1
|
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
|
2
|
Ushirozako G, Murayama N, Tsukiyama-Kohara K, Yamazaki H, Uno Y. Novel Tree Shrew Cytochrome P450 2Ds (CYP2D8a and CYP2D8b) Are Functional Drug-Metabolizing Enzymes that Metabolize Bufuralol and Dextromethorphan. Drug Metab Dispos 2024; 52:305-311. [PMID: 38262704 DOI: 10.1124/dmd.123.001603] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/25/2024] Open
Abstract
Tree shrews are a nonprimate species used in a range of biomedical studies. Recent genome analysis of tree shrews found that the sequence identities and the numbers of genes of cytochrome P450 (CYP or P450), an important family of drug-metabolizing enzymes, are similar to those of humans. However, tree shrew P450s have not yet been sufficiently identified and analyzed. In this study, novel CYP2D8a and CYP2D8b cDNAs were isolated from tree shrew liver and were characterized, along with human CYP2D6, dog CYP2D15, and pig CYP2D25. The amino acid sequences of these tree shrew CYP2Ds were 75%-78% identical to human CYP2D6, and phylogenetic analysis showed that they were more closely related to human CYP2D6 than rat CYP2Ds, similar to dog and pig CYP2Ds. For tree shrew CYP2D8b, two additional transcripts were isolated that contained different patterns of deletion. The gene and genome structures of CYP2Ds are generally similar in dogs, humans, pigs, and tree shrews. Tree shrew CYP2D8a mRNA was most abundantly expressed in liver, among the tissue types analyzed, similar to dog CYP2D15 and pig CYP2D25 mRNAs. Tree shrew CYP2D8b mRNA was also expressed in liver, but at a level 7.3-fold lower than CYP2D8a mRNA. Liver microsomes and recombinant protein of both tree shrew CYP2Ds metabolized bufuralol and dextromethorphan, selective substrates of human CYP2D6, but the activity level of CYP2D8a greatly exceeded that of CYP2D8b. These results suggest that tree shrew CYP2D8a and CYP2D8b are functional drug-metabolizing enzymes, of which CYP2D8a is the major CYP2D in liver. SIGNIFICANCE STATEMENT: Novel tree shrew CYP2D8a and CYP2D8b cDNAs were isolated from liver. Their amino acid sequences were 75%-78% identical to human CYP2D6. For CYP2D8b, two additional transcripts contained different patterns of deletion. Tree shrew CYP2D8a mRNA was abundantly expressed in liver, similar to dog CYP2D15 and pig CYP2D25 mRNAs. Recombinant tree shrew CYP2Ds catalyzed the oxidation of bufuralol and dextromethorphan. Tree shrew CYP2D8a and CYP2D8b are functional drug-metabolizing enzymes, of which CYP2D8a is the major CYP2D in liver.
Collapse
Affiliation(s)
- Genki Ushirozako
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima, Japan (G.U., K.T.-K., Y.U.); and Showa Pharmaceutical University, Machida, Tokyo, Japan (N.M., H.Y.)
| | - Norie Murayama
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima, Japan (G.U., K.T.-K., Y.U.); and Showa Pharmaceutical University, Machida, Tokyo, Japan (N.M., H.Y.)
| | - Kyoko Tsukiyama-Kohara
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima, Japan (G.U., K.T.-K., Y.U.); and Showa Pharmaceutical University, Machida, Tokyo, Japan (N.M., H.Y.)
| | - Hiroshi Yamazaki
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima, Japan (G.U., K.T.-K., Y.U.); and Showa Pharmaceutical University, Machida, Tokyo, Japan (N.M., H.Y.)
| | - Yasuhiro Uno
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima, Japan (G.U., K.T.-K., Y.U.); and Showa Pharmaceutical University, Machida, Tokyo, Japan (N.M., H.Y.)
