1
|
Liu Y, Li J, Zhu HJ. Regulation of carboxylesterases and its impact on pharmacokinetics and pharmacodynamics: an up-to-date review. Expert Opin Drug Metab Toxicol 2024; 20:377-397. [PMID: 38706437 PMCID: PMC11151177 DOI: 10.1080/17425255.2024.2348491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
INTRODUCTION Carboxylesterase 1 (CES1) and carboxylesterase 2 (CES2) are among the most abundant hydrolases in humans, catalyzing the metabolism of numerous clinically important medications, such as methylphenidate and clopidogrel. The large interindividual variability in the expression and activity of CES1 and CES2 affects the pharmacokinetics (PK) and pharmacodynamics (PD) of substrate drugs. AREAS COVERED This review provides an up-to-date overview of CES expression and activity regulations and examines their impact on the PK and PD of CES substrate drugs. The literature search was conducted on PubMed from inception to January 2024. EXPERT OPINION Current research revealed modest associations of CES genetic polymorphisms with drug exposure and response. Beyond genomic polymorphisms, transcriptional and posttranslational regulations can also significantly affect CES expression and activity and consequently alter PK and PD. Recent advances in plasma biomarkers of drug-metabolizing enzymes encourage the research of plasma protein and metabolite biomarkers for CES1 and CES2, which could lead to the establishment of precision pharmacotherapy regimens for drugs metabolized by CESs. Moreover, our understanding of tissue-specific expression and substrate selectivity of CES1 and CES2 has shed light on improving the design of CES1- and CES2-activated prodrugs.
Collapse
Affiliation(s)
- Yaping Liu
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan
| | - Jiapeng Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Hao-Jie Zhu
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
2
|
Gao H, Yang N, Yang L, Wang H, Zhang G, Ma X, Deng N. Advances and Perspectives in methods for identifying high platelet reactivity. Heliyon 2023; 9:e22214. [PMID: 38107326 PMCID: PMC10724541 DOI: 10.1016/j.heliyon.2023.e22214] [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: 05/08/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 12/19/2023] Open
Abstract
Antiplatelet therapy is the foundational treatment for the prevention and treatment of coronary and cerebrovascular ischemic events in patients with coronary heart disease, ischemic stroke, and transient ischemic attack (TIA). However, with more and more studies reporting an increased risk of thrombosis in some patients due to poor response to therapeutic agents, the selection of appropriate P2Y12 inhibitors has become a major challenge that needs to be addressed urgently. Currently, commonly used oral P2Y12 inhibitors include clopidogrel, ticagrelor, and prasugrel. Assessing patients' risk factors before the development of treatment regimens by effectively predicting the risk of high platelet reactivity with specific P2Y12 inhibitors in advance to avert the occurrence of major adverse cardiovascular and cerebrovascular events (MACCE) is the key point to the problem. Up to now, methods available for predicting platelet reactivity include genetic testing, platelet function testing, and risk scores. This review provides a summarization of the existent available identification methods and analyzes the advantages and drawbacks of different methods in specific clinical settings, intending to guide the rational clinical application of P2Y12 receptor inhibitors.
Collapse
Affiliation(s)
- Hua Gao
- Department of Pharmacy, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Nan Yang
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Libo Yang
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Hui Wang
- Department of Pharmacy, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Guoshan Zhang
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Xueping Ma
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Ning Deng
- Office of Drug Clinical Trial Organization, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| |
Collapse
|
3
|
Luo X, Lu F, Yin Z, Zhou Z, Wang Z, Zhang H. Hormetic effects of EGC and EGCG on CES1 activity and its rescue from oxidative stress in rat liver S9. Chem Biol Interact 2023; 382:110612. [PMID: 37353134 DOI: 10.1016/j.cbi.2023.110612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/17/2023] [Accepted: 06/21/2023] [Indexed: 06/25/2023]
Abstract
Carboxylesterase 1 (CES1) is a hydrolytic enzyme that plays an important role in the activation or deactivation of many therapeutic agents, thus affecting their pharmacokinetic and pharmacodynamic outcomes. Using rat liver S9 as an enzyme source and enalapril as a CES1 substrate, the present study examined effects of a number of flavonoids on the formation of enalaprilat (the active form of enalapril) produced by CES1-mediated hydrolysis. While a majority of flavonoids tested showed inhibition on CES1, an unexpected hormetic effect was observed for epigallocatechin (EGC) and epigallocatechin gallate (EGCG), i.e., stimulatory effect at low concentrations and enzyme inhibition at high concentrations. Further experiments revealed that oxidative stress caused by hydrogen peroxide, arachidonic acid plus iron, and oxidized low density lipoproteins (oxLOL) reduced CES1 activity in rat liver S9 and the loss of CES1 enzyme activity could be rescued largely by EGC or EGCG. In contrast, such effects were minimal in human liver S9, probably due to the presence of a higher ratio of reduced vs oxidized forms of glutathione. The above findings suggest that the polyphenolic nature of EGC or EGCG might be responsible for rescuing CES1 activity under oxidative stress. Because of the importance of CES1 in drug activation or deactivation and rat liver S9 as a versatile in vitro system used for drug metabolism studies and drug safety assessment, caution should be exercised to avoid potential biases for data interpretation and decision making when CES1 activity in rat liver S9 is evaluated with dependency on experimental conditions.
