PMID: 17093007 DOI: 10.1124/dmd.106.009423] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cytochrome P450 (P450) induction may have considerable implications for drug therapy. Therefore, understanding the induction potential of a new chemical entity at an early stage in discovery is crucial to reduce the risk of failure in the clinic and help the identification of noninducing chemical structures. Availability of human viable tissue often limits evaluation of induction potential in human hepatocytes. A solution is to increase the time period during which the hepatocytes remain viable. In this study we have investigated the induction of several P450 isozymes in long-term cultured hepatocytes compared with short-term cultured hepatocytes from the same individuals. Short- and long-term cultured primary hepatocytes isolated from each individual were cultured in a 96-well format and treated for 24 h with a range of prototypical P450 inducers and Merck Research Laboratories compounds. CYP3A4, 1A1, 1A2, 2B6, and 2C9 mRNA levels were measured using quantitative real-time reverse transcriptase-polymerase chain reaction (TaqMan) from the same cultured hepatocyte wells. CYP3A4, 1A1, 1A2, 2B6, and 2C9 were shown to be inducible in long-term cultured hepatocytes. The -fold induction varied between donors, and between short- and long-term cultured hepatocytes from the same donor. However, this variability can be controlled by normalizing data from each hepatocyte preparation to a positive control. The use of long-term cultured hepatocytes on 96-well plates has proven to be sensitive, robust, and convenient for assessing P450 induction potential of new compound entities during the drug discovery process.
Collapse
Affiliation(s)
- Georgina Meneses-Lorente
- Department of Medicinal Chemistry (Drug Metabolism Section), Merck Sharp and Dohme Research Laboratories, Harlow, Essex, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
18
|
Prueksaritanont T, Li C, Tang C, Kuo Y, Strong-Basalyga K, Carr B. Rifampin induces the in vitro oxidative metabolism, but not the in vivo clearance of diclofenac in rhesus monkeys. Drug Metab Dispos 2006; 34:1806-10. [PMID: 16928785 DOI: 10.1124/dmd.106.011643] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Effects of rifampin on in vitro oxidative metabolism and in vivo pharmacokinetics of diclofenac (DF), a prototypic CYP2C9 marker substrate, were investigated in rhesus monkeys. In monkey hepatocytes, rifampin markedly induced DF 4'-hydroxylase activity, with values for EC(50) of 0.2 to 0.4 microM and E(max) of 2- to 5-fold over control. However, pretreatment with rifampin did not alter the pharmacokinetics of DF obtained after either i.v. or intrahepatic portal vein (i.pv.) administration of DF to monkeys. At the dose studied, plasma concentrations of rifampin reached 10 microM, far exceeding the in vitro EC(50) values. Under similar treatment conditions, rifampin was previously shown to induce midazolam (MDZ) 1'-hydroxylation in rhesus monkey hepatocytes (EC(50) and E(max) values approximately 0.2 microM and approximately 2- to 3-fold, respectively), and markedly affected the in vivo pharmacokinetics of MDZ (>10-fold decreases in the i.pv. MDZ systemic exposure and its hepatic availability, F(h)) in this animal species. In monkey liver microsomes, DF underwent, predominantly, glucuronidation, and, modestly, oxidation; the intrinsic clearance (CL(int) = V(max)/K(m)) value for the glucuronidation pathway accounted for >95% (versus about 75% in human liver microsomes) of the total (glucuronidation + hydroxylation) intrinsic clearance value. In monkey hepatocytes, the hydroxylation also was a minor component (< or =10%) relative to the glucuronidation, supporting the liver microsomal finding. Collectively, our results suggest that the oxidative metabolism is not the major in vivo clearance mechanism of DF in either untreated or rifampin-treated monkeys and, conceivably, also in humans, raising a question about the utility of DF as an in vivo CYP2C9 probe.
