1
|
Asano D, Hamaue S, Zahir H, Shiozawa H, Nishiya Y, Kimura T, Kazui M, Yamamura N, Ikeguchi M, Shibayama T, Inoue SI, Shinozuka T, Watanabe T, Yahara C, Watanabe N, Yoshinari K. CYP2C8-Mediated Formation of a Human Disproportionate Metabolite of the Selective Na V1.7 Inhibitor DS-1971a, a Mixed Cytochrome P450 and Aldehyde Oxidase Substrate. Drug Metab Dispos 2022; 50:235-242. [PMID: 34930785 DOI: 10.1124/dmd.121.000665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022] Open
Abstract
Predicting human disproportionate metabolites is difficult, especially when drugs undergo species-specific metabolism mediated by cytochrome P450s (P450s) and/or non-P450 enzymes. This study assessed human metabolites of DS-1971a, a potent Nav1.7-selective blocker, by performing human mass balance studies and characterizing DS-1971a metabolites, in accordance with the Metabolites in Safety Testing guidance. In addition, we investigated the mechanism by which the major human disproportionate metabolite (M1) was formed. After oral administration of radiolabeled DS-1971a, the major metabolites in human plasma were P450-mediated monoxidized metabolites M1 and M2 with area under the curve ratios of 27% and 10% of total drug-related exposure, respectively; the minor metabolites were dioxidized metabolites produced by aldehyde oxidase and P450s. By comparing exposure levels of M1 and M2 between humans and safety assessment animals, M1 but not M2 was found to be a human disproportionate metabolite, requiring further characterization under the Metabolites in Safety Testing guidance. Incubation studies with human liver microsomes indicated that CYP2C8 was responsible for the formation of M1. Docking simulation indicated that, in the formation of M1 and M2, there would be hydrogen bonding and/or electrostatic interactions between the pyrimidine and sulfonamide moieties of DS-1971a and amino acid residues Ser100, Ile102, Ile106, Thr107, and Asn217 in CYP2C8, and that the cyclohexane ring of DS-1971a would be located near the heme iron of CYP2C8. These results clearly indicate that M1 is the predominant metabolite in humans and a human disproportionate metabolite due to species-specific differences in metabolism. SIGNIFICANCE STATEMENT: This report is the first to show a human disproportionate metabolite generated by CYP2C8-mediated primary metabolism. We clearly demonstrate that DS-1971a, a mixed aldehyde oxidase and cytochrome P450 substrate, was predominantly metabolized by CYP2C8 to form M1, a human disproportionate metabolite. Species differences in the formation of M1 highlight the regio- and stereoselective metabolism by CYP2C8, and the proposed interaction between DS-1971a and CYP2C8 provides new knowledge of CYP2C8-mediated metabolism of cyclohexane-containing substrates.
Collapse
Affiliation(s)
- Daigo Asano
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Syoya Hamaue
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Hamim Zahir
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Hideyuki Shiozawa
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Yumi Nishiya
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Takako Kimura
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Miho Kazui
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Naotoshi Yamamura
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Marie Ikeguchi
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Takahiro Shibayama
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Shin-Ichi Inoue
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Tsuyoshi Shinozuka
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Toshiyuki Watanabe
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Chizuko Yahara
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Nobuaki Watanabe
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| | - Kouichi Yoshinari
- Drug Metabolism and Pharmacokinetics Research Laboratories (D.A., H.S., Y.N., M.K., N.Y., Ta.S., S.I., C.Y., N.W.), Translational Science Department (M.I.), R&D Planning and Management Department (Ts.S.), and Medicinal Safety Research Laboratories (T.W.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; Organic and Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan (S.H., T.K.); Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc., Basking Ridge, New Jersey (H.Z.); and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan (K.Y.)
| |
Collapse
|
2
|
Ren Z, Bremer AA, Pawlyk AC. Drug development research in pregnant and lactating women. Am J Obstet Gynecol 2021; 225:33-42. [PMID: 33887238 DOI: 10.1016/j.ajog.2021.04.227] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 12/15/2022]
Abstract
Pregnant and lactating women are considered "therapeutic orphans" because they generally have been excluded from clinical drug research and the drug development process owing to legal, ethical, and safety concerns. Most medications prescribed for pregnant and lactating women are used "off-label" because most of the clinical approved medications do not have appropriate drug labeling information for pregnant and lactating women. Medications that lack human safety data on use during pregnancy and lactation may pose potential risks for adverse effects in pregnant and lactating women as well as risks of teratogenic effects to their unborn and newborn babies. Federal policy requiring the inclusion of women in clinical research and trials led to considerable changes in research design and practice. Despite more women being included in clinical research and trials, the inclusion of pregnant and lactating women in drug research and clinical trials remains limited. A recent revision to the "Common Rule" that removed pregnant women from the classification as a "vulnerable" population may change the culture of drug research and drug development in pregnant and lactating women. This review article provides an overview of medications studied by the Obstetric-Fetal Pharmacology Research Units Network and Centers and describes the challenges in current obstetrical pharmacology research and alternative strategies for future research in precision therapeutics in pregnant and lactating women. Implementation of the recommendations of the Task Force on Research Specific to Pregnant Women and Lactating Women can provide legislative requirements and opportunities for research focused on pregnant and lactating women.
