1
|
Yang L, Liao ZZ, Ran L, Xiao XH. Progress of arylacetamide deacetylase research in metabolic diseases. Front Oncol 2025; 15:1564419. [PMID: 40376582 PMCID: PMC12078129 DOI: 10.3389/fonc.2025.1564419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/31/2025] [Indexed: 05/18/2025] Open
Abstract
Arylacetamide deacetylase (AADAC), a microsomal serine esterase belonging to the polygenic hydrolase family, is predominantly localized in the liver and intestine. It plays a significant role in drug metabolism, lipid metabolism, and the pathogenesis of various diseases. In the context of drug metabolism, AADAC is vital for ensuring the safety of ester-based drugs. Its substrate specificity for short-chain acyl groups, along with genetic polymorphisms among individuals and species, influences drug-related processes. Regarding lipid metabolism, The lipase activity of AADAC is involved in the hydrolysis of cholesterol and triglycerides, lipid mobilization, and the assembly of lipoproteins. The expression of AADAC is regulated by multiple factors. It is associated with metabolic disorders; for instance, its decreased expression in the liver during obesity may impact triglyceride metabolism, and it may also have an indirect role in diabetes. In cardiovascular diseases, AADAC holds potential as a diagnostic marker. Its role in cancer is heterogeneous, being downregulated in certain cancers while upregulated in others, such as pancreatic and ovarian cancers, where it acts to inhibit cancer progression. Within the nervous system, AADAC may influence neurotransmitter regulation and drug metabolism. Currently, research on AADAC agonists is limited, and the development of inhibitors presents challenges, underscoring the necessity for further investigation in this area.
Collapse
Affiliation(s)
| | | | - Li Ran
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
2
|
Nakajima M, Yamazaki H, Yoshinari K, Kobayashi K, Ishii Y, Nakai D, Kamimura H, Kume T, Saito Y, Maeda K, Kusuhara H, Tamai I. Contribution of Japanese scientists to drug metabolism and disposition. Drug Metab Dispos 2025; 53:100071. [PMID: 40245580 DOI: 10.1016/j.dmd.2025.100071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Japanese researchers have played a pivotal role in advancing the field of drug metabolism and disposition, as demonstrated by their substantial contributions to the journal Drug Metabolism and Disposition (DMD) over the past 5 decades. This review highlights the historical and ongoing impact of Japanese scientists on DMD, celebrating their achievements in elucidating drug metabolism, membrane transport, pharmacokinetics, and toxicology. From the discovery of cytochrome P450 by Tsuneo Omura and Ryo Sato in 1962 to subsequent advances in drug transport research, Japan has maintained a leading position in the field. A geographical analysis of DMD publications reveals a notable increase in contributions from Japan during the 1980s, ranking second globally and maintaining this position through the 2000s. However, recent years have seen a slight decline in output, likely influenced by the COVID-19 pandemic and increased online journals as well as structural changes within academia and industry. Importantly, this trend is not unique to Japan. To sustain excellence and innovation in this field, it is crucial to strengthen funding for absorption, distribution, metabolism, excretion, and toxicity research and promote collaborations between academia, industry, and regulatory agencies. By prioritizing the translation of fundamental discoveries into drug development and clinical applications, scientists in this area can further advance global efforts toward achieving optimal drug efficacy and safety. This review underscores the enduring contributions of Japanese researchers to DMD and calls for renewed efforts to drive innovation and progress in this vital area of science. SIGNIFICANCE STATEMENT: Over the past 5 decades, Japanese scientists have made significant contributions to Drug Metabolism and Disposition through groundbreaking discoveries and advancements in the study of drug-metabolizing enzymes, transporters, pharmacokinetics analysis, and related areas. These contributions continue to shape the field, offering a foundation for future innovation in this area. We hope that the next generation of Japanese scientists will further solidify their global leadership in this area to advance drug development and proper pharmacotherapy.
Collapse
Affiliation(s)
- Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute, Kanazawa University, Kanazawa, Japan.
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Tokyo, Japan
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kaoru Kobayashi
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Tokyo, Japan
| | - Yuji Ishii
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Daisuke Nakai
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co, Ltd, Tokyo, Japan
| | | | | | - Yoshiro Saito
- National Institute of Health Sciences, Kanagawa, Japan
| | - Kazuya Maeda
- School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
3
|
Yerrakula G, Abraham S, John S, Zeharvi M, George SG, Senthil V, Maiz F, Rahman MH. Major implications of single nucleotide polymorphisms in human carboxylesterase 1 on substrate bioavailability. Biotechnol Genet Eng Rev 2024; 40:3174-3192. [PMID: 35946821 DOI: 10.1080/02648725.2022.2108997] [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: 05/29/2022] [Accepted: 07/26/2022] [Indexed: 11/02/2022]
Abstract
The number of studies and reviews conducted for the Carboxylesterase gene is limited in comparison with other enzymes. Carboxylesterase (CES) gene or human carboxylesterases (hCES) is a multigene protein belonging to the α/β-hydrolase family. Over the last decade, two major carboxylesterases (CES1 and CES2), located at 16q13-q22.1 on human chromosome 16 have been extensively studied as important mediators in the metabolism of a wide range of substrates. hCES1 is the most widely expressed enzyme in humans, and it is found in the liver. In this review, details regarding CES1 substrates include both inducers (e.g. Rifampicin) and inhibitors (e.g. Enalapril, Diltiazem, Simvastatin) and different types of hCES1 polymorphisms (nsSNPs) such as rs2244613 and rs71647871. along with their effects on various CES1 substrates were documented. Few instances where the presence of nsSNPs exerted a positive influence on certain substrates which are hydrolyzed via hCES1, such as anti-platelets like Clopidogrel when co-administered with other medications such as angiotensin-converting enzyme (ACE) inhibitors were also recorded. Remdesivir, an ester prodrug is widely used for the treatment of COVID-19, being a CES substrate, it is a potent inhibitor of CES2 and is hydrolyzed via CES1. The details provided in this review could give a clear-cut idea or information that could be used for further studies regarding the safety and efficacy of CES1 substrate.
Collapse
Affiliation(s)
- Goutham Yerrakula
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamilnadu
| | - Shyno Abraham
- Department of Pharmacy Practice, Krupanidhi college of Pharmacy, Bangalore
| | - Shiji John
- Department of Pharmacy Practice, Krupanidhi college of Pharmacy, Bangalore
| | - Mehrukh Zeharvi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University Alkharj, Saudia Arabia
| | | | - V Senthil
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamilnadu
| | - Fathi Maiz
- Department of Physics, Faculty of Science, King Khalid University, Abha, Saudi Arabia
- Laboratory of Thermal Processes, Center for Energy Research and Technology, Borj-Cedria, BP:95 Tunisia
| | - Md Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Gangwon-do, Wonju, Korea
| |
Collapse
|
4
|
Sun D, Liu Y, Zhu L, Xu Z, Zhang Y, Li H, Yang H, Cao X, Gu J. Pharmacokinetic/Pharmacodynamic Assessment of the Structural Refinement of Clopidogrel Focusing on the Balance between Bioactivation and Deactivation. Drug Metab Dispos 2024; 52:654-661. [PMID: 38729662 DOI: 10.1124/dmd.124.001699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/24/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
The delicate balance between ischemic and bleeding risks is a critical factor in antiplatelet therapy administration. Clopidogrel and prasugrel, belonging to the thienopyridine class of antiplatelet drugs, are known for their variability in individual responsiveness and high incidence of bleeding events, respectively. The present study is centered on the development and assessment of a range of deuterated thienopyridine derivatives, leveraging insights from structure-pharmacokinetic relationships of clopidogrel and prasugrel. Our approaches were grounded in the molecular framework of clopidogrel and incorporated the C2-pharmacophore design from prasugrel. The selection of ester or carbamate substituents at the C2-position facilitated the generation of the 2-oxointermediate through hydrolysis, akin to prasugrel, thereby bypassing the issue of CYP2C19 dependency. The bulky C2-pharmacophore in our approach distinguishes itself from prasugrel's acetyloxy substituent by exhibiting a moderated hydrolysis rate, resulting in a more gradual formation of the active metabolite. Excessive and rapid release of the active metabolite, believed to be linked with an elevated risk of bleeding, is thus mitigated. Our proposed structural modification retains the hydrolysis-sensitive methyl ester of clopidogrel but substitutes it with a deuterated methyl group, shown to effectively reduce metabolic deactivation. Three promising compounds demonstrated a pharmacokinetic profile similar to that of clopidogrel at four times the dose, while also augmenting its antiplatelet activity. SIGNIFICANCE STATEMENT: Inspired by the structure-pharmacokinetic relationship of clopidogrel and prasugrel, a range of clopidogrel derivatives were designed, synthesized, and assessed. Among them, three promising compounds have been identified, striking a delicate balance between efficacy and safety for antiplatelet therapy. Additionally, the ozagrel prodrug conjugate was discovered to exert a synergistic therapeutic effect alongside clopidogrel.
