1
|
Zhou J, Qin X, Zhou S, MacKenzie KR, Li F. CYP3A-Mediated Carbon-Carbon Bond Cleavages in Drug Metabolism. Biomolecules 2024; 14:1125. [PMID: 39334891 PMCID: PMC11430781 DOI: 10.3390/biom14091125] [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: 06/19/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Cytochrome P450 enzymes (P450s) play a critical role in drug metabolism, with the CYP3A subfamily being responsible for the biotransformation of over 50% of marked drugs. While CYP3A enzymes are known for their extensive catalytic versatility, one intriguing and less understood function is the ability to mediate carbon-carbon (C-C) bond cleavage. These uncommon reactions can lead to unusual metabolites and potentially influence drug safety and efficacy. This review focuses on examining examples of C-C bond cleavage catalyzed by CYP3A, exploring the mechanisms, physiological significance, and implications for drug metabolism. Additionally, examples of CYP3A-mediated ring expansion via C-C bond cleavages are included in this review. This work will enhance our understanding of CYP3A-catalyzed C-C bond cleavages and their mechanisms by carefully examining and analyzing these case studies. It may also guide future research in drug metabolism and drug design, improving drug safety and efficacy in clinical practice.
Collapse
Affiliation(s)
- Junhui Zhou
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (J.Z.); (X.Q.); (S.Z.); (K.R.M.)
| | - Xuan Qin
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (J.Z.); (X.Q.); (S.Z.); (K.R.M.)
- NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shenzhi Zhou
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (J.Z.); (X.Q.); (S.Z.); (K.R.M.)
| | - Kevin R. MacKenzie
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (J.Z.); (X.Q.); (S.Z.); (K.R.M.)
- NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Feng Li
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (J.Z.); (X.Q.); (S.Z.); (K.R.M.)
- NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
2
|
Hu XM, Hou YY, Teng XR, Liu Y, Li Y, Li W, Li Y, Ai CZ. Prediction of cytochrome P450-mediated bioactivation using machine learning models and in vitro validation. Arch Toxicol 2024; 98:1457-1467. [PMID: 38492097 DOI: 10.1007/s00204-024-03701-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/31/2024] [Indexed: 03/18/2024]
Abstract
Cytochrome P450 (P450)-mediated bioactivation, which can lead to the hepatotoxicity through the formation of reactive metabolites (RMs), has been regarded as the major problem of drug failures. Herein, we purposed to establish machine learning models to predict the bioactivation of P450. On the basis of the literature-derived bioactivation dataset, models for Benzene ring, Nitrogen heterocycle and Sulfur heterocycle were developed with machine learning methods, i.e., Random Forest, Random Subspace, SVM and Naïve Bayes. The models were assessed by metrics like "Precision", "Recall", "F-Measure", "AUC" (Area Under the Curve), etc. Random Forest algorithms illustrated the best predictability, with nice AUC values of 0.949, 0.973 and 0.958 for the test sets of Benzene ring, Nitrogen heterocycle and Sulfur heterocycle models, respectively. 2D descriptors like topological indices, 2D autocorrelations and Burden eigenvalues, etc. contributed most to the models. Furthermore, the models were applied to predict the occurrence of bioactivation of an external verification set. Drugs like selpercatinib, glafenine, encorafenib, etc. were predicted to undergo bioactivation into toxic RMs. In vitro, IC50 shift experiment was performed to assess the potential of bioactivation to validate the prediction. Encorafenib and tirbanibulin were observed of bioactivation potential with shifts of 3-6 folds or so. Overall, this study provided a reliable and robust strategy to predict the P450-mediated bioactivation, which will be helpful to the assessment of adverse drug reactions (ADRs) in clinic and the design of new candidates with lower toxicities.
