1
|
Mu J, Chen SS, Li SQ, Jin Q, Geng J, Zou LW. Discovery of lignans as the effective inhibitors of CES1A alleviate lipid droplets formation. J Enzyme Inhib Med Chem 2025; 40:2472817. [PMID: 40207794 PMCID: PMC11986867 DOI: 10.1080/14756366.2025.2472817] [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: 09/12/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 04/11/2025] Open
Abstract
ER carboxylesterase 1A (CES1A) is an important metabolic enzyme involved in lipid metabolism. Targeting the CES1A is a promising approach for diseases associated with disorders of lipid metabolism therapy. In this study, screening of 26 natural lignans, three of them were found displaying potent inhibition on CES1A and high specificity over other serine hydrolases. Inhibition kinetic analyses demonstrated that Schisandrin C and Anwuligan were mixed-type inhibitors, while Magnolol acts as a competitive inhibitor. Further investigation showed that they were cell permeable and exhibited minimal cytotoxicity and mitochondrial toxicity, as well as capable of inhibiting intracellular CES1A in living cells. Further investigation found that three Schisandras decreased the number of lipid droplets (LDs) in free fatty acid (FFA)-treated HepG2 cells. Collectively, our findings suggest that Schisandrin C is a potent and highly selective inhibitor of CES1A, which can be served as a promising lead compound.
Collapse
Affiliation(s)
- Jie Mu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Si-Si Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shi-Qing Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiang Jin
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jin Geng
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Li-Wei Zou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
2
|
Wagner AM, Lanier OL, Savk A, Peppas NA. Polybasic nanogels for intracellular co-delivery of paclitaxel and carboplatin: a novel approach to ovarian cancer therapy. RSC PHARMACEUTICS 2025; 2:553-569. [PMID: 39990011 PMCID: PMC11843545 DOI: 10.1039/d4pm00330f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Ovarian cancer is one of the leading causes of cancer-related deaths in women, with limited progress in treatments despite decades of research. Common treatment protocols rely on surgical removal of tumors and chemotherapy drugs, such as paclitaxel and carboplatin, which are capable of reaching cancer cells throughout the body. However, the effectiveness of these drugs is often limited due to toxic reactions in patients, nonspecific drug distribution affecting healthy cells, and the development of treatment resistance. In this study, we introduce a polybasic nanogel system composed of poly(diethylaminoethyl methacrylate-co-cyclohexyl methacrylate)-g-poly(ethylene glycol) designed for the targeted co-delivery of paclitaxel and carboplatin directly to ovarian cancer cells. These nanogel systems can respond to the cellular microenvironment to achieve controlled, on-demand drug release, reducing off-target effects and enhancing therapeutic uptake. Additionally, we investigated nanoparticle degradation and controlled drug release as a function of various crosslinkers, including tetraethylene glycol dimethacrylate, bis(2-methacryloyl)oxyethyl disulfide, poly(lactic acid)-b-poly(ethylene glycol)-b-poly(lactic acid)dimethacrylate, and polycaprolactone dimethacrylate. Our results, using OVCAR-3 human ovarian cancer cells, demonstrated that this dual-delivery system outperformed free drugs in inducing cancer cell death, representing a promising advance in the field of nanoparticle-based therapies for ovarian cancer. By loading two chemotherapeutic agents into a single, environmentally responsive particle, this approach shows the potential to overcome common resistance mechanisms and achieve more effective tumor suppression. In summary, by delivering chemotherapy more precisely, it may be possible to enhance therapeutic outcomes while minimizing toxicity and nonspecific drug distribution, ultimately improving patient quality of life.
Collapse
Affiliation(s)
- Angela M Wagner
- McKetta Department of Chemical Engineering, The University of Texas at Austin Austin TX USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin Austin TX USA
| | - Olivia L Lanier
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin Austin TX USA
- Department of Biomedical Engineering, The University of Texas at Austin Austin TX USA
- Department of Chemical and Biological Engineering, University of New Mexico Albuquerque NM USA
- Department of Biomedical Engineering, University of New Mexico Albuquerque NM USA
- Cancer Therapeutics Program, University of New Mexico Comprehensive Cancer Center Albuquerque NM USA
| | - Ani Savk
- McKetta Department of Chemical Engineering, The University of Texas at Austin Austin TX USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin Austin TX USA
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, The University of Texas at Austin Austin TX USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin Austin TX USA
- Department of Biomedical Engineering, The University of Texas at Austin Austin TX USA
- Department of Surgery and Perioperative Care, Dell Medical School, University of Texas at Austin Austin TX USA
- Department of Pediatrics, Dell Medical School, University of Texas at Austin Austin TX USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin Austin TX USA
| |
Collapse
|
3
|
Eleveld DJ, Colin PJ, Van den Berg JP, Koomen JV, Stoehr T, Struys MMRF. Development and analysis of a remimazolam pharmacokinetics and pharmacodynamics model with proposed dosing and concentrations for anaesthesia and sedation. Br J Anaesth 2025:S0007-0912(25)00192-8. [PMID: 40312166 DOI: 10.1016/j.bja.2025.02.038] [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: 08/11/2024] [Revised: 01/31/2025] [Accepted: 02/20/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Pharmacokinetic-pharmacodynamic (PK-PD) models of remimazolam and their covariate relationships are useful for understanding drug disposition and predicting drug effects. Although clinical studies have shown that remimazolam induction doses decline with advancing age, this property is not reflected in existing models. The purpose of this investigation was to develop a PK-PD model for remimazolam and perform covariate analysis to maximise its utility across broad, diverse populations and to evaluate its consistency with clinical observations of drug dosing. METHODS Arterial and venous concentrations of remimazolam and its metabolite, Modified Observer's Assessment of Alertness and Sedation score and bispectral index were determined in 20 studies. Final population models were developed with covariate analysis. Simulations of drug administration for sedation and anaesthesia for this and previously published models were compared with the results of clinical studies. RESULTS Model development proceeded from 933 individuals aged 6-93 yr and weight 21-171 kg. PK data from studies with extracorporeal membrane oxygenation and treatment in ICU were considered in a post hoc analysis. Simulations of target-controlled infusion with the final model targeting sedation (Modified Observer's Assessment of Alertness and Sedation score 2 or 3) or anaesthesia (bispectral index 50) showed drug administration declining with age consistent with clinical observations. CONCLUSIONS A PK-PD model for remimazolam was developed for a broad, diverse population. Dosing and target concentrations are proposed that are clinically useful for anaesthesia and sedation, especially for target-controlled infusion administration.
Collapse
Affiliation(s)
- Douglas J Eleveld
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Pieter J Colin
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Johannes P Van den Berg
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jeroen V Koomen
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Michel M R F Struys
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
4
|
Ribone SR, Estrin DA, Quevedo MA. Exploring human carboxylesterases 1 and 2 selectivity of two families of substrates at an atomistic level. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141069. [PMID: 40209868 DOI: 10.1016/j.bbapap.2025.141069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/12/2025]
Abstract
Human carboxylesterases (CES) are enzymes that play an important role in the metabolism and biotransformation of diverse substances. The two more relevant isoforms, CES1A1 and CES2A1, catalyze the hydrolysis of numerous approved drugs and prodrugs. The elucidation of CES isoform substrates specificity constitutes a very relevant medicinal chemistry issue. The general role pointed that the selectivity towards CES1A1 or CES2A1 depends on the size of the acyl and alkyl moieties present in the structure of the substrate, but several exceptions regarding substrate promiscuity towards both CES have been reported. In this work, a combination of classical molecular dynamics (MD) and hybrid quantum mechanics/molecular mechanics (QM/MM) simulations were applied with the purpose of studying the substrate selectivity of CES1A1 and CES2A1 on two sets of selected ligands: p-nitrophenyl ester derivatives (NPE) and pyrethroid stereoisomers (Pyr). The classical molecular modeling studies showed that the van der Waals (VDW) component of interaction, with the hydrophobic residues present on CES1A1 and CES2A1 subpocket 1 and subpocket 2, showed a significant contribution to the substrates-CES affinity properties. The hybrid QM/MM simulations exhibited that the rate-limiting step for the studied substrates reactions were related to the transition state (TS) with the higher steric hindrance molecular structure. In conclusion, it was possible to observe that the studied substrates generate the best possible interaction pattern with the residues from subpocket 1 and 2 in order to produce the corresponding affinity constant with the enzyme. Then, this interaction pattern drives the catalytic turn-over reaction through the presence or absence of a high steric hindrance center in the molecular structure of the rate-limiting reaction.
Collapse
Affiliation(s)
- Sergio R Ribone
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, Haya de la Torre esq. Medina Allende, Ciudad Universitaria, Córdoba 5000, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), Haya de la Torre esq. Medina Allende, Ciudad Universitaria, Córdoba 5000, Argentina
| | - Dario A Estrin
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Inorgánica, Analítica y Química Física, Intendente Güiraldes 2160, C1428EHA Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Fíisica de los Materiales, Medio Ambiente y Energía (INQUIMAE), Ciudad Universitaria, Pabellón 2, Ciudad Autónoma de Buenos Aires, C1428EHA Buenos Aires, Argentina
| | - Mario A Quevedo
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, Haya de la Torre esq. Medina Allende, Ciudad Universitaria, Córdoba 5000, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), Haya de la Torre esq. Medina Allende, Ciudad Universitaria, Córdoba 5000, Argentina.
| |
Collapse
|
5
|
Wang YG, Gan CP, Beukers-Korver J, Rosing H, Li WL, Wagenaar E, Lebre MC, Song JY, Pritchard C, Bin Ali R, Huijbers I, Beijnen JH, Schinkel AH. Intestinal human carboxylesterase 2 (CES2) expression rescues drug metabolism and most metabolic syndrome phenotypes in global Ces2 cluster knockout mice. Acta Pharmacol Sin 2025; 46:777-793. [PMID: 39496863 PMCID: PMC11845761 DOI: 10.1038/s41401-024-01407-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024]
Abstract
Carboxylesterase 2 (CES2) is expressed mainly in liver and intestine, but most abundantly in intestine. It hydrolyzes carboxylester, thioester, and amide bonds in many exogenous and endogenous compounds, including lipids. CES2 therefore not only plays an important role in the metabolism of many (pro-)drugs, toxins and pesticides, directly influencing pharmacology and toxicology in humans, but it is also involved in energy homeostasis, affecting lipid and glucose metabolism. In this study we investigated the pharmacological and physiological functions of CES2. We constructed Ces2 cluster knockout mice lacking all eight Ces2 genes (Ces2-/- strain) as well as humanized hepatic or intestinal CES2 transgenic strains in this Ces2-/- background. We showed that oral availability and tissue disposition of capecitabine were drastically increased in Ces2-/- mice, and tissue-specifically decreased by intestinal and hepatic human CES2 (hCES2) activity. The metabolism of the chemotherapeutic agent vinorelbine was strongly reduced in Ces2-/- mice, but only marginally rescued by hCES2 expression. On the other hand, Ces2-/- mice exhibited fatty liver, adipositis, hypercholesterolemia and diminished glucose tolerance and insulin sensitivity, but without body mass changes. Paradoxically, hepatic hCES2 expression rescued these metabolic phenotypes but increased liver size, adipose tissue mass and overall body weight, suggesting a "healthy" obesity phenotype. In contrast, intestinal hCES2 expression efficiently rescued all phenotypes, and even improved some parameters, including body weight, relative to the wild-type baseline values. Our results suggest that the induction of intestinal hCES2 may combat most, if not all, of the adverse effects of metabolic syndrome. These CES2 mouse models will provide powerful preclinical tools to enhance drug development, increase physiological insights, and explore potential solutions for metabolic syndrome-associated disorders.
Collapse
Affiliation(s)
- Yao-Geng Wang
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Chang-Pei Gan
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Joke Beukers-Korver
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Wen-Long Li
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Els Wagenaar
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Maria C Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Division of Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, the Netherlands
| | - Colin Pritchard
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Rahmen Bin Ali
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Ivo Huijbers
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Alfred H Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Zeng JL, Lan JX, Dai W, Liu SL, Huang H, Shu GZ, Huang LJ, Kang SS, Chen B, Hou W. A Review of Bavachinin and Its Derivatives as Multi-Therapeutic Agents. Chem Biodivers 2025:e202402762. [PMID: 39874061 DOI: 10.1002/cbdv.202402762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 01/30/2025]
Abstract
Extracting natural active ingredients from plants is an effective way to develop and screen modern drugs. Psoralea corylifolia (PC) is a leguminous plant whose seeds have long been used as a Traditional Chinese Medicine to treat psoriasis, rheumatism, dermatitis, and other diseases. To date, several main compounds, including coumarins, flavonoids, monoterpene phenols, and benzofurans, have been identified from the seeds of PC. Among them, bavachinin is a type of flavonoid with various biological activities. In this article, the biological activities and mechanisms of action of bavachinin and its derivatives are reviewed. It includes the pharmacokinetic characteristics of bavachinin and its derivatives, as well as its prominent anti-inflammatory, antitumor, antibacterial, and antiviral pharmacological activities and related metabolic studies. Bavachinin displayed these activities through different receptors, such as peroxisome proliferator-activated receptors (PPARs), as well as multiple signaling pathways and enzyme systems. In summary, bavachinin and its derivatives have potential drug development value in many fields, such as anti-inflammatory, antitumor, nervous system disease, and diabetes. We believe that this review will lay a foundation for bavachinin-based drug development throughout the world.