Collapse
Affiliation(s)
- Dong Sun
- Research Center for Drug Metabolism, School of Life Science (D.S., L.Z., Y.Z., H.L., H.Y., J.G.), Department of Pharmacology, School of Pharmacy Sciences (Y.L., Z.X., X.C.), and State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry (J.G.), Jilin University, Changchun, China; and Beijing Institute of Drug Metabolism, Beijing, China (J.G.)
| | - Yingze Liu
- Research Center for Drug Metabolism, School of Life Science (D.S., L.Z., Y.Z., H.L., H.Y., J.G.), Department of Pharmacology, School of Pharmacy Sciences (Y.L., Z.X., X.C.), and State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry (J.G.), Jilin University, Changchun, China; and Beijing Institute of Drug Metabolism, Beijing, China (J.G.)
| | - Lin Zhu
- Research Center for Drug Metabolism, School of Life Science (D.S., L.Z., Y.Z., H.L., H.Y., J.G.), Department of Pharmacology, School of Pharmacy Sciences (Y.L., Z.X., X.C.), and State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry (J.G.), Jilin University, Changchun, China; and Beijing Institute of Drug Metabolism, Beijing, China (J.G.)
| | - Zhiping Xu
- Research Center for Drug Metabolism, School of Life Science (D.S., L.Z., Y.Z., H.L., H.Y., J.G.), Department of Pharmacology, School of Pharmacy Sciences (Y.L., Z.X., X.C.), and State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry (J.G.), Jilin University, Changchun, China; and Beijing Institute of Drug Metabolism, Beijing, China (J.G.)
| | - Yuyao Zhang
- Research Center for Drug Metabolism, School of Life Science (D.S., L.Z., Y.Z., H.L., H.Y., J.G.), Department of Pharmacology, School of Pharmacy Sciences (Y.L., Z.X., X.C.), and State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry (J.G.), Jilin University, Changchun, China; and Beijing Institute of Drug Metabolism, Beijing, China (J.G.)
| | - Haipeng Li
- Research Center for Drug Metabolism, School of Life Science (D.S., L.Z., Y.Z., H.L., H.Y., J.G.), Department of Pharmacology, School of Pharmacy Sciences (Y.L., Z.X., X.C.), and State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry (J.G.), Jilin University, Changchun, China; and Beijing Institute of Drug Metabolism, Beijing, China (J.G.)
| | - Huan Yang
- Research Center for Drug Metabolism, School of Life Science (D.S., L.Z., Y.Z., H.L., H.Y., J.G.), Department of Pharmacology, School of Pharmacy Sciences (Y.L., Z.X., X.C.), and State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry (J.G.), Jilin University, Changchun, China; and Beijing Institute of Drug Metabolism, Beijing, China (J.G.)
| | - Xia Cao
- Research Center for Drug Metabolism, School of Life Science (D.S., L.Z., Y.Z., H.L., H.Y., J.G.), Department of Pharmacology, School of Pharmacy Sciences (Y.L., Z.X., X.C.), and State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry (J.G.), Jilin University, Changchun, China; and Beijing Institute of Drug Metabolism, Beijing, China (J.G.)
| | - Jingkai Gu
- Research Center for Drug Metabolism, School of Life Science (D.S., L.Z., Y.Z., H.L., H.Y., J.G.), Department of Pharmacology, School of Pharmacy Sciences (Y.L., Z.X., X.C.), and State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry (J.G.), Jilin University, Changchun, China; and Beijing Institute of Drug Metabolism, Beijing, China (J.G.)
| |
Collapse
|
5
|
Nagaoka M, Sakai Y, Nakajima M, Fukami T. Role of carboxylesterase and arylacetamide deacetylase in drug metabolism, physiology, and pathology. Biochem Pharmacol 2024; 223:116128. [PMID: 38492781 DOI: 10.1016/j.bcp.2024.116128] [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: 12/01/2023] [Revised: 01/20/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
Carboxylesterases (CES1 and CES2) and arylacetamide deacetylase (AADAC), which are expressed primarily in the liver and/or gastrointestinal tract, hydrolyze drugs containing ester and amide bonds in their chemical structure. These enzymes often catalyze the conversion of prodrugs, including the COVID-19 drugs remdesivir and molnupiravir, to their pharmacologically active forms. Information on the substrate specificity and inhibitory properties of these enzymes, which would be useful for drug development and toxicity avoidance, has accumulated. Recently,in vitroandin vivostudies have shown that these enzymes are involved not only in drug hydrolysis but also in lipid metabolism. CES1 and CES2 are capable of hydrolyzing triacylglycerol, and the deletion of their orthologous genes in mice has been associated with impaired lipid metabolism and hepatic steatosis. Adeno-associated virus-mediated human CES overexpression decreases hepatic triacylglycerol levels and increases fatty acid oxidation in mice. It has also been shown that overexpression of CES enzymes or AADAC in cultured cells suppresses the intracellular accumulation of triacylglycerol. Recent reports indicate that AADAC can be up- or downregulated in tumors of various organs, and its varied expression is associated with poor prognosis in patients with cancer. Thus, CES and AADAC not only determine drug efficacy and toxicity but are also involved in pathophysiology. This review summarizes recent findings on the roles of CES and AADAC in drug metabolism, physiology, and pathology.
Collapse
Affiliation(s)
- Mai Nagaoka
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshiyuki Sakai
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
6
|
Diaz-Vidal T, Romero-Olivas CB, Martínez-Pérez RB. Characterization, comparative, and functional analysis of arylacetamide deacetylase from Gnathostomata organisms. J Genet Eng Biotechnol 2022; 20:169. [PMID: 36542226 PMCID: PMC9772364 DOI: 10.1186/s43141-022-00443-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 11/12/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Arylacetamide deacetylase (AADAC) is a lipolytic enzyme involved in xenobiotic metabolism. The characterization in terms of activity and substrate preference has been limited to a few mammalian species. The potential role and catalytic activities of AADAC from other organisms are still poorly understood. Therefore, in this work, the physicochemical properties, proteomic analysis, and protein-protein interactions from Gnathostomata organisms were investigated. RESULTS The analysis were performed with 142 orthologue sequences with ~ 48-100% identity with human AADAC. The catalytic motif HGG[A/G] tetrapeptide block was conserved through all AADAC orthologues. Four variations were found in the consensus pentapeptide GXSXG sequence (GDSAG, GESAG, GDSSG, and GSSSG), and a novel motif YXLXP was found. The prediction of N-glycosylation sites projected 4, 1, 6, and 4 different patterns for amphibians, birds, mammals, and reptiles, respectively. The transmembrane regions of AADAC orthologues were not conserved among groups, and variations in the number and orientation of the active site and C-terminal carboxyl were observed among the sequences studied. The protein-protein interaction of AADAC orthologues were related to cancer, lipid, and xenobiotic metabolism genes. CONCLUSION The findings from this computational analysis offer new insight into one of the main enzymes involved in xenobiotic metabolism from mammals, reptiles, amphibians, and birds and its potential use in medical and veterinarian biotechnological approaches.