Collapse
|
19
|
Prueksaritanont T, Kuo Y, Tang C, Li C, Qiu Y, Lu B, Strong-Basalyga K, Richards K, Carr B, Lin JH. In vitro and in vivo CYP3A64 induction and inhibition studies in rhesus monkeys: a preclinical approach for CYP3A-mediated drug interaction studies. Drug Metab Dispos 2006; 34:1546-55. [PMID: 16782766 DOI: 10.1124/dmd.106.009878] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In this study, induction and inhibition of rhesus monkey CYP3A64 versus human CYP3A4 were characterized in vitro, and the corresponding pharmacokinetic consequences were evaluated in rhesus monkeys. In monkey hepatocytes, rifampin markedly induced CYP3A64 mRNA (EC50 = 0.5 microM; Emax = 6-fold) and midazolam (MDZ) 1'-hydroxylase activity (EC50 = 0.2 microM; Emax = 2-fold). Compound A (N-[2(R)-hydroxy-1(S)-indanyl-5-[2(S)-(1,1-dimethylethylaminocarbonyl)-4-[(furo[2,3-b]pyridin-5-yl)-methyl]piperazin-1-yl]-4(S)-hydroxy-2(R)-phenylmethylpentanamide), a known potent and mechanism-based inhibitor of CYP3A4, strongly inhibited the formation of 1'-hydroxy MDZ by recombinant CYP3A64 in a concentration- and time-dependent manner (KI = 0.25 microM; k(inact) = 0.4 min(-1)). Similar corresponding results also were obtained with human CYP3A4 in the presence of rifampin or compound A. In rhesus monkeys, MDZ exhibited a relatively high metabolic clearance (primarily via 1'-hydroxylation followed by glucuronidation) and a low hepatic availability (Fh = 16%). Consistent with the induction of hepatic metabolism of a high-clearance compound, pretreatment with rifampin (18 mg/kg p.o. for 5 days) did not significantly affect the i.v. kinetics of MDZ, but caused a pronounced reduction (approximately 10-fold) in the systemic exposure to MDZ and, consequently, its Fh following intrahepatic portal vein administration (i.pv.) of MDZ. A comparable extent of the pharmacokinetic interaction also was obtained after a 1.8 mg/kg rifampin dose. Also consistent with the in vitro CYP3A64 inhibition finding, compound A (6 mg/kg i.v.) markedly increased (10-fold) the i.pv. administered MDZ exposure. At the doses studied, plasma concentrations of rifampin or compound A reached or exceeded their respective in vitro EC50 or KI values. These findings suggest the potential applicability of the in vitro-in vivo relationship approach in rhesus monkeys for studying CYP3A-mediated interactions in humans.
Collapse
|
20
|
Baldwin SJ, Bramhall JL, Ashby CA, Yue L, Murdock PR, Hood SR, Ayrton AD, Clarke SE. Cytochrome P450 gene induction in rats ex vivo assessed by quantitative real-time reverse transcriptase-polymerase chain reaction (TaqMan). Drug Metab Dispos 2006; 34:1063-9. [PMID: 16531474 DOI: 10.1124/dmd.105.008185] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Drug-induced changes in expression of cytochrome P450 (P450) genes are a significant issue in the preclinical development of pharmaceuticals. For example, preclinically, P450 induction can affect safety studies by reducing the systemic exposure of a compound undergoing toxicological evaluation, thus limiting the exposure that can be safely investigated in patients. Therefore, the induction potential of candidate drugs has been studied as part of the drug development process, typically using protein and/or catalytic end points. However, measuring changes in the levels of mRNA using TaqMan technology offers the opportunity to investigate this issue with the advantages of better dynamic range and specific enzyme identification. Here, we describe the TaqMan application to study ex vivo the P450 gene induction in the rat. Initially, livers from rats dosed with the prototypic P450 inducers beta-napthoflavone (BNF), phenobarbital (PB), dexamethasone (DEX), and clofibric acid (CLO) were analyzed for mRNA levels of CYP1A1, 1A2, 2B1, 2B2, 2E1, 3A2, 3A23, and 4A1 and compared with control animals. The maximum fold induction of mRNA varied: 2500-fold for CYP1A1 with BNF, 680-fold for CYP2B1 with PB, 59-fold for CYP3A23 with DEX, and 16-fold for CYP4A1 with CLO. This method was then applied to estimate the inductive potential of putative drug candidates undergoing rodent toxicological evaluation. We present a summary of these data that demonstrates the sensitivity and specificity of the TaqMan assay to distinguish between inducers and noninducers and that offers a highly specific alternative to the quantification of drug effects on P450 expression using immunodetection and substrate metabolism.
