1
|
Li X, Meng Z, Gong S, Liang Y, Zhang Y, Xu X, Wang Z, Wang S. Synthesis of a new camphor-derived carboxylesterase-activated fluorescent probe for sensitive detection of dimethoate residues in agricultural products and its applications in biological systems. Food Chem 2025; 464:141625. [PMID: 39426261 DOI: 10.1016/j.foodchem.2024.141625] [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: 07/24/2024] [Revised: 09/25/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
In this paper, a camphor-derived enzyme activatable probe CPA was synthesized for detecting dimethoate pesticide. The ester bond of probe CPA could be selectively hydrolyzed and fragmented in the presence of carboxylesterase (CE), which caused a remarkably enhanced green fluorescence signal at 501 nm. Probe CPA could function as an effective fluorescent platform for the sequential detection of dimethoate due to the obvious inhibition effect of dimethoate on the activity of CE. The detection limit of probe CPA to dimethoate was computed to be 0.1104 μg/mL. Even more important, CPA was effectively utilized for quantitative determination of trace dimethoate residues in agricultural products including fresh vegetables and fruits with good accuracy. Furthermore, the probe CPA could realize the detection of dimethoate in living cells and zebrafish. This work is expected to provide a highly sensitive and accurate analytical method for detecting organophosphorus pesticide residues in food samples and biological systems.
Collapse
Affiliation(s)
- Xinyan Li
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Zhiyuan Meng
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Shuai Gong
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Yueyin Liang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Yan Zhang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Xu Xu
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Zhonglong Wang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China.
| | - Shifa Wang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
2
|
Wang H, Chen H, Cui X, Zhang Y, Zhou J, Chen X. Simultaneous determination of unecritinib (TQ-B3101) and its active metabolite crizotinib in rat plasma by LC-MS/MS:An application to pharmacokinetic studies. J Pharm Biomed Anal 2024; 246:116199. [PMID: 38744200 DOI: 10.1016/j.jpba.2024.116199] [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: 02/02/2024] [Revised: 03/31/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
Unecritinib (TQ-B3101) is a selective tyrosine kinase receptor inhibitor. In the study, in vitro metabolic experiments revealed that the hydrolysis of TQ-B3101 was mainly catalyzed by carboxylesterase 2 (CES2), followed by CES1. Next, a sensitive and reliable LC-MS/MS method was established for the simultaneous determination of TQ-B3101 and its metabolite crizotinib in rat plasma. To prevent in vitro hydrolysis of TQ-B3101, sodium fluoride, the CESs inhibitor at a concentration of 2 M, was immediately added after whole blood collection. Plasma samples were extracted by acetonitrile-induced protein precipitation method, and chromatographically separated on a Gemini C18 column (50 mm × 2.0 mm i.d., 5 μm) using gradient elution with a mobile phase of 0.1% formic acid and 5 mmol/L ammonium acetate with 0.1% formic acid. The retention times for TQ-B3101 and crizotinib were 2.61 and 2.38 min, respectively. The analytes were detected with tandem mass spectrometer by positive electrospray ionization, using the ion transitions at m/z 492.3 → 302.3 for TQ-B3101, m/z 450.3 → 260.3 for crizotinib, and m/z 494.0 → 394.3 for imatinib (internal standard). Method validation was conducted in the linear range of 1.00-800 ng/mL for the two analytes. The precision, accuracy and stabilities all met the acceptance criteria. The pharmacokinetic study indicated that TQ-B3101 was rapidly hydrolyzed to crizotinib with the elimination half-life of 1.11 h after a single gavage administration of 27 mg/kg to Sprague-Dawley rats, and the plasma exposure of TQ-B3101 was only 2.98% of that of crizotinib.