Collapse
Affiliation(s)
- Zhaoxia Ren
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD.
| | - Andrew A Bremer
- Pediatric Growth and Nutrition Branch, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD; Pregnancy and Perinatology Branch, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Aaron C Pawlyk
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| |
Collapse
|
3
|
Singh A, Zhao K, Bell C, Shah AJ. Effect of berberine on in vitro metabolism of sulfonylureas: A herb-drug interactions study. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34 Suppl 4:e8651. [PMID: 31721320 DOI: 10.1002/rcm.8651] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/25/2019] [Accepted: 11/04/2019] [Indexed: 06/10/2023]
Abstract
UNLABELLED Patients with type 2 diabetes may co-ingest herbal and prescription medicines to control their blood sugar levels. Competitive binding of drug and herb may mutually affect their metabolism. This can alter the level of drug and its kinetics in the body, potentially causing toxicities or loss of efficacy. Understanding how the metabolism of sulfonylureas like glyburide and gliclazide can be affected by the presence of berberine and vice versa can provide valuable information on the possible risk of toxicities caused by co-ingestion of drugs. METHODS Berberine and sulfonylureas (glyburide and gliclazide) were co-incubated with rat liver microsomes in the presence of a NADPH-regenerating system. The metabolites of berberine and sulfonylureas were analysed using liquid chromatography with high-resolution mass spectrometry in the positive ion mode. The role of individual isozymes in the metabolism of berberine, glyburide and gliclazide was investigated by using specific inhibitors. RESULTS In vitro metabolism of berberine led to the formation of demethyleneberberine (B1a) and its isomer B1b through demethylenation. Berberrubine (B2a) and its isomer B2b were formed through demethylation. The isozymes CYP3A and CYP2D were found to be involved in the metabolism of berberine. In vitro metabolism of glyburide and gliclazide led to the formation of hydroxylated metabolites. The isozymes CYP3A and CYP2C were found to be involved in the metabolism of glyburide. Gliclazide was metabolised by CYP2C. In vitro co-incubation of glyburide or gliclazide with berberine showed that each drug's metabolism was compromised as they share a common isozyme. A strong negative linear correlation of glyburide or gliclazide metabolite levels and the concentration of berberine confirmed the effect of berberine on the metabolism of sulfonylureas. CONCLUSIONS The metabolism of sulfonylureas and berberine was affected when these compounds were co-incubated with each other. This may be attributable to competitive binding of the herb and drug to the catalytic sites of the same isozymes.
Collapse
Affiliation(s)
- Amrinder Singh
- Department of Natural Sciences, Middlesex University, The Burroughs, London, NW4 4BT, UK
| | - Kaicun Zhao
- Department of Natural Sciences, Middlesex University, The Burroughs, London, NW4 4BT, UK
| | - Celia Bell
- Department of Natural Sciences, Middlesex University, The Burroughs, London, NW4 4BT, UK
| | - Ajit J Shah
- Department of Natural Sciences, Middlesex University, The Burroughs, London, NW4 4BT, UK
| |
Collapse
|
4
|
Blanco-Castañeda R, Galaviz-Hernández C, Souto PCS, Lima VV, Giachini FR, Escudero C, Damiano AE, Barragán-Zúñiga LJ, Martínez-Aguilar G, Sosa-Macías M. The role of xenobiotic-metabolizing enzymes in the placenta: a growing research field. Expert Rev Clin Pharmacol 2020; 13:247-263. [PMID: 32129110 DOI: 10.1080/17512433.2020.1733412] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The placenta is a temporary and unique organ that allows for the physical connection between a mother and fetus; this organ regulates the transport of gases and nutrients mediating the elimination of waste products contained in the fetal circulation. The placenta performs metabolic and excretion functions, on the basis of multiple enzymatic systems responsible for the oxidation, reduction, hydrolysis, and conjugation of xenobiotics. These mechanisms give the placenta a protective role that limits the fetal exposure to harmful compounds. During pregnancy, some diseases require uninterrupted treatment even if it is detrimental to the fetus. Drugs and other xenobiotics alter gene expression in the placenta with repercussions for the fetus and mother's well-being.Areas covered: This review provides a brief description of the human placental structure and function, the main drug and xenobiotic transporters and metabolizing enzymes, placenta-metabolized substrates, and alterations in gene expression that the exposure to xenobiotics may cause.Expert opinion: Research should be focused on the identification and validation of biological markers for the assessment of the harmful effects of some drugs in pregnancy, including the evaluation of polymorphisms and methylation patterns in chorionic villous samples and/or amniotic fluid.
Collapse
Affiliation(s)
| | | | - Paula C S Souto
- Laboratory of Vascular Biology, Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra Do Garcas, Brazil
| | - Victor Vitorino Lima
- Laboratory of Vascular Biology, Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra Do Garcas, Brazil
| | - Fernanda R Giachini
- Laboratory of Vascular Biology, Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra Do Garcas, Brazil
| | - Carlos Escudero
- Vascular Physiology Laboratory Group of Investigation in Tumor Angiogenesis (GIANT) Group of Research and Innovation in Vascular Health (GRIVAS Health) Basic Sciences Department Faculty of Sciences, Universidad Del Bio-Bio, Chillan, Chile
| | - Alicia E Damiano
- Laboratorio De Biología De La Reproducción, IFIBIO Houssay-UBA-CONICET, Buenos Aires, Argentina.,Departamento De Ciencias Biológicas, Facultad De Farmacia Y Bioquimica, Buenos Aires, UBA, Argentina
| | | | - Gerardo Martínez-Aguilar
- Unidad De Investigación Biomédica - Instituto Mexicano del Seguro Social (IMSS) Durango, Durango, México
| | - Martha Sosa-Macías
- Academia De Genómica, Instituto Politécnico Nacional-CIIDIR Durango, Durango, Mexico
| |
Collapse
|
5
|
Langer O. Pharmacological treatment of gestational diabetes mellitus: point/counterpoint. Am J Obstet Gynecol 2018; 218:490-499. [PMID: 29499921 DOI: 10.1016/j.ajog.2018.01.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 12/25/2022]
Abstract
Controversies persist over the most efficacious pharmacologic treatment for gestational diabetes mellitus. For purposes of accuracy in this article, the individual American College of Obstetricians and Gynecologists Practice Bulletin and American Diabetes Association Standards of Medical Care positions on each issue are quoted and then deliberated with evidence of counter claims presented in point/counterpoint. This is a review of all the relevant evidence for the most holistic picture possible. The main issues are (1) which diabetic drugs cross the placenta, (2) the quality of evidence and data source validity, (3) the rationale for the designation of glucose control as the primary outcome in gestational diabetes mellitus, and (4) which drugs (metformin, glyburide, or insulin) are most effective in improving secondary outcomes. The concept that 1 drug fits all, whether it be insulin, glyburide, or metformin, is a fallacy. Different drugs provide certain benefits but not all the benefits and not to all patients. In addition, the steps in the gestational diabetes mellitus management decision path and the current cost of the use of insulin, glyburide, or metformin are addressed. In the future, we must consider studying the potential of diabetic drugs that currently are used in nonpregnancy and incorporating the concept of precision medicine in the decision tree to maximize pregnancy outcomes.