Collapse
Affiliation(s)
- Sandra J Baldwin
- GlaxoSmithKline, Drug Metabolism and Pharmacokinetics, Ware, UK.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Hyland R, Jones B, van de Waterbeemd H. Utility of human/human-derived reagents in drug discovery and development: An industrial perspective. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2006; 21:179-183. [PMID: 21783655 DOI: 10.1016/j.etap.2005.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The shift to combinatorial chemistry and parallel synthesis in drug discovery has resulted in large numbers of compounds entering the lead seeking and lead development phases of the process. To support this, higher throughput computational (in silico) and in vitro approaches have become the forefront of the drug metabolism and pharmacokinetic (DMPK) input into drug discovery. This has been accompanied by a shift in focus from animal-derived data to human based studies, reflecting the realisation that extrapolation from animals to human has its limitations. In silico approaches may be regarded as human derived tools for DMPK, since models (template/pharmacophore and protein homology modelling), for example, for the human CYP enzymes, are widely used for identifying qualitatively enzyme/substrate interactions. Quantitative assessment of drug metabolism using human hepatocytes or sub-cellular fractions provide a valuable tool both for the screening out of high metabolic lability and in estimations of human intrinsic clearance. In terms of drug absorption, the human colon adenocarcinoma cell line, Caco-2, offers a versatile human derived system for measuring drug permeability, despite over expression of the efflux transporter P-glycoprotein (P-gp). The importance of P-gp can then be further assessed in recombinant systems expressing the human P-gp, where substrate affinity and inhibition potency can be measured, important factors when considering transporter mediated drug-drug interactions. The primary cause of pharmacokinetic-based drug-drug interactions (DDIs) is through enzyme inhibition or induction, with the CYP enzymes being of major importance. Human liver microsomes and hepatocytes are invaluable tools in assessment of DDI vulnerability of new chemical entities, having the capacity to identify enzymes responsible for specific routes of metabolism, and hence areas of vulnerability for a DDI. In addition, human-based screening tools can be used to identify the perpetrator of a DDI through enzyme inhibition/induction. Large differences in the nature of enzymes induced and the extent of induction when comparing animals to man are known. Thus, in vitro models allowing assessment of induction potential in human tissue, establishes some relevance to the clinical situation.
