1
|
Shigemura S, Kikuchi T, Ito Y, Morita O, Saito K. Development of a human FaFg prediction system for polyphenols using human induced pluripotent stem cell-derived small intestinal epithelial cells. Toxicol In Vitro 2025; 104:105998. [PMID: 39674233 DOI: 10.1016/j.tiv.2024.105998] [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: 07/26/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
Precise prediction of the fraction of compounds reaching the portal vein (FaFg) in humans, which could indicate the rate-limiting step of polyphenol metabolism, is particularly important for accurately evaluating the efficacy and safety of polyphenols. In this study, we aimed to develop a novel in vitro method to predict human FaFg of polyphenols using commercially available human induced pluripotent stem cell-derived small intestinal epithelial cells (hiPSC-SIECs). First, the chemicals were used at fixed test concentrations, considering their physicochemical properties and cytotoxicity. The apparent permeability coefficient (Papp) values of the six tested polyphenols in hiPSC-SIECs were considerably higher than those of the seven tested pharmaceuticals, resulting in a poor correlation between Papp in hiPSC-SIECs and human FaFg. A detailed assessment of the relationship between in vitro test concentration and metabolic activity suggested that the higher Papp value of polyphenols would be due to inadequate reflection of phase II metabolism in the human intestine. By optimizing test concentrations to reflect enzymatic metabolism in the human intestine, a good correlation was observed between the Papp values in hiPSC-SIECs and human FaFg for tested polyphenols and pharmaceuticals (R2 = 0.81). The developed method could be useful for precisely predicting human FaFg of polyphenols.
Collapse
Affiliation(s)
- Shunta Shigemura
- Kao Corporation, R&D, Safety Science Research, 2606 Akabane, Ichikai-Machi, Haga-Gun, Tochigi, Japan
| | - Takuya Kikuchi
- Kao Corporation, R&D, Safety Science Research, 2606 Akabane, Ichikai-Machi, Haga-Gun, Tochigi, Japan
| | - Yuichi Ito
- Kao Corporation, R&D, Safety Science Research, 2606 Akabane, Ichikai-Machi, Haga-Gun, Tochigi, Japan
| | - Osamu Morita
- Kao Corporation, R&D, Safety Science Research, 2606 Akabane, Ichikai-Machi, Haga-Gun, Tochigi, Japan
| | - Kazutoshi Saito
- Kao Corporation, R&D, Safety Science Research, 2606 Akabane, Ichikai-Machi, Haga-Gun, Tochigi, Japan.
| |
Collapse
|
2
|
Lv X, Wang Z, Wang Z, Yin H, Xia Y, Jiang L, Liu Y. Avapritinib Carries the Risk of Drug Interaction via Inhibition of UDP-Glucuronyltransferase (UGT) 1A1. Curr Drug Metab 2024; 25:197-204. [PMID: 38803186 DOI: 10.2174/0113892002288312240521092054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/04/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Avapritinib is the only drug for adult patients with PDGFRA exon 18 mutated unresectable or metastatic gastrointestinal stromal tumor (GIST). Although avapritinib has been approved by the FDA for four years, little is known about the risk of drug-drug interactions (DDIs) via UDP-glucuronyltransferases (UGTs) inhibition. OBJECTIVE The aim of the present study was to systematically evaluate the inhibitory effects of avapritinib against UGTs and to quantitatively estimate its potential DDIs risk in vivo. METHODS Recombinant human UGTs were employed to catalyze the glucuronidation of substrates in a range of concentrations of avapritinib. The kinetics analysis was performed to evaluate the inhibition types of avapritinib against UGTs. The quantitative prediction of DDIs was done using in vitro-in vivo extrapolation (IVIVE). RESULTS Avapritinib had a potent competitive inhibitory effect on UGT1A1. Quantitative prediction results showed that avapritinib administered at clinical doses might result in a 14.85% increase in area under the curve (AUC) of drugs primarily cleared by UGT1A1. Moreover, the Rgut value was calculated to be 18.44. CONCLUSION Avapritinib has the potential to cause intestinal DDIs via the inhibition of UGT1A1. Additional attention should be paid when avapritinib is coadministered with UGT1A1 substrates.