Collapse
|
6
|
Sahu R, Ahmed T, Sangana R, Punde R, Subudhi BB. Effect of Tinospora cordifolia aqua-alcoholic extract on pharmacokinetic of Glibenclamide in rat: An herb-drug interaction study. J Pharm Biomed Anal 2018; 151:310-316. [DOI: 10.1016/j.jpba.2018.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/03/2018] [Accepted: 01/07/2018] [Indexed: 12/14/2022]
|
7
|
Ilekis JV, Tsilou E, Fisher S, Abrahams VM, Soares MJ, Cross JC, Zamudio S, Illsley NP, Myatt L, Colvis C, Costantine MM, Haas DM, Sadovsky Y, Weiner C, Rytting E, Bidwell G. Placental origins of adverse pregnancy outcomes: potential molecular targets: an Executive Workshop Summary of the Eunice Kennedy Shriver National Institute of Child Health and Human Development. Am J Obstet Gynecol 2016; 215:S1-S46. [PMID: 26972897 DOI: 10.1016/j.ajog.2016.03.001] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 02/11/2016] [Accepted: 03/01/2016] [Indexed: 12/26/2022]
Abstract
Although much progress is being made in understanding the molecular pathways in the placenta that are involved in the pathophysiology of pregnancy-related disorders, a significant gap exists in the utilization of this information for the development of new drug therapies to improve pregnancy outcome. On March 5-6, 2015, the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health sponsored a 2-day workshop titled Placental Origins of Adverse Pregnancy Outcomes: Potential Molecular Targets to begin to address this gap. Particular emphasis was given to the identification of important molecular pathways that could serve as drug targets and the advantages and disadvantages of targeting these particular pathways. This article is a summary of the proceedings of that workshop. A broad number of topics were covered that ranged from basic placental biology to clinical trials. This included research in the basic biology of placentation, such as trophoblast migration and spiral artery remodeling, and trophoblast sensing and response to infectious and noninfectious agents. Research findings in these areas will be critical for the formulation of the development of future treatments and the development of therapies for the prevention of a number of pregnancy disorders of placental origin that include preeclampsia, fetal growth restriction, and uterine inflammation. Research was also presented that summarized ongoing clinical efforts in the United States and in Europe that has tested novel interventions for preeclampsia and fetal growth restriction, including agents such as oral arginine supplementation, sildenafil, pravastatin, gene therapy with virally delivered vascular endothelial growth factor, and oxygen supplementation therapy. Strategies were also proposed to improve fetal growth by the enhancement of nutrient transport to the fetus by modulation of their placental transporters and the targeting of placental mitochondrial dysfunction and oxidative stress to improve placental health. The roles of microRNAs and placental-derived exosomes, as well as messenger RNAs, were also discussed in the context of their use for diagnostics and as drug targets. The workshop discussed the aspect of safety and pharmacokinetic profiles of potential existing and new therapeutics that will need to be determined, especially in the context of the unique pharmacokinetic properties of pregnancy and the hurdles and pitfalls of the translation of research findings into practice. The workshop also discussed novel methods of drug delivery and targeting during pregnancy with the use of macromolecular carriers, such as nanoparticles and biopolymers, to minimize placental drug transfer and hence fetal drug exposure. In closing, a major theme that developed from the workshop was that the scientific community must change their thinking of the pregnant woman and her fetus as a vulnerable patient population for which drug development should be avoided, but rather be thought of as a deprived population in need of more effective therapeutic interventions.
Collapse
Affiliation(s)
- John V Ilekis
- Pregnancy and Perinatology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Department of Health and Human Services, Bethesda, MD.
| | - Ekaterini Tsilou
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Department of Health and Human Services, Bethesda, MD.
| | - Susan Fisher
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA
| | - Vikki M Abrahams
- Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine; New Haven, CT
| | - Michael J Soares
- Institute of Reproductive Health and Regenerative Medicine and Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - James C Cross
- Comparative Biology and Experimental Medicine, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Stacy Zamudio
- Department of Obstetrics and Gynecology, Hackensack University Medical Center, Hackensack, NJ
| | - Nicholas P Illsley
- Department of Obstetrics and Gynecology, Hackensack University Medical Center, Hackensack, NJ
| | - Leslie Myatt
- Center for Pregnancy and Newborn Research, University of Texas Health Science Center, San Antonio, TX
| | - Christine Colvis
- Therapeutics Discovery Program, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD
| | - Maged M Costantine
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX
| | - David M Haas
- Department of Obstetrics and Gynecology Indiana University, Indianapolis, IN
| | | | - Carl Weiner
- University of Kansas Medical Center, Kansas City, KS
| | - Erik Rytting
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX
| | - Gene Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
8
|
Shuster DL, Risler LJ, Prasad B, Calamia JC, Voellinger JL, Kelly EJ, Unadkat JD, Hebert MF, Shen DD, Thummel KE, Mao Q. Identification of CYP3A7 for glyburide metabolism in human fetal livers. Biochem Pharmacol 2014; 92:690-700. [PMID: 25450675 DOI: 10.1016/j.bcp.2014.09.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 09/02/2014] [Accepted: 09/29/2014] [Indexed: 02/04/2023]
Abstract
Glyburide is commonly prescribed for the treatment of gestational diabetes mellitus; however, fetal exposure to glyburide is not well understood and may have short- and long-term consequences for the health of the child. Glyburide can cross the placenta; fetal concentrations at term are nearly comparable to maternal levels. Whether or not glyburide is metabolized in the fetus and by what mechanisms has yet to be determined. In this study, we determined the kinetic parameters for glyburide depletion by CYP3A isoenzymes; characterized glyburide metabolism by human fetal liver tissues collected during the first or early second trimester of pregnancy; and identified the major enzyme responsible for glyburide metabolism in human fetal livers. CYP3A4 had the highest metabolic capacity towards glyburide, followed by CYP3A7 and CYP3A5 (Clint,u=37.1, 13.0, and 8.7ml/min/nmol P450, respectively). M5 was the predominant metabolite generated by CYP3A7 and human fetal liver microsomes (HFLMs) with approximately 96% relative abundance. M5 was also the dominant metabolite generated by CYP3A4, CYP3A5, and adult liver microsomes; however, M1-M4 were also present, with up to 15% relative abundance. CYP3A7 protein levels in HFLMs were highly correlated with glyburide Clint, 16α-OH DHEA formation, and 4'-OH midazolam formation. Likewise, glyburide Clint was highly correlated with 16α-OH DHEA formation. Fetal demographics as well as CYP3A5 and CYP3A7 genotype did not alter CYP3A7 protein levels or glyburide Clint. These results indicate that human fetal livers metabolize glyburide predominantly to M5 and that CYP3A7 is the major enzyme responsible for glyburide metabolism in human fetal livers.