Collapse
Affiliation(s)
- Jun Lin Zeng
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, P. R. China
- HuanKui Academy, Nanchang University, Nanchang, P. R. China
| | - Jin Xia Lan
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, P. R. China
| | - Wei Dai
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, P. R. China
| | - Sheng Lan Liu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, P. R. China
| | - Hao Huang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, P. R. China
| | - Guang Zhao Shu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, P. R. China
| | - Le Jun Huang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, P. R. China
| | - Si Shuang Kang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, P. R. China
| | - Bin Chen
- Department of Chinese Medicine, Jiangxi Management Vocational College, Nanchang, P. R. China
| | - Wen Hou
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, P. R. China
| |
Collapse
|
7
|
Felix MN, Waerner T, Lakatos D, Reisinger B, Fischer S, Garidel P. Polysorbates degrading enzymes in biotherapeutics - a current status and future perspectives. Front Bioeng Biotechnol 2025; 12:1490276. [PMID: 39867473 PMCID: PMC11760601 DOI: 10.3389/fbioe.2024.1490276] [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: 09/02/2024] [Accepted: 12/02/2024] [Indexed: 01/28/2025] Open
Abstract
Polysorbates, in particular polysorbate (PS) 20 and 80, are the most commonly used surfactants for stabilising biotherapeutics produced by biotechnological processes. PSs are derived from ethoxylated sorbitan (a derivative of sorbitol) esterified with fatty acids of varying chain length and degree of saturation. In the past, these surfactants have been reported to have specific liabilities. Chemical (oxidations and hydrolyses) and enzymatic degradations have been reported to affect the stability of PS in drug products. Specifically, the presence of trace amounts (sub-ppm) of certain host cell proteins (HCPs) can induce enzymatic PS degradation, which can lead to the release of free fatty acids during storage over time. Enzymatic polysorbate degradation may impair the functionality of the surfactant in stabilising therapeutic proteins, leading to the formation of visible and/or sub-visible particles in biopharmaceutical drug products. This review summarises the enzymes currently known to be involved in the degradation of polysorbate in mammalian biotechnological processes for therapeutic proteins. In recent years, advanced analytical methods have been developed to qualify and quantify the PS-degrading enzymes. Most of these assays are based on mass spectrometry with a preceding HCP enrichment approach. Efforts were made to measure the enzyme activity and correlate it with observed PS degradation. The impact on drug product quality attributes, including fatty acid solubility and phase separation, up to the formation of visible particles, and the potential induction of protein and protein/fatty acid mixed particles as well as the sensitivity of specific PS quality towards enzymatic degradation, was considered. Various drug substance (DS) mitigation strategies related to the occurrence of PS degrading enzymes are discussed as amongst them the generation of stable HCP knockout cell lines, which are also carefully analysed. The underlying opinion article reflects the undergoing discussions related to PS degrading enzymes and focusses on (i) impact on drug product, (ii) analytics for identification/quantification (characterisation) of the PS degrading enzymes, (iii) enzyme activity (iv) currently identified enzymes, and (v) potential mitigation strategies to avoid enzymatic PS degradation during DS manufacturing.
Collapse
Affiliation(s)
- Marius Nicolaus Felix
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Thomas Waerner
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Daniel Lakatos
- Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Bernd Reisinger
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Simon Fischer
- Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| | - Patrick Garidel
- Pharmaceutical Development Biologicals, TIP, Boehringer Ingelheim Pharma GmbH & Co., KG, Innovation Unit, Biberach an der Riss, Germany
| |
Collapse
|
8
|
Imai T, Isasaka M, Oyama Y, Takagi Y, Ohura K, Kotani S, Nakada Y. Functional Analysis of Modified Caco-2 Cells Carrying CES2A1 as a Model for Intestinal Absorption of Prodrugs. Biol Pharm Bull 2025; 48:162-171. [PMID: 40010717 DOI: 10.1248/bpb.b24-00645] [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] [Indexed: 02/28/2025]
Abstract
Carboxylesterase (CES) plays an important role in the metabolism of ester-containing drugs such as prodrugs and is highly expressed in the human intestine and liver. The ideal prodrug is barely hydrolyzed by human intestinal CES (CES2A1) but is extensively converted to an active drug by human hepatic CES (CES1A). It is, therefore, important to evaluate CES2A1-mediated hydrolysis during intestinal absorption. Unfortunately, Caco-2 cells, the most common enterocyte model for drug permeability, are not suitable for permeability studies of prodrugs due to their high and extremely low expression of CES1A and CES2A1, respectively. Previously, we have prepared CES2/Caco-2CES1KD cells with reduced human CES1A and highly expressed CES2A1. In the present study, the metabolic and transport properties of CES2/Caco-2CES1KD cells were characterized. The expression of transporters and metabolizing enzymes other than CESs was similar in CES2/Caco-2CES1KD and Caco-2 cells. However, the expression of CES2A1 in CES2/Caco-2CES1KD was about 7-10 fold higher than that of CES1A in Caco-2 cells and comparable to levels found in the human intestine. Hydrolysis during transport across cell monolayers was analyzed using ethyl and butyl esters of p-aminobenzoic acid (PABA). Ethyl PABA, a better substrate for CES1A than CES2A1, was similarly hydrolyzed in Caco-2 and CES2/Caco-2CES1KD cell monolayers due to the high expression of CES2A1 in CES2/Caco-2CES1KD cells. Butyl PABA, a good substrate for CES2A1, was substantially hydrolyzed in CES2/Caco-2CES1KD cell monolayers, in contrast to negligible hydrolysis in Caco-2 cell monolayers. N-Acetylation of PABA derived from PABA esters showed similar activity in Caco-2 and CES2/Caco-2CES1KD cell monolayers.
Collapse
Affiliation(s)
- Teruko Imai
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
- Daiichi University of Pharmacy, 22-1 Tamagawa-machi, Minami-ku, Fukuoka 815-8511, Japan
| | - Masanari Isasaka
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yusuke Oyama
- School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yusuke Takagi
- School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Kayoko Ohura
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
- Pharmacy Talent Development Department, Sogo Medical Co., Ltd., PRIO Fukuoka Building, 2‒9‒23 Daimyo, Chuo-ku, Fukuoka 810‒0041, Japan
| | - Shunsuke Kotani
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yuichiro Nakada
- Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-kita, Tondabayashi, Osaka 584-0066, Japan
| |
Collapse
|
9
|
Wang Z, McCalla Z, Lin L, Tornichio D, Agyemang Y, Bastulli JA, Zhang XS, Zhu HJ, Wang X. Impact of genetic polymorphisms and drug-drug interactions mediated by carboxylesterase 1 on remimazolam deactivation. Drug Metab Dispos 2025; 53:100023. [PMID: 39884809 DOI: 10.1124/dmd.124.001916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 01/22/2025] Open
Abstract
Remimazolam (Byfavo, Acacia Pharma), a recent Food and Drug Administration-approved ester-linked benzodiazepine, offers advantages in sedation, such as rapid onset and predictable duration, making it suitable for broad anesthesia applications. Its favorable pharmacological profile is primarily attributed to rapid hydrolysis, the primary metabolism pathway for its deactivation. Thus, understanding remimazolam hydrolysis determinants is essential for optimizing its clinical use. This study aimed to identify the enzyme(s) and tissue(s) responsible for remimazolam hydrolysis and to evaluate the influence of genetic polymorphisms and drug-drug interactions on its hydrolysis in the human liver. An initial incubation study with remimazolam and PBS, human serum, and the S9 fractions of human liver and intestine demonstrated that remimazolam was exclusively hydrolyzed by human liver S9 fractions. Subsequent incubation studies utilizing a carboxylesterase inhibitor (bis(4-nitrophenyl) phosphate), recombinant human carboxylesterase 1 (CES1) and carboxylesterase 2 confirmed that remimazolam is specifically hydrolyzed by CES1 in human liver. Furthermore, in vitro studies with wild-type CES1 and CES1 variants transfected cells revealed that certain genetic polymorphisms significantly impair remimazolam deactivation. Notably, the impact of CES1 G143E was verified using individual human liver samples. Moreover, our evaluation of the drug-drug interactions between remimazolam and several other substrates/inhibitors of CES1-including simvastatin, enalapril, clopidogrel, and sacubitril-found that clopidogrel significantly inhibited remimazolam hydrolysis at clinically relevant concentrations, with CES1 genetic variants potentially influencing the interactions. In summary, CES1 genetic variants and its interacting drugs are crucial factors contributing to interindividual variability in remimazolam hepatic hydrolysis, holding the potential to serve as biomarkers for optimizing remimazolam use. SIGNIFICANCE STATEMENT: This investigation demonstrates that remimazolam is deactivated by carboxylesterase 1 (CES1) in the human liver, with CES1 genetic variants and drug-drug interactions significantly influencing its metabolism. These findings emphasize the need to consider CES1 genetic variability and potential drug-drug interactions in remimazolam use, especially in personalized pharmacotherapy to achieve optimal anesthetic outcomes.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Zachary McCalla
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Li Lin
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Dominic Tornichio
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Yaw Agyemang
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - John A Bastulli
- Department of Surgery, Northeast Ohio Medical University, Rootstown, Ohio
| | - Xiaochun Susan Zhang
- Department of Pathology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Hao-Jie Zhu
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan
| | - Xinwen Wang
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio.
| |
Collapse
|
10
|
Wu XP, Yang N, Liu QQ, Zhu ZQ. A fluorescent probe for monitoring carboxylesterases in pulmonary cells under permissive hypercapnia condition. ANAL SCI 2024; 40:2241-2249. [PMID: 39242486 DOI: 10.1007/s44211-024-00659-8] [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: 06/18/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Abstract
Herein, by combining the benzofuranone-derived fluorophore and the carbamate recognition group, a fluorescent probe named BFO-CarE was developed for monitoring the carboxylesterase (CarE) level in pulmonary cells under the permissive hypercapnia condition. It showed a notable fluorescence response towards CarE at 570 nm under the excitation of 510 nm. The in-solution tests revealed the advantages of BFO-CarE including high sensitivity, high specificity, relatively rapid response, and high steadiness. It was also low-toxic upon the pulmonary cell lines. During the intracellular imaging in pulmonary cells, BFO-CarE achieved the monitoring of the CarE level in both inhibition and activation status. In particular, BFO-CarE realized the visualization of the affection of the permissive hypercapnia condition on the CarE level, which indicated the hypoxia tolerance of CarE. This work was informative for investigating the impact of hypoxia in pulmonary cells, and the corresponding anaesthesia-related approaches.
Collapse
Affiliation(s)
- Xiao-Ping Wu
- Department of Anesthesiology, School of Medicine, Affiliated Jinhua Hospital, Zhejiang University, Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Na Yang
- Department of Anesthesiology, School of Medicine, Affiliated Jinhua Hospital, Zhejiang University, Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Qing-Qing Liu
- Department of Anesthesiology, School of Medicine, Affiliated Jinhua Hospital, Zhejiang University, Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Zhong-Quan Zhu
- Department of Anesthesiology, School of Medicine, Affiliated Jinhua Hospital, Zhejiang University, Jinhua Municipal Central Hospital, Jinhua, 321000, China.
| |
Collapse
|
11
|
Murata M, Okada K, Takahashi M, Ueyama-Toba Y, Ito S, Mizuguchi H. Generation and application of CES1-knockout Tet-Off-regulated CYP3A4 and UGT1A1-expressing Caco-2 cells. Toxicol Lett 2024; 401:158-169. [PMID: 39383894 DOI: 10.1016/j.toxlet.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/07/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Caco-2 cells, a human colorectal adenocarcinoma cell line, are widely used to model small intestinal epithelial cells in the drug development process because they can predict drug absorption with high accuracy. However, Caco-2 cells have several issues. First, Caco-2 cells have little expression of cytochrome P450 3A4 (CYP3A4), which is a major drug-metabolizing enzyme in the human intestine. We previously developed Caco-2 cells whose expression of CYP3A4 can be controlled using doxycycline (Dox) (CYP3A4-Caco-2 cells) (Ichikawa et al., Sci. Rep, 2021). However, since the Tet-On system was used to regulate CYP3A4 expression in these cells, there was concern about drug-drug interactions. The second issue is that in the human small intestine, carboxylesterase 2 (CES2) is more highly expressed than carboxylesterase 1 (CES1), while in Caco-2 cells CES1 is more highly expressed. The third issue is the low level expression of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1), a phase II drug-metabolizing enzyme. In this study, we used genome-editing technology to establish CES1-knockout Caco-2 cells whose CYP3A4 and UGT1A1 expression can be regulated by the Tet-Off system. These cell lines would be useful in pharmaceutical researches, including intestinal toxicological studies, as an in vitro model for orally administered drugs.