Collapse
Affiliation(s)
- Xin Lv
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Zhen Wang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Zhe Wang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hang Yin
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Yangliu Xia
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Lili Jiang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Yong Liu
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| |
Collapse
|
3
|
El Houari A, Ecale F, Mercier A, Crapart S, Laparre J, Soulard B, Ramnath M, Berjeaud JM, Rodier MH, Crépin A. Development of an in vitro Model of Human Gut Microbiota for Screening the Reciprocal Interactions With Antibiotics, Drugs, and Xenobiotics. Front Microbiol 2022; 13:828359. [PMID: 35495704 PMCID: PMC9042397 DOI: 10.3389/fmicb.2022.828359] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Altering the gut microbiota can negatively affect human health. Efforts may be sustained to predict the intended or unintended effects of molecules not naturally produced or expected to be present within the organism on the gut microbiota. Here, culture-dependent and DNA-based approaches were combined to UHPLC-MS/MS analyses in order to investigate the reciprocal interactions between a constructed Human Gut Microbiota Model (HGMM) and molecules including antibiotics, drugs, and xenobiotics. Our HGMM was composed of strains from the five phyla commonly described in human gut microbiota and belonging to Firmicutes, Bacteroidetes, Proteobacteria, Fusobacteria, and Actinobacteria. Relevantly, the bacterial diversity was conserved in our constructed human gut model through subcultures. Uneven richness distribution was revealed and the sensitivity of the HGMM was mainly affected by antibiotic exposure rather than by drugs or xenobiotics. Interestingly, the constructed model and the individual cultured strains respond with the same sensitivity to the different molecules. UHPLC-MS/MS analyses revealed the disappearance of some native molecules in the supernatants of the HGMM as well as in those of the individual strains. These results suggest that biotransformation of molecules occurred in the presence of our gut microbiota model and the coupled approaches performed on the individual cultures may emphasize new bacterial strains active in these metabolic processes. From this study, the new HGMM appears as a simple, fast, stable, and inexpensive model for screening the reciprocal interactions between the intestinal microbiota and molecules of interest.
Collapse
Affiliation(s)
- Abdelaziz El Houari
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Florine Ecale
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Anne Mercier
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Stéphanie Crapart
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | | | | | | | - Jean-Marc Berjeaud
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Marie-Hélène Rodier
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France.,Laboratoire de Parasitologie et Mycologie, CHU de Poitiers, Poitiers, France
| | - Alexandre Crépin
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| |
Collapse
|
4
|
Mullapudi TVR, Ravi PR, Thipparapu G. UGT1A1 and UGT1A3 activity and inhibition in human liver and intestinal microsomes and a recombinant UGT system under similar assay conditions using selective substrates and inhibitors. Xenobiotica 2021; 51:1236-1246. [PMID: 34698602 DOI: 10.1080/00498254.2021.1998732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In vitro enzyme kinetics and inhibition data was compared for UGT1A1 and UGT1A3 isoforms under similar assay conditions using human liver microsomes (HLM), human intestinal microsomes (HIM) and recombinant UGT (rUGT) enzyme systems.UGT1A1 catalysed β-estradiol 3-β-D-glucuronide formation showed allosteric sigmoidal kinetics in all enzyme systems; while UGT1A3 catalysed CDCA 24-acyl-β-D-glucuronide formation exhibited Michaelis-Menten kinetics in HLM, substrate inhibition kinetics in HIM and rUGT systems. Corresponding Km or S50 concentrations of β-estradiol and CDCA were employed in the respective UGT inhibition studies.Atazanavir inhibited the production of β-estradiol 3-β-D-glucuronide with IC50 values of 0.54 µM and 0.16 µM in HLM and rUGT1A1, respectively. But its inhibition potential was not observed in HIM, indicating potential cross-talk with other high-affinity intestinal UGT isozymes. On the other hand, zafirlukast, a pan UGT inhibitor, exhibited moderate inhibition in HIM with an IC50 value of 16.70 µM. Lithocholic acid, inhibited the production of CDCA 24-acyl-β-D-glucuronide with IC50 values of 1.68, 1.84, and 12.42 µM in HLM, rUGT1A3, and HIM, respectively.These results indicated that HLM, HIM, and rUGTs may be used as complementary in vitro systems to evaluate hepatic and intestinal UGT mediated DDIs at the screening stage.