| |
Collapse
|
3
|
Ayuso M, Buyssens L, Stroe M, Valenzuela A, Allegaert K, Smits A, Annaert P, Mulder A, Carpentier S, Van Ginneken C, Van Cruchten S. The Neonatal and Juvenile Pig in Pediatric Drug Discovery and Development. Pharmaceutics 2020; 13:44. [PMID: 33396805 PMCID: PMC7823749 DOI: 10.3390/pharmaceutics13010044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Pharmacotherapy in pediatric patients is challenging in view of the maturation of organ systems and processes that affect pharmacokinetics and pharmacodynamics. Especially for the youngest age groups and for pediatric-only indications, neonatal and juvenile animal models can be useful to assess drug safety and to better understand the mechanisms of diseases or conditions. In this respect, the use of neonatal and juvenile pigs in the field of pediatric drug discovery and development is promising, although still limited at this point. This review summarizes the comparative postnatal development of pigs and humans and discusses the advantages of the juvenile pig in view of developmental pharmacology, pediatric diseases, drug discovery and drug safety testing. Furthermore, limitations and unexplored aspects of this large animal model are covered. At this point in time, the potential of the neonatal and juvenile pig as nonclinical safety models for pediatric drug development is underexplored.
Collapse
Affiliation(s)
- Miriam Ayuso
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium; (L.B.); (M.S.); (A.V.); (C.V.G.)
| | - Laura Buyssens
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium; (L.B.); (M.S.); (A.V.); (C.V.G.)
| | - Marina Stroe
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium; (L.B.); (M.S.); (A.V.); (C.V.G.)
| | - Allan Valenzuela
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium; (L.B.); (M.S.); (A.V.); (C.V.G.)
| | - Karel Allegaert
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; (K.A.); (P.A.)
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
- Department of Hospital Pharmacy, Erasmus MC Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Anne Smits
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
- Neonatal Intensive Care Unit, University Hospitals UZ Leuven, 3000 Leuven, Belgium
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; (K.A.); (P.A.)
| | - Antonius Mulder
- Department of Neonatology, University Hospital Antwerp, 2650 Edegem, Belgium;
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, 2610 Wilrijk, Belgium
| | | | - Chris Van Ginneken
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium; (L.B.); (M.S.); (A.V.); (C.V.G.)
| | - Steven Van Cruchten
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium; (L.B.); (M.S.); (A.V.); (C.V.G.)
| |
Collapse
|
4
|
Characterization of Porcine Hepatic and Intestinal Drug Metabolizing CYP450: Comparison with Human Orthologues from A Quantitative, Activity and Selectivity Perspective. Sci Rep 2019; 9:9233. [PMID: 31239454 PMCID: PMC6592956 DOI: 10.1038/s41598-019-45212-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Over the past two decades, the pig has gained attention as a potential model for human drug metabolism. Cytochrome P450 enzymes (CYP450), a superfamily of biotransformation enzymes, are pivotal in drug metabolism. Porcine CYP450 has been demonstrated to convert typical substrates of human CYP450. Nevertheless, knowledge and insight into porcine CYP450 quantity and substrate selectivity is scant, especially regarding intestinal CYP450. The current study aimed to map the quantities of hepatic and intestinal CYP450 in the conventional pig by using a proteomic approach. Moreover, the selectivity of the six most common used probe substrates (phenacetin, coumarin, midazolam, tolbutamide, dextromethorphan, and chlorzoxazone) for drug metabolizing enzyme subfamilies (CYP1A, CYP2A, CYP3A, CYP2C, CYP2D and CYP2E respectively), was investigated. Hepatic relative quantities were 4% (CYP1A), 31% (CYP2A), 14% (CYP3A), 10% (CYP2C), 28% (CYP2D) and 13% (CYP2E), whereas for the intestine only duodenal CYP450 could be determined with 88% for CYP3A and 12% for CYP2C. Furthermore, the results indicate that coumarin (CYP2A), midazolam (CYP3A), tolbutamide (CYP2C), and dextromethorphan (CYP2D) are as selective for porcine as for human CYP450. However, phenacetin (CYP1A2) and chlorzoxazone (CYP2E1) are less selective for the specific enzyme, despite similarities in selectivity towards the different enzymes involved compared to humans.