Collapse
Affiliation(s)
- Xiaoting Luo
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Feifei Lu
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Zhiyue Yin
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Zhiyun Zhou
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Zhongmin Wang
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Hongjian Zhang
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China.
| |
Collapse
|
4
|
Luo HY, Gao LC, Long HZ, Zhou ZW, Xu SG, Li FJ, Li HL, Cheng Y, Li CX, Peng XY, Li L, Chen R, Deng P. Association between the NEP rs701109 polymorphism and the clinical efficacy and safety of sacubitril/valsartan in Chinese patients with heart failure. Eur J Clin Pharmacol 2023; 79:663-670. [PMID: 36976322 DOI: 10.1007/s00228-023-03484-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
OBJECTIVE Sacubitril/valsartan is a commonly used medicine for treating heart failure (HF) patients, but the treatment effects significantly vary. Neprilysin (NEP) and carboxylesterase 1 (CES1) play an important role in the efficacy of sacubitril/valsartan. The purpose of this study was to explore the relationship between NEP and CES1 gene polymorphisms and the efficacy and safety of sacubitril/valsartan treatment in HF patients. METHODS Genotyping of 10 single nucleotide polymorphisms (SNPs) of the NEP and CES1 genes in 116 HF patients was performed by the Sequenom MassARRAY method, and logistic regression and haplotype analysis were used to evaluate the associations between SNPs and the clinical efficacy and safety of sacubitril/valsartan in HF patients. RESULTS A total of 116 Chinese patients with HF completed the whole trial, and T variations in rs701109 in NEP gene were an independent risk factor (P = 0.013, OR = 3.292, 95% CI:1.287-8.422) for the clinical efficacy of sacubitril/valsartan. Furthermore, haplotype analysis of 6 NEP SNPs (including rs701109) was performed and showed that the CGTACC and TGTACC haplotypes were significantly associated with clinical efficacy (OR = 0.095, 95%CI: 0.012-0.723, P = 0.003; OR = 5.586, 95% CI: 1.621-19.248, P = 0.005). Moreover, no association was found between SNPs of other selected genes in terms of efficacy in HF patients, and no association was observed between SNPs and symptomatic hypotension. CONCLUSION Our results suggest an association between rs701109 and sacubitril/valsartan response in HF patients. Symptomatic hypotension is not associated with the presence of NEP polymorphisms.
Collapse
Affiliation(s)
- Hong-Yu Luo
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China
| | - Li-Chen Gao
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China.