Collapse
Affiliation(s)
- Xin-Man Hu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China
| | - Yan-Yao Hou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China
| | - Xin-Ru Teng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Panjin, 124221, People's Republic of China
| | - Yu Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China
| | - Wei Li
- Translational Medicine Research Institute, College of Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, 136 Jiangyangzhong Road, Yangzhou, 225001, People's Republic of China.
| | - Yan Li
- Department of Materials Science and Chemical Engineering, Dalian University of Technology, Dalian, 116023, Liaoning, People's Republic of China
| | - Chun-Zhi Ai
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China.
| |
Collapse
|
3
|
Broberg MN, Ohlsson RT, Bondesson U, Pettersson C, Tidstedt B, Thevis M, Hedeland M. A multivariate data analysis approach for the investigation of in vitro derived metabolites of ACP-105 in comparison with human in vivo metabolites. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1231:123927. [PMID: 37972465 DOI: 10.1016/j.jchromb.2023.123927] [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: 10/11/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Selective androgen receptor modulators (SARMs) such as ACP-105 are prohibited in sports due to their anabolic properties. ACP-105 has in previous equine studies shown to undergo extensive metabolism, which makes its metabolite profile important to investigate in humans, since the metabolism is unknown in this species. The aims of the study were to systematically optimize in vitro microsome incubations for improved metabolite yield and to utilize a multivariate data analysis (MVDA) approach to aid the metabolite discovery. Microsomes together with S9 fractions were used at optimal conditions, both with and without phase II additives. Furthermore, the relevance of the in vitro derived metabolites was evaluated as analytical targets in doping control by comparison with results from a human post-administration urine sample collected after a single dose of 100 µg ACP-105. All samples were analyzed with liquid chromatography - Orbitrap mass spectrometry. The use of the systematical optimization and MVDA greatly simplified the search and a total of 18 in vitro metabolites were tentatively identified. The yield of the two main monohydroxylated isomers increased by 24 and 10 times, respectively. In the human urine sample, a total of seven metabolites of ACP-105, formed by a combination of hydroxylations and glucuronic acid conjugations, were tentatively identified. The main metabolites were two monohydroxylated forms that are suggested as analytical targets for human doping control after hydrolysis. All the in vivo metabolites could be detected with the MVDA approach on the in vitro models, demonstrating its usefulness for prediction of the in vivo metabolite profile.
Collapse
Affiliation(s)
- Malin Nilsson Broberg
- Department of Medicinal Chemistry, Uppsala University, Box 574, 75123 Uppsala, Sweden
| | | | - Ulf Bondesson
- Department of Medicinal Chemistry, Uppsala University, Box 574, 75123 Uppsala, Sweden
| | - Curt Pettersson
- Department of Medicinal Chemistry, Uppsala University, Box 574, 75123 Uppsala, Sweden
| | - Börje Tidstedt
- Department of Chemistry, Environment and Feed Hygiene, National Veterinary Institute (SVA), 75189 Uppsala, Sweden
| | - Mario Thevis
- Institute of Biochemistry, Center for Preventive Doping Research, German Sport University, 50933 Cologne, Germany
| | - Mikael Hedeland
- Department of Medicinal Chemistry, Uppsala University, Box 574, 75123 Uppsala, Sweden.
| |
Collapse
|
4
|
Chen B, Wang C, Fu Z, Yu H, Liu E, Gao X, Li J, Han L. RT-Ensemble Pred: A tool for retention time prediction of metabolites on different LC-MS systems. J Chromatogr A 2023; 1707:464304. [PMID: 37611386 DOI: 10.1016/j.chroma.2023.464304] [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: 08/03/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023]
Abstract
Liquid chromatography-mass spectrometry (LC-MS) could provide a large amount of information to assist in metabolites identification. Different liquid chromatographic methods (CMs) could produce different retention times to the same metabolite. To predict the retention time of local dataset by online datasets has become a trend, but the datasets downloaded from different databases were differences in quantity levels. And the imbalanced data could produce bad influence in model prediction. Thus, based on quantitative structure-retention relationships (QSRRs), an ensemble model, named RT-Ensemble Pred, has been successfully built to predict retention time of different LC-MS systems in this study. A total of 76, 807 metabolites (76, 909 retention times) have been collected across 9 CMs, and 19 natural products and 1 antifungal drug (20 retention times) have been collected to test the model applicability. An ensemble sampling was applied for the preprocessing procedure to solve the problem of imbalanced data. Based on the ensemble sampling, RT-Ensemble Pred could better utilize online datasets for the prediction of retention time. RT-Ensemble Pred was built based on the online datasets and tested by local dataset. The predictive accuracy of RT-Ensemble Pred was higher than the models without any sampling methods. The results showed that RT-Ensemble Pred could predict the metabolites which was not included in the database and the metabolites which were from new CMs. It could also be used for the prediction of other compounds beside metabolites. Furthermore, a tool of RT-Ensemble Pred was packed and can be freely downloaded at https://gitlab.com/mikic93/rt-ensemble-pred. It provides convenience for the users who need to predict the retention time of metabolites.