Collapse
Affiliation(s)
- Tania Diaz-Vidal
- grid.412890.60000 0001 2158 0196Present Address: Department of Chemical Engineering, University of Guadalajara, 44430 Guadalajara, Mexico
| | - Christian Berenice Romero-Olivas
- grid.466844.c0000 0000 9963 8346Present Address: Department of Biotechnology and Food Sciences, Instituto Tecnológico de Sonora, Ciudad Obregón, Mexico 85137
| | - Raúl Balam Martínez-Pérez
- grid.466844.c0000 0000 9963 8346Present Address: Department of Biotechnology and Food Sciences, Instituto Tecnológico de Sonora, Ciudad Obregón, Mexico 85137 ,grid.418270.80000 0004 0428 7635Industrial Biotechnology, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, 45019 Zapopan, Mexico
| |
Collapse
|
7
|
Zhu T, Wu Y, Li XM, Jia YM, Zhou H, Jiang LP, Tai T, Mi QY, Ji JZ, Xie HG. Vicagrel is hydrolyzed by Raf kinase inhibitor protein in human intestine. Biopharm Drug Dispos 2022; 43:247-254. [PMID: 36519186 DOI: 10.1002/bdd.2340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/12/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022]
Abstract
As an analog of clopidogrel and prasugrel, vicagrel is completely hydrolyzed to intermediate thiolactone metabolite 2-oxo-clopidogrel (also the precursor of active thiol metabolite H4) in human intestine, predominantly by AADAC and CES2; however, other unknown vicagrel hydrolases remain to be identified. In this study, recombinant human Raf kinase inhibitor protein (rhRKIP) and pooled human intestinal S9 (HIS9) fractions and microsome (HIM) preparations were used as the different enzyme sources; prasugrel as a probe drug for RKIP (a positive control), vicagrel as a substrate drug of interest, and the rate of the formation of thiolactone metabolites 2-oxo-clopidogrel and R95913 as metrics of hydrolase activity examined, respectively. In addition, an IC50 value of inhibition of rhRKIP-catalyzed vicagrel hydrolysis by locostatin was measured, and five classical esterase inhibitors with distinct esterase selectivity were used to dissect the involvement of multiple hydrolases in vicagrel hydrolysis. The results showed that rhRKIP hydrolyzed vicagrel in vitro, with the values of Km , Vmax , and CLint measured as 20.04 ± 1.99 μM, 434.60 ± 12.46 nM/min/mg protein, and 21.69 ± 0.28 ml/min/mg protein, respectively, and that an IC50 value of locostatin was estimated as 1.24 ± 0.04 mM for rhRKIP. In addition to locostatin, eserine and vinblastine strongly suppressed vicagrel hydrolysis in HIM. It is concluded that RKIP can catalyze the hydrolysis of vicagrel in the human intestine, and that vicagrel can be hydrolyzed by multiple hydrolases, such as RKIP, AADAC, and CES2, concomitantly.
Collapse
Affiliation(s)
- Ting Zhu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yu Wu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xue-Mei Li
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Meng Jia
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Huan Zhou
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Li-Ping Jiang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ting Tai
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qiong-Yu Mi
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jin-Zi Ji
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hong-Guang Xie
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Clinical Pharmacy, Nanjing Medical University School of Pharmacy, Nanjing, China
| |
Collapse
|
8
|
Gorycki P, Magee M, Ackerman P, Miao X, Moore K. Pharmacokinetics, Metabolism and Excretion of Radiolabeled Fostemsavir Administered with or without Ritonavir in Healthy Male Subjects. Xenobiotica 2022; 52:541-554. [PMID: 36083110 DOI: 10.1080/00498254.2022.2119179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The pharmacokinetics, elimination, and metabolism of fostemsavir (FTR), a prodrug of the HIV-1 attachment inhibitor temsavir (TMR), were investigated in healthy volunteers. FTR was administered with and without ritonavir (RTV), a protease inhibitor previously shown to boost TMR exposures. In vitro studies were also used to identify the enzymes responsible for the metabolism of TMR.Total recovery of the administered dose ranged from 78% to 89%. Approximately 44% to 58% of the dose was excreted in urine, 20% to 36% in feces, and 5% in bile, as TMR and metabolites. RTV had no effect on the recovery of radioactivity in any matrix.Compared to FTR alone, pretreatment of subjects with RTV increased the exposure of TMR by ∼66% and reduced the exposure of plasma total radioactivity by ∼68%.The major route of TMR elimination was through biotransformation. TMR, M28 (N-dealkylation), and M4 (amide hydrolysis) were the major circulating components in plasma. Pretreatment with RTV increased the amount of TMR present, decreased the amount of circulating M28, and M4 was unchanged.CYP3A4 metabolism accounted for 21% of the dose, forming multiple oxidative metabolites. This pathway was inhibited by coadministration of RTV.
Collapse
Affiliation(s)
| | - Mindy Magee
- ViiV Healthcare, Research Triangle Park, NC, USA
| | | | | | - Katy Moore
- ViiV Healthcare, Research Triangle Park, NC, USA
| |
Collapse
|
9
|
Sakai Y, Fukami T, Nagaoka M, Hirosawa K, Ichida H, Sato R, Suzuki K, Nakano M, Nakajima M. Arylacetamide deacetylase as a determinant of the hydrolysis and activation of abiraterone acetate in mice and humans. Life Sci 2021; 284:119896. [PMID: 34450168 DOI: 10.1016/j.lfs.2021.119896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/30/2021] [Accepted: 08/07/2021] [Indexed: 11/26/2022]
Abstract
AIM Abiraterone acetate for metastatic castration-resistant prostate cancer is an acetylated prodrug to be hydrolyzed to abiraterone. Abiraterone acetate is known to be hydrolyzed by pancreatic cholesterol esterase secreted into the intestinal lumen. This study aimed to investigate the possibility that arylacetamide deacetylase (AADAC) expressed in enterocytes contributes to the hydrolysis of abiraterone acetate based on its substrate preference. MATERIALS AND METHODS Abiraterone acetate hydrolase activity was measured using human intestinal (HIM) and liver microsomes (HLM) as well as recombinant AADAC. Correlation analysis between activity and AADAC expression was performed in 14 individual HIMs. The in vivo pharmacokinetics of abiraterone acetate was examined using wild-type and Aadac knockout mice administered abiraterone acetate with or without orlistat, a pancreatic cholesterol esterase inhibitor. KEY FINDINGS Recombinant AADAC showed abiraterone acetate hydrolase activity with similar Km value to HIM and HLM. The positive correlation between activity and AADAC levels in individual HIMs supported the responsibility of AADAC for abiraterone acetate hydrolysis. The area under the plasma concentration-time curve (AUC) of abiraterone after oral administration of abiraterone acetate in Aadac knockout mice was 38% lower than that in wild-type mice. The involvement of pancreatic cholesterol esterase in abiraterone formation was revealed by the decreased AUC of abiraterone by coadministration of orlistat. Orlistat potently inhibited AADAC, implying its potential as a perpetrator of drug-drug interactions. SIGNIFICANCE AADAC is responsible for the hydrolysis of abiraterone acetate in the intestine and liver, suggesting that concomitant use of abiraterone acetate and drugs potently inhibiting AADAC should be avoided.