Collapse
Affiliation(s)
- Hong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, PR China
| | - Huixian Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, PR China
| | - Xinran Cui
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yuchen Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, PR China
| | - Jialan Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China
| | - Xiaoyan Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, PR China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
3
|
Senkina J, Knapp S. Incorporation of an Isohexide Subunit into the Endochin-like Quinolone Scaffold. Molecules 2024; 29:3615. [PMID: 39125020 PMCID: PMC11314205 DOI: 10.3390/molecules29153615] [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: 06/15/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
In order to improve the drug-likeness qualities, the antimalarial endochin-like quinolone (ELQ) scaffold has been modified by replacing the 4-(trifluoromethoxy)phenyl portion with an isoidide unit that is further adjustable by varying the distal O-substituents. As expected, the water solubilities of the new analogs are greatly improved, and the melting points are lower. However, the antimalarial potency of the new analogs is reduced to EC50 > 1 millimolar, a result ascribable to the hydrophilic nature of the new substitution.
Collapse
Affiliation(s)
| | - Spencer Knapp
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ 08854, USA;
| |
Collapse
|
4
|
Gao P, Ha-Duong T, Nicolas J. Coarse-Grained Model-Assisted Design of Polymer Prodrug Nanoparticles with Enhanced Cytotoxicity: A Combined Theoretical and Experimental Study. Angew Chem Int Ed Engl 2024; 63:e202316056. [PMID: 38345287 DOI: 10.1002/anie.202316056] [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: 10/24/2023] [Indexed: 03/12/2024]
Abstract
To achieve drug release from polymer prodrug nanoparticles, the drug-polymer linker must be accessible for cleavage to release the drug, which can occur under certain physiological conditions (e.g., presence of specific enzymes). Supramolecular organization of polymer prodrug nanoparticles is crucial as it greatly affects the location of the linker, its surface exposure/solvation and thus its cleavage to release the drug. Since experimental access to these data is not straightforward, new methodologies are critically needed to access this information and to accelerate the development of more effective polymer prodrug nanoparticles, and replace the time-consuming and resource-intensive traditional trial-and-error strategy. In this context, we reported here the use of a coarse-grained model to assist the design of polymer prodrug nanoparticles with enhanced cytotoxicity. By choosing the solvent accessible surface area as the critical parameter for predicting drug release and hence cytotoxicity of polymer prodrug nanoparticles, we developed an optimized polymer-drug linker with enhanced hydrophilicity and solvation. Our hypothesis was then experimentally validated by the synthesis of the corresponding polymer prodrugs based on two different drugs (gemcitabine and paclitaxel), which demonstrated greater performances in terms of drug release and cytotoxicity on two cancer cell lines. Interestingly, our methodology can be easily applied to other polymer prodrug structures, which would contribute to the development of more efficient drug delivery systems via in silico screening.
Collapse
Affiliation(s)
- Ping Gao
- Université Paris-Saclay, CNRS, BioCIS, Orsay, 91400, France
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay, 91400, France
| | - Tâp Ha-Duong
- Université Paris-Saclay, CNRS, BioCIS, Orsay, 91400, France
| | - Julien Nicolas
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay, 91400, France
| |
Collapse
|
5
|
Chiu WA. Invited Perspective: Uneven Progress Addressing Population Variability in Human Health Risk Assessment. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:31305. [PMID: 38498339 PMCID: PMC10947099 DOI: 10.1289/ehp13461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/03/2023] [Accepted: 02/06/2024] [Indexed: 03/20/2024]
Affiliation(s)
- Weihsueh A. Chiu
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas, USA
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
6
|
Pelkonen O, Abass K, Parra Morte JM, Panzarea M, Testai E, Rudaz S, Louisse J, Gundert-Remy U, Wolterink G, Jean-Lou CM D, Coecke S, Bernasconi C. Metabolites in the regulatory risk assessment of pesticides in the EU. FRONTIERS IN TOXICOLOGY 2023; 5:1304885. [PMID: 38188093 PMCID: PMC10770266 DOI: 10.3389/ftox.2023.1304885] [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: 09/30/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
A large majority of chemicals is converted into metabolites through xenobiotic-metabolising enzymes. Metabolites may present a spectrum of characteristics varying from similar to vastly different compared with the parent compound in terms of both toxicokinetics and toxicodynamics. In the pesticide arena, the role of metabolism and metabolites is increasingly recognised as a significant factor particularly for the design and interpretation of mammalian toxicological studies and in the toxicity assessment of pesticide/metabolite-associated issues for hazard characterization and risk assessment purposes, including the role of metabolites as parts in various residues in ecotoxicological adversities. This is of particular relevance to pesticide metabolites that are unique to humans in comparison with metabolites found in in vitro or in vivo animal studies, but also to disproportionate metabolites (quantitative differences) between humans and mammalian species. Presence of unique or disproportionate metabolites may underlie potential toxicological concerns. This review aims to present the current state-of-the-art of comparative metabolism and metabolites in pesticide research for hazard and risk assessment, including One Health perspectives, and future research needs based on the experiences gained at the European Food Safety Authority.