Collapse
Affiliation(s)
- Biying Chen
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai, Tianjin 301617, PR China
| | - Chenxi Wang
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai, Tianjin 301617, PR China
| | - Zhifei Fu
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai, Tianjin 301617, PR China
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai, Tianjin 301617, PR China
| | - Erwei Liu
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai, Tianjin 301617, PR China
| | - Xiumei Gao
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai, Tianjin 301617, PR China
| | - Jie Li
- Tianjin Key Laboratory of Clinical Multi-omics, Airport Economy Zone, Tianjin, China.
| | - Lifeng Han
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai, Tianjin 301617, PR China.
| |
Collapse
|
5
|
Gumieniczek A, Berecka-Rycerz A. Metabolism and Chemical Degradation of New Antidiabetic Drugs: A Review of Analytical Approaches for Analysis of Glutides and Gliflozins. Biomedicines 2023; 11:2127. [PMID: 37626624 PMCID: PMC10452759 DOI: 10.3390/biomedicines11082127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
The drug metabolism and drug degradation pathways may overlap, resulting in the formation of similar constituents. Therefore, the metabolism data can be helpful for deriving safe levels of degradation impurities and improving the quality of respective pharmaceutical products. The present article contains considerations on possible links between metabolic and degradation pathways for new antidiabetic drugs such as glutides, gliflozins, and gliptins. Special attention was paid to their reported metabolites and identified degradation products. At the same time, many interesting analytical approaches to conducting metabolism as well as degradation experiments were mentioned, including chromatographic methods and radioactive labeling of the drugs. The review addresses the analytical approaches elaborated for examining the metabolism and degradation pathways of glutides, i.e., glucagon like peptide 1 (GLP-1) receptor agonists, and gliflozins, i.e., sodium glucose co-transporter 2 (SGLT2) inhibitors. The problems associated with the chromatographic analysis of the peptide compounds (glutides) and the polar drugs (gliflozins) were addressed. Furthermore, issues related to in vitro experiments and the use of stable isotopes were discussed.
Collapse
Affiliation(s)
- Anna Gumieniczek
- Department of Medicinal Chemistry, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| | | |
Collapse
|
6
|
Xue Z, Zeng J, Yin X, Li Y, Meng B, Zhao Y, Fang X, Gong X, Dai X. Investigation on acquired palbociclib resistance by LC-MS based multi-omics analysis. Front Mol Biosci 2023; 10:1116398. [PMID: 36743215 PMCID: PMC9892630 DOI: 10.3389/fmolb.2023.1116398] [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: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Palbociclib is a specific CDK4/6 inhibitor that has been widely applied in multiple types of tumors. Different from cytotoxic drugs, the anticancer mechanism of palbociclib mainly depends on cell cycle inhibition. Therefore, the resistance mechanism is different. For clinical cancer patients, drug resistance is inevitable for almost all cancer therapies including palbociclib. We have trained palbociclib resistant cells in vitro to simulate the clinical situation and applied LC-MS multi-omics analysis methods including proteomic, metabolomic, and glycoproteomic techniques, to deeply understand the underly mechanism behind the resistance. As a result of proteomic analysis, the resistant cells were found to rely on altered metabolic pathways to keep proliferation. Metabolic processes related to carbohydrates, lipids, DNA, cellular proteins, glucose, and amino acids were observed to be upregulated. Most dramatically, the protein expressions of COX-1 and NDUFB8 have been detected to be significantly overexpressed by proteomic analysis. When a COX-1 inhibitor was hired to combine with palbociclib, a synergistic effect could be obtained, suggesting the altered COX-1 involved metabolic pathway is an important reason for the acquired palbociclib resistance. The KEGG pathway of N-glycan biosynthesis was identified through metabolomics analysis. N-glycoproteomic analysis was therefore included and the global glycosylation was found to be elevated in the palbociclib-resistant cells. Moreover, integration analysis of glycoproteomic data allowed us to detect a lot more proteins that have been glycosylated with low abundances, these proteins were considered to be overwhelmed by those highly abundant proteins during regular proteomic LC-MS detection. These low-abundant proteins are mainly involved in the cellular biology processes of cell migration, the regulation of chemotaxis, as well as the glycoprotein metabolic process which offered us great more details on the roles played by N-glycosylation in drug resistance. Our result also verified that N-glycosylation inhibitors could enhance the cell growth inhibition of palbociclib in resistant cells. The high efficiency of the integrated multi-omics analysis workflow in discovering drug resistance mechanisms paves a new way for drug development. With a clear understanding of the resistance mechanism, new drug targets and drug combinations could be designed to resensitize the resistant tumors.