Collapse
Affiliation(s)
- Yoshiyuki Sakai
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute, Kakuma-machi, Kanazawa 920-1192, Japan.
| | - Mai Nagaoka
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Keiya Hirosawa
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Hiroyuki Ichida
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Rei Sato
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Kohei Suzuki
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; WPI Nano Life Science Institute, Kakuma-machi, Kanazawa 920-1192, Japan
| |
Collapse
|
10
|
Schilling U, Dingemanse J, Ufer M. Pharmacokinetics and Pharmacodynamics of Approved and Investigational P2Y12 Receptor Antagonists. Clin Pharmacokinet 2021; 59:545-566. [PMID: 32056160 DOI: 10.1007/s40262-020-00864-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Coronary artery disease remains the major cause of mortality worldwide. Antiplatelet drugs such as acetylsalicylic acid and P2Y12 receptor antagonists are cornerstone treatments for the prevention of thrombotic events in patients with coronary artery disease. Clopidogrel has long been the gold standard but has major pharmacological limitations such as a slow onset and long duration of effect, as well as weak platelet inhibition with high inter-individual pharmacokinetic and pharmacodynamic variability. There has been a strong need to develop potent P2Y12 receptor antagonists with more favorable pharmacological properties. Prasugrel and ticagrelor are more potent and have a faster onset of action; however, they have shown an increased bleeding risk compared with clopidogrel. Cangrelor is highly potent and has a very rapid onset and offset of effect; however, its indication is limited to P2Y12 antagonist-naïve patients undergoing percutaneous coronary intervention. Two novel P2Y12 receptor antagonists are currently in clinical development, namely vicagrel and selatogrel. Vicagrel is an analog of clopidogrel with enhanced and more efficient formation of its active metabolite. Selatogrel is characterized by a rapid onset of action following subcutaneous administration and developed for early treatment of a suspected acute myocardial infarction. This review article describes the clinical pharmacology profile of marketed P2Y12 receptor antagonists and those under development focusing on pharmacokinetic, pharmacodynamic, and drug-drug interaction liability.
Collapse
Affiliation(s)
- Uta Schilling
- Department of Clinical Pharmacology, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, 4123, Allschwil, Switzerland.
| | - Jasper Dingemanse
- Department of Clinical Pharmacology, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Mike Ufer
- Department of Clinical Pharmacology, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| |
Collapse
|
11
|
Fukami T, Yokoi T, Nakajima M. Non-P450 Drug-Metabolizing Enzymes: Contribution to Drug Disposition, Toxicity, and Development. Annu Rev Pharmacol Toxicol 2021; 62:405-425. [PMID: 34499522 DOI: 10.1146/annurev-pharmtox-052220-105907] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Most clinically used drugs are metabolized in the body via oxidation, reduction, or hydrolysis reactions, which are considered phase I reactions. Cytochrome P450 (P450) enzymes, which primarily catalyze oxidation reactions, contribute to the metabolism of over 50% of clinically used drugs. In the last few decades, the function and regulation of P450s have been extensively studied, whereas the characterization of non-P450 phase I enzymes is still incomplete. Recent studies suggest that approximately 30% of drug metabolism is carried out by non-P450 enzymes. This review summarizes current knowledge of non-P450 phase I enzymes, focusing on their roles in controlling drug efficacy and adverse reactions as an important aspect of drug development. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, and WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, and WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| |
Collapse
|
12
|
Honda S, Fukami T, Hirosawa K, Tsujiguchi T, Zhang Y, Nakano M, Uehara S, Uno Y, Yamazaki H, Nakajima M. Differences in Hydrolase Activities in the Liver and Small Intestine between Marmosets and Humans. Drug Metab Dispos 2021; 49:718-728. [PMID: 34135089 DOI: 10.1124/dmd.121.000513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/11/2021] [Indexed: 11/22/2022] Open
Abstract
For drug development, species differences in drug-metabolism reactions present obstacles for predicting pharmacokinetics in humans. We characterized the species differences in hydrolases among humans and mice, rats, dogs, and cynomolgus monkeys. In this study, to expand the series of such studies, we attempted to characterize marmoset hydrolases. We measured hydrolase activities for 24 compounds using marmoset liver and intestinal microsomes, as well as recombinant marmoset carboxylesterase (CES) 1, CES2, and arylacetamide deacetylase (AADAC). The contributions of CES1, CES2, and AADAC to hydrolysis in marmoset liver microsomes were estimated by correcting the activities by using the ratios of hydrolase protein levels in the liver microsomes and those in recombinant systems. For six out of eight human CES1 substrates, the activities in marmoset liver microsomes were lower than those in human liver microsomes. For two human CES2 substrates and three out of seven human AADAC substrates, the activities in marmoset liver microsomes were higher than those in human liver microsomes. Notably, among the three rifamycins, only rifabutin was hydrolyzed by marmoset tissue microsomes and recombinant AADAC. The activities for all substrates in marmoset intestinal microsomes tended to be lower than those in liver microsomes, which suggests that the first-pass effects of the CES and AADAC substrates are due to hepatic hydrolysis. In most cases, the sums of the values of the contributions of CES1, CES2, and AADAC were below 100%, which indicated the involvement of other hydrolases in marmosets. In conclusion, we clarified the substrate preferences of hydrolases in marmosets. SIGNIFICANCE STATEMENT: This study confirmed that there are large differences in hydrolase activities between humans and marmosets by characterizing marmoset hydrolase activities for compounds that are substrates of human CES1, CES2, or arylacetamide deacetylase. The data obtained in this study may be useful for considering whether marmosets are appropriate for examining the pharmacokinetics and efficacies of new chemical entities in preclinical studies.
Collapse
Affiliation(s)
- Shiori Honda
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Keiya Hirosawa
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Takuya Tsujiguchi
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Yongjie Zhang
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Shotaro Uehara
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Yasuhiro Uno
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Hiroshi Yamazaki
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.H., T.F., K.H., T.T., Ma.N., Mi.N.), WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Y.Z., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Y.Z.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Japan (S.U., H.Y.); Central Institute for Experimental Animals, Kawasaki, Japan (S.U.); Shin Nippon Biomedical Laboratories, Ltd., Kainan, Japan (Y.U.); and Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan (Y.U.)
| |
Collapse
|
13
|
Honda S, Fukami T, Tsujiguchi T, Zhang Y, Nakano M, Nakajima M. Hydrolase activities of cynomolgus monkey liver microsomes and recombinant CES1, CES2, and AADAC. Eur J Pharm Sci 2021; 161:105807. [PMID: 33722734 DOI: 10.1016/j.ejps.2021.105807] [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: 09/18/2020] [Revised: 02/12/2021] [Accepted: 03/09/2021] [Indexed: 11/28/2022]
Abstract
The cynomolgus monkey is a nonhuman primate that is often used for pharmacokinetic and toxicokinetic studies of new chemical entities. Species differences in drug metabolism are obstacles for the extrapolation of animal data to humans. This study aimed to characterize hydrolase activities for typical compounds by cynomolgus monkey liver microsomes and recombinant monkey carboxylesterases (CES1 and CES2) and arylacetamide deacetylase (AADAC) compared with the activities in humans. To estimate the contribution of each hydrolase, the ratios of the expression level of each hydrolase in the liver microsomes and recombinant systems were used. For almost all of the tested human CES1 substrates, hydrolase activities in cynomolgus monkey liver microsomes tended to be lower than those in human liver microsomes, and recombinant cynomolgus monkey CES1 showed catalytic activity, but not for all substrates. For human CES2 substrates, hydrolase activities in cynomolgus monkey liver were higher than those in human liver microsomes, and recombinant monkey CES2 was responsible for their hydrolysis. Among human AADAC substrates, phenacetin was mainly hydrolyzed by monkey AADAC, whereas indiplon and ketoconazole were hydrolyzed by AADAC and other unknown enzymes. Flutamide was hydrolyzed by monkey CES2, not by AADAC. Rifamycins were hardly hydrolyzed in monkey liver microsomes. In conclusion, this study characterized the hydrolase activities of cynomolgus monkeys compared with those in humans. The findings would be helpful for pharmacokinetic or toxicokinetic studies of new chemical entities whose main metabolic pathway is hydrolysis.