| | - Hui-Zhi Long
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China
| | - Zi-Wei Zhou
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China
| | - Shuo-Guo Xu
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China
| | - Feng-Jiao Li
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China
| | - Hong-Li Li
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China
| | - Yan Cheng
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China
| | - Cai-Xia Li
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xing-Yu Peng
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Liang Li
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ran Chen
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China
| | - Ping Deng
- School of Pharmacy, Department of Pharmacy, Phase I Clinical Trial Centre, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, 410004, China
| |
Collapse
|
5
|
Loucks CM, Lin JJ, Trueman JN, Drögemöller BI, Wright GEB, Chang WC, Li KH, Yoshida EM, Ford JA, Lee SS, Crotty P, Kim RB, Al-Judaibi B, Schwarz UI, Ramji A, Farivar JF, Tam E, Walston LL, Ross CJD, Carleton BC. Patient-specific genetic factors predict treatment failure in sofosbuvir-treated patients with chronic hepatitis C. Liver Int 2022; 42:796-808. [PMID: 35107877 DOI: 10.1111/liv.15175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/12/2022] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS According to pivotal clinical trials, cure rates for sofosbuvir-based antiviral therapy exceed 96%. Treatment failure is usually assumed to be because of virological resistance-associated substitutions or clinical risk factors, yet the role of patient-specific genetic factors has not been well explored. We determined if patient-specific genetic factors help predict patients likely to fail sofosbuvir treatment in real-world treatment situations. METHODS We recruited sofosbuvir-treated patients with chronic hepatitis C from five Canadian treatment sites, and performed a case-control pharmacogenomics study assessing both previously published and novel genetic polymorphisms. Specifically studied were variants predicted to impair CES1-dependent production of sofosbuvir's active metabolite, interferon-λ signalling variants expected to impact a patient's immune response to the virus and an HLA variant associated with increased spontaneous and treatment-induced viral clearance. RESULTS Three hundred and fifty-nine sofosbuvir-treated patients were available for analyses after exclusions, with 34 (9.5%) failing treatment. We identified CES1 variants as novel predictors for treatment failure in European patients (rs115629050 or rs4513095; odds ratio (OR): 5.43; 95% confidence interval (CI): 1.64-18.01; P = .0057), replicated associations with IFNL4 variants predicted to increase interferon-λ signalling (eg rs12979860; OR: 2.25; 95% CI: 1.25-4.06; P = .0071) and discovered a novel association with a coding variant predicted to enhance the activity of IFNL4's receptor (rs2834167 in IL10RB; OR: 1.81; 95% CI: 1.01-3.24; P = .047). CONCLUSIONS Ultimately, this work demonstrates that patient-specific genetic factors could be used as a tool to identify patients at higher risk of treatment failure and allow for these patients to receive effective therapy sooner.
Collapse
Affiliation(s)
- Catrina M Loucks
- BC Children's Hospital Research Institute, Vancouver, Canada.,Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, Canada.,Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Jennifer J Lin
- BC Children's Hospital Research Institute, Vancouver, Canada.,Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, Canada.,Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Jessica N Trueman
- BC Children's Hospital Research Institute, Vancouver, Canada.,Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Britt I Drögemöller
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Galen E B Wright
- Department of Pharmacy and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Wan-Chun Chang
- BC Children's Hospital Research Institute, Vancouver, Canada.,Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Kathy H Li
- BC Children's Hospital Research Institute, Vancouver, Canada.,Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Eric M Yoshida
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Jo-Ann Ford
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Samuel S Lee
- Liver Unit, University of Calgary Cumming School of Medicine, Calgary, Canada
| | - Pam Crotty
- Liver Unit, University of Calgary Cumming School of Medicine, Calgary, Canada
| | - Richard B Kim
- Division of Clinical Pharmacology, Department of Medicine, Western University, London, Canada
| | - Bandar Al-Judaibi
- Division of Transplantation, University of Rochester, Rochester, New York, USA.,Department of Liver Transplantation and Hepatobiliary Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Ute I Schwarz
- Division of Clinical Pharmacology, Department of Medicine, Western University, London, Canada
| | - Alnoor Ramji
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, Canada
| | | | | | | | - Colin J D Ross
- BC Children's Hospital Research Institute, Vancouver, Canada.,Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, Canada.,Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, Canada.,Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, Canada.,Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, Canada.,Pharmaceutical Outcomes Program (POPi), British Columbia Children's Hospital, Vancouver, Canada
| |
Collapse
|
6
|
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
|
7
|