Collapse
|
5
|
In vitro Phase I- and Phase II-Drug Metabolism in The Liver of Juvenile and Adult Göttingen Minipigs. Pharm Res 2017; 34:750-764. [DOI: 10.1007/s11095-017-2101-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 01/06/2017] [Indexed: 10/20/2022]
|
6
|
Hu SX, Mazur CA, Feenstra KL, Lorenz JK, Merritt DA. Assessment of inhibition of porcine hepatic cytochrome P450 enzymes by 48 commercial drugs. Vet J 2016; 211:26-31. [PMID: 27053015 DOI: 10.1016/j.tvjl.2016.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 01/26/2016] [Accepted: 03/13/2016] [Indexed: 11/30/2022]
Abstract
Drug interactions due to inhibition of hepatic cytochrome P450 (CYP450) enzymes are not well understood in veterinary medicine. Forty-eight commercial porcine medicines were selected to evaluate their potential inhibition on porcine hepatic CYP450 enzymes at their commercial doses and administration routes. Those drugs were first assessed through a single point inhibitory assay at 3 µM in porcine liver microsomes for six specific CYP450 metabolisms (phenacetin o-deethylation, coumarin 7-hydroxylation, tolbutamide 4-hydroxylation, bufuralol 1-hydroxylation, chlorozoxazone 6-hydroxylation and midazolam 1'-hydroxylation). When the inhibition was > 10% in the single point inhibitory assay, IC50 values (inhibitory concentrations that decrease biotransformation of selected substrate by 50%) were determined. Overall, 17 drugs showed in vitro inhibition on one or more porcine hepatic CYP450 metabolisms with different IC50 values. The potential in vivo porcine hepatic CYP450 inhibition by those drugs was assessed by combining the in vitro data and in vivo Cmax (maximum plasma concentrations from pharmacokinetic studies of the porcine medicines at their commercial doses and administration routes). Three drugs showed high potential inhibition to one or two porcine hepatic CYP450 isoforms at their commercial doses and administration routes, while seven drugs had medium risk and seven had low risk of such in vivo inhibition. These data are useful to prevent potential drug interactions in veterinary medical practice.
Collapse
Affiliation(s)
- Steven X Hu
- Veterinary Medicine Research and Development, Zoetis, Inc, 333 Portage Street, Kalamazoo, MI 49007, USA.
| | - Chase A Mazur
- Veterinary Medicine Research and Development, Zoetis, Inc, 333 Portage Street, Kalamazoo, MI 49007, USA
| | - Kenneth L Feenstra
- Veterinary Medicine Research and Development, Zoetis, Inc, 333 Portage Street, Kalamazoo, MI 49007, USA
| | - Julie K Lorenz
- Veterinary Medicine Research and Development, Zoetis, Inc, 333 Portage Street, Kalamazoo, MI 49007, USA
| | - Dawn A Merritt
- Veterinary Medicine Research and Development, Zoetis, Inc, 333 Portage Street, Kalamazoo, MI 49007, USA
| |
Collapse
|
7
|
Comparison of minipig, dog, monkey and human drug metabolism and disposition. J Pharmacol Toxicol Methods 2014; 74:80-92. [PMID: 25545337 DOI: 10.1016/j.vascn.2014.12.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/02/2014] [Accepted: 12/16/2014] [Indexed: 02/06/2023]
Abstract
INTRODUCTION This article gives an overview of the drug metabolism and disposition (ADME) characteristics of the most common non-rodent species used in toxicity testing of drugs (minipigs, dogs, and monkeys) and compares these to human characteristics with regard to enzymes mediating the metabolism of drugs and the transport proteins which contribute to the absorption, distribution and excretion of drugs. METHODS Literature on ADME and regulatory guidelines of relevance in drug development of small molecules has been gathered. RESULTS Non-human primates (monkeys) are the species that is closest to humans in terms of genetic homology. Dogs have an advantage due to the ready availability of comprehensive background data for toxicological safety assessment and dogs are easy to handle. Pigs have been used less than dogs and monkeys as a model in safety assessment of drug candidates. However, when a drug candidate is metabolised by aldehyde oxidase (AOX1), N-acetyltransferases (NAT1 and NAT2) or cytochrome (CYP2C9-like) enzymes which are not expressed in dogs, but are present in pigs, this species may be a better choice than dogs, provided that adequate exposure can be obtained in pigs. Conversely, pigs might not be the right choice if sulfation, involving 3-phospho-adenosyl-5-phosphosulphate sulphotransferase (PAPS) is an important pathway in the human metabolism of a drug candidate. DISCUSSION In general, the species selection should be based on comparison between in vitro studies with human cell-based systems and animal-cell-based systems. Results from pharmacokinetic studies are also important for decision-making by establishing the obtainable exposure level in the species. Access to genetically humanized mouse models and highly sensitive analytical methods (accelerator mass spectrometry) makes it possible to improve the chance of finding all metabolites relevant for humans before clinical trials have been initiated and, if necessary, to include another animal species before long term toxicity studies are initiated. In conclusion, safety testing can be optimized by applying knowledge about species ADME differences and utilising advanced analytical techniques.