Collapse
Affiliation(s)
- Olavi Pelkonen
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Khaled Abass
- Department of Environmental Health Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research (SIMR), University of Sharjah, Sharjah, United Arab Emirates
- Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
| | | | | | - Emanuela Testai
- Mechanisms, Biomarkers and Models Unit, Environment and Health Department, Istituto Superiore di Sanità, Rome, Italy
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
| | - Jochem Louisse
- EFSA, European Food Safety Authority, Parma, Italy
- Wageningen Food Safety Research (WFSR), Wageningen, Netherlands
| | - Ursula Gundert-Remy
- Institute of Clinical Pharmacology and Toxicology, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gerrit Wolterink
- Centre for Prevention, Lifestyle and Health, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | | | - Sandra Coecke
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | |
Collapse
|
7
|
Xu M, Zhang L, Lin L, Qiang Z, Liu W, Yang J. Cisplatin increases carboxylesterases through increasing PXR mediated by the decrease of DEC1. J Biomed Res 2023; 37:431-447. [PMID: 37990879 PMCID: PMC10687532 DOI: 10.7555/jbr.37.20230047] [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: 03/05/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 11/23/2023] Open
Abstract
cis-Diamminedichloroplatinum (CDDP) is widely used for the treatment of various solid cancers. Here we reported that CDDP increased the expression and enzymatic activities of carboxylesterase 1 (CES1) and carboxylesterase 2 (CES2), along with the upregulation of pregnane X receptor (PXR) and the downregulation of differentiated embryonic chondrocyte-expressed gene 1 (DEC1) in human hepatoma cells, primary mouse hepatocytes, mouse liver and intestine. The overexpression or knockdown of PXR alone upregulated or downregulated the CES1 and CES2 expression, respectively. The increases in CES1 and CES2 expression levels induced by CDDP abolished or enhanced by PXR knockdown or overexpression, implying that CDDP induces carboxylesterases through the activation of PXR. Likewise, the overexpression or knockdown of DEC1 alone significantly decreased or increased PXR and its targets. Moreover, the increases of PXR and its targets induced by CDDP were abolished or alleviated by the overexpression or knockdown of DEC1. The overexpression or knockdown of DEC1 affected the response of PXR to CDDP, but not vice versa, suggesting that CDDP increases carboxylesterases by upregulating PXR mediated by the decrease of DEC1. In addition, CDDP did not increase DEC1 mRNA degradation but suppressed DEC1 promoter reporter activity, indicating that it suppresses DEC1 transcriptionally. The combined use of CDDP and irinotecan had a synergistic effect on two cell lines, especially when CDDP was used first.