Makhaeva GF, Lushchekina SV, Boltneva NP, Serebryakova OG, Kovaleva NV, Rudakova EV, Elkina NA, Shchegolkov EV, Burgart YV, Stupina TS, Terentiev AA, Radchenko EV, Palyulin VA, Saloutin VI, Bachurin SO, Richardson RJ. Novel potent bifunctional carboxylesterase inhibitors based on a polyfluoroalkyl-2-imino-1,3-dione scaffold. Eur J Med Chem 2021; 218:113385. [PMID: 33831780 DOI: 10.1016/j.ejmech.2021.113385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 01/04/2023]
Abstract
An expanded series of alkyl 2-arylhydrazinylidene-3-oxo-3-polyfluoroalkylpropionates (HOPs) 3 was obtained via Cu(OAc)2-catalyzed azo coupling. All were nanomolar inhibitors of carboxylesterase (CES), while moderate or weak inhibitors of acetylcholinesterase and butyrylcholinesterase. Steady-state kinetics studies showed that HOPs 3 are mixed type inhibitors of the three esterases. Molecular docking studies demonstrated that two functional groups in the structure of HOPs, trifluoromethyl ketone (TFK) and ester groups, bind to the CES active site suggesting subsequent reactions: formation of a tetrahedral adduct, and a slow hydrolysis reaction. The results of molecular modeling allowed us to explain some structure-activity relationships of CES inhibition by HOPs 3: their selectivity toward CES in comparison with cholinesterases and the high selectivity of pentafluoroethyl-substituted HOP 3p to hCES1 compared to hCES2. All compounds were predicted to have good intestinal absorption and blood-brain barrier permeability, low cardiac toxicity, good lipophilicity and aqueous solubility, and reasonable overall drug-likeness. HOPs with a TFK group and electron-donor substituents in the arylhydrazone moiety were potent antioxidants. All compounds possessed low cytotoxicity and low acute toxicity. Overall, a new promising type of bifunctional CES inhibitors has been found that are able to interact with the active site of the enzyme with the participation of two functional groups. The results indicate that HOPs have the potential to be good candidates as human CES inhibitors for biomedicinal applications.
Collapse
Affiliation(s)
- Galina F Makhaeva
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Sofya V Lushchekina
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia; Emanuel Institute of Biochemical Physics Russian Academy of Sciences, Moscow, 119334, Russia
| | - Natalia P Boltneva
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Olga G Serebryakova
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Nadezhda V Kovaleva
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Elena V Rudakova
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Natalia A Elkina
- Postovsky Institute of Organic Synthesis, Urals Branch of Russian Academy of Sciences, Ekaterinburg, 620990, Russia
| | - Evgeny V Shchegolkov
- Postovsky Institute of Organic Synthesis, Urals Branch of Russian Academy of Sciences, Ekaterinburg, 620990, Russia
| | - Yanina V Burgart
- Postovsky Institute of Organic Synthesis, Urals Branch of Russian Academy of Sciences, Ekaterinburg, 620990, Russia
| | - Tatyana S Stupina
- Institute of Problems of Chemical Physics Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Alexey A Terentiev
- Institute of Problems of Chemical Physics Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Eugene V Radchenko
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia; Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Vladimir A Palyulin
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia; Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Victor I Saloutin
- Postovsky Institute of Organic Synthesis, Urals Branch of Russian Academy of Sciences, Ekaterinburg, 620990, Russia
| | - Sergey O Bachurin
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Rudy J Richardson
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA; Center of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
8
|
Pei T, Yang J, Hu C, Chen X, Gong S, Hu X, Li L, Zhang L. Pharmacokinetics and Bioequivalence of Clopidogrel Hydrogen Sulfate Tablets in Fed and Fasted Conditions: An Open-Label, Randomized, Semireplicated Crossover Study in Healthy Chinese Volunteers. Clin Pharmacol Drug Dev 2020; 9:813-820. [PMID: 32452659 DOI: 10.1002/cpdd.804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/07/2020] [Indexed: 11/11/2022]
Abstract
Clopidogrel is an antiplatelet drug with high intraindividual variability in systemic exposure and efficacy. It has been used for treating atherosclerosis and acute coronary syndrome and in preventing stent restenosis and thrombotic complications after stent implantation in clinical practice for nearly 20 years. In this study we aimed to evaluate the bioequivalence of 2 clopidogrel hydrogen sulfate formulations (75-mg tablets) under fed (n = 66) and fasted (n = 66) conditions by using the reference-scaled average bioequivalence method. An open-label, randomized, 3-sequence and 3-period crossover (3×3), semireplicated study was designed and conducted. Clopidogrel concentration of plasma samples was measured by high-precision liquid chromatography and tandem mass spectrometry. The pharmacokinetic parameters were derived by a noncompartmental model. In the fed condition the geometric least-squares mean ratios of peak concentration (Cmax ) and area under the concentration-time curve (AUC0-t ) were, respectively, 103.38% and 98.97%, and the corresponding 90%CIs were 95.68% to 111.70% and 94.67% to 103.47%. In the fasted condition the geometric least squares mean ratios of Cmax and AUC0-t were, respectively, 106.53% and 105.77%, and the corresponding 90%CIs were 97.62% to 116.25% and 96.96% to 115.38%. According to the criteria for bioequivalence (80.00% to 125.00%), the test formulations of clopidogrel and Plavix were determined to be bioequivalent.