Collapse
Affiliation(s)
- T V Radhakrishna Mullapudi
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, India.,Drug Metabolism and Pharmacokinetics, PharmaJen Laboratories Private Limited, A209 Technology Business Incubator, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, India
| | - Punna Rao Ravi
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, India
| | - Ganapathi Thipparapu
- Drug Metabolism and Pharmacokinetics, PharmaJen Laboratories Private Limited, A209 Technology Business Incubator, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, India
| |
Collapse
|
5
|
UGT1A Gene Family Members Serve as Potential Targets and Prognostic Biomarkers for Pancreatic Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6673125. [PMID: 34595239 PMCID: PMC8478536 DOI: 10.1155/2021/6673125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 07/21/2021] [Accepted: 08/21/2021] [Indexed: 12/24/2022]
Abstract
Background Pancreatic cancer (PC) is one of the most common cancers worldwide, with high mortality. The UGT1A gene family plays important roles in pharmacology and toxicology, contributing to interindividual differences in drug disposition. However, mRNA expression and prognostic value of the UGT1A gene family in PC have not been identified. Methods Oncomine, GEPIA2, DAVID 6.8, Metascape, Kaplan-Meier plotter, cBioPortal, GeneMANIA, TRRUST v2, TIMER, and R software were used in our study. Results The transcriptional levels of UGT1A1/3/6/8/9/10 in PC tissues were significantly higher than those in normal tissues. These results were further validated using five pairs of PC tumor tissues and adjacent nontumor tissues. A significant correlation was found between the expression of UGT1A1/6/10 and the pathological stage of PC. PC patients with lower transcriptional levels of UGT1A1/4/5/6/10 were associated with a better prognosis. The differentially expressed UGT1A gene family functions were primarily related to the glucuronidation pathway, cytokine-cytokine receptor interactions, and the ILK signaling pathway. Our data suggest that HNF1A, AHR, and CDX2 are key transcription factors for the UGT1A gene family. Furthermore, the expression levels of UGT1A1/3/8/9/10 were positively correlated with the activities of tumor-infiltrating immune cells, especially B cells. The expression levels of UGT1A6/9 were negatively correlated with macrophage infiltration levels. Conclusions These results suggest that the UGT1A gene family could serve as a potential prognostic biomarker and target for PC. However, future studies are required to validate our findings and promote the clinical utility of the UGT1A gene family in PC.
Collapse
|
6
|
Wang X, Wang Z, Fan X, Yan M, Jiang L, Xia Y, Cao J, Liu Y. Comparison of the drug-drug interactions potential of ibrutinib and acalabrutinib via inhibition of UDP-glucuronosyltransferase. Toxicol Appl Pharmacol 2021; 424:115595. [PMID: 34038714 DOI: 10.1016/j.taap.2021.115595] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 11/26/2022]
Abstract
Ibrutinib and acalabrutinib are two Bruton's tyrosine kinase (BTK) inhibitors which have gained Food and Drug Administration (FDA) approval for the treatment of various B cell malignancies. Herein, we investigated the effects of the two drugs on UDP-glucuronosyltransferase (UGT) activities to evaluate their potential risk for drug-drug interactions (DDIs) via UGT inhibition. Our data indicated that ibrutinib exerted broad inhibition on most of UGTs, including a potent competitive inhibition against UGT1A1 with a Ki value of 0.90 ± 0.03 μM, a noncompetitive inhibition against UGT1A3 and UGT1A7 with Ki values of 0.88 ± 0.03 μM and 2.52 ± 0.23 μM, respectively, while acalabrutinib only exhibited weak UGT inhibition towards all tested UGT isoforms. DDI risk prediction suggested that the inhibition against UGT1A1 and UGT1A3 by ibrutinib might bring a potential DDIs risk, while acalabrutinib was unlikely to trigger clinically significant UGT-mediated DDIs due to its weak effects. Our study raises an alarm bell about potential DDI risk associated with ibrutinib, however, the extrapolation from in vitro data to in vivo drug interactions should be taken with caution, and additional systemic study is needed.
Collapse
Affiliation(s)
- Xiaoyu Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhe Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Xiaoyu Fan
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Mingrui Yan
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Yangliu Xia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Jun Cao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian 116044, China.
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| |
Collapse
|
7
|
De Vriese K, Pollier J, Goossens A, Beeckman T, Vanneste S. Dissecting cholesterol and phytosterol biosynthesis via mutants and inhibitors. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:241-253. [PMID: 32929492 DOI: 10.1093/jxb/eraa429] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/11/2020] [Indexed: 06/11/2023]
Abstract
Plants stand out among eukaryotes due to the large variety of sterols and sterol derivatives that they can produce. These metabolites not only serve as critical determinants of membrane structures, but also act as signaling molecules, as growth-regulating hormones, or as modulators of enzyme activities. Therefore, it is critical to understand the wiring of the biosynthetic pathways by which plants generate these distinct sterols, to allow their manipulation and to dissect their precise physiological roles. Here, we review the complexity and variation of the biosynthetic routes of the most abundant phytosterols and cholesterol in the green lineage and how different enzymes in these pathways are conserved and diverged from humans, yeast, and even bacteria. Many enzymatic steps show a deep evolutionary conservation, while others are executed by completely different enzymes. This has important implications for the use and specificity of available human and yeast sterol biosynthesis inhibitors in plants, and argues for the development of plant-tailored inhibitors of sterol biosynthesis.