Collapse
Affiliation(s)
- Ruth Hyland
- Pfizer Global Research & Development, PDM, Sandwich, Kent CT13 9NJ, UK
| | | | | |
Collapse
|
22
|
Gibson CR, Lin C, Singh R, Brown CM, Richards K, Brunner J, Michel K, Adelsberger J, Carlini E, Boothe-Genthe C, Raab C, Luu M, Michael A, Parikh M, Ciecko P, Subramanian R, Krolikowski P, Rodrigues AD, Baillie TA, Rushmore TH. Induction of CYP1A in the beagle dog by an inhibitor of kinase insert domain-containing receptor: differential effects in vitro and in vivo on mRNA and functional activity. Drug Metab Dispos 2005; 33:1044-51. [PMID: 15833927 DOI: 10.1124/dmd.105.003913] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Compound I [3-[5-(4-methanesulfonyl-piperazin-1-ylmethyl)-1H-indol-2-yl]-1H-quinolin-2-one] is a potent inhibitor of human kinase insert domain-containing receptor (KDR kinase), which is under investigation for the treatment of cancer. Bile duct-cannulated male beagle dogs were administered 6 mg/kg compound I q.d. for 14 days. There was an approximately 2.5-fold decrease in the mean plasma area under the curve of I on days 7 and 14 (approximately 11.3 microM . h), relative to day 1 (28.2 microM . h). In the dog, compound I was eliminated by metabolism, with a major pathway being aromatic hydroxylation and subsequent sulfation to form the metabolite M3. Metabolic profiling suggested that the pathway leading to the formation of the sulfated conjugate M3 was induced upon multiple dosing of I. Studies conducted in vitro suggested that CYP1A1/2 was responsible for the formation of the hydroxylated metabolite, which is sulfated to yield M3. Additional studies confirmed induction of CYP1A protein and activity in the livers of dogs treated with I. However, studies in a dog hepatocyte model of induction showed a surprising decrease both in CYP1A mRNA and enzymatic activity in the presence of I, emphasizing the need to consider the results from a variety of in vitro and in vivo studies in deriving an understanding of the metabolic fate of a drug candidate. It is concluded that the autoinduction observed after multiple treatments with compound I occurs since compound I is both an inducer and a substrate for dog CYP1A.
Collapse
Affiliation(s)
- Christopher R Gibson
- Department of Drug Metabolism, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Goodsaid FM, Palamanda JR, Montgomery D, Mandakas G, Gu C, Li Z, You X, Norton L, Smith R, Chu I, Soares T, Alton K, Kishnani NS, Rosenblum IY. Assessment of temporal biochemical and gene transcription changes in rat liver cytochrome P450: utility of real-time quantitative RT-PCR. Pharm Res 2004; 20:1373-80. [PMID: 14567630 DOI: 10.1023/a:1025793707794] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE A conventional approach to assess cytochrome P450 (CYP) induction in preclinical animal models involves daily dosing for a least a week followed by Western blot and/or enzyme activity analysis. To evaluate the potential benefit of a third more specific and sensitive assay, real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), with the objective of reducing the duration of the conventional 1-week study, we simultaneously assessed gene expression by qRT-PCR along with Western blots and enzyme activity assays as a time course in an in vivo model. METHODS Rats were dosed daily for 8 days with model inducers of CYP1A, CYP2B, CYP3A, or CYP4A. Liver P450 levels were measured after 0.5, 1, 2, 4, and 8 days of dosing by qRT-PCR, Western blot, and enzyme activity. RESULTS CYP1A, CYP3A, and CYP4A genes were maximally induced very rapidly (0.5-1 day), whereas the CYP2B gene was maximally induced after a lag time of 4 days. In all cases, fold changes in induction detected by qRT-PCR were greater than fold changes in protein levels and enzyme activities. CONCLUSIONS Maximal persistent and larger fold changes observed by qRT-PCR either preceded or occurred simultaneously with maximal sustained fold changes in protein levels as measured by Western blots and enzyme activity assays. Our data show that qRT-PCR provides increased sensitivity and specificity over conventional assays and may be key information for reliable assessment of drug-related changes in CYP induction during the transition from discovery to toxicology studies.