Collapse
Affiliation(s)
- Michika Murata
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Kentaro Okada
- Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; Laboratory of Functional Organoid for Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Masaki Takahashi
- GenoMembrane Co., Ltd., 2-3-18 Namamugi, Tsurumi-ku, Yokohama, Kanagawa 230-0052, Japan
| | - Yukiko Ueyama-Toba
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; Laboratory of Functional Organoid for Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Sumito Ito
- GenoMembrane Co., Ltd., 2-3-18 Namamugi, Tsurumi-ku, Yokohama, Kanagawa 230-0052, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; Laboratory of Functional Organoid for Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan; Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
12
|
Wang DD, Wang ZZ, Liu WC, Qian XK, Zhu YD, Wang TG, Pan SM, Zou LW. Pyrazolone compounds could inhibit CES1 and ameliorates fat accumulation during adipocyte differentiation. Bioorg Chem 2024; 150:107536. [PMID: 38878751 DOI: 10.1016/j.bioorg.2024.107536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 07/21/2024]
Abstract
Carboxylesterase 1 (CES1), a member of the serine hydrolase superfamily, is involved in a wide range of xenobiotic and endogenous substances metabolic reactions in mammals. The inhibition of CES1 could not only alter the metabolism and disposition of related drugs, but also be benefit for treatment of metabolic disorders, such as obesity and fatty liver disease. In the present study, we aim to develop potential inhibitors of CES1 and reveal the preferred inhibitor structure from a series of synthetic pyrazolones (compounds 1-27). By in vitro high-throughput screening method, we found compounds 25 and 27 had non-competitive inhibition on CES1-mediated N-alkylated d-luciferin methyl ester (NLMe) hydrolysis, while compound 26 competitively inhibited CES1-mediated NLMe hydrolysis. Additionally, Compounds 25, 26 and 27 can inhibit CES1-mediated fluorescent probe hydrolysis in live HepG2 cells with effect. Besides, compounds 25, 26 and 27 could effectively inhibit the accumulation of lipid droplets in mouse adipocytes cells. These data not only provided study basis for the design of newly CES1 inhibitors. The present study not only provided the basis for the development of lead compounds for novel CES1 inhibitors with better performance, but also offered a new direction for the explore of candidate compounds for the treatment of hyperlipidemia and related diseases.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China.
| | - Zhen-Zhen Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Wen-Cai Liu
- Asymchem Biotechnology (Tianjin) Co., Ltd, Tianjin 300457, China
| | - Xing-Kai Qian
- Translational Medicine Research Center, Guizhou Medical University, University Town, Guian New District, Guizhou 550025, China.
| | - Ya-Di Zhu
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, China
| | - Tie-Gang Wang
- Tangshan Boshide Medical Devices Co., Ltd, Tangshan 063599, China
| | - Shu-Mei Pan
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Li-Wei Zou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
13
|
Cavallero A, Donadel G, Puccini P, Gervasi PG, Gabisonia K, Longo V, Gabriele M. New insight on porcine carboxylesterases expression and activity in lung tissues. Res Vet Sci 2024; 175:105314. [PMID: 38823354 DOI: 10.1016/j.rvsc.2024.105314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/04/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Over the course of the last twenty years, there has been a growing recognition of the pig's potential as a valuable model for studying human drug metabolism. This study aimed to investigate the expression, enzymatic activity, inhibitory susceptibility, and cellular localization of carboxylesterases (CES) in porcine lung tissue not yet explored. Our results showed that CESs hydrolysis activity followed Michaelis-Menten kinetics in both cytosolic and microsomal fractions of porcine lung tissues (N = 8), with comparable hydrolysis rates for tested substrates, namely 4-nitrophenyl acetate (pNPA), 4-methylumbelliferyl acetate (4-MUA), and fluorescein diacetate (FD). We also determined the CESs hydrolysis activity in a representative sample of the porcine liver that, as expected, displayed higher activity than the lung ones. The study demonstrated variable levels of enzyme activities and interindividual variability in both porcine lung fractions. Inhibition studies used to assess the CESs' involvement in the hydrolysis of pNPA, 4-MUA, and FD suggested that CESs may be the enzymes primarily involved in the metabolism of ester compounds in the pig lung tissue. Overall, this study provides insight into the distribution and diversity of CES isoforms involved in substrate hydrolysis across different cellular fractions (cytosol and microsomes) in porcine lungs.
Collapse
Affiliation(s)
- Andrea Cavallero
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Giorgia Donadel
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Paola Puccini
- Chiesi Farmaceutici S.P.A., via Palermo 26/A, Parma, Italy
| | - Pier Giovanni Gervasi
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Khatia Gabisonia
- Interdisciplinary Center "Health Science", Scuola Superiore Sant'Anna, c/o Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Vincenzo Longo
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy
| | - Morena Gabriele
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
14
|
Nandi S, Swain R, Habibullah S, Sahoo RN, Nayak AK, Mallick S. Lipid-Gelucire based rectal delivery of ramipril prodrug exhibits significant lowering of intra-ocular pressure in normotensive rabbit: sustained structural relaxation release kinetics and IVIVC. Pharm Dev Technol 2024; 29:468-476. [PMID: 38662798 DOI: 10.1080/10837450.2024.2345807] [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: 02/27/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024]
Abstract
Carboxylesterase enzymes convert a prodrug ramipril into the biologically active metabolite ramiprilat. It is prescribed for controlling ocular hypertension after oral administration. High concentrations of carboxylesterase enzymes in rectal and colon tissue can transform ramipril significantly to ramiprilat. Sustained rectal delivery of ramipril has been developed for intra-ocular pressure lowering effect using a normotensive rabbit model. Rectal suppositories have been formulated using a matrix base of HPMC K100-PEG 400-PEG 6000, incorporating varying amounts of Gelucire by the fusion moulding method. The presence of Gelucire in the suppository exhibited sustained structural relaxation-based release kinetics of RM compared to its absence. Intravenous and oral administration of ramipril has decreased IOP in the treated rabbit up to 90 and 360 min, respectively. Treated rabbits with suppositories have revealed decreased IOP for an extended period compared to the above. Formulation containing GEL 3% reduced intra-ocular pressure to 540 min, with the highest area under the decreased IOP curve. Compared to oral, the pharmacodynamic bioavailability of ramipril has been improved significantly using a sustained-release rectal suppository. A rectal suppository for sustained delivery of ramipril could be used to lower IOP significantly.
Collapse
Affiliation(s)
- Souvik Nandi
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Rakesh Swain
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Sk Habibullah
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Rudra Narayan Sahoo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Amit Kumar Nayak
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Subrata Mallick
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| |
Collapse
|
15
|
Uno Y, Yamato O, Yamazaki H. Transcript abundance of hepatic drug-metabolizing enzymes in two dog breeds compared with 14 species including humans. Drug Metab Pharmacokinet 2024; 55:101002. [PMID: 38452615 DOI: 10.1016/j.dmpk.2024.101002] [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: 12/02/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 03/09/2024]
Abstract
Drug-metabolizing enzymes are important in drug development and therapy, but have not been fully identified and characterized in many species, lines, and breeds. Liver transcriptomic data were analyzed for phase I cytochromes P450, flavin-containing monooxygenases, and carboxylesterases and phase II UDP-glucuronosyltransferases, sulfotransferases, and glutathione S-transferases. Comparisons with a variety of species (humans, rhesus macaques, African green monkeys, baboons, common marmosets, cattle, sheep, pigs, cats, dogs, rabbits, tree shrews, rats, mice, and chickens) revealed both general similarities and differences in the transcript abundances of drug-metabolizing enzymes. Similarly, Beagle and Shiba dogs were examined by next-generation sequencing (RNA-seq). Consequently, no substantial differences in transcript abundance were noted in different breeds of pigs and dogs and in different lines of mice and rats. Therefore, the expression profiles of hepatic drug-metabolizing enzyme transcripts appear to be similar in Shiba and Beagle dogs and pig breeds and the rat and mouse lines analyzed, although some differences were found in other species.
Collapse
Affiliation(s)
- Yasuhiro Uno
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima, 890-0065, Japan.
| | - Osamu Yamato
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima, 890-0065, Japan
| | - Hiroshi Yamazaki
- Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan.
| |
Collapse
|
16
|
Wang X, Gao J, Fan C, Gao Y, Yang X, Chen L. New Near-Infrared Fluorescence Imaging Platform with Large Stokes Shift for Carboxylesterase 2 Detection in Thyroid Cancer and Inflammatory Diseases Diagnosis. Anal Chem 2024; 96:3772-3779. [PMID: 38372636 DOI: 10.1021/acs.analchem.3c04399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Development of new near-infrared fluorophores is one of the eternal themes in the field of biosensing and biological imaging. In this work, we constructed a novel fluorophore platform MOR by replacing methylindole of hemicyanine fluorophore (CyR) with benzoxazole to acquire better fluorescence characteristics. Based on the platform, a near infrared (NIR) fluorescent probe MOR-CES2 was synthesized for the specific "off-on" response to carboxylesterase 2 (CES2). The probe exhibited excellent properties including near-infrared emission (735 nm), large Stokes shift (105 nm), high sensitivity (LOD, 0.3 ng/mL), and rapid response (15 min). The successful application of MOR-CES2 in biological imaging of CES2 in mice with thyroid cancer and inflammatory bowel disease demonstrated that the probe could identify cancer cells and tissues and sensitively respond to inflammation. The results proved the potency of MOR-CES2 as an efficient imaging tool to assist in the surgical resection of CES2-related tumors.
Collapse
Affiliation(s)
- Xiaochun Wang
- Liaoning Key Laboratory of Development and Utilization for Natural Products Active Molecules, School of Chemistry and Life Science, Anshan Normal University, Anshan 114007, China
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jian Gao
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Chuanfeng Fan
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yingkai Gao
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xintong Yang
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Lingxin Chen
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Research Center for Coastal Environmental Engineering and Technology, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
- College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing 312000, China
| |
Collapse
|
17
|
Takani D, Takahashi M, Hosokawa M. Synthesis and evaluation of indomethacin prodrugs with a diester structure that are metabolically activated by human carboxylesterases. Xenobiotica 2024; 54:10-17. [PMID: 38142303 DOI: 10.1080/00498254.2023.2298270] [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: 11/07/2023] [Accepted: 12/19/2023] [Indexed: 12/25/2023]
Abstract
1. Carboxylesterase (CES) has been studied extensively, mostly with substrates in the monoester structures. We investigated the relationship between indomethacin diester prodrugs and metabolic activation by microsomes and recombinant human CES.2. Eight indomethacin diester prodrugs were synthesised in two steps. They were used as substrates and hydrolysis rates were calculated.3. As a result, the major hydrolysis enzyme was CES. The hydrolysis rate of recombinant CES2A1 was comparable to that of recombinant CES1A1.4. In this study, by changing the structure of the prodrug to a diester structure, it was found that CES2 activity was equivalent to CES1 activity.5. It should be noted that the use of diester prodrugs in prodrug discovery, where organ-specific hydrolysis reactions are expected, may not yield the expected results.
Collapse
Affiliation(s)
- Daisuke Takani
- Faculty of Pharmacy, Chiba Institute of Science, Choshi, Chiba, Japan
| | - Masato Takahashi
- Faculty of Pharmacy, Chiba Institute of Science, Choshi, Chiba, Japan
| | - Masakiyo Hosokawa
- Faculty of Pharmacy, Chiba Institute of Science, Choshi, Chiba, Japan
| |
Collapse
|
18
|
Darnotuk ES, Siniavin AE, Shastina NS, Luyksaar SI, Inshakova AM, Bondareva NE, Zolotov SA, Lubenec NL, Sheremet AB, Logunov DY, Zigangirova NA, Gushchin VA, Gintsburg AL. Synthesis and Antiviral Activity of Novel β-D-N4-Hydroxycytidine Ester Prodrugs as Potential Compounds for the Treatment of SARS-CoV-2 and Other Human Coronaviruses. Pharmaceuticals (Basel) 2023; 17:35. [PMID: 38256869 PMCID: PMC10821229 DOI: 10.3390/ph17010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
The spread of COVID-19 infection continues due to the emergence of multiple transmissible and immune-evasive variants of the SARS-CoV-2 virus. Although various vaccines have been developed and several drugs have been approved for the treatment of COVID-19, the development of new drugs to combat COVID-19 is still necessary. In this work, new 5'-O-ester derivatives of N4-hydroxycytidine based on carboxylic acids were developed and synthesized by Steglich esterification. The antiviral activity of the compounds was assessed in vitro-inhibiting the cytopathic effect of HCoV-229E, and three variants of SARS-CoV-2, on huh-7 and Vero E6 cells. Data have shown that most synthesized derivatives exhibit high activity against coronaviruses. In addition, the relationship between the chemical structure of the compounds and their antiviral effect has been established. The obtained results show that the most active compound was conjugate SN_22 based on 3-methyl phenoxyacetic acid. The results of this study indicate the potential advantage of the chemical strategies used to modify NHC as a promising avenue to be explored in vivo, which could lead to the development of drugs with improved pharmacological properties that potently inhibit SARS-CoV-2.