Collapse
Affiliation(s)
- Shiori Honda
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan.
| | - Takuya Tsujiguchi
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yongjie Zhang
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| |
Collapse
|
14
|
Chen KJ, Plaunt AJ, Leifer FG, Kang JY, Cipolla D. Recent advances in prodrug-based nanoparticle therapeutics. Eur J Pharm Biopharm 2021; 165:219-243. [PMID: 33979661 DOI: 10.1016/j.ejpb.2021.04.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 03/10/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022]
Abstract
Extensive research into prodrug modification of active pharmaceutical ingredients and nanoparticle drug delivery systems has led to unprecedented levels of control over the pharmacological properties of drugs and resulted in the approval of many prodrug or nanoparticle-based therapies. In recent years, the combination of these two strategies into prodrug-based nanoparticle drug delivery systems (PNDDS) has been explored as a way to further advance nanomedicine and identify novel therapies for difficult-to-treat indications. Many of the PNDDS currently in the clinical development pipeline are expected to enter the market in the coming years, making the rapidly evolving field of PNDDS highly relevant to pharmaceutical scientists. This review paper is intended to introduce PNDDS to the novice reader while also updating those working in the field with a comprehensive summary of recent efforts. To that end, first, an overview of FDA-approved prodrugs is provided to familiarize the reader with their advantages over traditional small molecule drugs and to describe the chemistries that can be used to create them. Because this article is part of a themed issue on nanoparticles, only a brief introduction to nanoparticle-based drug delivery systems is provided summarizing their successful application and unfulfilled opportunities. Finally, the review's centerpiece is a detailed discussion of rationally designed PNDDS formulations in development that successfully leverage the strengths of prodrug and nanoparticle approaches to yield highly effective therapeutic options for the treatment of many diseases.
Collapse
|
15
|
Hirosawa K, Fukami T, Tashiro K, Sakai Y, Kisui F, Nakano M, Nakajima M. Role of Human Arylacetamide Deacetylase (AADAC) on Hydrolysis of Eslicarbazepine Acetate and Effects of AADAC Genetic Polymorphisms on Hydrolase Activity. Drug Metab Dispos 2021; 49:322-329. [PMID: 33446525 DOI: 10.1124/dmd.120.000295] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/07/2021] [Indexed: 02/13/2025] Open
Abstract
Human arylacetamide deacetylase (AADAC) plays a role in the detoxification or activation of drugs and is sometimes involved in the incidence of toxicity by catalyzing hydrolysis reactions. AADAC prefers compounds with relatively small acyl groups, such as acetyl groups. Eslicarbazepine acetate, an antiepileptic drug, is a prodrug rapidly hydrolyzed to eslicarbazepine. We sought to clarify whether AADAC might be responsible for the hydrolysis of eslicarbazepine acetate. Eslicarbazepine acetate was efficiently hydrolyzed by human intestinal and liver microsomes and recombinant human AADAC. The hydrolase activities in human intestinal and liver microsomes were inhibited by epigallocatechin gallate, a specific inhibitor of AADAC, by 82% and 88% of the control, respectively. The hydrolase activities in liver microsomes from 25 human livers were significantly correlated (r = 0.87, P < 0.001) with AADAC protein levels, suggesting that the enzyme AADAC is responsible for the hydrolysis of eslicarbazepine acetate. The effects of genetic polymorphisms of AADAC on eslicarbazepine acetate hydrolysis were examined by using the constructed recombinant AADAC variants with T74A, V172I, R248S, V281I, N366K, or X400Q. AADAC variants with R248S or X400Q showed lower activity than wild type (5% or 21%, respectively), whereas those with V172I showed higher activity than wild type (174%). Similar tendencies were observed in the other four substrates of AADAC; that is, p-nitrophenyl acetate, ketoconazole, phenacetin, and rifampicin. Collectively, we found that eslicarbazepine acetate is specifically and efficiently hydrolyzed by human AADAC, and several AADAC polymorphic alleles would be a factor affecting the enzyme activity and drug response. SIGNIFICANCE STATEMENT: This is the first study to clarify that arylacetamide deacetylase (AADAC) is responsible for the activation of eslicarbazepine acetate, an antiepileptic prodrug, to eslicarbazepine, an active form, in the human liver and intestines. In addition, we found that several AADAC polymorphic alleles would be a factor affecting the enzyme activity and drug response.
Collapse
Affiliation(s)
- Keiya Hirosawa
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.H., T.F., K.T., Y.S., F.K., Ma.N., Mi.N.); and WPI Nano Life Science Institute, Kakuma-machi, Kanazawa, Japan (T.F., Ma.N., Mi.N.)
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.H., T.F., K.T., Y.S., F.K., Ma.N., Mi.N.); and WPI Nano Life Science Institute, Kakuma-machi, Kanazawa, Japan (T.F., Ma.N., Mi.N.)
| | - Kiyomichi Tashiro
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.H., T.F., K.T., Y.S., F.K., Ma.N., Mi.N.); and WPI Nano Life Science Institute, Kakuma-machi, Kanazawa, Japan (T.F., Ma.N., Mi.N.)
| | - Yoshiyuki Sakai
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.H., T.F., K.T., Y.S., F.K., Ma.N., Mi.N.); and WPI Nano Life Science Institute, Kakuma-machi, Kanazawa, Japan (T.F., Ma.N., Mi.N.)
| | - Fumiya Kisui
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.H., T.F., K.T., Y.S., F.K., Ma.N., Mi.N.); and WPI Nano Life Science Institute, Kakuma-machi, Kanazawa, Japan (T.F., Ma.N., Mi.N.)
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.H., T.F., K.T., Y.S., F.K., Ma.N., Mi.N.); and WPI Nano Life Science Institute, Kakuma-machi, Kanazawa, Japan (T.F., Ma.N., Mi.N.)
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.H., T.F., K.T., Y.S., F.K., Ma.N., Mi.N.); and WPI Nano Life Science Institute, Kakuma-machi, Kanazawa, Japan (T.F., Ma.N., Mi.N.)
| |
Collapse
|
16
|
Sun HZ, Qin GQ, Wang FG, Bai Y, Zhang Z, Fang ZZ. Hydroxylated polychlorinated biphenyls (OH-PCBs) exert strong inhibitory effects towards human carboxylesterases (CESs). THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 745:141140. [PMID: 32736114 DOI: 10.1016/j.scitotenv.2020.141140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/12/2020] [Accepted: 07/19/2020] [Indexed: 06/11/2023]
Abstract
Polychlorinated biphenyls (PCBs) have been reported to pose a severe risk towards human health, and hydroxylated polychlorinated biphenyls (OH-PCBs) were potential substances basis for PCBs' toxicity. This study aims to determine the inhibition of OH-PCBs towards human carboxylesterases (CESs), including CES1 and CES2. For phenotypic analysis of CES1 and CES2 activity, we used the hydrolysis metabolism of 2-(2-benzoyl3-methoxyphenyl) benzothiazole (BMBT) and fluorescein diacetate (FD) catalyzed by human liver microsomes (HLMs) as the probe reactions. Preliminary inhibition screening showed that the inhibition potential of OH-PCBs towards CES1 and CES2 increased with the increased numbers of chlorine atoms in OH-PCBs. Both 2'-OH-PCB61 and 2'-OH-PCB65 showed concentration-dependent inhibition towards both CES1 and CES2. Lineweaver-Burk plots showed that 2'-OH-PCB61 and 2'-OH-PCB65 exerted non-competitive inhibition towards CES1 and competitive inhibition towards CES2. The inhibition kinetics parameters (Ki) were 6.8 μM and 7.0 μM for 2'-OH-PCB61 and 2'-OH-PCB65 towards CES1, respectively. The inhibition kinetics parameters (Ki) were 1.4 μM and 1.0 μM for 2'-OH-PCB61 and 2'-OH-PCB65 towards CES2, respectively. In silico docking methods elucidate the contribution of hydrogen bonds and hydrophobic contacts towards the binding of 2'-OH-PCB61 and 2'-OH-PCB65 with CES1 and CES2. All these results will provide a new perspective for elucidation of toxicity mechanism of PCBs and OH-PCBs.