Collapse
|
8
|
Hu SX. Impact of age on hepatic cytochrome P450 of domestic male Camborough-29 pigs. J Vet Pharmacol Ther 2014; 38:150-9. [DOI: 10.1111/jvp.12163] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/07/2014] [Indexed: 01/19/2023]
Affiliation(s)
- S. X. Hu
- Zoetis Inc; Veterinary Medicine Research and Development; Kalamazoo MI USA
| |
Collapse
|
9
|
Sjögren E, Abrahamsson B, Augustijns P, Becker D, Bolger MB, Brewster M, Brouwers J, Flanagan T, Harwood M, Heinen C, Holm R, Juretschke HP, Kubbinga M, Lindahl A, Lukacova V, Münster U, Neuhoff S, Nguyen MA, Peer AV, Reppas C, Hodjegan AR, Tannergren C, Weitschies W, Wilson C, Zane P, Lennernäs H, Langguth P. In vivo methods for drug absorption – Comparative physiologies, model selection, correlations with in vitro methods (IVIVC), and applications for formulation/API/excipient characterization including food effects. Eur J Pharm Sci 2014; 57:99-151. [PMID: 24637348 DOI: 10.1016/j.ejps.2014.02.010] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 02/15/2014] [Accepted: 02/17/2014] [Indexed: 01/11/2023]
|
10
|
A Physiologically Based Pharmacokinetic Model of the Minipig: Data Compilation and Model Implementation. Pharm Res 2012. [DOI: 10.1007/s11095-012-0911-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
11
|
Ganderup NC, Harvey W, Mortensen JT, Harrouk W. The minipig as nonrodent species in toxicology--where are we now? Int J Toxicol 2012; 31:507-28. [PMID: 23134714 DOI: 10.1177/1091581812462039] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Over the past 3 decades minipigs have moved from being an obscure alternative to dogs and nonhuman primates to being a standard animal model in regulatory toxicity studies. This article covers the use of minipigs as a model in the context of nonclinical drug safety and provides an overview of the minipig's developmental history and relates minipigs to other animal species commonly used in toxicology; and the minipig's translational power is supported by 43 case studies of marketed drug products covered. Special focus is given to criteria for selecting minipigs in nonclinical programs supporting the development of new medicines; the use of swine in the assessment of food additives, agrochemicals, and pesticides; as well as a regulatory perspective on the use of minipigs in Food and Drug Administration (FDA)-regulated products. This article presents the main points conveyed at a symposium held at the 2010 American College of Toxicology meeting in Baltimore, Maryland.