Collapse
Affiliation(s)
- Minqin Xu
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Lihua Zhang
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Lan Lin
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zhiyi Qiang
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wei Liu
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jian Yang
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
8
|
Luo X, Lu F, Yin Z, Zhou Z, Wang Z, Zhang H. Hormetic effects of EGC and EGCG on CES1 activity and its rescue from oxidative stress in rat liver S9. Chem Biol Interact 2023; 382:110612. [PMID: 37353134 DOI: 10.1016/j.cbi.2023.110612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/17/2023] [Accepted: 06/21/2023] [Indexed: 06/25/2023]
Abstract
Carboxylesterase 1 (CES1) is a hydrolytic enzyme that plays an important role in the activation or deactivation of many therapeutic agents, thus affecting their pharmacokinetic and pharmacodynamic outcomes. Using rat liver S9 as an enzyme source and enalapril as a CES1 substrate, the present study examined effects of a number of flavonoids on the formation of enalaprilat (the active form of enalapril) produced by CES1-mediated hydrolysis. While a majority of flavonoids tested showed inhibition on CES1, an unexpected hormetic effect was observed for epigallocatechin (EGC) and epigallocatechin gallate (EGCG), i.e., stimulatory effect at low concentrations and enzyme inhibition at high concentrations. Further experiments revealed that oxidative stress caused by hydrogen peroxide, arachidonic acid plus iron, and oxidized low density lipoproteins (oxLOL) reduced CES1 activity in rat liver S9 and the loss of CES1 enzyme activity could be rescued largely by EGC or EGCG. In contrast, such effects were minimal in human liver S9, probably due to the presence of a higher ratio of reduced vs oxidized forms of glutathione. The above findings suggest that the polyphenolic nature of EGC or EGCG might be responsible for rescuing CES1 activity under oxidative stress. Because of the importance of CES1 in drug activation or deactivation and rat liver S9 as a versatile in vitro system used for drug metabolism studies and drug safety assessment, caution should be exercised to avoid potential biases for data interpretation and decision making when CES1 activity in rat liver S9 is evaluated with dependency on experimental conditions.
Collapse
Affiliation(s)
- Xiaoting Luo
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Feifei Lu
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Zhiyue Yin
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Zhiyun Zhou
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Zhongmin Wang
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China
| | - Hongjian Zhang
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, 215123, China.
| |
Collapse
|
9
|
Wang X, Zhang J, Zheng K, Du Q, Wang G, Huang J, Zhou Y, Li Y, Jin H, He J. Discovering metabolic vulnerability using spatially resolved metabolomics for antitumor small molecule-drug conjugates development as a precise cancer therapy strategy. J Pharm Anal 2023; 13:776-787. [PMID: 37577390 PMCID: PMC10422108 DOI: 10.1016/j.jpha.2023.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Against tumor-dependent metabolic vulnerability is an attractive strategy for tumor-targeted therapy. However, metabolic inhibitors are limited by the drug resistance of cancerous cells due to their metabolic plasticity and heterogeneity. Herein, choline metabolism was discovered by spatially resolved metabolomics analysis as metabolic vulnerability which is highly active in different cancer types, and a choline-modified strategy for small molecule-drug conjugates (SMDCs) design was developed to fool tumor cells into indiscriminately taking in choline-modified chemotherapy drugs for targeted cancer therapy, instead of directly inhibiting choline metabolism. As a proof-of-concept, choline-modified SMDCs were designed, screened, and investigated for their druggability in vitro and in vivo. This strategy improved tumor targeting, preserved tumor inhibition and reduced toxicity of paclitaxel, through targeted drug delivery to tumor by highly expressed choline transporters, and site-specific release by carboxylesterase. This study expands the strategy of targeting metabolic vulnerability and provides new ideas of developing SMDCs for precise cancer therapy.