Collapse
Affiliation(s)
- Elizaveta S. Darnotuk
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
- Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119571 Moscow, Russia
| | - Andrei E. Siniavin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Natal’ya S. Shastina
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
- Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119571 Moscow, Russia
| | - Sergey I. Luyksaar
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
| | - Anna M. Inshakova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
- Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119571 Moscow, Russia
| | - Natalia E. Bondareva
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
| | - Sergey A. Zolotov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
| | - Nadezhda L. Lubenec
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
| | - Anna B. Sheremet
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
| | - Denis Y. Logunov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
| | - Nailya A. Zigangirova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
| | - Vladimir A. Gushchin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
| | - Alexander L. Gintsburg
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.S.D.); (N.S.S.); (S.I.L.); (A.M.I.); (N.E.B.); (S.A.Z.); (N.L.L.); (A.B.S.); (D.Y.L.); (N.A.Z.); (A.L.G.)
| |
Collapse
|
19
|
Fan Y, Zhang T, Song Y, Sang Z, Zeng H, Liu P, Wang P, Ge G. Rationally Engineered hCES2A Near-Infrared Fluorogenic Substrate for Functional Imaging and High-Throughput Inhibitor Screening. Anal Chem 2023; 95:15665-15672. [PMID: 37782032 DOI: 10.1021/acs.analchem.3c02873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Human carboxylesterase 2A (hCES2A) is an important endoplasmic reticulum (ER)-resident enzyme that is responsible for the hydrolytic metabolism or activation of numerous ester-bearing drugs and environmental toxins. The previously reported hCES2A fluorogenic substrates suffer from limited emission wavelength, low specificity, and poor localization accuracy, thereby greatly limiting the in situ functional imaging of hCES2A and drug discovery. Herein, a rational ligand design strategy was adopted to construct a highly specific near-infrared (NIR) substrate for hCES2A. Following scaffold screening and recognition group optimization, HTCF was identified as a desirable NIR fluorophore with excellent photophysical properties and high ER accumulation ability, while several HTCF esters held a high potential to be good hCES2A substrates. Further investigations revealed that TP-HTCF (the tert-pentyl ester of HTCF) was an ideal substrate with ultrahigh sensitivity, excellent specificity, and a substantial signal-to-noise ratio. Upon the addition of hCES2A, TP-HTCF could be rapidly hydrolyzed to release HTCF, a chemically stable product that emitted bright fluorescent signals at around 670 nm. A TP-HTCF-based biochemical assay was then established for the high-throughput screening of potent and cell-active hCES2A inhibitors from an in-house compound library. Furthermore, TP-HTCF displayed high imaging resolution for imaging hCES2A in living cells as well as mouse liver slices and tumor-xenograft mice. Collectively, this study demonstrates a rational strategy for developing highly specific fluorogenic substrates for an ER-resident target enzyme, while TP-HTCF can act as a practical tool for sensing hCES2A in living systems.
Collapse
Affiliation(s)
- Yufan Fan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tiantian Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yunqing Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhipei Sang
- School of Pharmaceutical Sciences, Hainan University, Haikou, Hainan 570228, China
| | - Hairong Zeng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peiqi Liu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
20
|
Ohura K, Nakada Y, Imai T. Bioconversion and P-gp-Mediated Transport of Depot Fluphenazine Prodrugs after Intramuscular Injection. J Pharm Sci 2023; 112:1975-1984. [PMID: 37019360 DOI: 10.1016/j.xphs.2023.03.018] [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: 02/17/2023] [Revised: 03/26/2023] [Accepted: 03/26/2023] [Indexed: 04/07/2023]
Abstract
Fluphenazine (FPZ) decanoate, an ester-type prodrug formulated as a long-acting injection (LAI), is used in the treatment of schizophrenia. FPZ enanthate was also developed as an LAI formulation, but is no longer in use clinically because of the short elimination half-life of FPZ, the parent drug, after intramuscular injection. In the present study, the hydrolysis of FPZ prodrugs was evaluated in human plasma and liver to clarify the reason for this difference in elimination half-lives. FPZ prodrugs were hydrolyzed in human plasma and liver microsomes. The rate of hydrolysis of FPZ enanthate in human plasma and liver microsomes was 15-fold and 6-fold, respectively, faster than that of FPZ decanoate. Butyrylcholinesterase (BChE) and human serum albumin (HSA) present in human plasma, and two carboxylesterase (CES) isozymes, hCE1 and hCE2, expressed in ubiquitous organs including liver, were mainly responsible for the hydrolysis of FPZ prodrugs. FPZ prodrugs may not be bioconverted in human skeletal muscle at the injection site because of lack of expression of BChE and CESs in muscle. Interestingly, although FPZ was a poor substrate for human P-glycoprotein, FPZ caproate was a good substrate. In conclusion, it is suggested that the shorter elimination half-life of FPZ following administration of FPZ enanthate compared with FPZ decanoate can be attributed to the more rapid hydrolysis of FPZ enanthate by BChE, HSA and CESs.
Collapse
Affiliation(s)
- Kayoko Ohura
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan; Headquarters for Admissions and Education, Kumamoto University, Kumamoto, Japan
| | | | - Teruko Imai
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan; Daiichi University of Pharmacy, Fukuoka, Japan.
| |
Collapse
|
21
|
Nakashima S, Sato R, Fukami T, Kudo T, Hashiba S, Morinaga G, Nakano M, Ludwig-Schwellinger E, Matsui A, Ishiguro N, Ebner T, Nakajima M. Characterization of Enzymes Involved in Nintedanib Metabolism in Humans. Drug Metab Dispos 2023; 51:733-742. [PMID: 36927840 DOI: 10.1124/dmd.122.001113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/03/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Nintedanib, which is used to treat idiopathic pulmonary fibrosis and non-small cell lung cancer, is metabolized to a pharmacologically inactive carboxylate derivative, BIBF1202, via hydrolysis and subsequently by glucuronidation to BIBF1202 acyl-glucuronide (BIBF1202-G). Since BIBF1202-G contains an ester bond, it can be hydrolytically cleaved to BIBF1202. In this study, we sought to characterize these metabolic reactions in the human liver and intestine. Nintedanib hydrolysis was detected in human liver microsomes (HLMs) (Clearance [CL int]: 102.8 ± 18.9 µL/min per mg protein) but not in small intestinal preparations. CES1 was suggested to be responsible for nintedanib hydrolysis according to experiments using recombinant hydrolases and hydrolase inhibitors as well as proteomic correlation analysis using 25 individual HLM. BIBF1202 glucuronidation in HLM (3.6 ± 0.3 µL/min per mg protein) was higher than that in human intestinal microsomes (1.5 ± 0.06 µL/min per mg protein). UGT1A1 and gastrointestinal UGT1A7, UGT1A8, and UGT1A10 were able to mediate BIBF1202 glucuronidation. The impact of UGT1A1 on glucuronidation was supported by the finding that liver microsomes from subjects homozygous for the UGT1A1*28 allele showed significantly lower activity than those from subjects carrying the wild-type UGT1A1 allele. Interestingly, BIBF1202-G was converted to BIBF1202 in HLS9 at 70-fold higher rates than the rates of BIBF1202 glucuronidation. An inhibition study and proteomic correlation analysis suggested that β-glucuronidase is responsible for hepatic BIBF1202-G deglucuronidation. In conclusion, the major metabolic reactions of nintedanib in the human liver and intestine were quantitatively and thoroughly elucidated. This information could be helpful to understand the inter- and intraindividual variability in the efficacy of nintedanib. SIGNIFICANCE STATEMENT: To our knowledge, this is the first study to characterize the enzymes responsible for each step of nintedanib metabolism in the human body. This study found that β-glucuronidase may contribute to BIBF1202-G deglucuronidation.
Collapse
Affiliation(s)
- Shimon Nakashima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Rei Sato
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Takashi Kudo
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Shiori Hashiba
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Gaku Morinaga
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Eva Ludwig-Schwellinger
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Akiko Matsui
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Naoki Ishiguro
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Thomas Ebner
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences (S.N., R.S., T.F., S.H., Ma.N., Mi.N.) and WPI Nano Life Science Institute (WPI-NanoLSI) (T.F., Ma.N., Mi.N.), Kanazawa University, Kanazawa, Japan; Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan (T.K., G.M., A.M., N.I.); and Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (E.L.-S., T.E.)
| |
Collapse
|
22
|
Zhao Y, Li Y, Wang F, Gan X, Zheng T, Chen M, Wei L, Chen J, Yu C. CES1-Triggered Liver-Specific Cargo Release of CRISPR/Cas9 Elements by Cationic Triadic Copolymeric Nanoparticles Targeting Gene Editing of PCSK9 for Hyperlipidemia Amelioration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2300502. [PMID: 37083231 DOI: 10.1002/advs.202300502] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/25/2023] [Indexed: 05/03/2023]
Abstract
The broad application of clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 genome editing tools is hindered by challenges in the efficient delivery of its two components into specific cells and intracytoplasmic release. Herein, a novel copolymer for delivery of Cas9-mRNA/ single-guide RNA (Cas9-mRNA/sgRNA) in vitro and vivo, using carboxylesterase-responsive cationic triadic copolymeric nanoparticles targeted proprotein convertase subtilisin/kexin type 9 (PCSK9) for hyperlipidemia amelioration is reported. A dimethyl biguanide derivative is designed and synthesized to form cationic block, and copolymerization onto prepolymer with propyl methacrylate, to fabricate a triadic copolymer mPEG-b-P(Met/n-PMA). The copolymer can self-assemble with Cas9-mRNA/sgRNA, indicating the excellent potential of nanoparticles to form a delivery carrier. This vehicle can efficiently release RNA in response to the hepatocytes carboxylesterase for genome editing. It was demonstrated that the mPEG-b-P(Met/n-PMA)/Cas9 mRNA/sgRNA nanoparticles effectively accumulated in hepatocytes, lead to the inhibition of PCSK9, and lowered the levels of Low-density lipoprotein cholesterol and total cholesterol in mouse serum down 20% of nontreatment. Interestingly, the nanoparticles even enable multiple functions in the regulation of blood glucose and weight. This study establishes a novel method to achieve complex CRISPR components stable loading, safe delivery, and fixed-point release, which expand the application of CRISPR delivery systems.
Collapse
Affiliation(s)
- Yunfei Zhao
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yun Li
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Fan Wang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Xuelan Gan
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Tianye Zheng
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Mengyue Chen
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Li Wei
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jun Chen
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Chao Yu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, P. R. China
| |
Collapse
|
23
|
Yang Y, Xiong Y, Zhu G, Sun M, Zou K, Zhao Y, Zhang Y, Xu Z, Li Y, Zhu W, Jia Q, Li B, Ge G. Discovery of seven-membered ring berberine analogues as highly potent and specific hCES2A inhibitors. Chem Biol Interact 2023; 378:110501. [PMID: 37080375 DOI: 10.1016/j.cbi.2023.110501] [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: 02/14/2023] [Revised: 03/23/2023] [Accepted: 04/18/2023] [Indexed: 04/22/2023]
Abstract
Human carboxylesterase 2A (hCES2A) is a key serine hydrolase responsible for the metabolic clearance of large number of compounds bearing the ester- or amide-bond(s). Inhibition of hCES2A can relieve the chemotherapy-induced toxicity and alter the pharmacokinetic bahaviors of some orally administrate esters-containing agents. However, most of the hCES2A inhibitors show poor cell-membrane permeability and poor specificity. Herein, guided by the structure activity relationships (SAR) of fifteen natural alkaloids against hCES2A, fifteen new seven-membered ring berberine analogues were designed and synthesized, and their anti-hCES2A activities were evaluated. Among all tested compounds, compound 28 showed potent anti-hCES2A effect (IC50 = 1.66 μM) and excellent selectivity over hCES1A (IC50 > 100 μM). The SAR analysis revealed that the seven-membered ring of these berberine analogues was a crucial moiety for hCES2A inhibition, while the secondary amine group of the ring-C is important for improving their specificity over other serine hydrolases. Inhibition kinetic analyses and molecular dynamic simulation demonstrated that 28 strongly inhibited hCES2A in a mixed-inhibition manner, with an estimated Ki value of 1.035 μM. Moreover, 28 could inhibit intracellular hCES2A in living HepG2 cells and exhibited suitable metabolic stability. Collectively, the SAR of seven-membered ring berberine analogues as hCES2A inhibitors were studied, while compound 28 acted as a promising candidate for developing highly selective hCES2A inhibitors.