Collapse
Affiliation(s)
- Diana L Shuster
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195, USA.
| | - Linda J Risler
- Department of Pharmacy, University of Washington, Box 357630, Seattle, Washington 98195, USA.
| | - Bhagwat Prasad
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195, USA.
| | - Justina C Calamia
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195, USA.
| | - Jenna L Voellinger
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195, USA.
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195, USA.
| | - Jashvant D Unadkat
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195, USA.
| | - Mary F Hebert
- Department of Pharmacy, University of Washington, Box 357630, Seattle, Washington 98195, USA; Department of Obstetrics and Gynecology, University of Washington, Box 356460, Seattle, Washington 98195, USA.
| | - Danny D Shen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195, USA; Department of Pharmacy, University of Washington, Box 357630, Seattle, Washington 98195, USA.
| | - Kenneth E Thummel
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195, USA.
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195, USA.
| |
Collapse
|
9
|
Rytting E, Wang X, Vernikovskaya DI, Zhan Y, Bauer C, Abdel-Rahman SM, Ahmed MS, Nanovskaya TN. Metabolism and disposition of bupropion in pregnant baboons (Papio cynocephalus). Drug Metab Dispos 2014; 42:1773-9. [PMID: 25097227 PMCID: PMC4164976 DOI: 10.1124/dmd.114.058255] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 08/05/2014] [Indexed: 01/09/2023] Open
Abstract
Recent in vitro data obtained in our laboratory revealed similarities between baboons and humans in the biotransformation of bupropion (BUP) by both hepatic and placental microsomes. These data supported the use of baboons to study BUP biotransformation during pregnancy. The aim of this investigation was to determine the pharmacokinetics of BUP in baboons during pregnancy and postpartum, as well as fetal exposure to the drug after intravenous administration. Pregnant baboons (n = 5) received a single intravenous bolus dose of bupropion hydrochloride (1 mg/kg) at gestational ages 94-108 days (midpregnancy), 142-156 days (late pregnancy), and 6 weeks postpartum. Blood and urine samples were collected for 12 and 24 hours, respectively. The concentrations of BUP, hydroxybupropion (OH-BUP), threohydrobupropion, and erythrohydrobupropion in plasma were determined by liquid chromatography-tandem mass spectrometry. Relative to the postpartum period, the average midpregnancy clearance of BUP trended higher (3.6 ± 0.15 versus 2.7 ± 0.28 l/h per kg) and the average C(max) (294 ± 91 versus 361 ± 64 ng/ml) and the area under the curve (AUC) of BUP values (288 ± 22 versus 382 ± 42 h·ng/ml) trended lower. AUC(OH-BUP) also tended to be lower midpregnancy compared with postpartum (194 ± 76 versus 353 ± 165 h·ng/ml). Whereas the observed trend toward increased clearance of BUP during baboon pregnancy could be associated with a pregnancy-induced increase in its biotransformation, the trend toward increased renal elimination of OH-BUP may overshadow any corresponding change in the hydroxylation activity of CYP2B.
Collapse
Affiliation(s)
- Erik Rytting
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, Galveston, Texas (E.R., X.W., D.I.V., Y.Z., M.S.A., T.N.N.); Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas (C.B.); and Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospital, Kansas City, Missouri (S.M.A.-R.)
| | - Xiaoming Wang
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, Galveston, Texas (E.R., X.W., D.I.V., Y.Z., M.S.A., T.N.N.); Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas (C.B.); and Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospital, Kansas City, Missouri (S.M.A.-R.)
| | - Daria I Vernikovskaya
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, Galveston, Texas (E.R., X.W., D.I.V., Y.Z., M.S.A., T.N.N.); Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas (C.B.); and Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospital, Kansas City, Missouri (S.M.A.-R.)
| | - Ying Zhan
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, Galveston, Texas (E.R., X.W., D.I.V., Y.Z., M.S.A., T.N.N.); Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas (C.B.); and Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospital, Kansas City, Missouri (S.M.A.-R.)
| | - Cassondra Bauer
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, Galveston, Texas (E.R., X.W., D.I.V., Y.Z., M.S.A., T.N.N.); Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas (C.B.); and Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospital, Kansas City, Missouri (S.M.A.-R.)
| | - Susan M Abdel-Rahman
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, Galveston, Texas (E.R., X.W., D.I.V., Y.Z., M.S.A., T.N.N.); Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas (C.B.); and Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospital, Kansas City, Missouri (S.M.A.-R.)
| | - Mahmoud S Ahmed
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, Galveston, Texas (E.R., X.W., D.I.V., Y.Z., M.S.A., T.N.N.); Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas (C.B.); and Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospital, Kansas City, Missouri (S.M.A.-R.)
| | - Tatiana N Nanovskaya
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, Galveston, Texas (E.R., X.W., D.I.V., Y.Z., M.S.A., T.N.N.); Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas (C.B.); and Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospital, Kansas City, Missouri (S.M.A.-R.)