Collapse
Affiliation(s)
- Zhichao Xue
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Jiaming Zeng
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang, China
| | - Xinchi Yin
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Yongshu Li
- Shenzhen Institute for Technology Innovation, National Institute of Metrology Shenzhen, Shenzhen, China
| | - Bo Meng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Yang Zhao
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Xiang Fang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Xiaoyun Gong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China,*Correspondence: Xiaoyun Gong, ; Xinhua Dai,
| | - Xinhua Dai
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China,*Correspondence: Xiaoyun Gong, ; Xinhua Dai,
| |
Collapse
|
7
|
Usman S, Razis AFA, Shaari K, Azmai MNA, Saad MZ, Isa NM, Nazarudin MF. Polystyrene microplastics induce gut microbiome and metabolome changes in Javanese medaka fish ( Oryzias javanicus Bleeker, 1854). Toxicol Rep 2022; 9:1369-1379. [PMID: 36518379 PMCID: PMC9742877 DOI: 10.1016/j.toxrep.2022.05.001] [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: 03/03/2022] [Revised: 04/22/2022] [Accepted: 05/01/2022] [Indexed: 02/07/2023] Open
Abstract
Microplastics (MPs) have become emerging pollutants of public health concern, due to their impact on aqua-terrestrial ecosystems and integration into the food web, with evidence of human exposure and unrevealed health implications. There is a paucity of information regarding the effects of MPs exposure on the gut system using metagenomic and metabolomic approaches. In this study, Javanese medaka fish was exposed to 5 µm beads of polystyrene microplastics (PS-MPs) suspensions, at concentrations of 100 μg/L (MP-LOW), 500 μg/L (MP-MED), and 1000 μg/L (MP-HIGH), for a duration of 21 days, and evaluated for gut microbiome and metabolome responses. The results revealed a significant reduction (p < 0.05) in richness and diversity of the gut microbiome in the MP-HIGH group, and identification of 7 bacterial genera as differential features by the Linear discriminant analysis Effect Size (LEfSe). The gut metabolic profile revealed upregulation of 9 metabolites related to energy metabolism, via tricarboxylic acid cycle (TCA), creatine pathway, and urea cycle, as determined by the pathway analysis. Furthermore, positive correlation was found between the genus Aeromonas and glucose, lactate, and creatine metabolites. The study revealed that PS-MPs exposure resulted in altered bacterial microbiome and metabolic disorder related to energy metabolism. It further provided additional data on gut bacterial genera and metabolites associated with MPs toxicity in aquatic organism, which will inevitably enable its future health risks assessment in animals and possibly humans.