Collapse
Affiliation(s)
- Yun Yang
- School of Pharmacy, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Yuan Xiong
- School of Pharmacy, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guanghao Zhu
- School of Pharmacy, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mengru Sun
- School of Pharmacy, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Kun Zou
- School of Pharmacy, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yitian Zhao
- School of Pharmacy, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yong Zhang
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiming Li
- School of Pharmacy, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weiliang Zhu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qi Jia
- School of Pharmacy, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Bo Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Guangbo Ge
- School of Pharmacy, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
24
|
Hydrolysis of dibutyl phthalate and di(2-ethylhexyl) phthalate in human liver, small intestine, kidney, and lung: An in vitro analysis using organ subcellular fractions and recombinant carboxylesterases. Chem Biol Interact 2023; 372:110353. [PMID: 36657734 DOI: 10.1016/j.cbi.2023.110353] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/28/2022] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
Phthalates are widely used plasticizers that are primarily and rapidly metabolized to monoester phthalates in mammals. In the present study, the hydrolysis of dibutyl phthalate (DBP) and di(2-ethylhexyl) phthalate (DEHP) in the human liver, small intestine, kidney, and lung was examined by the catalytic, kinetic, and inhibition analyses using organ microsomal and cytosolic fractions and recombinant carboxylesterases (CESs). The Vmax (y-intercept) values based on the Eadie-Hofstee plots of DBP hydrolysis were liver > small intestine > kidney > lung in microsomes, and liver > small intestine > lung > kidney in cytosol, respectively. The CLint values (x-intercept) were small intestine > liver > kidney > lung in both microsomes and cytosol. The Vmax and CLint or CLmax values of DEHP hydrolysis were small intestine > liver > kidney > lung in both microsomes and cytosol. Bis(4-nitrophenyl) phosphate (BNPP) effectively inhibited the activities of DBP and DEHP hydrolysis in the microsomes and cytosol of liver, small intestine, kidney, and lung. Although physostigmine also potently inhibited DBP and DEHP hydrolysis activities in both the microsomes and cytosol of the small intestine and kidney, the inhibitory effects in the liver and lung were weak. In recombinant CESs, the Vmax values of DBP hydrolysis were CES1 (CES1b, CES1c) > CES2, whereas the CLmax values were CES2 > CES1 (CES1b, CES1c). On the other hand, the Vmax and CLmax values of DEHP hydrolysis were CES2 > CES1 (CES1b, CES1c). These results suggest an extensive organ-dependence of DBP and DEHP hydrolysis due to CES expression, and that CESs are responsible for the metabolic activation of phthalates.
Collapse
|
25
|
Chen Z, Yu J, Sun K, Song J, Chen L, Jiang Y, Wang Z. Rational design of a turn-on near-infrared fluorescence probe for the highly sensitive and selective monitoring of carboxylesterase 2 in living systems. Analyst 2023; 148:876-887. [PMID: 36661088 DOI: 10.1039/d2an01874h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In vivo selective fluorescence imaging of carboxylesterase 2 (CES2) remains a great challenge because existing fluorescence probes can potentially suffer from interference by other hydrolases. In addition, some fluorescent probes that have been separately reported for measuring CES2 activity in vitro are affected by autofluorescence and absorption of the biological matrix due to their limited emission wavelength or short Stokes shift. Herein, based on the substrate preference and catalytic performance of CES2, a novel and NIR fluorescent probe was developed, in which a hemi-cyanine dye ester derivative was used as the basic fluorescent group. In the presence of CES2, the probe was hydrolyzed to expose the fluorophore CZX-OH (λabs ∼ 675 nm, λem ∼ 850 nm), which led to a notable red-shift in the fluorescence (∼175 nm) spectrum. Confocal imaging of cells and live mice demonstrated that the fluorescent signal of this probe was related to the real activities of CES2 in cancer cells. All these results will powerfully promote the screening of CES2 regulators and the analysis of CES2-related physiological and pathological processes.
Collapse
Affiliation(s)
- Zhixin Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Jiaying Yu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Kai Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Jia Song
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Lucheng Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Yong Jiang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Zhifei Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| |
Collapse
|
26
|
Dai X, Yu F, Jiang Z, Dong B, Kong X. A fast fluorescent probe for tracing endoplasmic reticulum-located carboxylesterase in living cells. LUMINESCENCE 2022; 37:2067-2073. [PMID: 36200455 DOI: 10.1002/bio.4392] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 12/14/2022]
Abstract
Carboxylesterase (CEs), mainly localized in endoplasmic reticulum (ER), are responsible for hydrolyzing compounds containing various ester bonds. They have been closely associated with drug metabolism and cellular homeostasis. Although some CE fluorescent probes have been developed, there are still a lack of probes that could target to the ER. Here, we developed a novel fluorescent probe CR with a specific ER anchor for monitoring CEs. In CR, p-toluenesulfonamide was chosen for precise ER targeting. A simple acetyl moiety was used as the CE response site and fluorescence modulation unit. During the spectral tests, CR displayed a fast response speed (within 10 s) towards CEs. In addition, it showed high sensitivity [limit of detection (LOD) = 5.1 × 10-3 U/ml] and high selectivity with CEs. In biological imaging, probe CR could especially locate in the ER in HepG2 cells. After cells were treated with orilistat, CR succeeded in monitoring the changes in the CEs. Importantly, CR also had the ability to trace the changes in CEs in a tunicamycin-induced ER stress model. Therefore, probe CR could be a powerful molecular tool for further investigating the functions of CEs in the ER.
Collapse
Affiliation(s)
- Xiaoyu Dai
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| | - Faqi Yu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| | - Zekun Jiang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| | - Baoli Dong
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| | - Xiuqi Kong
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| |
Collapse
|
27
|
Yuk IH, Koulis T, Doshi N, Gregoritza K, Hediger C, Lebouc-Haefliger V, Giddings J, Khan TA. Formulation mitigations for particle formation induced by enzymatic hydrolysis of polysorbate 20 in protein-based drug products: insights from a full-factorial longitudinal study. AAPS OPEN 2022. [DOI: 10.1186/s41120-022-00064-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Hydrolytic degradation of the polysorbate 20 (PS20) surfactant in protein-based liquid formulations releases free fatty acids (FFAs), which can accumulate to form particles in drug products during real-time (long-term) storage. To identify formulation conditions that mitigate the risk of particle formation, we conducted a longitudinal study using purified recombinant monoclonal antibody (mAb) formulated in 24 conditions. In this real-time stability study at 5 °C, three key formulation parameters—mAb concentration, initial PS20 concentration, and pH—were varied across representative ranges in a full-factorial design. A longitudinal regression analysis was used to evaluate the effects of these parameters and their interactions on PS20 degradation (via measurements of PS20, FFAs, and PS20 ester distribution) and on particle formation (via visible particle observations and subvisible particle counts). The time-dependent onset of visible particles trended with the rise in subvisible particle counts and FFA levels and fall in PS20 concentration. In the ranges studied here, lower mAb concentration and higher initial PS20 concentration delayed the onset of particles, whereas pH had a negligible effect. These observations were consistent with the general trends predicted by our previously published FFA solubility model. Taken together, these findings highlight the complex relationships between formulation parameters, PS20 degradation, and particle formation.
Collapse
|
28
|
Zhao J, Tang Z, Selvaraju M, Johnson KA, Douglas JT, Gao PF, Petrassi HM, Wang MZ, Wang J. Cellular Target Deconvolution of Small Molecules Using a Selection-Based Genetic Screening Platform. ACS CENTRAL SCIENCE 2022; 8:1424-1434. [PMID: 36313155 PMCID: PMC9615120 DOI: 10.1021/acscentsci.2c00609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Indexed: 05/04/2023]
Abstract
Small-molecule drug target identification is an essential and often rate-limiting step in phenotypic drug discovery and remains a major challenge. Here, we report a novel platform for target identification of activators of signaling pathways by leveraging the power of a clustered regularly interspaced short palindromic repeats (CRISPR) knockout library. This platform links the expression of a suicide gene to the small-molecule-activated signaling pathway to create a selection system. With this system, loss-of-function screening using a CRISPR single-guide (sg) RNA library positively enriches cells in which the target has been knocked out. The identities of the drug targets and other essential genes required for the activity of small molecules of interest are then uncovered by sequencing. We tested this platform on BDW568, a newly discovered type-I interferon signaling activator, and identified stimulator of interferon genes (STING) as its target and carboxylesterase 1 (CES1) to be a key metabolizing enzyme required to activate BDW568 for target engagement. The platform we present here can be a general method applicable for target identification for a wide range of small molecules that activate different signaling pathways.
Collapse
Affiliation(s)
- Junxing Zhao
- Department
of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Zhichao Tang
- Department
of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Manikandan Selvaraju
- Department
of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Kristen A. Johnson
- Calibr,
Scripps Research Institute, La Jolla, California 92037, United States
| | - Justin T. Douglas
- Nuclear
Magnetic Resonance Laboratory, University
of Kansas, Lawrence, Kansas 66047, United States
| | - Philip F. Gao
- Protein
Production Group, University of Kansas, Lawrence, Kansas 66047, United States
| | - H. Michael Petrassi
- Calibr,
Scripps Research Institute, La Jolla, California 92037, United States
| | - Michael Zhuo Wang
- Department
of Pharmaceutical Chemistry, University
of Kansas, Lawrence, Kansas 66047, United States
| | - Jingxin Wang
- Department
of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| |
Collapse
|
29
|
Imai T, Isozaki M, Ohura K. Esterases Involved in the Rapid Bioconversion of Esmolol after Intravenous Injection in Humans. Biol Pharm Bull 2022; 45:1544-1552. [PMID: 36184514 DOI: 10.1248/bpb.b22-00468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Esmolol is indicated for the acute and temporary control of ventricular rate due to its rapid onset of action and elimination at a rate greater than cardiac output. This rapid elimination is achieved by the hydrolysis of esmolol to esmolol acid. It has previously been reported that esmolol is hydrolyzed in the cytosol of red blood cells (RBCs). In order to elucidate the metabolic tissues and enzymes involved in the rapid elimination of esmolol, a hydrolysis study was performed using different fractions of human blood and liver. Esmolol was slightly hydrolyzed by washed RBCs and plasma proteins while it was extensively hydrolyzed in plasma containing white blood cells and platelets. The negligible hydrolysis of esmolol in RBCs is supported by its poor hydrolysis by esterase D, the sole cytosolic esterase in RBCs. In human liver microsomes, esmolol was rapidly hydrolyzed according to Michaelis-Menten kinetics, and its hepatic clearance, calculated by the well-stirred model, was limited by hepatic blood flow. An inhibition study and a hydrolysis study using individual recombinant esterases showed that human carboxylesterase 1 isozyme (hCE1) is the main metabolic enzyme of esmolol in both white blood cells and human liver. These studies also showed that acyl protein thioesterase 1 (APT1) is involved in the cytosolic hydrolysis of esmolol in the liver. The hydrolysis of esmolol by hCE1 and APT1 also results in its pulmonary metabolism, which might be a reason for its high total clearance (170-285 mL/min/kg bodyweight), 3.5-fold greater than cardiac output (80.0 mL/min/kg bodyweight).
Collapse
Affiliation(s)
- Teruko Imai
- Graduate School of Pharmaceutical Sciences, Kumamoto University.,Daiichi University of Pharmacy
| | - Mizuki Isozaki
- Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Kayoko Ohura
- Graduate School of Pharmaceutical Sciences, Kumamoto University.,Headquarters for Admissions and Education, Kumamoto University
| |
Collapse
|
30
|
Liu J, Yao B, Gao L, Zhang Y, Huang S, Wang X. Emerging role of carboxylesterases in nonalcoholic fatty liver disease. Biochem Pharmacol 2022; 205:115250. [PMID: 36130649 DOI: 10.1016/j.bcp.2022.115250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/02/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is increasingly recognized as a global public health problem. Carboxylesterases (CESs), as potential influencing factors of NAFLD, are very important to improve clinical outcomes. This review aims to deeply understand the role of CESs in the progression of NAFLD and proposes that CESs can be used as potential targets for NAFLD treatment. We first introduced CESs and analyzed the relationship between CESs and hepatic lipid metabolism and inflammation. Then, we further reviewed the regulation of nuclear receptors on CESs, including PXR, CAR, PPARα, HNF4α and FXR, which may influence the progression of NAFLD. Finally, we evaluated the advantages and disadvantages of existing NAFLD animal models and summarized the application of CES-related animal models in NAFLD research. In general, this review provides an overview of the relationship between CESs and NAFLD and discusses the role and potential value of CESs in the treatment and prevention of NAFLD.