Collapse
Affiliation(s)
- Kjell De Vriese
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark, Ghent, Belgium
- VIB Center for Plant Systems Biology, VIB, Technologiepark, Ghent, Belgium
| | - Jacob Pollier
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark, Ghent, Belgium
- VIB Center for Plant Systems Biology, VIB, Technologiepark, Ghent, Belgium
- VIB Metabolomics Core, Technologiepark, Ghent, Belgium
| | - Alain Goossens
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark, Ghent, Belgium
- VIB Center for Plant Systems Biology, VIB, Technologiepark, Ghent, Belgium
| | - Tom Beeckman
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark, Ghent, Belgium
- VIB Center for Plant Systems Biology, VIB, Technologiepark, Ghent, Belgium
| | - Steffen Vanneste
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark, Ghent, Belgium
- VIB Center for Plant Systems Biology, VIB, Technologiepark, Ghent, Belgium
- Laboratory of Plant Growth Analysis, Ghent University Global Campus, Songdomunhwa-Ro, Yeonsu-gu, Incheon, Republic of Korea
- Department of Plants and Crops, Ghent University, Ghent, Belgium
| |
Collapse
|
8
|
Docci L, Umehara K, Krähenbühl S, Fowler S, Parrott N. Construction and Verification of Physiologically Based Pharmacokinetic Models for Four Drugs Majorly Cleared by Glucuronidation: Lorazepam, Oxazepam, Naloxone, and Zidovudine. AAPS JOURNAL 2020; 22:128. [DOI: 10.1208/s12248-020-00513-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
|
9
|
Vaillancourt J, Turcotte V, Caron P, Villeneuve L, Lacombe L, Pouliot F, Lévesque É, Guillemette C. Glucuronidation of Abiraterone and Its Pharmacologically Active Metabolites by UGT1A4, Influence of Polymorphic Variants and Their Potential as Inhibitors of Steroid Glucuronidation. Drug Metab Dispos 2020; 48:75-84. [PMID: 31727674 DOI: 10.1124/dmd.119.088229] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/08/2019] [Indexed: 02/13/2025] Open
Abstract
Abiraterone (Abi) acetate (AA) is a prodrug of Abi, a CYP17A1 inhibitor used to treat patients with advanced prostate cancer. Abi is a selective steroidal inhibitor that blocks the biosynthesis of androgens. It undergoes extensive biotransformation by steroid pathways, leading to the formation of pharmacologically active Δ4-Abi (D4A) and 5α-Abi. This study aimed to characterize the glucuronidation pathway of Abi and its two active metabolites. We show that Abi, its metabolites, and another steroidal inhibitor galeterone (Gal) undergo secondary metabolism to form glucuronides (G) in human liver microsomes with minor formation by intestine and kidney microsomal preparations. The potential clinical relevance of this pathway is supported by the detection by liquid chromatography-tandem mass spectrometry of Abi-G, D4A-G, and 5α-Abi-G in patients under AA therapy. A screening of UGT enzymes reveals that UGT1A4 is the main enzyme involved. This is supported by inhibition experiments using a selective UGT1A4 inhibitor hecogenin. A number of common and rare nonsynonymous variants significantly abrogate the UGT1A4-mediated formation of Abi-G, D4A-G, and 5α-Abi-G in vitro. We also identify Gal, Abi, and its metabolites as highly potent inhibitors of steroid inactivation by the UGT pathway with submicromolar inhibitor constant values. They reduce the glucuronidation of both the adrenal precursors and potent androgens in human liver, prostate cancer cells, and by recombinant UGTs involved in their inactivation. In conclusion, tested CYP17A1 inhibitors are metabolized through UGT1A4, and germline variations affecting this metabolic pathway may also influence drug metabolism. SIGNIFICANCE STATEMENT: The antiandrogen abiraterone (Abi) is a selective steroidal inhibitor of the cytochrome P450 17α-hydroxy/17,20-lyase, an enzyme involved in the biosynthesis of androgens. Abi is metabolized to pharmacologically active metabolites by steroidogenic enzymes. We demonstrate that Abi and its metabolites are glucuronidated in the liver and that their glucuronide derivatives are detected at variable levels in circulation of treated prostate cancer patients. UDP-glucuronosyltransferase (UGT)1A4 is the primary enzyme involved, and nonsynonymous germline variations affect this metabolic pathway in vitro, suggesting a potential influence of drug metabolism and action in patients. Their inhibitory effect on drug and steroid glucuronidation raises the possibility that these pharmacological compounds might affect the UGT-associated drug-metabolizing system and pre-receptor control of androgen metabolism in patients.