Collapse
Affiliation(s)
- Federico M Goodsaid
- Department of Genetic and Molecular Toxicology, Schering-Plough Research Institute, PO Box 32, Lafayette, New Jersey 07848, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bjornsson TD, Callaghan JT, Einolf HJ, Fischer V, Gan L, Grimm S, Kao J, King SP, Miwa G, Ni L, Kumar G, McLeod J, Obach RS, Roberts S, Roe A, Shah A, Snikeris F, Sullivan JT, Tweedie D, Vega JM, Walsh J, Wrighton SA. The conduct of in vitro and in vivo drug-drug interaction studies: a Pharmaceutical Research and Manufacturers of America (PhRMA) perspective. Drug Metab Dispos 2003; 31:815-32. [PMID: 12814957 DOI: 10.1124/dmd.31.7.815] [Citation(s) in RCA: 550] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Current regulatory guidances do not address specific study designs for in vitro and in vivo drug-drug interaction studies. There is a common desire by regulatory authorities and by industry sponsors to harmonize approaches, to allow for a better assessment of the significance of findings across different studies and drugs. There is also a growing consensus for the standardization of cytochrome P450 (P450) probe substrates, inhibitors and inducers and for the development of classification systems to improve the communication of risk to health care providers and to patients. While existing guidances cover mainly P450-mediated drug interactions, the importance of other mechanisms, such as transporters, has been recognized more recently, and should also be addressed. This article was prepared by the Pharmaceutical Research and Manufacturers of America (PhRMA) Drug Metabolism and Clinical Pharmacology Technical Working Groups and represents the current industry position. The intent is to define a minimal best practice for in vitro and in vivo pharmacokinetic drug-drug interaction studies targeted to development (not discovery support) and to define a data package that can be expected by regulatory agencies in compound registration dossiers.
Collapse
|
25
|
Seegers U, Potschka H, Löscher W. Lack of effects of prolonged treatment with phenobarbital or phenytoin on the expression of P-glycoprotein in various rat brain regions. Eur J Pharmacol 2002; 451:149-55. [PMID: 12231384 DOI: 10.1016/s0014-2999(02)02235-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
P-glycoprotein is an ATP-dependent drug transport protein that is predominantly found in the apical membranes of various epithelial cell types in the body, including the blood luminar membrane of the brain capillary endothelial cells that make up the blood-brain barrier. Increased P-glycoprotein expression in the blood-brain barrier has been described in epileptogenic brain tissue of patients with pharmacoresistant epilepsy, suggesting that overexpression of P-glycoprotein may be involved in multidrug resistance of epilepsy. The mechanisms underlying the overexpression of P-glycoprotein in brain tissue of epileptic patients are not clear. Two antiepileptic drugs, phenobarbital and phenytoin, have been reported to up-regulate P-glycoprotein in cell cultures, so that chronic treatment with antiepileptic drugs may enhance P-glycoprotein expression in the blood-brain barrier. To directly address this possibility, we treated rats with phenobarbital or phenytoin over 11 days and subsequently determined expression of P-glycoprotein by immunohistochemistry in endothelium and parenchyma of several brain regions, including regions of the temporal lobe, which is often involved in pharmacoresistant types of epilepsy. Except for a moderate increase in the intensity of P-glycoprotein expression in the piriform/parietal cortex and cerebellum of phenobarbital-treated rats, no significant P-glycoprotein increases were seen after prolonged treatment with phenobarbital or phenytoin in any brain region examined. In view of recent findings that seizures lead to a transient induction of P-glycoprotein in the brain of rats, it seems reasonable to suggest that the overexpression of P-glycoprotein in brain regions of patients with intractable epilepsy is a consequence of uncontrolled seizures rather than of chronic treatment with antiepileptic drugs.
Collapse
Affiliation(s)
- Ulrike Seegers
- Department of Pharmacology, Toxicology and Pharmacy, School of Veterinary Medicine, Bünteweg 17 Building 218, D-30559 Hannover, Germany
| | | | | |
Collapse
|
26
|
Gibson GG, Plant NJ, Swales KE, Ayrton A, El-Sankary W. Receptor-dependent transcriptional activation of cytochrome P4503A genes: induction mechanisms, species differences and interindividual variation in man. Xenobiotica 2002; 32:165-206. [PMID: 11958559 DOI: 10.1080/00498250110102674] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
1. The importance of CYP3A enzymes in drug metabolism and toxicology has yielded a wealth of information on the structure, function and regulation of this subfamily and recent research emphasis has been placed on the human forms, namely CYP3A4, CYP3A5, CYP3A7 and CYP3A43. 2. The current review will focus on the receptor-dependency of CYP3A regulation and includes consideration of the regulatory roles of the glucocorticoid (GR), pregnane X (PXR) and constitutive androstane (CAR) receptors. 3. Emphasis has been placed on the topics of expression and substrate specificity, assessment of induction, species differences in induction, CYP3A promoter sequences and regulation of gene expression, structural and functional aspects of receptor-mediated, CYP3A gene activation, receptor variants and interindividual variation in human CYP3A expression, the latter encompassing environmental, physiological and genetic aspects. 4. An outline of future research needs will be discussed in the context of receptor-mediated molecular mechanisms of CYP3A gene regulation and the impact on interindividual variations in CYP3A expression. 5. Taken collectively, this review highlights the importance of understanding the molecular mechanisms of CYP3A induction as a means of rationalizing human responses to many clinically used drugs, in addition to providing a mechanistically coherent platform to understand and predict interindividual variations in response and drug-drug interactions.