Collapse
Affiliation(s)
- Xiangyi Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- NMPA Key Laboratory of Safety Research and Evaluation of Innovative Drug, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jin Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- NMPA Key Laboratory of Safety Research and Evaluation of Innovative Drug, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kailu Zheng
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Qianqian Du
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Guocai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- NMPA Key Laboratory of Safety Research and Evaluation of Innovative Drug, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jianpeng Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- NMPA Key Laboratory of Safety Research and Evaluation of Innovative Drug, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yanhe Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- NMPA Key Laboratory of Safety Research and Evaluation of Innovative Drug, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yan Li
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hongtao Jin
- NMPA Key Laboratory of Safety Research and Evaluation of Innovative Drug, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jiuming He
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- NMPA Key Laboratory of Safety Research and Evaluation of Innovative Drug, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
10
|
Elbagoury RM, Shenouda MA, Elnakib HE, Wober J, Abadi AH, Ahmed NS. Design, synthesis, and metabolite identification of Tamoxifen esterase-activatable prodrugs. Bioorg Chem 2023; 131:106303. [PMID: 36455483 DOI: 10.1016/j.bioorg.2022.106303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/13/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
Tamoxifen (TAM) is used in treatment of hormonal dependent breast cancer, both in premenopausal and postmenopausal women. TAM is intrinsically metabolized by CYP450 enzymes to more active metabolites. Recent reports identified CYP2D6, an enzyme involved in the conversion of TAM to the more potent 4-OH-TAM, is encoded by theCYP2D6gene, which is highly polymorphic. Women with inactive alleles are poor metabolizers; in many cases they suffer acquired TAM resistance. Herein we report synthesis and biological evaluation of novel TAM analogues. The novel analogues are designed to elude CYP2D6 metabolism. Hydrolysis of the carbamate moiety on ring C is mediated via carboxylesterases. Compound 3d [E/Z Benzyl-carbamic acid4-{2-benzyl-1-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]-but-1-enyl}-phenyl ester] showed GI50 = 0.09 µM on MCF-7 and GI50 = 1.84 µM on MDA-MB231 cell lines. To further validate our hypothesis, metabolites of selected novel analogues were determined in vitro under different incubation conditions. The hydroxylated analogues were obtained under non CYP2D6 dependent conditions. Compound 8d, a benzyl carbamate derivative, was the least-stable analog and showed the highest rate of metabolism among all tested analogues. Our in silico model showed the novel flexible analogues can still adopt an antiestrogenic binding profile occupying the same pocket as 4-OH-TAM.
Collapse
Affiliation(s)
- Rahma M Elbagoury
- Faculty of Pharmacy and Biotechnology, Department of Pharmaceutical Chemistry, German University in Cairo, Cairo, Egypt
| | - Miriam A Shenouda
- Faculty of Pharmacy and Biotechnology, Department of Pharmaceutical Chemistry, German University in Cairo, Cairo, Egypt
| | - Heba E Elnakib
- Faculty of Pharmacy and Biotechnology, Department of Pharmaceutical Chemistry, German University in Cairo, Cairo, Egypt
| | - Jannette Wober
- Faculty of Biology, Institute of Zoology, Technische Universität Dresden, 01062 Dresden, Germany
| | - Ashraf H Abadi
- Faculty of Pharmacy and Biotechnology, Department of Pharmaceutical Chemistry, German University in Cairo, Cairo, Egypt
| | - Nermin S Ahmed
- Faculty of Pharmacy and Biotechnology, Department of Pharmaceutical Chemistry, German University in Cairo, Cairo, Egypt.