Collapse
Affiliation(s)
- Hong-Zhi Sun
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.
| | - Guo-Qiang Qin
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Fei-Ge Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Yu Bai
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Zhipeng Zhang
- General Surgery Department, Peking University Third Hospital, Beijing, 100191, China
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China.
| |
Collapse
|
17
|
Liu YZ, Pan LH, Bai Y, Yang K, Dong PP, Fang ZZ. Per- and polyfluoroalkyl substances exert strong inhibition towards human carboxylesterases. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 263:114463. [PMID: 32283456 DOI: 10.1016/j.envpol.2020.114463] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/11/2020] [Accepted: 03/24/2020] [Indexed: 06/11/2023]
Abstract
PFASs are highly persistent in both natural and living environment, and pose a significant risk for wildlife and human beings. The present study was carried out to determine the inhibitory behaviours of fourteen PFASs on metabolic activity of two major isoforms of carboxylesterases (CES). The probe substrates 2-(2-benzoyl-3-methoxyphenyl) benzothiazole (BMBT) for CES1 and fluorescein diacetate (FD) for CES2 were utilized to determine the inhibitory potentials of PFASs on CES in vitro. The results demonstrated that perfluorododecanoic acid (PFDoA), perfluorotetradecanoic acid (PFTA) and perfluorooctadecanoic acid (PFOcDA) strongly inhibited CES1 and CES2. The half inhibition concentration (IC50) value of PFDoA, PFTA and PFOcDA for CES1 inhibition was 10.6 μM, 13.4 μM and 12.6 μM, respectively. The IC50 for the inhibition of PFDoA, PFTA and PFOcDA towards CES2 were calculated to be 9.56 μM, 17.2 μM and 8.73 μM, respectively. PFDoA, PFTA and PFOcDA exhibited noncompetitive inhibition towards both CES1 and CES2. The inhibition kinetics parameters (Ki) were 27.7 μM, 26.9 μM, 11.9 μM, 4.04 μM, 29.1 μM, 27.4 μM for PFDoA-CES1, PFTA-CES1, PFOcDA-CES1, PFDoA-CES2, PFTA-CES2, PFOcDA-CES2, respectively. In vitro-in vivo extrapolation (IVIVE) predicted that when the plasma concentrations of PFDoA, PFTA and PFOcDA were greater than 2.77 μM, 2.69 μM and 1.19 μM, respectively, it might interfere with the metabolic reaction catalyzed by CES1 in vivo; when the plasma concentrations of PFDoA, PFTA and PFOcDA were greater than 0.40 μM, 2.91 μM, 2.74 μM, it might interfere with the metabolic reaction catalyzed by CES2 in vivo. Molecular docking was used to explore the interactions between PFASs and CES. In conclusion, PFASs were found to cause inhibitory effects on CES in vitro, and this finding would provide an important experimental basis for further in vivo testing of PFASs focused on CES inhibition endpoints.
Collapse
Affiliation(s)
- Yong-Zhe Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China
| | - Li-Hua Pan
- Department of Pharmacy, Tianjin Xiqing Hospital, Tianjin, 300000, China
| | - Yu Bai
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Kun Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China
| | - Pei-Pei Dong
- College of Pharmacy, College (Institute) of Integrative Medicine, Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, 300070, China; National Demonstration Center for Experimental Preventive Medicine Education, Tianjin Medical University, Tianjin, 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China.
| |
Collapse
|
18
|
Nishiya Y, Suzuki E, Ishizuka T, Kazui M, Sakurai H, Nakai D. Identification of non-P450 enzymes involved in the metabolism of new drugs: Their significance in drug interaction evaluation and prodrug disposition. Drug Metab Pharmacokinet 2020; 35:45-55. [PMID: 31926835 DOI: 10.1016/j.dmpk.2019.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 09/29/2019] [Accepted: 11/02/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Yumi Nishiya
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd, Tokyo, Japan.
| | - Eiko Suzuki
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd, Tokyo, Japan
| | - Tomoko Ishizuka
- Clinical Pharmacology Department, Daiichi Sankyo Co., Ltd, Tokyo, Japan
| | - Miho Kazui
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd, Tokyo, Japan
| | - Hidetaka Sakurai
- General Administration Department, Daiichi Sankyo RD Novare Co., Ltd, Tokyo, Japan
| | - Daisuke Nakai
- Biomarker & Translational Research Department, Daiichi Sankyo Co., Ltd, Tokyo, Japan
| |
Collapse
|
19
|
Strain and sex differences in drug hydrolase activities in rodent livers. Eur J Pharm Sci 2020; 142:105143. [DOI: 10.1016/j.ejps.2019.105143] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/07/2019] [Accepted: 11/09/2019] [Indexed: 01/07/2023]
|
20
|
Li Y, Meng Q, Yang M, Liu D, Hou X, Tang L, Wang X, Lyu Y, Chen X, Liu K, Yu AM, Zuo Z, Bi H. Current trends in drug metabolism and pharmacokinetics. Acta Pharm Sin B 2019; 9:1113-1144. [PMID: 31867160 PMCID: PMC6900561 DOI: 10.1016/j.apsb.2019.10.001] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/23/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
Pharmacokinetics (PK) is the study of the absorption, distribution, metabolism, and excretion (ADME) processes of a drug. Understanding PK properties is essential for drug development and precision medication. In this review we provided an overview of recent research on PK with focus on the following aspects: (1) an update on drug-metabolizing enzymes and transporters in the determination of PK, as well as advances in xenobiotic receptors and noncoding RNAs (ncRNAs) in the modulation of PK, providing new understanding of the transcriptional and posttranscriptional regulatory mechanisms that result in inter-individual variations in pharmacotherapy; (2) current status and trends in assessing drug-drug interactions, especially interactions between drugs and herbs, between drugs and therapeutic biologics, and microbiota-mediated interactions; (3) advances in understanding the effects of diseases on PK, particularly changes in metabolizing enzymes and transporters with disease progression; (4) trends in mathematical modeling including physiologically-based PK modeling and novel animal models such as CRISPR/Cas9-based animal models for DMPK studies; (5) emerging non-classical xenobiotic metabolic pathways and the involvement of novel metabolic enzymes, especially non-P450s. Existing challenges and perspectives on future directions are discussed, and may stimulate the development of new research models, technologies, and strategies towards the development of better drugs and improved clinical practice.
Collapse
Affiliation(s)
- Yuhua Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
- The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qiang Meng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Mengbi Yang
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Xiangyu Hou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lan Tang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xin Wang
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuanfeng Lyu
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ai-Ming Yu
- UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Zhong Zuo
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Huichang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
21
|
Laizure SC, Parker RB. Is genetic variability in carboxylesterase-1 and carboxylesterase-2 drug metabolism an important component of personalized medicine? Xenobiotica 2019; 50:92-100. [DOI: 10.1080/00498254.2019.1678078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- S. Casey Laizure
- Department of Clinical Pharmacy & Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Robert B Parker
- Department of Clinical Pharmacy & Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
22
|
Gabriele M, Puccini P, Lucchi M, Aprile V, Gervasi PG, Longo V. Arylacetamide Deacetylase Enzyme: Presence and Interindividual Variability in Human Lungs. Drug Metab Dispos 2019; 47:961-965. [PMID: 31235486 DOI: 10.1124/dmd.119.087031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
Human arylacetamide deacetylase (AADAC) is a single microsomal serine esterase involved in the hydrolysis of many acetyl-containing drugs. To date, the presence and activity of the AADAC enzyme in human lungs has been scarcely examined. We investigated its gene and protein expression as well as interindividual variations in AADAC activities in a large number of human lungs (n = 25) using phenacetin as a selective substrate. The kinetic parameters K m and V max were determined. Our findings highlighted a high interindividual variability in both AADAC mRNA levels and hydrolysis activities. Furthermore, for the first time we demonstrated the presence of the AADAC protein in various lung samples by means of immunoblot analysis. As a comparison, phenacetin hydrolysis was detected in pooled human liver microsomes. Lung activities were much lower than those found in the liver. However, similar K m values were found, which suggests that this hydrolysis could be due to the same enzyme. Pulmonary phenacetin hydrolysis proved to be positively correlated with AADAC mRNA (*P < 0.05) and protein (*P < 0.05) levels. Moreover, the average values of AADAC activity in smokers was significantly higher than in nonsmoker subjects (*P < 0.05), and this might have an important role in the administration of some drugs. These findings add more information to our knowledge of pulmonary enzymes and could be particularly useful in the design and preclinical development of inhaled drugs. SIGNIFICANCE STATEMENT: This study investigated the presence and activity of the AADAC enzyme in several human lungs. Our results highlight high interindividual variability in both AADAC gene and protein expression as well as in phenacetin hydrolysis activity. These findings add more information to our knowledge of pulmonary enzymes and could be particularly useful in the design and preclinical development of inhaled drugs.