Collapse
Affiliation(s)
- G G Gibson
- Molecular Toxicology Group, School of Biomedical and Life Sciences, University of Surrey, Guildford, Surrey, UK.
| | | | | | | | | |
Collapse
|
27
|
Matsubara T. Safety Evaluation and Drug Development based on Biological Fate of Drugs —Efforts Made to Overcome Drug Interaction in Drug Development—. Drug Metab Pharmacokinet 2002; 17:379-94. [PMID: 15618690 DOI: 10.2133/dmpk.17.379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. Assay methods to detect drug interaction in toxicological samples were established by determining cytochrome P450 content and its activity in liver samples. The O-dealkylation reaction of 7-alkoxycoumarin was indicated to reflect changes in the molecular forms of P450s, and the enzyme induction or inhibition in the toxicological samples was easily detected by using the established methods. 2. During toxicological studies of 450191-S or the sleep inducer rilmazafone, a phenobarbital type-induction of hepatic drug metabolizing enzymes was observed in animals, and the doses required for the induction differed markedly between rats and dogs. Enzyme induction was caused by some specific metabolites of 450191-S, and the plasma concentrations of these metabolites were comparable when the enzyme induction was developed in both animals. 3. A nonsteroidal antiinflammatory compound 480156-S showed a slight or no effect on microsomal drug metabolizing activity in rats. On the other hand, repeated administration of this compound to humans resulted in a marked decrease in the oxidative metabolism of 480156-S, followed by a marked increase in the plasma concentrations of the compound. When volunteers were given 480156-S followed by several drugs, such as tolubutamide, the plasma clearance was delayed remarkably, indicating a severe drug interaction. 4. Cytochrome P450 belonging to the CYP2C family was indicated to participate in the oxidative metabolism of 480156-S in both rat and human liver microsomes. The preincubation of microsomes with 480156-S caused a concentration-dependent inhibition of CYP2C-dependent tolubutamide hydroxylation reaction in both rats and humans. There was a marked species difference in the susceptibility to the inhibitory effect of 480156-S, and the concentration required to inhibit rat CYP2C was almost 10 times higher than that required in humans. 5. The cephem antibiotics having N-methyltetrazolethiol (NMTT) at the 3'-position substituent were demonstrated to inhibit mitochondrial low K(m) aldehyde dehydrogenase (ALDH), and produced disulfiram-like (Antabuse) reaction during alcohol metabolism. Pharmacokinetic studies indicated that NMTT released from the antibiotics in bile duct or intestine cause the inhibitory action followed by the development of disulfiram-like reaction. 6. Attempts had been made to develop new cephem antibiotics lacking the disulfiram-like reaction by changing the chemical structure of 3'-position substituents, and a hydroxyethyltetrazolethiol was found not to inhibit the enzyme. Based on this result, together with the antibacterial activity, we have developed a new oxacephem antibiotic flomoxef (6315-S). Flomoxef showed no disulfiram-like reaction both in rats and human.
Collapse
Affiliation(s)
- Takashi Matsubara
- Department of Chemical Biology, Osaka City University Medical School, Japan.
| |
Collapse
|