Collapse
|
12
|
Improvac does not modify the expression and activities of the major drug metabolizing enzymes cytochrome P450 3A and 2C in pigs. Vaccine 2012; 30:3515-8. [DOI: 10.1016/j.vaccine.2012.03.072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 03/21/2012] [Accepted: 03/22/2012] [Indexed: 11/22/2022]
|
13
|
Thörn HA, Lundahl A, Schrickx JA, Dickinson PA, Lennernäs H. Drug metabolism of CYP3A4, CYP2C9 and CYP2D6 substrates in pigs and humans. Eur J Pharm Sci 2011; 43:89-98. [DOI: 10.1016/j.ejps.2011.03.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 02/08/2011] [Accepted: 03/18/2011] [Indexed: 11/28/2022]
|
14
|
Drug Metabolism in Hemorrhagic Shock: Pharmacokinetics of Selective Markers of Cytochrome-P450 2C9, 2D6, and 3A4 Enzyme Activities in a Porcine Model. J Surg Res 2011; 167:e231-43. [DOI: 10.1016/j.jss.2010.06.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 06/10/2010] [Accepted: 06/22/2010] [Indexed: 11/21/2022]
|
15
|
Bode G, Clausing P, Gervais F, Loegsted J, Luft J, Nogues V, Sims J. The utility of the minipig as an animal model in regulatory toxicology. J Pharmacol Toxicol Methods 2010; 62:196-220. [DOI: 10.1016/j.vascn.2010.05.009] [Citation(s) in RCA: 309] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 05/21/2010] [Accepted: 05/24/2010] [Indexed: 11/26/2022]
|
16
|
Pegolo S, Giantin M, Dacasto M, Montesissa C, Capolongo F. Testosterone hydroxylation in bovine liver: enzyme kinetic and inhibition study. Xenobiotica 2010; 40:255-61. [DOI: 10.3109/00498250903540875] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
17
|
Implications of hepatic cytochrome P450-related biotransformation processes in veterinary sciences. Eur J Pharmacol 2008; 585:502-9. [DOI: 10.1016/j.ejphar.2008.03.013] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Revised: 03/05/2008] [Accepted: 03/06/2008] [Indexed: 02/07/2023]
|
18
|
Jacobs A. Use of nontraditional animals for evaluation of pharmaceutical products. Expert Opin Drug Metab Toxicol 2007; 2:345-9. [PMID: 16863438 DOI: 10.1517/17425255.2.3.345] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Although the International Conference on Harmonization Guideline ICH M3 indicates the use of nonrodents for some studies of pharmaceutical products, the specific nonrodent species is not specified. Dogs are used most frequently; however, there may be reasons why dogs are not the best model for a particular drug. Minipigs are being used increasingly for evaluation of toxicity, especially for dermally applied drugs, and for various efficacy models. Hamsters may be used for the evaluation of intraoral drugs and for carcinogenicity studies. Less commonly, pharmaceutical manufacturers may choose on their own to use marmosets, when a nonhuman primate is considered critical to evaluation, or to use ferrets for specific purposes. When nontraditional species are used, there may be less historical information available and unique issues of their care, and differences in physiology and anatomy and susceptibility to infection need to be understood. Nonmammalian test species, such as zebrafish and Caenorhabditis elegans may be used by drug sponsors in screening assays, but are not yet ready for use in pivotal toxicology studies because of the difficulty in extrapolating to mammalian species. Use of nontraditional animal species may be proposed by a drug sponsor to a reviewing division with supporting data and reasons for using a particular species.
Collapse
Affiliation(s)
- Abigail Jacobs
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA.
| |
Collapse
|
19
|
Cumming P, Møller M, Benda K, Minuzzi L, Jakobsen S, Jensen SB, Pakkenberg B, Stark AK, Gramsbergen JB, Andreasen MF, Olsen AK. A PET study of effects of chronic 3,4-methylenedioxymethamphetamine (MDMA, “ecstasy”) on serotonin markers in Göttingen minipig brain. Synapse 2007; 61:478-87. [PMID: 17415793 DOI: 10.1002/syn.20377] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The psychostimulant 3,4-methylendioxymethamphetamine (MDMA, "ecstasy") evokes degeneration of telencephalic serotonin innervations in rodents, nonhuman primates, and human recreational drug users. However, there has been no alternative to nonhuman primates for studies of the cognitive and neurochemical consequences of serotonin depletion in a large-bodied animal. Therefore, we used positron emission tomography (PET) with [(11)C]DASB to map the distribution of plasma membrane serotonin transporters in brain of Göttingen minipigs, first in a baseline condition, and again at 2 weeks after treatment with MDMA (i.m.), administered at a range of doses. In parallel PET studies, [(11)C]WAY-100635 was used to map the distribution of serotonin 5HT(1A) receptors. The acute MDMA treatment in awake pigs evoked 1 degrees C of hyperthermia. MDMA at total doses greater than 20 mg/kg administered over 2-4 days reduced the binding potential (pB) of [(11)C]DASB for serotonin transporters in porcine brain. A mean total dose of 42 mg/kg MDMA in four animals evoked a mean 32% decrease in [(11)C]DASB pB in mesencephalon and diencephalon, and a mean 53% decrease in telencephalic structures. However, this depletion of serotonin innervations was not associated with consistent alterations in the binding of [(11)C]WAY-100635 to serotonin 5HT(1A) receptors. Stereological cell counting of serotonin-positive neurons, which numbered 95,000 in the dorsal raphé nucleus of normal animals, was unaffected in MDMA-treated group. group.