Collapse
Affiliation(s)
- Morena Gabriele
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Paola Puccini
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Marco Lucchi
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Vittorio Aprile
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Pier Giovanni Gervasi
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| | - Vincenzo Longo
- National Research Council, Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Pisa, Italy (M.G., P.G.G., V.L.); Chiesi Farmaceutici S.p.A., Parma, Italy (P.P.); and Division of Thoracic Surgery, Department of Surgical Medical Molecular Pathology and Critical Care, University Hospital of Pisa, Pisa, Italy (M.L., V.A.)
| |
Collapse
|
23
|
Li X, Sun J, Guo Z, Zhong D, Chen X. Carboxylesterase 2 and Intestine Transporters Contribute to the Low Bioavailability of Allisartan, a Prodrug of Exp3174 for Hypertension Treatment in Humans. Drug Metab Dispos 2019; 47:843-853. [PMID: 31076412 DOI: 10.1124/dmd.118.085092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 05/08/2019] [Indexed: 02/13/2025] Open
Abstract
Exp3174 is an active metabolite of losartan for the treatment of hypertension. Allisartan (ALS3) is a marketed ester prodrug of Exp3174 to reduce bioavailability variation of losartan in China. However, ALS3 exhibited a lower oral absorption than losartan in humans. In this study, the enzymes and transporters involved in ALS3 and Exp3174 disposition were investigated to clarify the mechanisms. ALS3 underwent extensive hydrolysis to Exp3174 in S9 of Caco-2 cells, human intestine microsomes (HIM), recombinant carboxylesterase (rCES) 1, and rCES2. ALS3 exhibited similar affinity in HIM and rCES2 with K m values of 6.92 and 6.77 μM, respectively, indicating that ALS3 is mainly hydrolyzed to Exp3174 in human intestine by CES2. Transport assays of ALS3 and Exp3174 suggested that ALS3 and Exp3174 are substrates of P-glycoprotein, breast cancer resistance protein, and multidrug resistance protein 2 with poor permeability. Organic anion-transporting polypeptide 2B1 showed higher affinity and clearance toward ALS3 (K m 0.75 μM and intrinsic clearance 215 μl/min/mg) than those of Exp3174 (K m 7.85 μM and intrinsic clearance 16.1 μl/min/mg), indicating that ALS3 is preferred to be uptaken into intestinal epithelia. Hydrolysis of ALS3 was increased from approximately 30% to 55% in CES2-transfected human embryonic kidney 293-OATP2B1 cells, indicating the possible interplay between OATP2B1 and CES2. The influx and efflux of ALS3 across Caco-2 cells increased the potential of ALS3 hydrolysis to Exp3174, and the produced Exp3174 was rapidly pumped out, which led to undetectable ALS3 and Exp3174 in basolateral (receiver) side in Caco-2 cells. Overall, our study provided supportive evidences that the interplay between CES2 and transporters in intestine contributes to the low bioavailability of ALS3 in humans.
Collapse
Affiliation(s)
- Xiuli Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.L., Z.G., D.Z., X.C.); and Shenzhen Salubris Pharmaceutical, Guangdong, China (J.S.)
| | - Jingchao Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.L., Z.G., D.Z., X.C.); and Shenzhen Salubris Pharmaceutical, Guangdong, China (J.S.)
| | - Zitao Guo
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.L., Z.G., D.Z., X.C.); and Shenzhen Salubris Pharmaceutical, Guangdong, China (J.S.)
| | - Dafang Zhong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.L., Z.G., D.Z., X.C.); and Shenzhen Salubris Pharmaceutical, Guangdong, China (J.S.)
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.L., Z.G., D.Z., X.C.); and Shenzhen Salubris Pharmaceutical, Guangdong, China (J.S.)
| |
Collapse
|
24
|
Vicagrel enhances aspirin-induced inhibition of both platelet aggregation and thrombus formation in rodents due to its decreased metabolic inactivation. Biomed Pharmacother 2019; 115:108906. [DOI: 10.1016/j.biopha.2019.108906] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/12/2019] [Accepted: 04/22/2019] [Indexed: 02/07/2023] Open
|
25
|
Fu Q, Yang K, Hu RX, Du Z, Hu CM, Zhang X. Evaluation of the inhibition of human carboxylesterases (CESs) by the active ingredients from Schisandra chinensis. Xenobiotica 2019; 49:1260-1268. [PMID: 30486721 DOI: 10.1080/00498254.2018.1548718] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Qiang Fu
- Department of Cardiac Surgery, The General Hospital of Tianjin Medical University, Tianjin, China
| | - Kai Yang
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Rui-Xia Hu
- National Demonstration Center for Experimental Preventive Medicine Education (Tianjin Medical University), Tianjin, China
| | - Zuo Du
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Cui-Min Hu
- Tianjin Life Science Research Center, Department of Microbiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xibo Zhang
- Department of Hepatopancreatobiliary Surgery, Tianjin Nankai Hospital, Tianjin, China
| |
Collapse
|
26
|
Liu C, Zhang Y, Chen W, Lu Y, Li W, Liu Y, Lai X, Gong Y, Liu X, Li Y, Chen X, Li X, Sun H, Yang J, Zhong D. Pharmacokinetics and pharmacokinetic/pharmacodynamic relationship of vicagrel, a novel thienopyridine P2Y12 inhibitor, compared with clopidogrel in healthy Chinese subjects following single oral dosing. Eur J Pharm Sci 2019; 127:151-160. [DOI: 10.1016/j.ejps.2018.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 08/31/2018] [Accepted: 10/11/2018] [Indexed: 12/18/2022]
|
27
|
Máchal J, Hlinomaz O. Efficacy of P2Y12 Receptor Blockers After Myocardial Infarction and Genetic Variability of their Metabolic Pathways. Curr Vasc Pharmacol 2018; 17:35-40. [DOI: 10.2174/1570161116666180206110657] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/18/2017] [Accepted: 11/07/2017] [Indexed: 01/15/2023]
Abstract
Background: Various antiplatelet drugs are used following Acute Coronary Syndromes
(ACS). Of them, adenosine diphosphate receptor P2Y12 inhibitors clopidogrel, prasugrel and ticagrelor
are currently used for post-ACS long-term treatment. Although they act on the same receptor, they differ
in pharmacodynamics and pharmacokinetics. Several enzymes and transporters involved in the metabolism
of P2Y12 inhibitors show genetic variability with functional impact. This includes Pglycoprotein,
carboxylesterase 1 and, most notably, CYP2C19 that is important in clopidogrel activation.
Common gain-of-function or loss-of-function alleles of CYP2C19 gene are associated with lower
or higher platelet reactivity that may impact clinical outcomes of clopidogrel treatment. Prasugrel is
considered to be less dependent on CYP2C19 variability as it is also metabolized by other CYP450 isoforms.
Some studies, however, showed the relevance of CYP2C19 variants for platelet reactivity during
prasugrel treatment as well. Ticagrelor is metabolized mainly by CYP3A4, which does not show functionally
relevant genetic variability. Its concentrations may be modified by the variants of Pglycoprotein
gene ABCB1. While no substantial difference between the clinical efficacy of prasugrel
and ticagrelor has been documented, both of them have been shown to be superior to clopidogrel in
post-ACS treatment. This can be partially explained by lower variability at each step of their metabolism.