Collapse
Affiliation(s)
- Joanie Vaillancourt
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center - Université Laval and Faculty of Pharmacy (J.V., V.T., P.C., L.V., C.G.), CHU de Québec Research Center - Université Laval, Division of Urology, Faculty of Medicine, Surgery Department (L.L., F.P.), and CHU de Québec Research Center - Université Laval, Division of Hematology-Oncology, Faculty of Medicine (E.L.), Laval University, Québec, Canada
| | - Véronique Turcotte
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center - Université Laval and Faculty of Pharmacy (J.V., V.T., P.C., L.V., C.G.), CHU de Québec Research Center - Université Laval, Division of Urology, Faculty of Medicine, Surgery Department (L.L., F.P.), and CHU de Québec Research Center - Université Laval, Division of Hematology-Oncology, Faculty of Medicine (E.L.), Laval University, Québec, Canada
| | - Patrick Caron
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center - Université Laval and Faculty of Pharmacy (J.V., V.T., P.C., L.V., C.G.), CHU de Québec Research Center - Université Laval, Division of Urology, Faculty of Medicine, Surgery Department (L.L., F.P.), and CHU de Québec Research Center - Université Laval, Division of Hematology-Oncology, Faculty of Medicine (E.L.), Laval University, Québec, Canada
| | - Lyne Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center - Université Laval and Faculty of Pharmacy (J.V., V.T., P.C., L.V., C.G.), CHU de Québec Research Center - Université Laval, Division of Urology, Faculty of Medicine, Surgery Department (L.L., F.P.), and CHU de Québec Research Center - Université Laval, Division of Hematology-Oncology, Faculty of Medicine (E.L.), Laval University, Québec, Canada
| | - Louis Lacombe
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center - Université Laval and Faculty of Pharmacy (J.V., V.T., P.C., L.V., C.G.), CHU de Québec Research Center - Université Laval, Division of Urology, Faculty of Medicine, Surgery Department (L.L., F.P.), and CHU de Québec Research Center - Université Laval, Division of Hematology-Oncology, Faculty of Medicine (E.L.), Laval University, Québec, Canada
| | - Frédéric Pouliot
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center - Université Laval and Faculty of Pharmacy (J.V., V.T., P.C., L.V., C.G.), CHU de Québec Research Center - Université Laval, Division of Urology, Faculty of Medicine, Surgery Department (L.L., F.P.), and CHU de Québec Research Center - Université Laval, Division of Hematology-Oncology, Faculty of Medicine (E.L.), Laval University, Québec, Canada
| | - Éric Lévesque
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center - Université Laval and Faculty of Pharmacy (J.V., V.T., P.C., L.V., C.G.), CHU de Québec Research Center - Université Laval, Division of Urology, Faculty of Medicine, Surgery Department (L.L., F.P.), and CHU de Québec Research Center - Université Laval, Division of Hematology-Oncology, Faculty of Medicine (E.L.), Laval University, Québec, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center - Université Laval and Faculty of Pharmacy (J.V., V.T., P.C., L.V., C.G.), CHU de Québec Research Center - Université Laval, Division of Urology, Faculty of Medicine, Surgery Department (L.L., F.P.), and CHU de Québec Research Center - Université Laval, Division of Hematology-Oncology, Faculty of Medicine (E.L.), Laval University, Québec, Canada
| |
Collapse
|
10
|
Lee H, Heo JK, Lee GH, Park SY, Jang SN, Kim HJ, Kwon MJ, Song IS, Liu KH. Ginsenoside Rc Is a New Selective UGT1A9 Inhibitor in Human Liver Microsomes and Recombinant Human UGT Isoforms. Drug Metab Dispos 2019; 47:1372-1379. [PMID: 31578207 DOI: 10.1124/dmd.119.087965] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/25/2019] [Indexed: 02/13/2025] Open
Abstract
Ginseng is known to have inhibitory effects on UGT1A9 activity. However, little is known about the inhibitory effects of ginsenosides, the major active compounds in ginseng, on UGT1A9 activity. In vitro investigation of UGT1A9 inhibition by ginsenosides was carried out using human liver microsomes (HLMs). Among 10 ginsenosides, ginsenoside Rc was the strongest inhibitor of UGT1A9-mediated mycophenolic acid glucuronidase activity. Further inhibition kinetic studies using HLMs suggested that ginsenoside Rc competitively and noncompetitively inhibited UGT1A9-mediated propofol and mycophenolic acid glucuronidation activities, with K i values of 2.83 and 3.31 μM, respectively. Next, to investigate whether the inhibitory effect of ginsenoside Rc is specific to the UGT1A9 isoform, we studied the inhibitory potency of ginsenoside Rc on nine human uridine diphospho-glucuronosyltransferase (UGT) activities using recombinant human UGT isoforms. Ginsenoside Rc exhibited a 12.9-fold selectivity (which was similar to niflumic acid at 12.5-fold) for UGT1A9 inhibition. Ginsenoside Rc at 50 μM also inhibited none of the other UGT isoform-specific activities above 12.0%, except for UGT1A9 (>91.5%) in HLMs, indicating that ginsenoside Rc might be used as a selective UGT1A9 inhibitor in reaction phenotyping studies of new chemical entities. Considering lower plasma concentrations (0.01 μM) of ginsenoside Rc in healthy subjects and no induction potential on UGT isoforms, ginsenoside Rc does not cause pharmacokinetic drug interactions with other coadministered drugs metabolized by UGT1A9. SIGNIFICANCE STATEMENT: Ginsenoside Rc selectively inhibited UGT1A9-mediated propofol and mycophenolic acid glucuronidation activities in human liver microsomes and recombinant uridine diphospho-glucuronosyltransferase (UGT) isoforms. It exhibited a 12.9-fold selectivity for UGT1A9 inhibition. Therefore, ginsenoside Rc might be used as a selective UGT1A9 inhibitor in reaction phenotyping studies of new chemical entities, such as niflumic acid.