Collapse
Affiliation(s)
- Tong Pei
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Jing Yang
- Department of Cardiology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Chaoying Hu
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Xiaoping Chen
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Shili Gong
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Xiao Hu
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Lin Li
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Makhaeva GF, Rudakova EV, Kovaleva NV, Lushchekina SV, Boltneva NP, Proshin AN, Shchegolkov EV, Burgart YV, Saloutin VI. Cholinesterase and carboxylesterase inhibitors as pharmacological agents. Russ Chem Bull 2019. [DOI: 10.1007/s11172-019-2507-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
10
|
Yee SW, Brackman DJ, Ennis EA, Sugiyama Y, Kamdem LK, Blanchard R, Galetin A, Zhang L, Giacomini KM. Influence of Transporter Polymorphisms on Drug Disposition and Response: A Perspective From the International Transporter Consortium. Clin Pharmacol Ther 2018; 104:803-817. [PMID: 29679469 DOI: 10.1002/cpt.1098] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/10/2018] [Accepted: 04/11/2018] [Indexed: 12/21/2022]
Abstract
Advances in genomic technologies have led to a wealth of information identifying genetic polymorphisms in membrane transporters, specifically how these polymorphisms affect drug disposition and response. This review describes the current perspective of the International Transporter Consortium (ITC) on clinically important polymorphisms in membrane transporters. ITC suggests that, in addition to previously recommended polymorphisms in ABCG2 (BCRP) and SLCO1B1 (OATP1B1), polymorphisms in the emerging transporter, SLC22A1 (OCT1), be considered during drug development. Collectively, polymorphisms in these transporters are important determinants of interindividual differences in the levels, toxicities, and response to many drugs.
Collapse
Affiliation(s)
- Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Deanna J Brackman
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Elizabeth A Ennis
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Yokohama, Japan
| | - Landry K Kamdem
- Department of Pharmaceutical Sciences, Harding University College of Pharmacy, Searcy, Arkansas, USA
| | | | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, UK
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, San Francisco, California, USA.,Institute of Human Genetics, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
11
|
Boltneva N, Makhaeva G, Shchegol’kov E, Burgart Y, Saloutin V. Selective Carboxylesterase Inhibitors for Improving Efficacy, Safety and Rational use of Ester-Containing Drugs. ACTA ACUST UNITED AC 2018. [DOI: 10.18097/bmcrm00026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In clinical practice, a large number of prodrugs and active drugs containing an ester, carbamate or amide moiety are used. Carboxylesterase (CaE, EC 3.1.1.1) is the key enzyme of hydrolytic metabolism of such drugs in the body, it largely determines their pharmacokinetics, bioavailability, efficacy and possible toxic effects. Using CaE selective inhibitors as components of combined drug therapy it is possible us to regulate the rate of hydrolytic transformation of ester-containing drugs and opens the possibility of their rational use. The development of effective and selective CaE inhibitors suitable for in vivo application is a new promising approach in medicinal chemistry and pharmacology that allows to improve the efficacy, bioavailability and reduce the side effects of ester-containing drugs.
Collapse
Affiliation(s)
- N.P. Boltneva
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, Chernogolovka, Russia
| | - G.F. Makhaeva
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, Chernogolovka, Russia
| | - E.V. Shchegol’kov
- Postovsky Institute of Organic Synthesis, Urals Branch of Russian Academy of Sciences, Yekaterinburg, Russia
| | - Ya.V. Burgart
- Postovsky Institute of Organic Synthesis, Urals Branch of Russian Academy of Sciences, Yekaterinburg, Russia
| | - V.I. Saloutin
- Postovsky Institute of Organic Synthesis, Urals Branch of Russian Academy of Sciences, Yekaterinburg, Russia
| |
Collapse
|