Collapse
Affiliation(s)
- Jie Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Liangcai Gao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China.
| |
Collapse
|
31
|
Formulation and administration of ramipril prodrug for improving bioactivity significantly: In vitro and in vivo correlation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
32
|
Kailass K, Sadovski O, Zipfel WR, Beharry AA. Two-Photon Photodynamic Therapy Targeting Cancers with Low Carboxylesterase 2 Activity Guided by Ratiometric Fluorescence. J Med Chem 2022; 65:8855-8868. [PMID: 35700557 DOI: 10.1021/acs.jmedchem.1c01965] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human carboxylesterase 2 (hCES2) converts anticancer prodrugs, such as irinotecan, into their active metabolites via phase I drug metabolism. Owing to interindividual variability, hCES2 serves as a predictive marker of patient response to hCES2-activated prodrug-based therapy, whereby a low intratumoral hCES2 activity leads to therapeutic resistance. Despite the ability to identify nonresponders, effective treatments for resistant patients are needed. Clinically approved photodynamic therapy is an attractive alternative for irinotecan-resistant patients. Here, we describe the application of our hCES2-selective small-molecule ratiometric fluorescent chemosensor, Benz-AP, as a single theranostic agent given its discovered functionality as a photosensitizer. Benz-AP produces singlet oxygen and induces photocytotoxicity in cancer cells in a strong negative correlation with hCES2 activity. Two-photon excitation of Benz-AP produces fluorescence, singlet oxygen, and photocytotoxicity in tumor spheroids. Overall, Benz-AP serves as a novel theranostic agent with selective photocytotoxicity in hCES2-prodrug resistant cancer cells, making Benz-AP a promising agent for in vivo applications.
Collapse
Affiliation(s)
- Karishma Kailass
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Oleg Sadovski
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Warren R Zipfel
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Andrew A Beharry
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| |
Collapse
|
33
|
Lim IH, Lee SJ, Shin BS, Kang HG. Ilaprazole and Clopidogrel Resistance in Acute Stroke Patients. Biomedicines 2022; 10:biomedicines10061366. [PMID: 35740386 PMCID: PMC9219695 DOI: 10.3390/biomedicines10061366] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 01/01/2023] Open
Abstract
Clopidogrel, an antiplatelet agent used for secondary prevention of cerebrovascular diseases, is often taken with proton pump inhibitors (PPIs). Generally, the combined use of clopidogrel and PPIs causes adverse drug–drug interactions. VerifyNow is a quick and convenient method to confirm clopidogrel resistance (CR), which compromises adequate antithrombotic effects. We aimed to confirm CR, identify its factors, and determine the influence of the combination of ilaprazole and clopidogrel on clopidogrel using VerifyNow. In this retrospective study, we examined patients who were receiving clopidogrel after three months, starting within one week from the onset of cerebral infarction symptoms. Clinical records, imaging records, and diagnostic laboratory results, including P2Y12 reaction units (PRU), were compared and analyzed to check for CR. Additionally, the groups treated with either both ilaprazole and clopidogrel or with medications other than ilaprazole were comparatively analyzed. CR was defined as a PRU ≥240 after clopidogrel for three months. Among factors influencing CR by affecting clopidogrel metabolism, positive statistical correlations with age and alcohol consumption were confirmed. The diagnostic tests revealed a lower glomerular filtration rate and platelet count of the CR-positive group. This finding proved that the combination therapy of ilaprazole and clopidogrel is safe, as it does not interfere with the metabolism of clopidogrel.
Collapse
Affiliation(s)
- In Hwan Lim
- Department of Pharmacology, School of Medicine, Wonkwang University, Iksan 54538, Korea;
| | - Seung Jae Lee
- Department of Chemistry, Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54907, Korea;
| | - Byoung-Soo Shin
- Department of Neurology, Jeonbuk National University, Jeonju 54907, Korea;
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54907, Korea
- Biomedical Research Institute, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Korea
| | - Hyun Goo Kang
- Department of Neurology, Jeonbuk National University, Jeonju 54907, Korea;
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54907, Korea
- Biomedical Research Institute, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Korea
- Correspondence: ; Tel.: +82-63-250-1590
| |
Collapse
|
34
|
Shervington LA, Ingham O. Investigating the Stability of Six Phenolic TMZ Ester Analogues, Incubated in the Presence of Porcine Liver Esterase and Monitored by HPLC. Molecules 2022; 27:2958. [PMID: 35566308 PMCID: PMC9103334 DOI: 10.3390/molecules27092958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/30/2022] Open
Abstract
Previous published data from our group showed the encouraging in vitro activities of six phenolic temozolomide (TMZ) ester analogues (ES8-ES12 and ES14) with up to a five-fold increase in potency compared to TMZ against glioblastoma multiform cell lines and TMZ-resistant O6-methylguanine-DNA methyl transferase (MGMT)-positive primary cells. This study investigated the stabilities of the six phenolic TMZ ester analogues in the presence of porcine liver esterase (PLE) as a hydrolytic enzyme, using high-performance liquid chromatography (HPLC), monitored by a diode-array detector (DAD). Determining the rates of hydrolysis of the esters provided a useful insight into the feasibility of progressing them to the next phase of drug development. Fifty percent of TMZ esters consisting of para nitro, chloro, phenyl and tolyl groups (ES9, ES10, ES12 and ES14) were hydrolysed within the first 4.2 min of PLE exposure, while the TMZ esters consisting of para methoxy and nitrile groups (ES8 and ES11) demonstrated increased stability, with 50% hydrolysis achieved in 7.3 and 13.7 min, respectively. In conclusion, the survival of these phenolic TMZ esters on route to the target site of a brain tumor would be a challenge, mainly due to the undesirable rapid rate of hydrolysis. These findings therefore pose a question regarding the effectiveness of these esters in an in vivo setting.
Collapse
Affiliation(s)
- Leroy A. Shervington
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK
| | - Oliver Ingham
- Kindeva Drug Delivery, Bakewell Road, Loughborough LE11 5RB, UK;
| |
Collapse
|
35
|
Swindell WR, Bojanowski K, Chaudhuri RK. Transcriptomic Analysis of Fumarate Compounds Identifies Unique Effects of Isosorbide Di-(Methyl Fumarate) on NRF2, NF-kappaB and IRF1 Pathway Genes. Pharmaceuticals (Basel) 2022; 15:ph15040461. [PMID: 35455458 PMCID: PMC9026097 DOI: 10.3390/ph15040461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/09/2022] [Accepted: 04/10/2022] [Indexed: 11/16/2022] Open
Abstract
Dimethyl fumarate (DMF) has emerged as a first-line therapy for relapsing-remitting multiple sclerosis (RRMS). This treatment, however, has been limited by adverse effects, which has prompted development of novel derivatives with improved tolerability. We compared the effects of fumarates on gene expression in astrocytes. Our analysis included diroximel fumarate (DRF) and its metabolite monomethyl fumarate (MMF), along with a novel compound isosorbide di-(methyl fumarate) (IDMF). Treatment with IDMF resulted in the largest number of differentially expressed genes. The effects of DRF and MMF were consistent with NRF2 activation and NF-κB inhibition, respectively. IDMF responses, however, were concordant with both NRF2 activation and NF-κB inhibition, and we confirmed IDMF-mediated NF-κB inhibition using a reporter assay. IDMF also down-regulated IRF1 expression and IDMF-decreased gene promoters were enriched with IRF1 recognition sequences. Genes altered by each fumarate overlapped significantly with those near loci from MS genetic association studies, but IDMF had the strongest overall effect on MS-associated genes. These results show that next-generation fumarates, such as DRF and IDMF, have effects differing from those of the MMF metabolite. Our findings support a model in which IDMF attenuates oxidative stress via NRF2 activation, with suppression of NF-κB and IRF1 contributing to mitigation of inflammation and pyroptosis.
Collapse
Affiliation(s)
- William R. Swindell
- Department of Internal Medicine, The Jewish Hospital, Cincinnati, OH 45236, USA
- Correspondence:
| | - Krzysztof Bojanowski
- Sunny BioDiscovery Inc., Santa Paula, CA 93060, USA;
- Symbionyx Pharmaceuticals Inc., Boonton, NJ 07005, USA;
| | - Ratan K. Chaudhuri
- Symbionyx Pharmaceuticals Inc., Boonton, NJ 07005, USA;
- Sytheon Ltd., Boonton, NJ 07005, USA
| |
Collapse
|
36
|
Diurnal Variation in Biomarkers of Exposure to Endocrine-Disrupting Chemicals and Their Association with Oxidative Damage in Norwegian Adults: The EuroMix Study. TOXICS 2022; 10:toxics10040181. [PMID: 35448442 PMCID: PMC9028082 DOI: 10.3390/toxics10040181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/27/2022] [Accepted: 04/02/2022] [Indexed: 11/23/2022]
Abstract
Much evidence on the adverse health effects of endocrine-disrupting chemicals (EDCs) has accumulated during recent decades. EDCs are commonly found in various foods and personal care products (PCP). Data documenting a diurnally varying EDC metabolism in humans is scarce. This study examined (i) the time-of-day effect on the diurnal magnitude and variance of urinary biomarkers of exposure to EDCs, and (ii) the association between EDC exposures and oxidative damage in a Norwegian adult subpopulation. This was a cross-sectional panel study using biobanked samples from the EuroMix project. During a typical weekday, participants were asked to collect all day’s urine voids and record dietary and PCP habitual uses in a diary. Collected time stamps of urine voids were classified into three distinct periods in the day (morning 6 a.m.−12 p.m., mid-day 12 p.m.−6 p.m., evening 6 p.m.−6 a.m.). Questionnaires regarding demographic characteristics, personal care product usage, and dietary habits were completed. Urinary levels of EDCs (phthalates, parabens, and bisphenols) were measured using mass spectrometry and adjusted for urinary volume using specific gravity. Urinary 4-hydroxynonenal (4HNE), a lipid peroxidation marker, was measured using an immunoassay kit. Linear mixed-effect models identified EDCs under the influence of a diurnal variation effect that was adjusted for dietary habits and PCP use and examined associations between EDC and 4HNE. p-values were FDR-adjusted. Most phthalates appeared to be diurnally varying with higher urinary levels towards the evening (q < 0.001) than those measured during mid-day; this strong diurnal variation effect was not present for parabens and bisphenols. Significant (q < 0.001) positive associations were observed between all phthalates, parabens, and bisphenols (except bisphenol S) and 4HNE. This study’s findings highlighted the diurnal variation of excretion for certain EDC, but not for others, in real-life conditions. The degree of EDC chronotoxicity in distinct diurnal windows of the day warrants further investigation with longitudinal human studies.
Collapse
|
37
|
Li J, Shi J, Xiao J, Tran L, Wang X, Zhu HJ. Contributions of Cathepsin A and Carboxylesterase 1 to the Hydrolysis of Tenofovir Alafenamide in the Human Liver, and the Effect of CES1 Genetic Variation on Tenofovir Alafenamide Hydrolysis. Drug Metab Dispos 2022; 50:243-248. [PMID: 34933885 PMCID: PMC8969131 DOI: 10.1124/dmd.120.000323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 12/10/2021] [Indexed: 11/22/2022] Open
Abstract
The prodrug tenofovir alafenamide (TAF) is a first-line antiviral agent for the treatment of chronic hepatitis B infection. TAF activation involves multiple steps, and the first step is an ester hydrolysis reaction catalyzed by hydrolases. This study was to determine the contributions of carboxylesterase 1 (CES1) and cathepsin A (CatA) to TAF hydrolysis in the human liver. Our in vitro incubation studies showed that both CatA and CES1 catalyzed TAF hydrolysis in a pH-dependent manner. At their physiologic pH environment, the activity of CatA (pH 5.2) was approximately 1,000-fold higher than that of CES1 (pH 7.2). Given that the hepatic protein expression of CatA was approximately 200-fold lower than that of CES1, the contribution of CatA to TAF hydrolysis in the human liver was estimated to be much greater than that of CES1, which is contrary to the previous perception that CES1 is the primary hepatic enzyme hydrolyzing TAF. The findings were further supported by a TAF incubation study with the CatA inhibitor telaprevir and the CES1 inhibitor bis-(p-nitrophenyl) phosphate. Moreover, an in vitro study revealed that the CES1 variant G143E (rs71647871) is a loss-of-function variant for CES1-mediated TAF hydrolysis. In summary, our results suggest that CatA may play a more important role in the hepatic activation of TAF than CES1. Additionally, TAF activation in the liver could be affected by CES1 genetic variation, but the magnitude of impact appears to be limited due to the major contribution of CatA to hepatic TAF activation. SIGNIFICANCE STATEMENT: Contrary to the general perception that carboxylesterase 1 (CES1) is the major enzyme responsible for tenofovir alafenamide (TAF) hydrolysis in the human liver, the present study demonstrated that cathepsin A may play a more significant role in TAF hepatic hydrolysis. Furthermore, the CES1 variant G143E (rs71647871) was found to be a loss-of-function variant for CES1-mediated TAF hydrolysis.