| |
Collapse
|
10
|
Ravindran S, Jadhav A, Surve P, Lonsane G, Honrao P, Nanda B. Technologies and strategies to characterize and quantitate metabolites in drug discovery and development. Biomed Chromatogr 2014; 28:1547-53. [DOI: 10.1002/bmc.3309] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/10/2014] [Accepted: 07/18/2014] [Indexed: 11/07/2022]
Affiliation(s)
- Selvan Ravindran
- Departments of Bio-Analytical and Biotransformation, Drug Metabolism and Pharmacokinetics; Sai Life Sciences Limited; Building 1, Plot No 2, Chrysalis Enclave, International Bio-Tech Park, Phase-II, Hinjewadi Pune- 411 057 Maharashtra India
| | - Amol Jadhav
- Departments of Bio-Analytical and Biotransformation, Drug Metabolism and Pharmacokinetics; Sai Life Sciences Limited; Building 1, Plot No 2, Chrysalis Enclave, International Bio-Tech Park, Phase-II, Hinjewadi Pune- 411 057 Maharashtra India
| | - Prashant Surve
- Departments of Bio-Analytical and Biotransformation, Drug Metabolism and Pharmacokinetics; Sai Life Sciences Limited; Building 1, Plot No 2, Chrysalis Enclave, International Bio-Tech Park, Phase-II, Hinjewadi Pune- 411 057 Maharashtra India
| | - Ganesh Lonsane
- Departments of Bio-Analytical and Biotransformation, Drug Metabolism and Pharmacokinetics; Sai Life Sciences Limited; Building 1, Plot No 2, Chrysalis Enclave, International Bio-Tech Park, Phase-II, Hinjewadi Pune- 411 057 Maharashtra India
| | - Pradnya Honrao
- Departments of Bio-Analytical and Biotransformation, Drug Metabolism and Pharmacokinetics; Sai Life Sciences Limited; Building 1, Plot No 2, Chrysalis Enclave, International Bio-Tech Park, Phase-II, Hinjewadi Pune- 411 057 Maharashtra India
| | - Bidyut Nanda
- Departments of Bio-Analytical and Biotransformation, Drug Metabolism and Pharmacokinetics; Sai Life Sciences Limited; Building 1, Plot No 2, Chrysalis Enclave, International Bio-Tech Park, Phase-II, Hinjewadi Pune- 411 057 Maharashtra India
| |
Collapse
|
11
|
Wang X, Paul JA, Nanovskaya TN, Hankins GDV, Ahmed MS. Quantitative determination of telavancin in pregnant baboon plasma by solid-phase extraction and LC-ESI-MS. J Pharm Biomed Anal 2014; 98:107-12. [PMID: 24905291 PMCID: PMC4127372 DOI: 10.1016/j.jpba.2014.04.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 04/22/2014] [Accepted: 04/28/2014] [Indexed: 11/15/2022]
Abstract
The increasing incidence and severity of methicillin- and vancomycin-resistant infections during pregnancy prompted further development of telavancin. The understanding of the pharmacokinetics of telavancin during pregnancy is critical to optimize dosing. Due to ethical and safety concerns the study is conducted on the pregnant baboons. A method using solid-phase extraction coupled with liquid chromatography-single quadrupole mass spectrometry for the quantitative determination of telavancin in baboon plasma samples was developed and validated. Teicoplanin was used as an internal standard. Telavancin was extracted from baboon plasma samples by using Waters Oasis(®) MAX 96-Well SPE plate and achieved extraction recovery was >66% with variation <12%. Telavancin was separated on Waters Symmetry C18 column with gradient elution. Two SIM channels were monitored at m/z 823 and m/z 586 to achieve quantification with simultaneous confirmation of telavancin identification in baboon plasma samples. The linearity was assessed in the range of 0.188μg/mL to75.0μg/mL, with a correlation coefficient of 0.998. The relative standard deviation of this method was <11% for within- and between-run assays, and the accuracy ranged between 96% and 114%.
Collapse
Affiliation(s)
- Xiaoming Wang
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA
| | - Jonathan A Paul
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA
| | - Tatiana N Nanovskaya
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA
| | - Gary D V Hankins
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA
| | - Mahmoud S Ahmed
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA.
| |
Collapse
|
12
|
Zhang X, Wang X, Vernikovskaya DI, Fokina VM, Nanovskaya TN, Hankins GDV, Ahmed MS. Quantitative determination of metformin, glyburide and its metabolites in plasma and urine of pregnant patients by LC-MS/MS. Biomed Chromatogr 2014; 29:560-9. [PMID: 25164921 DOI: 10.1002/bmc.3314] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 05/09/2014] [Accepted: 07/22/2014] [Indexed: 11/09/2022]
Abstract
This report describes the development and validation of an LC-MS/MS method for the quantitative determination of glyburide (GLB), its five metabolites (M1, M2a, M2b, M3 and M4) and metformin (MET) in plasma and urine of pregnant patients under treatment with a combination of the two medications. The extraction recovery of the analytes from plasma samples was 87-99%, and that from urine samples was 85-95%. The differences in retention times among the analytes and the wide range of the concentrations of the medications and their metabolites in plasma and urine patient samples required the development of three LC methods. The lower limit of quantitation (LLOQ) of the analytes in plasma samples was as follows: GLB, 1.02 ng/mL; its five metabolites, 0.100-0.113 ng/mL; and MET, 4.95 ng/mL. The LLOQ in urine samples was 0.0594 ng/mL for GLB, 0.984-1.02 ng/mL for its five metabolites and 30.0 µg/mL for MET. The relative deviation of this method was <14% for intra-day and inter-day assays in plasma and urine samples, and the accuracy was 86-114% in plasma, and 94-105% in urine. The method described in this report was successfully utilized for determining the concentrations of the two medications in patient plasma and urine.