It is probable that factors influencing the pharmacokinetics of both drugs, including genetic factors,
may predict the clinical efficacy of antiplatelet treatment in personalized medicine.
</P><P>
Conclusion: We summarize the pharmacokinetics and pharmacogenetics of P2Y12 inhibitors with respect
to their clinical effects in post-myocardial infarction treatment.
Collapse
Affiliation(s)
- Jan Máchal
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Ota Hlinomaz
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
28
|
Imai S, Ichikawa T, Sugiyama C, Nonaka K, Yamada T. Contribution of Human Liver and Intestinal Carboxylesterases to the Hydrolysis of Selexipag In Vitro. J Pharm Sci 2018; 108:1027-1034. [PMID: 30267780 DOI: 10.1016/j.xphs.2018.09.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 09/07/2018] [Accepted: 09/14/2018] [Indexed: 02/01/2023]
Abstract
In liver microsomes, selexipag (NS-304; ACT-293987) mainly undergoes hydrolytic removal of the sulfonamide moiety by carboxylesterase 1 (CES1) to yield the pharmacologically active metabolite MRE-269 (ACT-333679). However, it is not known how much CES in the liver and intestine contributes to the hydrolysis of selexipag or how selexipag is metabolized in the intestine, including by hydrolysis. To obtain a better understanding of selexipag metabolism in humans, we determined the percentage contribution of CES1 and carboxylesterase 2 (CES2) to the hydrolysis of selexipag and 7 of its analogs with different sulfonamide moieties and evaluated its nonhydrolytic metabolism in human liver microsomes and human intestinal microsomes (HIMS). For selexipag, the percentage contributions of CES1 and CES2 in human liver microsomes were 77.0% and 9.99%, respectively, while the percentage contribution of CES2 in HIMS was 100%. In HIMS, the rate of hydrolysis of selexipag was the lowest among the compounds tested, and no difference between the presence and absence of nicotinamide adenine dinucleotide phosphate was noted. We infer from these results that selexipag is likely to be hydrolyzed by CES2 as well as CES1, and only selexipag itself and the MRE-269 produced by hydrolysis in the intestine would be absorbed after oral administration.
Collapse
Affiliation(s)
- Shunji Imai
- Pharmacokinetics and Safety Assessment Department, Discovery Research Laboratories, Nippon Shinyaku Co., Ltd, Kyoto, Japan.
| | - Tomohiko Ichikawa
- Pharmacokinetics and Safety Assessment Department, Discovery Research Laboratories, Nippon Shinyaku Co., Ltd, Kyoto, Japan
| | - Chihiro Sugiyama
- Pharmacokinetics and Safety Assessment Department, Discovery Research Laboratories, Nippon Shinyaku Co., Ltd, Kyoto, Japan
| | - Kiyoko Nonaka
- Pharmacokinetics and Safety Assessment Department, Discovery Research Laboratories, Nippon Shinyaku Co., Ltd, Kyoto, Japan
| | - Tetsuhiro Yamada
- Pharmacokinetics and Safety Assessment Department, Discovery Research Laboratories, Nippon Shinyaku Co., Ltd, Kyoto, Japan
| |
Collapse
|
29
|
Jiang J, Chen X, Zhong D. Arylacetamide Deacetylase Is Involved in Vicagrel Bioactivation in Humans. Front Pharmacol 2017; 8:846. [PMID: 29209217 PMCID: PMC5701912 DOI: 10.3389/fphar.2017.00846] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/06/2017] [Indexed: 11/26/2022] Open
Abstract
Vicagrel, a structural analog of clopidogrel, is now being developed as a thienopyridine antiplatelet agent in a phase II clinical trial in China. Some studies have shown that vicagrel undergoes complete first-pass metabolism in human intestine, generating the hydrolytic metabolite 2-oxo-clopidogrel via carboxylesterase-2 (CES2) and subsequently the active metabolite H4 via CYP450s. This study aimed to identify hydrolases other than CES2 that are involved in the bioactivation of vicagrel in human intestine. This study is the first to determine that human arylacetamide deacetylase (AADAC) is involved in 2-oxo-clopidogrel production from vicagrel in human intestine. In vitro hydrolytic kinetics were determined in human intestine microsomes and recombinant human CES and AADAC. The calculated contribution of CES2 and AADAC to vicagrel hydrolysis was 44.2 and 53.1% in human intestine, respectively. The AADAC-selective inhibitors vinblastine and eserine effectively inhibited vicagrel hydrolysis in vitro. In addition to CES2, human intestine AADAC was involved in vicagrel hydrolytic activation before it entered systemic circulation. In addition, simvastatin efficiently inhibited the production of both 2-oxo-clopidogrel and active H4; further clinical trials are needed to determine whether the hydrolytic activation of vicagrel is influenced by coadministration with simvastatin. This study deepens the understanding of the bioactivation and metabolism properties of vicagrel in humans, which can help further understand the bioactivation mechanism of vicagrel and the variations in the treatment responses to vicagrel and clopidogrel.
Collapse
Affiliation(s)
- Jinfang Jiang
- State Key Laboratory of Drug Research, Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Chen
- State Key Laboratory of Drug Research, Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Dafang Zhong
- State Key Laboratory of Drug Research, Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
30
|
Yoshida T, Fukami T, Kurokawa T, Gotoh S, Oda A, Nakajima M. Difference in substrate specificity of carboxylesterase and arylacetamide deacetylase between dogs and humans. Eur J Pharm Sci 2017; 111:167-176. [PMID: 28966098 DOI: 10.1016/j.ejps.2017.09.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/11/2017] [Accepted: 09/25/2017] [Indexed: 01/09/2023]
Abstract
Carboxylesterase (CES) and arylacetamide deacetylase (AADAC) are the major enzymes responsible for the hydrolysis of various clinical drugs. Our recent study demonstrated that the identity of the responsible hydrolase can be roughly surmised based on the chemical structures of compounds in humans. Dogs are used for preclinical studies in drug development, but the substrate specificities of dog CES and AADAC remain to be clarified. The purpose of this study is to characterize their substrate specificities. We prepared recombinant dog CES1, CES2, and AADAC. p-Nitrophenyl acetate, a general substrate for esterases, was hydrolyzed by dog CES1 and AADAC, while it was not hydrolyzed by CES2. CES2 protein was not substantially detected in the recombinant system or in the dog liver and intestinal microsomes by Western blot using anti-human CES2 antibodies. In silico analyses demonstrated slight differences in the three-dimensional structures of dog CES2 and human CES2, indicating that dog CES2 might be unstable or inactive. By evaluating the hydrolase activities of 22 compounds, which are known to be substrates of human CES and/or AADAC, we found that the activities of dog recombinant CES1 and AADAC as well as dog tissue preparations for nearly all compounds were lower than those of human enzymes. The dog enzymes that were responsible for the hydrolysis of most compounds corresponded to the human enzymes, but the following differences were observed: oseltamivir, irinotecan, and rifampicin were not hydrolyzed in the dog liver or by any of the recombinant esterases and procaine, a human CES2 substrate, was hydrolyzed by dog CES1. In conclusion, the present study could provide new finding to facilitate our understanding of species differences in drug hydrolysis, which can facilitate drug development and drug safety evaluation.
Collapse
Affiliation(s)
- Tomohiro Yoshida
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| | - Takaya Kurokawa
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Saki Gotoh
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Akifumi Oda
- Biophysical Chemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| |
Collapse
|
31
|
Identification of enzymes responsible for nitrazepam metabolism and toxicity in human. Biochem Pharmacol 2017; 140:150-160. [DOI: 10.1016/j.bcp.2017.06.114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/07/2017] [Indexed: 12/16/2022]
|
32
|
Wang DD, Zou LW, Jin Q, Hou J, Ge GB, Yang L. Recent progress in the discovery of natural inhibitors against human carboxylesterases. Fitoterapia 2017; 117:84-95. [DOI: 10.1016/j.fitote.2017.01.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/13/2017] [Accepted: 01/21/2017] [Indexed: 01/22/2023]
|