Collapse
Affiliation(s)
- Jiapeng Li
- Department of Clinical Pharmacy (J.L., H.-J. Z.), Department of Pharmaceutical Sciences (J.X.), and College of Pharmacy (L.T.), University of Michigan, Ann Arbor, Michigan; Alliance Pharma, Inc, Malvern, Pennsylvania (J.S.); and Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (X.W.)
| | - Jian Shi
- Department of Clinical Pharmacy (J.L., H.-J. Z.), Department of Pharmaceutical Sciences (J.X.), and College of Pharmacy (L.T.), University of Michigan, Ann Arbor, Michigan; Alliance Pharma, Inc, Malvern, Pennsylvania (J.S.); and Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (X.W.)
| | - Jingcheng Xiao
- Department of Clinical Pharmacy (J.L., H.-J. Z.), Department of Pharmaceutical Sciences (J.X.), and College of Pharmacy (L.T.), University of Michigan, Ann Arbor, Michigan; Alliance Pharma, Inc, Malvern, Pennsylvania (J.S.); and Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (X.W.)
| | - Lana Tran
- Department of Clinical Pharmacy (J.L., H.-J. Z.), Department of Pharmaceutical Sciences (J.X.), and College of Pharmacy (L.T.), University of Michigan, Ann Arbor, Michigan; Alliance Pharma, Inc, Malvern, Pennsylvania (J.S.); and Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (X.W.)
| | - Xinwen Wang
- Department of Clinical Pharmacy (J.L., H.-J. Z.), Department of Pharmaceutical Sciences (J.X.), and College of Pharmacy (L.T.), University of Michigan, Ann Arbor, Michigan; Alliance Pharma, Inc, Malvern, Pennsylvania (J.S.); and Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (X.W.)
| | - Hao-Jie Zhu
- Department of Clinical Pharmacy (J.L., H.-J. Z.), Department of Pharmaceutical Sciences (J.X.), and College of Pharmacy (L.T.), University of Michigan, Ann Arbor, Michigan; Alliance Pharma, Inc, Malvern, Pennsylvania (J.S.); and Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (X.W.)
| |
Collapse
|
38
|
Chen Y, Capello M, Rios Perez MV, Vykoukal JV, Roife D, Kang Y, Prakash LR, Katayama H, Irajizad E, Fleury A, Ferri-Borgogno S, Baluya DL, Dennison JB, Do KA, Fiehn O, Maitra A, Wang H, Chiao PJ, Katz MHG, Fleming JB, Hanash SM, Fahrmann JF. CES2 sustains HNF4α expression to promote pancreatic adenocarcinoma progression through an epoxide hydrolase-dependent regulatory loop. Mol Metab 2022; 56:101426. [PMID: 34971802 PMCID: PMC8841288 DOI: 10.1016/j.molmet.2021.101426] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/16/2021] [Accepted: 12/23/2021] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Intra-tumoral expression of the serine hydrolase carboxylesterase 2 (CES2) contributes to the activation of the pro-drug irinotecan in pancreatic ductal adenocarcinoma (PDAC). Given other potential roles of CES2, we assessed its regulation, downstream effects, and contribution to tumor development in PDAC. METHODS Association between the mRNA expression of CES2 in pancreatic tumors and overall survival was assessed using The Cancer Genome Atlas. Cell viability, clonogenic, and anchorage-independent growth assays as well as an orthotopic mouse model of PDAC were used to evaluate the biological relevance of CES2 in pancreatic cancer. CES2-driven metabolic changes were determined by untargeted and targeted metabolomic analyses. RESULTS Elevated tumoral CES2 mRNA expression was a statistically significant predictor of poor overall survival in PDAC patients. Knockdown of CES2 in PDAC cells reduced cell viability, clonogenic capacity, and anchorage-independent growth in vitro and attenuated tumor growth in an orthotopic mouse model of PDAC. Mechanistically, CES2 was found to promote the catabolism of phospholipids resulting in HNF4α activation through a soluble epoxide hydrolase (sEH)-dependent pathway. Targeting of CES2 via siRNA or small molecule inhibitors attenuated HNF4α protein expression and reduced gene expression of classical/progenitor markers and increased basal-like markers. Targeting of the CES2-sEH-HNF4α axis using small molecule inhibitors of CES2 or sEH reduced cell viability. CONCLUSIONS We establish a novel regulatory loop between CES2 and HNF4α to sustain the progenitor subtype and promote PDAC progression and highlight the potential utility of CES2 or sEH inhibitors for the treatment of PDAC as part of non-irinotecan-containing regimens.
Collapse
Affiliation(s)
- Yihui Chen
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michela Capello
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mayrim V Rios Perez
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jody V Vykoukal
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Roife
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ya'an Kang
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laura R Prakash
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hiroyuki Katayama
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ehsan Irajizad
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alia Fleury
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sammy Ferri-Borgogno
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dodge L Baluya
- Departments of Center for Radiation Oncology Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer B Dennison
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim-Anh Do
- Departments of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Oliver Fiehn
- UC Davis Genome Center - Metabolomics, University of California, Davis, 95616, CA, USA
| | - Anirban Maitra
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA; Departments of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huamin Wang
- Departments of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul J Chiao
- Departments of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew H G Katz
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Samir M Hanash
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Johannes F Fahrmann
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
39
|
Investigation of the effect of verapamil on the regional absorption of sofosbuvir from rabbit intestine in situ. Daru 2022; 30:49-58. [PMID: 35023081 PMCID: PMC9114277 DOI: 10.1007/s40199-021-00429-1] [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: 07/25/2021] [Accepted: 12/05/2021] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Sofosbuvir, a nucleotide antiviral drug, is a Biopharmaceutics Classification System (BCS) class III prodrug suffering from limited intestinal absorption due to its high hydrophilicity and low intestinal permeability. This research aims to investigate the luminal stability of Sofosbuvir, the influence of anatomical site on its intestinal absorption and the effects of verapamil on such absorption. METHOD The study utilized in situ rabbit intestinal perfusion technique to examine absorption of Sofosbuvir from duodenum, jejunum, ileum and ascending colon. This was conducted both with and without verapamil. RESULTS The luminal stability study showed that Sofosbuvir was subjected to premature degradation with varying fractions degraded from the different intestinal segments. The in situ perfusion data showed incomplete absorption of Sofosbuvir from small and large intestinal segments. The recorded values of the absorptive clearance per unit length (Pe.A/L) of Sofosbuvir were 0.026, 0.0075, 0.0026, & 0.054 ml/min.cm for duodenum, jejunum, ileum, and ascending colon, respectively. The Pe.A/L values were ordered as colon > duodenum > jejunum > ileum. This is the opposite rank of P-gp content in the different intestinal segments. The recorded values of the length required for complete Sofosbuvir absorption (L95%) were 29.58, 128.47, 949.2 and, 13.63 cm for duodenum, jejunum, ileum, and ascending colon, respectively. Co-perfusion with verapamil significantly increased Pe.A/L and reduced the L95% of Sofosbuvir from both jejunum and ileum (P-value < 0.05). CONCLUSION The results indicated that the absorptive clearance of Sofosbuvir was site dependent and associated with the content of P-glycoprotein, in addition to the expected drug interactions that can occur in polymedicated hepatitis C virus (HCV) infected patients.
Collapse
|
40
|
Johnson JL, Huang J, Rooney M, Gu C. Optimal pH 8.5 to 9 for the Hydrolysis of Vixotrigine and Other Basic Substrates of Carboxylesterase-1 in Human Liver Microsomes. Xenobiotica 2021; 52:105-112. [PMID: 34904522 DOI: 10.1080/00498254.2021.2018629] [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] [Indexed: 10/19/2022]
Abstract
Vixotrigine is a voltage- and use-dependent sodium channel blocker under investigation for the potential treatment of neuropathic pain. One of the major in vivo metabolic pathways of vixotrigine in humans is the hydrolysis of the carboxamide to form the carboxylic acid metabolite M14.The in vitro formation of M14 in human hepatocytes was inhibited by the carboxylesterase (CES) inhibitor Bis(4-nitrophenyl) phosphate in a concentration-dependent manner. The hydrolysis reaction was identified to be catalyzed by recombinant human CES1b.Initial observation of only trace level formation of M14 in human liver microsomes at pH 7.4 caused us to doubt the involvement of CES1, an enzyme localized at the endoplasmic reticulum and the dominant carboxylesterase in human liver. Further investigation has revealed that optimal pH for the hydrolysis of vixotrigine and two other basic substrates of CES1, methylphenidate and oseltamivir, in human liver microsomes was pH 8.5 to 9 which is higher than their respective pKa(base), suggesting that neutral form of basic substrates is probably preferred for CES1 catalysis in liver microsomes.
Collapse
Affiliation(s)
- Joshua L Johnson
- Drug Metabolism and Pharmacokinetics.,Current affiliation of JLJ: Drug Metabolism and Pharmacokinetics, Takeda, San Diego, CA, USA
| | | | - Michael Rooney
- Clinical Pharmacology and Pharmacometrics, Biogen, Cambridge, MA, USA
| | | |
Collapse
|
41
|
Dätwyler P, Jiang X, Wagner B, Varga N, Mühlethaler T, Hostettler K, Rabbani S, Schwardt O, Ernst B. Prodrugs of E-selectin Antagonists with Enhanced Pharmacokinetic Properties. ChemMedChem 2021; 17:e202100634. [PMID: 34870892 DOI: 10.1002/cmdc.202100634] [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: 09/28/2021] [Indexed: 11/10/2022]
Abstract
Because of their large polar surface area, carbohydrates often exhibit insufficient pharmacokinetic properties. Specifically, the carboxylic acid function of the tetrasaccharide sialyl Lewisx , a pharmacophore crucial for the formation of a salt bridge with selectins, prevents oral availability. A common approach is the transfer of carboxylic acid into ester prodrugs. Once the prodrug is either actively or passively absorbed, the active principle is released by hydrolysis. In the present study, ester prodrugs of selectin antagonists with aliphatic promoieties were synthesized and their potential for oral availability was investigated in vitro and in vivo. The addition of lipophilic ester moieties to overcome insufficient lipophilicity improved passive permeation into enterocytes, however at the same time supported efflux back to the small intestines as well as oxidation into non-hydrolysable metabolites. In summary, our examples demonstrate that different modifications of carbohydrates can result in opposing effects and have to be studied in their entirety.
Collapse
Affiliation(s)
- Philipp Dätwyler
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Xiaohua Jiang
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Beatrice Wagner
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Norbert Varga
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Tobias Mühlethaler
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Katja Hostettler
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Said Rabbani
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Oliver Schwardt
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Beat Ernst
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| |
Collapse
|
42
|
Shi J, Xiao J, Wang X, Jung SM, Bleske BE, Markowitz JS, Patrick KS, Zhu HJ. Plasma Carboxylesterase 1 Predicts Methylphenidate Exposure: A Proof-of-Concept Study Using Plasma Protein Biomarker for Hepatic Drug Metabolism. Clin Pharmacol Ther 2021; 111:878-885. [PMID: 34743324 DOI: 10.1002/cpt.2486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/27/2021] [Indexed: 12/11/2022]
Abstract
Hepatic drug-metabolizing enzymes (DMEs) play critical roles in determining the pharmacokinetics and pharmacodynamics of numerous therapeutic agents. As such, noninvasive biomarkers capable of predicting DME expression in the liver have the potential to be used to personalize pharmacotherapy and improve drug treatment outcomes. In the present study, we quantified carboxylesterase 1 (CES1) protein concentrations in plasma samples collected during a methylphenidate pharmacokinetics study. CES1 is a prominent hepatic enzyme responsible for the metabolism of many medications containing small ester moieties, including methylphenidate. The results revealed a significant inverse correlation between plasma CES1 protein concentrations and the area under the concentration-time curves (AUCs) of plasma d-methylphenidate (P = 0.014, r = -0.617). In addition, when plasma CES1 protein levels were normalized to the plasma concentrations of 24 liver-enriched proteins to account for potential interindividual differences in hepatic protein release rate, the correlation was further improved (P = 0.003, r = -0.703), suggesting that plasma CES1 protein could explain ~ 50% of the variability in d-methylphenidate AUCs in the study participants. A physiologically-based pharmacokinetic modeling simulation revealed that the CES1-based individualized dosing strategy might significantly reduce d-methylphenidate exposure variability in pediatric patients relative to conventional trial and error fixed dosing regimens. This proof-of-concept study indicates that the plasma protein of a hepatic DME may serve as a biomarker for predicting its metabolic function and the pharmacokinetics of its substrate drugs.
Collapse
Affiliation(s)
- Jian Shi
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Jingcheng Xiao
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Xinwen Wang
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Sun Min Jung
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Barry E Bleske
- Department of Pharmacy Practice and Administrative Sciences, The University of New Mexico, Albuquerque, New Mexico, USA
| | - John S Markowitz
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida, USA
| | - Kennerly S Patrick
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Hao-Jie Zhu
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
43
|
Liu J, Shang X, Huang S, Xu Y, Lu J, Zhang Y, Liu Z, Wang X. Construction and Characterization of CRISPR/Cas9 Knockout Rat Model of Carboxylesterase 2a Gene. Mol Pharmacol 2021; 100:480-490. [PMID: 34503976 DOI: 10.1124/molpharm.121.000357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/30/2021] [Indexed: 11/22/2022] Open
Abstract
Carboxylesterase (CES) 2, an important metabolic enzyme, plays a critical role in drug biotransformation and lipid metabolism. Although CES2 is very important, few animal models have been generated to study its properties and functions. Rat Ces2 is similar to human CES2A-CES3A-CES4A gene cluster, with highly similar gene structure, function, and substrate. In this report, CRISPR-associated protein-9 (CRISPR/Cas9) technology was first used to knock out rat Ces2a, which is a main subtype of Ces2 mostly distributed in the liver and intestine. This model showed the absence of CES2A protein expression in the liver. Further pharmacokinetic studies of diltiazem, a typical substrate of CES2A, confirmed the loss of function of CES2A both in vivo and in vitro. At the same time, the expression of CES2C and CES2J protein in the liver decreased significantly. The body and liver weight of Ces2a knockout rats also increased, but the food intake did not change. Moreover, the deficiency of Ces2a led to obesity, insulin resistance, and liver fat accumulation, which are consistent with the symptoms of nonalcoholic fatty liver disease (NAFLD). Therefore, this rat model is not only a powerful tool to study drug metabolism mediated by CES2 but also a good disease model to study NAFLD. SIGNIFICANCE STATEMENT: Human carboxylesterase (CES) 2 plays a key role in the first-pass hydrolysis metabolism of most oral prodrugs as well as lipid metabolism. In this study, CRISPR/Cas9 technology was used to knock out Ces2a gene in rats for the first time. This model can be used not only in the study of drug metabolism and pharmacokinetics but also as a disease model of nonalcoholic fatty liver disease (NAFLD) and other metabolic disorders.