Collapse
Affiliation(s)
- Xing Zhang
- Maternal-Fetal Pharmacology and Bio-development Laboratories, Department of Obstetrics and Gynecology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-0587, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Varma MVS, Scialis RJ, Lin J, Bi YA, Rotter CJ, Goosen TC, Yang X. Mechanism-based pharmacokinetic modeling to evaluate transporter-enzyme interplay in drug interactions and pharmacogenetics of glyburide. AAPS JOURNAL 2014; 16:736-48. [PMID: 24839071 DOI: 10.1208/s12248-014-9614-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 04/26/2014] [Indexed: 11/30/2022]
Abstract
The purpose of this study is to characterize the involvement of hepato-biliary transport and cytochrome-P450 (CYP)-mediated metabolism in the disposition of glyburide and predict its pharmacokinetic variability due to drug interactions and genetic variations. Comprehensive in vitro studies suggested that glyburide is a highly permeable drug with substrate affinity to multiple efflux pumps and to organic anion transporting polypeptide (OATP)1B1 and OATP2B1. Active hepatic uptake was found to be significantly higher than the passive uptake clearance (15.8 versus 5.3 μL/min/10(6)-hepatocytes), using the sandwich-cultured hepatocyte model. In vitro, glyburide is metabolized (intrinsic clearance, 52.9 μL/min/mg-microsomal protein) by CYP3A4, CYP2C9, and CYP2C8 with fraction metabolism of 0.53, 0.36, and 0.11, respectively. Using these in vitro data, physiologically based pharmacokinetic models, assuming rapid-equilibrium between blood and liver compartments or permeability-limited hepatic disposition, were built to describe pharmacokinetics and evaluate drug interactions. Permeability-limited model successfully predicted glyburide interactions with rifampicin and other perpetrator drugs. Conversely, model assuming rapid-equilibrium mispredicted glyburide interactions, overall, suggesting hepatic uptake as the primary rate-determining process in the systemic clearance of glyburide. Further modeling and simulations indicated that the impairment of CYP2C9 function has a minimal effect on the systemic exposure, implying discrepancy in the contribution of CYP2C9 to glyburide clearance.
Collapse
Affiliation(s)
- Manthena V S Varma
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Groton, Connecticut, USA,
| | | | | | | | | | | | | |
Collapse
|
14
|
Impact of genetic polymorphisms of cytochrome P450 2 C (CYP2C) enzymes on the drug metabolism and design of antidiabetics. Chem Biol Interact 2011; 194:159-67. [DOI: 10.1016/j.cbi.2011.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 08/26/2011] [Accepted: 08/29/2011] [Indexed: 01/01/2023]
|
15
|
Wang X, Abdelrahman DR, Fokina VM, Hankins GDV, Ahmed MS, Nanovskaya TN. Metabolism of bupropion by baboon hepatic and placental microsomes. Biochem Pharmacol 2011; 82:295-303. [PMID: 21570381 DOI: 10.1016/j.bcp.2011.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 04/19/2011] [Accepted: 04/27/2011] [Indexed: 11/20/2022]
Abstract
The aim of this investigation was to determine the biotransformation of bupropion by baboon hepatic and placental microsomes, identify the enzyme(s) catalyzing the reaction(s) and determine its kinetics. Bupropion was metabolized by baboon hepatic and placental microsomes to hydroxybupropion (OH-BUP), threo- (TB) and erythrohydrobupropion (EB). OH-bupropion was the major metabolite formed by hepatic microsomes (Km 36±6 μM, Vmax 258±32 pmol mg protein(-1) min(-1)), however the formation of OH-BUP by placental microsomes was below the limit of quantification. The apparent Km values of bupropion for the formation of TB and EB by hepatic and placental microsomes were similar. The selective inhibitors of CYP2B6 (ticlopidine and phencyclidine) and monoclonal antibodies raised against human CYP2B6 isozyme caused 80% inhibition of OH-BUP formation by baboon hepatic microsomes. The chemical inhibitors of aldo-keto reductases (flufenamic acid), carbonyl reductases (menadione), and 11β-hydroxysteroid dehydrogenases (18β-glycyrrhetinic acid) significantly decreased the formation of TB and EB by hepatic and placental microsomes. Data indicate that CYP2B of baboon hepatic microsomes is responsible for biotransformation of bupropion to OH-BUP, while hepatic and placental short chain dehydrogenases/reductases and to a lesser extent aldo-keto reductases are responsible for the reduction of bupropion to TB and EB.
Collapse
Affiliation(s)
- Xiaoming Wang
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA
| | | | | | | | | | | |
Collapse
|
16
|
Earhart AD, Patrikeeva S, Wang X, Abdelrahman DR, Hankins GDV, Ahmed MS, Nanovskaya T. Transplacental transfer and metabolism of bupropion. J Matern Fetal Neonatal Med 2010; 23:409-16. [PMID: 19658039 DOI: 10.1080/14767050903168424] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE In order to evaluate the potential use of bupropion as smoking cessation therapy during pregnancy, the aim of this investigation was to determine transplacental transfer and metabolism of bupropion and its distribution among placental tissue and maternal and fetal circuits of the dually perfused placental lobule. METHODS Placentas obtained from healthy term pregnancies were perfused with bupropion at two concentrations 150 ng/ml and 450 ng/ml, along with the marker compound antipyrine 20 microg/ml. Radioactive isotopes of the two drugs were co-transfused to enhance their detection limits. Concentrations of bupropion and its metabolite were determined by liquid chromatography and liquid scintillation spectrometry. RESULTS The fetal/maternal concentration ratio of bupropion was 1.07 +/- 0.22. Following 4 h of its perfusion, 48 +/- 6% of bupropion was retained by placental tissue, 32 +/- 5% remained in the maternal circuit, and 20 +/- 6% was transferred to the fetal circuit. A metabolite of bupropion, threohydrobupropion, was identified. CONCLUSIONS Bupropion was transferred from the maternal to fetal circuit and was biotransformed by placental tissue enzymes to its metabolite threohydrobupropion. Bupropion and its metabolite did not affect placental tissue viability or functional parameters. These data suggest that bupropion has the potential of being used for smoking cessation during pregnancy and should be further investigated for its safety and efficacy.