Collapse
Affiliation(s)
- Sunusi Usman
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Ahmad Faizal Abdull Razis
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Khozirah Shaari
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Mohammad Noor Amal Azmai
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Aquatic Animal Health and Therapeutics Laboratory (Aqua Health), Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Mohd Zamri Saad
- Aquatic Animal Health and Therapeutics Laboratory (Aqua Health), Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Veterinary Laboratory Diagnosis, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Nurulfiza M. Isa
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines and Biomolecules (VacBio), Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Muhammad Farhan Nazarudin
- Aquatic Animal Health and Therapeutics Laboratory (Aqua Health), Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
8
|
Qin X, Hakenjos JM, MacKenzie KR, Barzi M, Chavan H, Nyshadham P, Wang J, Jung SY, Guner JZ, Chen S, Guo L, Krishnamurthy P, Bissig KD, Palmer S, Matzuk MM, Li F. Metabolism of a Selective Serotonin and Norepinephrine Reuptake Inhibitor Duloxetine in Liver Microsomes and Mice. Drug Metab Dispos 2022; 50:128-139. [PMID: 34785568 PMCID: PMC8969139 DOI: 10.1124/dmd.121.000633] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/12/2021] [Indexed: 11/25/2022] Open
Abstract
Duloxetine (DLX) is a dual serotonin and norepinephrine reuptake inhibitor, widely used for the treatment of major depressive disorder. Although DLX has shown good efficacy and safety, serious adverse effects (e.g., liver injury) have been reported. The mechanisms associated with DLX-induced toxicity remain elusive. Drug metabolism plays critical roles in drug safety and efficacy. However, the metabolic profile of DLX in mice is not available, although mice serve as commonly used animal models for mechanistic studies of drug-induced adverse effects. Our study revealed 39 DLX metabolites in human/mouse liver microsomes and mice. Of note, 13 metabolites are novel, including five N-acetyl cysteine adducts and one reduced glutathione (GSH) adduct associated with DLX. Additionally, the species differences of certain metabolites were observed between human and mouse liver microsomes. CYP1A2 and CYP2D6 are primary enzymes responsible for the formation of DLX metabolites in liver microsomes, including DLX-GSH adducts. In summary, a total of 39 DLX metabolites were identified, and species differences were noticed in vitro. The roles of CYP450s in DLX metabolite formation were also verified using human recombinant cytochrome P450 (P450) enzymes and corresponding chemical inhibitors. Further studies are warranted to address the exact role of DLX metabolism in its adverse effects in vitro (e.g., human primary hepatocytes) and in vivo (e.g., Cyp1a2-null mice). SIGNIFICANCE STATEMENT: This current study systematically investigated Duloxetine (DLX) metabolism and bioactivation in liver microsomes and mice. This study provided a global view of DLX metabolism and bioactivation in liver microsomes and mice, which are very valuable to further elucidate the mechanistic study of DLX-related adverse effects and drug-drug interaction from metabolic aspects.
Collapse
Affiliation(s)
- Xuan Qin
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - John M Hakenjos
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Kevin R MacKenzie
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Mercedes Barzi
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Hemantkumar Chavan
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Pranavanand Nyshadham
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Jin Wang
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Sung Yun Jung
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Joie Z Guner
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Si Chen
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Lei Guo
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Partha Krishnamurthy
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Karl-Dimiter Bissig
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Stephen Palmer
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Martin M Matzuk
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| | - Feng Li
- Center for Drug Discovery, Department of Pathology & Immunology (X.Q., J.M.H., K.R.M., P.N., J.Z.G., S.P., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (K.R.M., F.L.), Department of Pharmacology & Chemical Biology (K.R.M., J.W., M.M.M., F.L.), and Department of Molecular & Cellular Biology (S.Y.J., K.-D.B., F.L.), Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina (M.B., K.-D.B.); Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (H.C., P.K.); and Division of Biochemical Toxicology, National Center for Toxicological Research/US Food and Drug Administration (FDA), Jefferson, Arkansas (S.C., L.G.)
| |
Collapse
|
9
|
Yang Y, Yang Q, Luo S, Zhang Y, Lian C, He H, Zeng J, Zhang G. Comparative Analysis Reveals Novel Changes in Plasma Metabolites and Metabolomic Networks of Infants With Retinopathy of Prematurity. Invest Ophthalmol Vis Sci 2022; 63:28. [PMID: 35060995 PMCID: PMC8787637 DOI: 10.1167/iovs.63.1.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Advances in mass spectrometry have provided new insights into the role of metabolomics in the etiology of several diseases. Studies on retinopathy of prematurity (ROP), for example, overlooked the role of metabolic alterations in disease development. We employed comprehensive metabolic profiling and gold-standard metabolic analysis to explore major metabolites and metabolic pathways, which were significantly affected in early stages of pathogenesis toward ROP. Methods This was a multicenter, retrospective, matched-pair, case-control study. We collected plasma from 57 ROP cases and 57 strictly matched non-ROP controls. Non-targeted ultra-high-performance liquid chromatography-tandem mass spectroscopy (UPLC-MS/MS) was used to detect the metabolites. Machine learning was employed to reveal the most affected metabolites and pathways in ROP development. Results Compared with non-ROP controls, we found a significant metabolic perturbation in the plasma of ROP cases, which featured an increase in the levels of lipids, nucleotides, and carbohydrate metabolites and lower levels of peptides. Machine leaning enabled us to distinguish a cluster of metabolic pathways (glycometabolism, redox homeostasis, lipid metabolism, and arginine pathway) were strongly correlated with the development of ROP. Moreover, the severity of ROP was associated with the levels of creatinine and ribitol; also, overactivity of aerobic glycolysis and lipid metabolism was noted in the metabolic profile of ROP. Conclusions The results suggest a strong correlation between metabolic profiling and retinal neovascularization in ROP pathogenesis. These findings provide an insight into the identification of novel metabolic biomarkers for the diagnosis and prevention of ROP, but the clinical significance requires further validation.