Collapse
Affiliation(s)
- Paul Cumming
- PET Centre and Centre for Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Suzuki S, Satoh T, Yoshino H, Kobayashi E. Impact of warm ischemic time on microsomal P450 isoforms in a porcine model of therapeutic liver resection. Life Sci 2004; 76:39-46. [PMID: 15501478 DOI: 10.1016/j.lfs.2004.06.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2003] [Accepted: 06/07/2004] [Indexed: 11/22/2022]
Abstract
Human microsomes and hepatocytes obtained from non-transplantable livers of brain-dead donors are very useful in predicting the in vivo metabolism of xenobiotics in humans. Fresh liver specimens obtained from therapeutic liver resection are also useful for research in cases where non-transplantable livers are not readily available. In the present study, the effect of warm ischemic duration, in the course of hepatic surgery, on the activities of liver cytochrome P450 (CYP) CYP1A, CYP2C, CYP2D, CYP2E1 and CYP3A were evaluated in a porcine model. Partial occlusion (portal vein and hepatic artery occlusion) decreased the activities of CYP2C, CYP2E and CYP3A, but not those of CYP1A and CYP2D. CYP3A, known to account for an average 30% of total P450 content in the human liver was the most susceptible to the warm ischemia. These results demonstrate that the activities of CYP isoforms, particularly those of CYP3A, are markedly affected by warm ischemia; it is, therefore, essential that care should be exercised when using microsomes prepared from surgically removed livers.
Collapse
Affiliation(s)
- Satoshi Suzuki
- Research Laboratories, HAB Research Organization, Cornea Center Bldg. 3rd Floor, Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa, Chiba 272-8523, Japan.
| | | | | | | |
Collapse
|
21
|
Sakuma T, Shimojima T, Miwa K, Kamataki T. Cloning CYP2D21 and CYP3A22 cDNAs from liver of miniature pigs. Drug Metab Dispos 2004; 32:376-8. [PMID: 15039288 DOI: 10.1124/dmd.32.4.376] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To compare the identity of the primary structure of drug-metabolizing cytochrome P450 between miniature pigs and humans, two cDNA clones, coding for miniature pig CYP2D21 and CYP3A22, were isolated. The deduced amino acid sequences of CYP2D21 and CYP3A22 were 78.3 and 75.0% identical to human CYP2D6 and CYP3A4, respectively. These values were nearly the same as those of bovine, dog, and some rodent isoforms, and 12.2 to 18.4% lower than those of nonhuman primates such as cynomolgus monkeys, Japanese monkey, and marmosets. These data indicate that miniature pig P450s are genetically not so close as monkey P450s to human P450s as previously expected. The recombinant CYP2D21 enzyme, however, showed bufuralol 1'-hydroxylase activity, suggesting that miniature pig CYP2D21 is capable of metabolizing some of the same substrates associated with human CYP2D6 despite its low identity to human counterparts.
Collapse
Affiliation(s)
- Tsutomu Sakuma
- Department of Toxicology, Faculty of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University, 2630 Sugitani, Toyama, Toyama 930-0194, Japan.
| | | | | | | |
Collapse
|