Collapse
Affiliation(s)
- Angela D Earhart
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas 77555-0587, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Zhou L, Naraharisetti SB, Liu L, Wang H, Lin YS, Isoherranen N, Unadkat JD, Hebert MF, Mao Q. Contributions of human cytochrome P450 enzymes to glyburide metabolism. Biopharm Drug Dispos 2010; 31:228-42. [PMID: 20437462 DOI: 10.1002/bdd.706] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Glyburide (GLB) is a widely used oral sulfonylurea for the treatment of gestational diabetes. The therapeutic use of GLB is often complicated by a substantial inter-individual variability in the pharmacokinetics and pharmacodynamics of the drug in human populations, which might be caused by inter-individual variations in factors such as GLB metabolism. Therefore, there has been a continued interest in identifying human cytochrome P450 (CYP) isoforms that play a major role in the metabolism of GLB. However, contrasting data are available in the present literature in this regard. The present study systematically investigated the contributions of various human CYP isoforms (CYP3A4, CYP3A5, CYP2C8, CYP2C9 and CYP2C19) to in vitro metabolism of GLB. GLB depletion and metabolite formation in human liver microsomes were most significantly inhibited by the CYP3A inhibitor ketoconazole compared with the inhibitors of other CYP isoforms. Furthermore, multiple correlation analysis between GLB depletion and individual CYP activities was performed, demonstrating a significant correlation between GLB depletion and the CYP3A probe activity in 16 individual human liver microsomal preparations, but not between GLB depletion and the CYP2C19, CYP2C8 or CYP2C9 probe activity. By using recombinant supersomes overexpressing individual human CYP isoforms, it was found that GLB could be depleted by all the enzymes tested; however, the intrinsic clearance (V(max)/K(m)) of CYP3A4 for GLB depletion was 4-17 times greater than that of other CYP isoforms. These results confirm that human CYP3A4 is the major enzyme involved in the in vitro metabolism of GLB.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wang X, Abdelrahman DR, Zharikova OL, Patrikeeva SL, Hankins GDV, Ahmed MS, Nanovskaya TN. Bupropion metabolism by human placenta. Biochem Pharmacol 2010; 79:1684-90. [PMID: 20109440 PMCID: PMC2847018 DOI: 10.1016/j.bcp.2010.01.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 01/20/2010] [Accepted: 01/20/2010] [Indexed: 11/23/2022]
Abstract
Smoking during pregnancy is the largest modifiable risk factor for pregnancy-related morbidity and mortality. The success of bupropion for smoking cessation warrants its investigation for the treatment of pregnant patients. Nevertheless, the use of bupropion for the treatment of pregnant smokers requires additional data on its bio-disposition during pregnancy. Therefore, the aim of this investigation was to determine the metabolism of bupropion in placentas obtained from nonsmoking and smoking women, identify metabolites formed and the enzymes catalyzing their formation, as well as the kinetics of the reaction. Data obtained revealed that human placentas metabolized bupropion to hydroxybupropion, erythro- and threohydrobupropion. The rates for formation of erythro- and threohydrobupropion exceeded that for hydroxybupropion by several folds, were dependent on the concentration of bupropion and exhibited saturation kinetics with an apparent K(m) value of 40microM. Human placental 11beta-hydroxysteroid dehydrogenases were identified as the major carbonyl-reducing enzymes responsible for the reduction of bupropion to threo- and erythrohydrobupropion in microsomal fractions. On the other hand, CYP2B6 was responsible for the formation of OH-bupropion. These data suggest that both placental microsomal carbonyl-reducing and oxidizing enzymes are involved in the metabolism of bupropion.
Collapse
Affiliation(s)
- Xiaoming Wang
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 77555-0587, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Earhart AD, Patrikeeva S, Wang X, Reda Abdelrahman D, Hankins GDV, Ahmed MS, Nanovskaya T. Transplacental transfer and metabolism of bupropion. J Matern Fetal Neonatal Med 2010. [DOI: 10.3109/14767050903168424] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
20
|
Zhou SF, Liu JP, Chowbay B. Polymorphism of human cytochrome P450 enzymes and its clinical impact. Drug Metab Rev 2009; 41:89-295. [PMID: 19514967 DOI: 10.1080/03602530902843483] [Citation(s) in RCA: 536] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pharmacogenetics is the study of how interindividual variations in the DNA sequence of specific genes affect drug response. This article highlights current pharmacogenetic knowledge on important human drug-metabolizing cytochrome P450s (CYPs) to understand the large interindividual variability in drug clearance and responses in clinical practice. The human CYP superfamily contains 57 functional genes and 58 pseudogenes, with members of the 1, 2, and 3 families playing an important role in the metabolism of therapeutic drugs, other xenobiotics, and some endogenous compounds. Polymorphisms in the CYP family may have had the most impact on the fate of therapeutic drugs. CYP2D6, 2C19, and 2C9 polymorphisms account for the most frequent variations in phase I metabolism of drugs, since almost 80% of drugs in use today are metabolized by these enzymes. Approximately 5-14% of Caucasians, 0-5% Africans, and 0-1% of Asians lack CYP2D6 activity, and these individuals are known as poor metabolizers. CYP2C9 is another clinically significant enzyme that demonstrates multiple genetic variants with a potentially functional impact on the efficacy and adverse effects of drugs that are mainly eliminated by this enzyme. Studies into the CYP2C9 polymorphism have highlighted the importance of the CYP2C9*2 and *3 alleles. Extensive polymorphism also occurs in other CYP genes, such as CYP1A1, 2A6, 2A13, 2C8, 3A4, and 3A5. Since several of these CYPs (e.g., CYP1A1 and 1A2) play a role in the bioactivation of many procarcinogens, polymorphisms of these enzymes may contribute to the variable susceptibility to carcinogenesis. The distribution of the common variant alleles of CYP genes varies among different ethnic populations. Pharmacogenetics has the potential to achieve optimal quality use of medicines, and to improve the efficacy and safety of both prospective and currently available drugs. Further studies are warranted to explore the gene-dose, gene-concentration, and gene-response relationships for these important drug-metabolizing CYPs.