Collapse
Affiliation(s)
- Yuhang Yang
- Shenzhen Eye Hospital, Shenzhen Key Ophthalmic Laboratory, The Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Qian Yang
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Sisi Luo
- Shenzhen Key Prevention and Control Laboratory of Birth Defects Prevention and Control, Shenzhen Maternal and Child Health Hospital, The Affiliated Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Yinsheng Zhang
- School of Management and E-Business, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Chaohui Lian
- Shenzhen Key Prevention and Control Laboratory of Birth Defects Prevention and Control, Shenzhen Maternal and Child Health Hospital, The Affiliated Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Honghui He
- Shenzhen Eye Hospital, Shenzhen Key Ophthalmic Laboratory, The Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Jian Zeng
- Shenzhen Eye Hospital, Shenzhen Key Ophthalmic Laboratory, The Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Guoming Zhang
- Shenzhen Eye Hospital, Shenzhen Key Ophthalmic Laboratory, The Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| |
Collapse
|
10
|
Wang YK, Li WQ, Xia S, Guo L, Miao Y, Zhang BK. Metabolic Activation of the Toxic Natural Products From Herbal and Dietary Supplements Leading to Toxicities. Front Pharmacol 2021; 12:758468. [PMID: 34744736 PMCID: PMC8564355 DOI: 10.3389/fphar.2021.758468] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
Currently, herbal and dietary supplements have been widely applied to prevent and treat various diseases. However, the potential toxicities and adverse reactions of herbal and dietary supplements have been increasingly reported, and have gradually attracted widespread attention from clinical pharmacists and physicians. Metabolic activation of specific natural products from herbal and dietary supplements is mediated by hepatic cytochrome P450 or intestinal bacteria, and generates chemical reactive/toxic metabolites that bind to cellular reduced glutathione or macromolecules, and form reactive metabolites-glutathione/protein/DNA adducts, and these protein/DNA adducts can result in toxicities. The present review focuses on the relation between metabolic activation and toxicities of natural products, and provides updated, comprehensive and critical comment on the toxic mechanisms of reactive metabolites. The key inductive role of metabolic activation in toxicity is highlighted, and frequently toxic functional groups of toxic natural products were summarized. The biotransformation of drug cytochrome P450 or intestinal bacteria involved in metabolic activation were clarified, the reactive metabolites-protein adducts were selected as biomarkers for predicting toxicity. And finally, further perspectives between metabolic activation and toxicities of natural products from herbal and dietary supplements are discussed, to provide a reference for the reasonable and safe usage of herbal and dietary supplements.
Collapse
Affiliation(s)
- Yi-Kun Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wen Qun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Shuang Xia
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Guo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Miao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Bi-Kui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
11
|
MacKenzie KR, Zhao M, Barzi M, Wang J, Bissig KD, Maletic-Savatic M, Jung SY, Li F. Metabolic profiling of norepinephrine reuptake inhibitor atomoxetine. Eur J Pharm Sci 2020; 153:105488. [PMID: 32712217 PMCID: PMC7506503 DOI: 10.1016/j.ejps.2020.105488] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/25/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022]
Abstract
Atomoxetine (ATX), a selective and potent inhibitor of the presynaptic norepinephrine transporter, is used mainly to treat attention-deficit hyperactivity disorder. Although multiple adverse effects associated with ATX have been reported including severe liver injuries, the mechanisms of ATX-related toxicity remain largely unknown. Metabolism frequently contributes to adverse effects of a drug through reactive metabolites, and the bioactivation status of ATX is still not investigated yet. Here, we systematically investigated ATX metabolism, bioactivation, species difference in human, mouse, and rat liver microsomes (HLM, MLM, and RLM) and in mice using metabolomic approaches as mice and rats are commonly used animal models for the studies of drug toxicity. We identified thirty one ATX metabolites and adducts in LMs and mice, 16 of which are novel. In LMs, we uncovered two methoxyamine-trapped aldehydes, two cyclization metabolites, detoluene-ATX, and ATX-N-hydroxylation for the first time. Detoluene-ATX and one cyclization metabolite were also observed in mice. Using chemical inhibitors and recombinant CYP enzymes, we demonstrated that CYP2C8 and CYP2B6 mainly contribute to the formation of aldehyde; CYP2D6 is the dominant enzyme for the formation of ATX cyclization and detoluene-ATX; CYP3A4 is major enzyme responsible for the hydroxylamine formation. The findings concerning aldehydes should be very useful to further elucidate the mechanistic aspects of adverse effects associated with ATX from metabolic angles. Additionally, the species differences for each metabolite should be helpful to investigate the contribution of specific metabolites to ATX toxicity and possible drug-drug interactions in suitable models.