| |
Collapse
|
11
|
Gu QL, Xue FL, Zheng ZL, Wang HN, Guan YP, Wen YZ, Ye F, Huang M, Huang WQ, Wang ZX, Li JL. Nongenetic and genetic predictors of haemodynamic instability induced by propofol and opioids: A retrospective clinical study. Br J Clin Pharmacol 2023; 89:209-221. [PMID: 35939394 DOI: 10.1111/bcp.15480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/28/2022] Open
Abstract
AIM Propofol and opioids are commonly used in anaesthesia, but are highly susceptible to haemodynamic instability, thereby threatening the patient's surgical safety and prognosis. The purpose of this study was to investigate the predictors of haemodynamic instability and establish its predictive model. METHODS A total of 150 Chinese patients undergoing thyroid or breast surgery participated in the study, with target-controlled infusion concentrations of propofol, opioids dosage, heart rate (HR), mean arterial pressure (MAP) and Narcotrend Index recorded at key points throughout the procedure. The Agena MassARRAY system was used to genotype candidate single nucleotide polymorphisms related to pharmacodynamics and pharmacokinetics of propofol and opioids. RESULTS Among nongenetic factors, baseline HR (R = -.579, P < .001) and baseline MAP (R = -.725, P < .001) had a significant effect on the haemodynamic instability. Among genetic factors, the CT/CC genotype of GABRB1 rs4694846 (95% confidence interval [CI]: -11.309 to -3.155), AA/AG of OPRM1 rs1799971 (95%CI: 0.773 to 10.290), AA of CES2 rs8192925 (95%CI: 1.842 to 9.090) were associated with higher HR instability; the AA/GG genotype of NR1I2 rs6438550 (95%CI: 0.351 to 7.761), AA of BDNF rs2049046 (95%CI: -9.039 to -0.640) and GG of GABBR2 rs1167768 (95%CI: -10.146 to -1.740) were associated with higher MAP instability. The predictive models of HR and MAP fluctuations were developed, accounting for 45.0 and 59.2% of variations, respectively. CONCLUSION We found that cardiovascular fundamentals and genetic variants of GABRB1, GABBR2, OPRM1, BDNF, CES2 and NR1I2 are associated with cardiovascular susceptibility, which can provide a reference for haemodynamic management in clinical anaesthesia.
Collapse
Affiliation(s)
- Qing-Ling Gu
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fa-Ling Xue
- Department of Anaesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhuo-Ling Zheng
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hai-Ni Wang
- Department of Pharmacy, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Yan-Ping Guan
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yong-Zi Wen
- Junzhi Biomedical Research Laboratory (Foshan) Co., Ltd., Foshan, Guangdong, China
| | - Fang Ye
- Department of Anaesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Min Huang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wen-Qi Huang
- Department of Anaesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhong-Xing Wang
- Department of Anaesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jia-Li Li
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Bordat A, Boissenot T, Ibrahim N, Ferrere M, Levêque M, Potiron L, Denis S, Garcia-Argote S, Carvalho O, Abadie J, Cailleau C, Pieters G, Tsapis N, Nicolas J. A Polymer Prodrug Strategy to Switch from Intravenous to Subcutaneous Cancer Therapy for Irritant/Vesicant Drugs. J Am Chem Soc 2022; 144:18844-18860. [PMID: 36193551 PMCID: PMC9585574 DOI: 10.1021/jacs.2c04944] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Chemotherapy is almost exclusively administered via the
intravenous
(IV) route, which has serious limitations (e.g., patient discomfort,
long hospital stays, need for trained staff, high cost, catheter failures,
infections). Therefore, the development of effective and less costly
chemotherapy that is more comfortable for the patient would revolutionize
cancer therapy. While subcutaneous (SC) administration has the potential
to meet these criteria, it is extremely restrictive as it cannot be
applied to most anticancer drugs, such as irritant or vesicant ones,
for local toxicity reasons. Herein, we report a facile, general, and
scalable approach for the SC administration of anticancer drugs through
the design of well-defined hydrophilic polymer prodrugs. This was
applied to the anticancer drug paclitaxel (Ptx) as a worst-case scenario
due to its high hydrophobicity and vesicant properties (two factors
promoting necrosis at the injection site). After a preliminary screening
of well-established polymers used in nanomedicine, polyacrylamide
(PAAm) was chosen as a hydrophilic polymer owing to its greater physicochemical,
pharmacokinetic, and tumor accumulation properties. A small library
of Ptx-based polymer prodrugs was designed by adjusting the nature
of the linker (ester, diglycolate, and carbonate) and then evaluated
in terms of rheological/viscosity properties in aqueous solutions,
drug release kinetics in PBS and in murine plasma, cytotoxicity on
two different cancer cell lines, acute local and systemic toxicity,
pharmacokinetics and biodistribution, and finally their anticancer
efficacy. We demonstrated that Ptx-PAAm polymer prodrugs could be
safely injected subcutaneously without inducing local toxicity while
outperforming Taxol, the commercial formulation of Ptx, thus opening
the door to the safe transposition from IV to SC chemotherapy.