Collapse
Affiliation(s)
- Shu-Feng Zhou
- School of Health Sciences, RMIT University, Bundoora, Victoria, Australia.
| | | | | |
Collapse
|
21
|
Zhou SF, Zhou ZW, Huang M. Polymorphisms of human cytochrome P450 2C9 and the functional relevance. Toxicology 2009; 278:165-88. [PMID: 19715737 DOI: 10.1016/j.tox.2009.08.013] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 08/18/2009] [Accepted: 08/21/2009] [Indexed: 12/19/2022]
Abstract
Human cytochrome P450 2C9 (CYP2C9) accounts for ∼20% of hepatic total CYP content and metabolizes ~15% clinical drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and many nonsteroidal anti-inflammatory agents (NSAIDs). CYP2C9 is highly polymorphic, with at least 33 variants of CYP2C9 (*1B through *34) being identified so far. CYP2C9*2 is frequent among Caucasians with ~1% of the population being homozygous carriers and 22% are heterozygous. The corresponding figures for the CYP2C9*3 allele are 0.4% and 15%, respectively. There are a number of clinical studies addressing the impact of CYP2C9 polymorphisms on the clearance and/or therapeutic response of therapeutic drugs. These studies have highlighted the importance of the CYP2C9*2 and *3 alleles as a determining factor for drug clearance and drug response. The CYP2C9 polymorphisms are relevant for the efficacy and adverse effects of numerous NSAIDs, sulfonylurea antidiabetic drugs and, most critically, oral anticoagulants belonging to the class of vitamin K epoxide reductase inhibitors. Warfarin has served as a practical example of how pharmacogenetics can be utilized to achieve maximum efficacy and minimum toxicity. For many of these drugs, a clear gene-dose and gene-effect relationship has been observed in patients. In this regard, CYP2C9 alleles can be considered as a useful biomarker in monitoring drug response and adverse effects. Genetic testing of CYP2C9 is expected to play a role in predicting drug clearance and conducting individualized pharmacotherapy. However, prospective clinical studies with large samples are warranted to establish gene-dose and gene-effect relationships for CYP2C9 and its substrate drugs.
Collapse
Affiliation(s)
- Shu-Feng Zhou
- School of Health Sciences, RMIT University, Victoria 3083, Australia.
| | | | | |
Collapse
|
22
|
Zharikova OL, Fokina VM, Nanovskaya TN, Hill RA, Mattison DR, Hankins GDV, Ahmed MS. Identification of the major human hepatic and placental enzymes responsible for the biotransformation of glyburide. Biochem Pharmacol 2009; 78:1483-90. [PMID: 19679108 DOI: 10.1016/j.bcp.2009.08.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 07/31/2009] [Accepted: 08/04/2009] [Indexed: 02/01/2023]
Abstract
One of the factors affecting the pharmacokinetics (PK) of a drug during pregnancy is the activity of hepatic and placental metabolizing enzymes. Recently, we reported on the biotransformation of glyburide by human hepatic and placental microsomes to six metabolites that are structurally identical between the two tissues. Two of the metabolites, 4-trans-(M1) and 3-cis-hydroxycyclohexyl glyburide (M2b), were previously identified in plasma and urine of patients treated with glyburide and are pharmacologically active. The aim of this investigation was to identify the major human hepatic and placental CYP450 isozymes responsible for the formation of each metabolite of glyburide. This was achieved by the use of chemical inhibitors selective for individual CYP isozymes and antibodies raised against them. The identification was confirmed by the kinetic constants for the biotransformation of glyburide by cDNA-expressed enzymes. The data revealed that the major hepatic isozymes responsible for the formation of each metabolite are as follows: CYP3A4 (ethylene-hydroxylated glyburide (M5), 3-trans-(M3) and 2-trans-(M4) cyclohexyl glyburide); CYP2C9 (M1, M2a (4-cis-) and M2b); CYP2C8 (M1 and M2b); and CYP2C19 (M2a). Human placental microsomal CYP19/aromatase was the major isozyme responsible for the biotransformation of glyburide to predominantly M5. The formation of significant amounts of M5 by CYP19 in the placenta could render this metabolite more accessible to the fetal circulation. The multiplicity of enzymes biotransforming glyburide and the metabolites formed underscores the potential for its drug interactions in vivo.
Collapse
Affiliation(s)
- Olga L Zharikova
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX, 77555-0587, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Yan R, Nanovskaya TN, Zharikova OL, Mattison DR, Hankins GDV, Ahmed MS. Metabolism of 17alpha-hydroxyprogesterone caproate by hepatic and placental microsomes of human and baboons. Biochem Pharmacol 2008; 75:1848-57. [PMID: 18329004 DOI: 10.1016/j.bcp.2008.01.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Revised: 01/24/2008] [Accepted: 01/25/2008] [Indexed: 10/22/2022]
Abstract
Recent data from our laboratory revealed the formation of an unknown metabolite of 17 hydroxyprogesterone caproate (17-HPC), used for treatment of preterm deliveries, during its perfusion across the dually perfused human placental lobule. Previously, we demonstrated that the drug is not hydrolyzed, neither in vivo nor in vitro, to progesterone and caproate. Therefore, the hypothesis for this investigation is that 17-HPC is actively metabolized by human and baboon (Papio cynocephalus) hepatic and placental microsomes. Baboon hepatic and placental microsomes were investigated to validate the nonhuman primate as an animal model for drug use during pregnancy. Data presented here indicate that human and baboon hepatic microsomes formed several mono-, di-, and tri-hydroxylated derivatives of 17-HPC. However, microsomes of human and baboon placentas metabolized 17-HPC to its mono-hydroxylated derivatives only in quantities that were a fraction of those formed by their respective livers, except for two metabolites (M16' and M17') that are unique for placenta and contributed to 25% and 75% of the total metabolites formed by human and baboon, respectively. The amounts of metabolites formed, relative to each other, by human and baboon microsomes were different suggesting that the affinity of 17-HPC to CYP enzymes and their activity could be species-dependent.
Collapse
Affiliation(s)
- Ru Yan
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0587, USA
| | | | | | | | | | | |
Collapse
|