Collapse
Affiliation(s)
- Kevin R MacKenzie
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mingkun Zhao
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mercedes Barzi
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Karl-Dimiter Bissig
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mirjana Maletic-Savatic
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Feng Li
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Skillman B, Kerrigan S. CYP450-Mediated metabolism of suvorexant and investigation of metabolites in forensic case specimens. Forensic Sci Int 2020; 312:110307. [PMID: 32473525 DOI: 10.1016/j.forsciint.2020.110307] [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: 11/20/2019] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 01/10/2023]
Abstract
Suvorexant (Belsomra®) is a sedative hypnotic that was approved for use in 2015. It has a novel mechanism of action and was the first dual orexin receptor antagonist (DORA) to be approved for the treatment of sleep disorders. Sedative hypnotics often feature prominently in forensic investigations such as impaired driving and drug-facilitated sexual assault (DFSA) cases. As such, suvorexant is a drug of interest and its identification in forensic toxicology investigations is of significance. However, limited studies have been published to date and the disposition or importance of its metabolites has been largely uninvestigated. In this report, we investigate the enzymes responsible for metabolism and explore the prevalence of metabolites in blood from a series of thirteen forensic investigations. Recombinant cytochrome P450 enzymes (rCYPs) were used to generate phase I metabolites for suvorexant in vitro, and metabolites were identified using liquid chromatography-quadrupole/time-of-flight-mass spectrometry (LC-Q/TOF-MS). Four rCYP isoenzymes (3A4, 2C19, 2D6, and 2C9) were found to contribute to suvorexant metabolism. The only metabolite identified in blood or plasma arose from hydroxylation of the benzyl triazole moiety (M9). This metabolite was identified in seventeen blood and plasma specimens from twelve medicolegal death investigations and one impaired driving investigation. In the absence of a commercially available reference material, the metabolite was confirmed using rCYP-generated in vitro controls using high resolution mass spectrometry.
Collapse
Affiliation(s)
- Britni Skillman
- Sam Houston State University, Department of Forensic Science, 1003 Bowers Blvd., Huntsville, TX 77341, United States
| | - Sarah Kerrigan
- Sam Houston State University, Department of Forensic Science, 1003 Bowers Blvd., Huntsville, TX 77341, United States.