Collapse
Affiliation(s)
- Hyunyoung Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Jae-Kyung Heo
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Ga-Hyun Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - So-Young Park
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Su-Nyeong Jang
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Hyun-Ji Kim
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Mi Jeong Kwon
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Im-Sook Song
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Kwang-Hyeon Liu
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
11
|
Potential of herb-drug / herb interactions between substrates and inhibitors of UGTs derived from herbal medicines. Pharmacol Res 2019; 150:104510. [DOI: 10.1016/j.phrs.2019.104510] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/14/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022]
|
12
|
Speer JE, Wang Y, Fallon JK, Smith PC, Allbritton NL. Evaluation of human primary intestinal monolayers for drug metabolizing capabilities. J Biol Eng 2019; 13:82. [PMID: 31709009 PMCID: PMC6829970 DOI: 10.1186/s13036-019-0212-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The intestinal epithelium is a major site of drug metabolism in the human body, possessing enterocytes that house brush border enzymes and phase I and II drug metabolizing enzymes (DMEs). The enterocytes are supported by a porous extracellular matrix (ECM) that enables proper cell adhesion and function of brush border enzymes, such as alkaline phosphatase (ALP) and alanyl aminopeptidase (AAP), phase I DMEs that convert a parent drug to a more polar metabolite by introducing or unmasking a functional group, and phase II DMEs that form a covalent conjugate between a functional group on the parent compound or sequential metabolism of phase I metabolite. In our effort to develop an in vitro intestinal epithelium model, we investigate the impact of two previously described simple and customizable scaffolding systems, a gradient cross-linked scaffold and a conventional scaffold, on the ability of intestinal epithelial cells to produce drug metabolizing proteins as well as to metabolize exogenously added compounds. While the scaffolding systems possess a range of differences, they are most distinguished by their stiffness with the gradient cross-linked scaffold possessing a stiffness similar to that found in the in vivo intestine, while the conventional scaffold possesses a stiffness several orders of magnitude greater than that found in vivo. RESULTS The monolayers on the gradient cross-linked scaffold expressed CYP3A4, UGTs 2B17, 1A1 and 1A10, and CES2 proteins at a level similar to that in fresh crypts/villi. The monolayers on the conventional scaffold expressed similar levels of CYP3A4 and UGTs 1A1 and 1A10 DMEs to that found in fresh crypts/villi but significantly decreased expression of UGT2B17 and CES2 proteins. The activity of CYP3A4 and UGTs 1A1 and 1A10 was inducible in cells on the gradient cross-linked scaffold when the cells were treated with known inducers, whereas the CYP3A4 and UGT activities were not inducible in cells grown on the conventional scaffold. Both monolayers demonstrate esterase activity but the activity measured in cells on the conventional scaffold could not be inhibited with a known CES2 inhibitor. Both monolayer culture systems displayed similar ALP and AAP brush border enzyme activity. When cells on the conventional scaffold were incubated with a yes-associated protein (YAP) inhibitor, CYP3A4 activity was greatly enhanced suggesting that mechano-transduction signaling can modulate drug metabolizing enzymes. CONCLUSIONS The use of a cross-linked hydrogel scaffold for expansion and differentiation of primary human intestinal stem cells dramatically impacts the induction of CYP3A4 and maintenance of UGT and CES drug metabolizing enzymes in vitro making this a superior substrate for enterocyte culture in DME studies. This work highlights the influence of mechanical properties of the culture substrate on protein expression and the activity of drug metabolizing enzymes as a critical factor in developing accurate assay protocols for pharmacokinetic studies using primary intestinal cells. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Jennifer E. Speer
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - John K. Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607 USA
| | - Philip C. Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607 USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607 USA
| |
Collapse
|
13
|