Collapse
Affiliation(s)
- Jie Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Xuyang Shang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Shengbo Huang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Yuan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Jian Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Yuanjin Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Zongjun Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| |
Collapse
|
44
|
Hoffmann C, Evcüman S, Neumaier F, Zlatopolskiy BD, Humpert S, Bier D, Holschbach M, Schulze A, Endepols H, Neumaier B. [ 18F]ALX5406: A Brain-Penetrating Prodrug for GlyT1-Specific PET Imaging. ACS Chem Neurosci 2021; 12:3335-3346. [PMID: 34449193 DOI: 10.1021/acschemneuro.1c00284] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Selective inhibition of glycine transporter 1 (GlyT1) has emerged as a potential approach to alleviate N-methyl-d-aspartate receptor (NMDAR) hypofunction in patients with schizophrenia and cognitive decline. ALX5407 is a potent and selective inhibitor of GlyT1 derived from the metabolic intermediate sarcosine (N-methylglycine) that showed antipsychotic potential in a number of animal models. Whereas clinical application of ALX5407 is limited by adverse effects on motor performance and respiratory function, a suitably radiolabeled drug could represent a promising PET tracer for the visualization of GlyT1 in the brain. Herein, [18F]ALX5407 and the corresponding methyl ester, [18F]ALX5406, were prepared by alcohol-enhanced copper mediated radiofluorination and studied in vitro in rat brain slices and in vivo in normal rats. [18F]ALX5407 demonstrated accumulation consistent with the distribution of GlyT1 in in vitro autoradiographic studies but no brain uptake in μPET experiments in naı̈ve rats. In contrast, the methyl ester [18F]ALX5406 rapidly entered the brain and was enzymatically transformed into [18F]ALX5407, resulting in a regional accumulation pattern consistent with GlyT1 specific binding. We conclude that [18F]ALX5406 is a promising and easily accessible PET probe for preclinical in vivo imaging of GlyT1 in the brain.
Collapse
Affiliation(s)
- Chris Hoffmann
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine, and University Hospital Cologne, 50937 Cologne, Germany
| | - Sibel Evcüman
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Felix Neumaier
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine, and University Hospital Cologne, 50937 Cologne, Germany
| | - Boris D. Zlatopolskiy
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine, and University Hospital Cologne, 50937 Cologne, Germany
| | - Swen Humpert
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Dirk Bier
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Marcus Holschbach
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Annette Schulze
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Heike Endepols
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine, and University Hospital Cologne, 50937 Cologne, Germany
- Nuclear Medicine Department, University of Cologne, Faculty of Medicine, and University Hospital Cologne, 50937 Cologne, Germany
| | - Bernd Neumaier
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine, and University Hospital Cologne, 50937 Cologne, Germany
- Max Planck Institute of Metabolism Research, 50931 Cologne, Germany
| |
Collapse
|
45
|
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
|
46
|
Dikmen G. Investigation of non-covalent complex formation between 2-(4-hydroxyphenylazo) benzoic acid and α-Cyclodextrin in solid and solution forms. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Singh A, Gao M, Beck MW. Human carboxylesterases and fluorescent probes to image their activity in live cells. RSC Med Chem 2021; 12:1142-1153. [PMID: 34355180 PMCID: PMC8292992 DOI: 10.1039/d1md00073j] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Human carboxylesterases (CESs) are serine hydrolases that are responsible for the phase I metabolism of an assortment of ester, amide, thioester, carbonate, and carbamate containing drugs. CES activity is known to be influenced by a variety of factors including single nucleotide polymorphisms, alternative splicing, and drug-drug interactions. These different factors contribute to interindividual variability of CES activity which has been demonstrated to influence clinical outcomes among people treated with CES-substrate therapeutics. Detailed exploration of the factors that influence CES activity is emerging as an important area of research. The use of fluorescent probes with live cell imaging techniques can selectively visualize the real-time activity of CESs and have the potential to be useful tools to help reveal the impacts of CES activity variations on human health. This review summarizes the properties of the five known human CESs including factors reported to or that could potentially influence their activity before discussing the design aspects and use considerations of CES fluorescent probes in general in addition to highlighting several well-characterized probes.
Collapse
Affiliation(s)
- Anchal Singh
- Department of Chemistry and Biochemistry, Eastern Illinois University Charleston IL 61920 USA +1 217 581 6227
| | - Mingze Gao
- Department of Biological Sciences, Eastern Illinois University Charleston IL 61920 USA
| | - Michael W Beck
- Department of Chemistry and Biochemistry, Eastern Illinois University Charleston IL 61920 USA +1 217 581 6227
| |
Collapse
|
48
|
Di Consiglio E, Darney K, Buratti FM, Turco L, Vichi S, Testai E, Lautz LS, Dorne JLCM. Human Variability in Carboxylesterases and carboxylesterase-related Uncertainty Factors for Chemical Risk Assessment. Toxicol Lett 2021; 350:162-170. [PMID: 34256091 DOI: 10.1016/j.toxlet.2021.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
Carboxylesterases (CES) are an important class of enzymes involved in the hydrolysis of a range of chemicals and show large inter-individual variability in vitro. An extensive literature search was performed to identify in vivo probe substrates for CES1 and CES2 together with their protein content and enzymatic activity. Human pharmacokinetic (PK) data on Cmax, clearance, and AUC were extracted from 89 publications and Bayesian meta-analysis was performed using a hierarchical model to derive CES-related variability distributions and related uncertainty factors (UF). The CES-related variability indicated that 97.5% of healthy adults are covered by the kinetic default UF (3.16), except for clopidogrel and dabigatran etexilate. Clopidogrel is metabolised for a small amount by the polymorphic CYP2C19, which can have an impact on the overall pharmacokinetics, while the variability seen for dabigatran etexilate might be due to differences in the absorption, since this can be influenced by food intake. The overall CES-related variability was moderate to high in vivo (<CV 50%), which might be due to possible polymorphism in the enzyme but also to the small sample size available per chemical. The presented CES-related variability can be used in combination with in vitro data to derive pathway-specific distributions.
Collapse
Affiliation(s)
- E Di Consiglio
- Istituto Superiore di Sanità, Environment & Health Department, Viale Regina Elena 299, Roma, Italy
| | - K Darney
- Risk Assessment Department, French Agency for Food, Environmental and Occupational Health & Safety (Anses), 14 rue Pierre et Marie Curie, Maisons-Alfort, F-94701, France.
| | - F M Buratti
- Istituto Superiore di Sanità, Environment & Health Department, Viale Regina Elena 299, Roma, Italy
| | - L Turco
- Istituto Superiore di Sanità, Environment & Health Department, Viale Regina Elena 299, Roma, Italy
| | - S Vichi
- Istituto Superiore di Sanità, Environment & Health Department, Viale Regina Elena 299, Roma, Italy
| | - E Testai
- Istituto Superiore di Sanità, Environment & Health Department, Viale Regina Elena 299, Roma, Italy
| | - L S Lautz
- Risk Assessment Department, French Agency for Food, Environmental and Occupational Health & Safety (Anses), 14 rue Pierre et Marie Curie, Maisons-Alfort, F-94701, France; Wageningen Food Safety Research, Akkermaalsbos 2, 6708WB, Wageningen, the Netherlands
| | - J L C M Dorne
- European Food Safety Authority, Via Carlo Magno 1A, 43126, Parma, Italy
| |
Collapse
|
49
|
Matwiejczuk N, Galicka A, Brzóska MM. Review of the safety of application of cosmetic products containing parabens. J Appl Toxicol 2021; 40:176-210. [PMID: 31903662 DOI: 10.1002/jat.3917] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022]
Abstract
Cosmetics are a source of lifetime exposure to various substances including parabens, being the most popular synthetic preservatives. Because the use of cosmetics shows an increasing trend and some adverse health outcomes of parabens present in these products have been reported, the present review focused on the safety of dermal application of these compounds. Special attention has been paid to the absorption of parabens and their retention in the human body in the intact form, as well as to their toxicological characteristics. Particular emphasis has been placed on the estrogenic potential of parabens. Based on the available published data of the concentrations of parabens in various kinds of cosmetics, the average ranges of systemic exposure dose (SED) for methylparaben, ethylparaben, propylparaben, and butylparaben have been calculated. Safety evaluations [margin of safety (MoS)] for these compounds, based on their aggregate exposure, have also been performed. Moreover, evidence for the negative impact of methylparaben on skin cells has been provided, and the main factors that may intensify dermal absorption of parabens and their impact on the skin have been described. Summarizing, the use of single cosmetics containing parabens should not pose a hazard for human health; however, using excessive quantities of cosmetic preparations containing these compounds may lead to the development of unfavorable health outcomes. Due to the real risk of estrogenic effects, as a result of exposure to parabens in cosmetics, simultaneous use of many cosmetic products containing these preservatives should be avoided.
Collapse
Affiliation(s)
- Natalia Matwiejczuk
- Department of Medical Chemistry, Medical University of Białystok, Bialystok, Poland
| | - Anna Galicka
- Department of Medical Chemistry, Medical University of Białystok, Bialystok, Poland
| | | |
Collapse
|
50
|
Wang X, Her L, Xiao J, Shi J, Wu AH, Bleske BE, Zhu H. Impact of carboxylesterase 1 genetic polymorphism on trandolapril activation in human liver and the pharmacokinetics and pharmacodynamics in healthy volunteers. Clin Transl Sci 2021; 14:1380-1389. [PMID: 33660934 PMCID: PMC8301577 DOI: 10.1111/cts.12989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 01/20/2023] Open
Abstract
Trandolapril, an angiotensin-converting enzyme inhibitor prodrug, needs to be activated by carboxylesterase 1 (CES1) in the liver to exert its intended therapeutic effect. A previous in vitro study demonstrated that the CES1 genetic variant G143E (rs71647871) abolished CES1-mediated trandolapril activation in cells transfected with the variant. This study aimed to determine the effect of the G143E variant on trandolapril activation in human livers and the pharmacokinetics (PKs) and pharmacodynamics (PDs) in human subjects. We performed an in vitro incubation study to assess trandolapril activation in human livers (5 G143E heterozygotes and 97 noncarriers) and conducted a single-dose (1 mg) PK and PD study of trandolapril in healthy volunteers (8 G143E heterozygotes and 11 noncarriers). The incubation study revealed that the mean trandolapril activation rate in G143E heterozygous livers was 42% of those not carrying the variant (p = 0.0015). The clinical study showed that, relative to noncarriers, G143E carriers exhibited 20% and 15% decreases, respectively, in the peak concentration (Cmax ) and area under the curve from 0 to 72 h (AUC0-72 h ) of the active metabolite trandolaprilat, although the differences were not statistically significant. Additionally, the average maximum reductions of systolic blood pressure and diastolic blood pressure in carriers were ~ 22% and 23% less than in noncarriers, respectively, but the differences did not reach a statistically significant level. In summary, the CES1 G143E variant markedly impaired trandolapril activation in the human liver under the in vitro incubation conditions; however, this variant had only a modest impact on the PK and PD of trandolapril in healthy human subjects.
Collapse
Affiliation(s)
- Xinwen Wang
- Department of Clinical PharmacyUniversity of MichiganAnn ArborMichiganUSA
- Present address:
Department of Pharmaceutical SciencesNortheast Ohio Medical UniversityRootstownOhioUSA
| | - Lucy Her
- Department of Clinical PharmacyUniversity of MichiganAnn ArborMichiganUSA
| | - Jingcheng Xiao
- Department of Clinical PharmacyUniversity of MichiganAnn ArborMichiganUSA
| | - Jian Shi
- Department of Clinical PharmacyUniversity of MichiganAnn ArborMichiganUSA
| | - Audrey H. Wu
- Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Barry E. Bleske
- Department of Pharmacy Practice and Administrative SciencesThe University of New MexicoAlbuquerqueNew MexicoUSA
| | - Hao‐Jie Zhu
- Department of Clinical PharmacyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|