Collapse
Affiliation(s)
- Alexandre Bordat
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Tanguy Boissenot
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Nada Ibrahim
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Marianne Ferrere
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Manon Levêque
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Léa Potiron
- Imescia, Université Paris-Saclay, 91400 Saclay, France
| | - Stéphanie Denis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Sébastien Garcia-Argote
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris-Saclay, CEA, INRAE, Gif-sur-Yvette F-91191, France
| | - Olivia Carvalho
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris-Saclay, CEA, INRAE, Gif-sur-Yvette F-91191, France
| | - Jérôme Abadie
- Laboniris, Départment de Biology, Pathologie et Sciences de l'Aliment, Oniris, F-44307 Nantes, France
| | - Catherine Cailleau
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Grégory Pieters
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris-Saclay, CEA, INRAE, Gif-sur-Yvette F-91191, France
| | - Nicolas Tsapis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Julien Nicolas
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| |
Collapse
|
13
|
Deciphering the species differences in CES1A-mediated hydrolytic metabolism by using a bioluminescence substrate. Chem Biol Interact 2022; 368:110197. [PMID: 36174736 DOI: 10.1016/j.cbi.2022.110197] [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: 07/29/2022] [Revised: 09/04/2022] [Accepted: 09/22/2022] [Indexed: 11/21/2022]
Abstract
Carboxylesterases 1A (CES1A) is a key enzyme responsible for the hydrolytic metabolism of a great deal of endogenous and exogenous substrates bearing ester- or amide-bond(s). This study aimed to decipher the species difference in CES1A-mediated hydrolytic metabolism by using a newly developed bioluminescence CES1A sensor (termed NLMe) as the probe substrate, while the liver microsomes from six different mammalian species (human, cynomolgus monkey, dog, minipig, rat and mouse) were used as the enzyme sources. Metabolite profiling demonstrated that all tested liver microsomes from various species could catalyze NLMe hydrolysis, but significant difference in hydrolytic rate was observed. Kinetic plots of NLMe hydrolysis in liver microsomes from different species showed that the inherent clearance rates (Clint) of NLMe in human liver microsomes (HLM), cynomolgus monkey liver microsomes (CyLM), and pig liver microsome (PLM) were comparable, while the Clint values of NLMe in dog liver microsomes (DLM), mouse liver microsomes (MLM), and rat liver microsomes (RLM) were relatively small. Moreover, chemical inhibition assays showed that NLMe hydrolysis in all tested liver microsomes could be competently inhibited by BNPP (a potent broad-spectrum inhibitor of CES), but CUA (a selective inhibitor of human CES1A) only inhibited NLMe hydrolysis in human liver microsomes and dog liver microsomes. In summary, the species differences in CES1A-catalyzed NLMe hydrolysis were carefully investigated from the views of the similarities in metabolite profile, hydrolytic kinetics and inhibitor response. All these findings provide new insights into the species differences in CES1A-mediated hydrolytic metabolism and suggest that it is necessary for the pharmacologists to choose appropriate animal models to replace humans for evaluating the in vivo effects of CES1A inhibitors.
Collapse
|
14
|
Model systems and organisms for addressing inter- and intra-species variability in risk assessment. Regul Toxicol Pharmacol 2022; 132:105197. [DOI: 10.1016/j.yrtph.2022.105197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
|