Lv X, Xia Y, Finel M, Wu J, Ge G, Yang L. Recent progress and challenges in screening and characterization of UGT1A1 inhibitors. Acta Pharm Sin B 2019; 9:258-278. [PMID: 30972276 PMCID: PMC6437557 DOI: 10.1016/j.apsb.2018.09.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/16/2018] [Accepted: 08/27/2018] [Indexed: 02/07/2023] Open
Abstract
Uridine-diphosphate glucuronosyltransferase 1A1 (UGT1A1) is an important conjugative enzyme in mammals that is responsible for the conjugation and detoxification of both endogenous and xenobiotic compounds. Strong inhibition of UGT1A1 may trigger adverse drug/herb-drug interactions, or result in metabolic disorders of endobiotic metabolism. Therefore, both the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have recommended assaying the inhibitory potential of drugs under development on the human UGT1A1 prior to approval. This review focuses on the significance, progress and challenges in discovery and characterization of UGT1A1 inhibitors. Recent advances in the development of UGT1A1 probes and their application for screening UGT1A1 inhibitors are summarized and discussed in this review for the first time. Furthermore, a long list of UGT1A1 inhibitors, including information on their inhibition potency, inhibition mode, and affinity, has been prepared and analyzed. Challenges and future directions in this field are highlighted in the final section. The information and knowledge that are presented in this review provide guidance for rational use of drugs/herbs in order to avoid the occurrence of adverse effects via UGT1A1 inhibition, as well as presenting methods for rapid screening and characterization of UGT1A1 inhibitors and for facilitating investigations on UGT1A1-ligand interactions.
Collapse
|
14
|
Tsai MT, Tarng DC. Beyond a Measure of Liver Function-Bilirubin Acts as a Potential Cardiovascular Protector in Chronic Kidney Disease Patients. Int J Mol Sci 2018; 20:ijms20010117. [PMID: 30597982 PMCID: PMC6337523 DOI: 10.3390/ijms20010117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/17/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023] Open
Abstract
Bilirubin is a well-known neurotoxin in newborn infants; however, current evidence has shown that a higher serum bilirubin concentration in physiological ranges is associated with a lower risk for the development and progression of both chronic kidney disease (CKD) and cardiovascular disease (CVD) in adults. The protective mechanisms of bilirubin in CVD, CKD, and associated mortality may be ascribed to its antioxidant and anti-inflammatory properties. Bilirubin further improves insulin sensitivity, reduces low-density lipoprotein cholesterol levels and inhibits platelet activation in at-risk individuals. These effects are expected to maintain normal vascular homeostasis and thus reduce the incidence of CKD and the risks of cardiovascular complications and death. In this review, we highlight the recent advances in the biological actions of bilirubin in the pathogenesis of CVD and CKD progression, and further propose that targeting bilirubin metabolism could be a potential approach to ameliorate morbidity and mortality in CKD patients.
Collapse
Affiliation(s)
- Ming-Tsun Tsai
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Department and Institute of Physiology, National Yang-Ming University, Taipei 11217, Taiwan.
| |
Collapse
|
15
|
Osborne MJ, Coutinho de Oliveira L, Volpon L, Zahreddine HA, Borden KLB. Overcoming Drug Resistance through the Development of Selective Inhibitors of UDP-Glucuronosyltransferase Enzymes. J Mol Biol 2018; 431:258-272. [PMID: 30428301 DOI: 10.1016/j.jmb.2018.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/18/2018] [Accepted: 11/05/2018] [Indexed: 12/26/2022]
Abstract
Drug resistance is a major cause of cancer-related mortality. Glucuronidation of drugs via elevation of UDP-glucuronosyltransferases (UGT1As) correlates with clinical resistance. The nine UGT1A family members have broad substrate specificities attributed to their variable N-terminal domains and share a common C-terminal domain. Development of UGT1As as pharmacological targets has been hampered by toxicity of pan-UGT inhibitors and by difficulty in isolating pure N-terminal domains or full-length proteins. Here, we developed a strategy to target selected UGT1As which exploited the biochemical tractability of the C-domain and its ability to allosterically communicate with the catalytic site. By combining NMR fragment screening with in vitro glucuronidation assays, we identified inhibitors selective for UGT1A4. Significantly, these compounds selectively restored sensitivity in resistant cancer cells only for substrates of the targeted UGT1A. This strategy represents a crucial first step toward developing compounds to overcome unwanted glucuronidation thereby reversing resistance in patients.
Collapse
Affiliation(s)
- Michael J Osborne
- Institute of Research in Immunology and Cancer (IRIC), Department of Pathology and Cell Biology, Université de Montréal, Pavilion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Luciana Coutinho de Oliveira
- Institute of Research in Immunology and Cancer (IRIC), Department of Pathology and Cell Biology, Université de Montréal, Pavilion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Laurent Volpon
- Institute of Research in Immunology and Cancer (IRIC), Department of Pathology and Cell Biology, Université de Montréal, Pavilion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Hiba Ahmad Zahreddine
- Institute of Research in Immunology and Cancer (IRIC), Department of Pathology and Cell Biology, Université de Montréal, Pavilion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC, Canada
| | - Katherine L B Borden
- Institute of Research in Immunology and Cancer (IRIC), Department of Pathology and Cell Biology, Université de Montréal, Pavilion Marcelle-Coutu, Chemin Polytechnique, Montreal, QC, Canada.
| |
Collapse
|
16
|
Fujiwara R, Yoda E, Tukey RH. Species differences in drug glucuronidation: Humanized UDP-glucuronosyltransferase 1 mice and their application for predicting drug glucuronidation and drug-induced toxicity in humans. Drug Metab Pharmacokinet 2017; 33:9-16. [PMID: 29079228 DOI: 10.1016/j.dmpk.2017.10.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/05/2017] [Accepted: 10/03/2017] [Indexed: 12/11/2022]
Abstract
More than 20% of clinically used drugs are glucuronidated by a microsomal enzyme UDP-glucuronosyltransferase (UGT). Inhibition or induction of UGT can result in an increase or decrease in blood drug concentration. To avoid drug-drug interactions and adverse drug reactions in individuals, therefore, it is important to understand whether UGTs are involved in metabolism of drugs and drug candidates. While most of glucuronides are inactive metabolites, acyl-glucuronides that are formed from compounds with a carboxylic acid group can be highly toxic. Animals such as mice and rats are widely used to predict drug metabolism and drug-induced toxicity in humans. However, there are marked species differences in the expression and function of drug-metabolizing enzymes including UGTs. To overcome the species differences, mice in which certain drug-metabolizing enzymes are humanized have been recently developed. Humanized UGT1 (hUGT1) mice were created in 2010 by crossing Ugt1-null mice with human UGT1 transgenic mice in a C57BL/6 background. hUGT1 mice can be promising tools to predict human drug glucuronidation and acyl-glucuronide-associated toxicity. In this review article, studies of drug metabolism and toxicity in the hUGT1 mice are summarized. We further discuss research and strategic directions to advance the understanding of drug glucuronidation in humans.
Collapse
Affiliation(s)
- Ryoichi Fujiwara
- Department of Pharmaceutics, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| | - Emiko Yoda
- Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Robert H Tukey
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Yuan XY, He XL, Zou H, Zou RY. [Repeated yellowing of the skin and sclera for 2 years]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:77-80. [PMID: 28100328 PMCID: PMC7390122 DOI: 10.7499/j.issn.1008-8830.2017.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/20/2016] [Indexed: 06/06/2023]
Abstract
A two-year-old girl was admitted due to repeated yellowing of the skin and sclera for 2 years and had no other specific symptoms or signs. The use of phenobarbital could relieve the symptoms of jaundice. Multiple examinations showed increased indirect bilirubin levels, and the results of aminotransferases and liver imaging were normal. There was no evidence of hemolysis. The analysis of UGT1A1 gene in her family found that this child had double homozygous mutation of c.211G>A(G71R) and c.1456T>G(Y486D), which had been reported as the pathogenic mutation for Gilbert syndrome. Her parents carried double heterozygous mutation of G71R and Y486D and had no symptom of jaundice. The child was diagnosed as having Gilbert syndrome. It is concluded that as for patients with unconjugated hyperbilirubinemia which cannot be explained by liver damage and hemolysis, their family history should be investigated in detail and gene analysis should be performed as early as possible, in order to identify congenital bilirubin metabolic disorders.
Collapse
Affiliation(s)
- Xiao-Ye Yuan
- Department of Pediatric Hematology and Oncology, Hunan People's Hospital, Changsha 410005, China.
| | | | | | | |
Collapse
|
18
|
Xu M, Dong P, Tian X, Wang C, Huo X, Zhang B, Wu L, Deng S, Ma X. Drug interaction study of natural steroids from herbs specifically toward human UDP-glucuronosyltransferase (UGT) 1A4 and their quantitative structure activity relationship (QSAR) analysis for prediction. Pharmacol Res 2016; 110:139-150. [DOI: 10.1016/j.phrs.2016.05.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/26/2016] [Accepted: 05/06/2016] [Indexed: 12/11/2022]
|