| |
Collapse
|
13
|
Yang YY, Wu ZY, Zhang H, Yin SJ, Xia FB, Zhang Q, Wan JB, Gao JL, Yang FQ. LC-MS-based multivariate statistical analysis for the screening of potential thrombin/factor Xa inhibitors from Radix Salvia Miltiorrhiza. Chin Med 2020; 15:38. [PMID: 32351617 PMCID: PMC7183602 DOI: 10.1186/s13020-020-00320-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 04/20/2020] [Indexed: 02/03/2023] Open
Abstract
Background The dry root and rhizome of Salvia miltiorrhiza Bunge, or Danshen, is a well-known traditional Chinese medicine with anticoagulant activity. Taking into account that thrombin (THR) and factor Xa (FXa) play crucial roles in the coagulation cascade, it is reasonable and meaningful to screening THR and/or FXa inhibitors from Danshen. Methods Four extracts [butanol (BA), ethyl acetate (EA) and remained extract (RE) from 75% ethanol extract, and water extract (WE)] of Danshen were prepared, and their THR/FXa inhibitory activities were assessed in vitro. Then, the active EA extract was further separated by silica-gel column chromatography (SC), and its fractions (SC1–SC5) were analyzed by LC–MS. The principal component analysis (PCA) and orthogonal partial least squares discriminate analysis (OPLS-DA) were employed for predicting the specific marker compounds. The chemical structures of targeted compounds were identified by LC–MS/MS and their interactions with THR/FXa were analyzed by the molecular docking analysis. Results Danshen EA extract showed strong activity against THR and FXa, and its fractions (SC1–SC5) exhibited obvious difference in inhibitory activity against these two enzymes. Furthermore, four marker compounds with potential THR/FXa inhibitory activity were screened by PCA and OPLS-DA, and were identified as cryptotanshinone, tanshinone I, dihydrotanshinone I and tanshinone IIA. The molecular docking study showed that all these four tanshinones can interact with some key amino acid residues of the THR/FXa active cavities, such as HIS57 and SER195, which were considered to be promising candidates targeting THR and/or FXa with low binding energy (< − 7 kcal mol−1). Conclusions LC–MS combined with multivariate statistical analysis can effectively screen potential THR/FXa inhibitory components in Danshen.
Collapse
Affiliation(s)
- Yi-Yao Yang
- 1School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 401331 People's Republic of China
| | - Zhao-Yu Wu
- 1School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 401331 People's Republic of China
| | - Hao Zhang
- 1School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 401331 People's Republic of China
| | - Shi-Jun Yin
- 1School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 401331 People's Republic of China
| | - Fang-Bo Xia
- 3State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, People's Republic of China
| | - Qian Zhang
- 1School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 401331 People's Republic of China
| | - Jian-Bo Wan
- 3State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, People's Republic of China
| | - Jian-Li Gao
- 2Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053 People's Republic of China
| | - Feng-Qing Yang
- 1School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 401331 People's Republic of China
| |
Collapse
|
14
|
Gawlik M, Savic V, Jovanovic M, Skibiński R. Mimicking of Phase I Metabolism Reactions of Molindone by HLM and Photocatalytic Methods with the Use of UHPLC-MS/MS. Molecules 2020; 25:E1367. [PMID: 32192164 PMCID: PMC7144366 DOI: 10.3390/molecules25061367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 01/30/2023] Open
Abstract
Establishing the metabolism pathway of the drug undergoing the hepatic biotransformation pathway is one of the most important aspects in the preclinical discovery process since the presence of toxic or reactive metabolites may result in drug withdrawal from the market. In this study, we present the structural elucidation of six, not described yet, metabolites of an antipsychotic molecule: molindone. The elucidation of metabolites was supported with a novel photocatalytical approach with the use of WO3 and WS2 assisted photochemical reactions. An UHPLC-ESI-Q-TOF combined system was used for the registration of all obtained metabolite profiles as well as to record the high resolution fragmentation spectra of the observed transformation products. As a reference in the in vitro metabolism simulation method, the incubation with human liver microsomes was used. Chemometric comparison of the obtained profiles pointed out the use of the WO3 approach as being more convenient in the field of drug metabolism studies. Moreover, the photocatalysis was used in the direction of the main drug metabolite synthesis in order to further isolation and characterization.
Collapse
Affiliation(s)
- Maciej Gawlik
- Department of Medicinal Chemistry, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| | - Vladimir Savic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (V.S.); (M.J.)
| | - Milos Jovanovic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (V.S.); (M.J.)
| | - Robert Skibiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| |
Collapse
|
15
|
Yang Y, Wu Z, Li S, Yang M, Xiao X, Lian C, Wen W, He H, Zeng J, Wang J, Zhang G. Targeted Blood Metabolomic Study on Retinopathy of Prematurity. Invest Ophthalmol Vis Sci 2020; 61:12. [PMID: 32049343 PMCID: PMC7326483 DOI: 10.1167/iovs.61.2.12] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/20/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose This study aims at exploring alterations of major metabolites and metabolic pathways in retinopathy of prematurity (ROP) infants and identifying biomarkers that may merit early diagnosis of ROP. Methods We analyzed targeted metabolites from 81 premature infants (<34 weeks of gestational age), including 40 ROP cases (15 males and 25 females, birth weight 1.263 ± 0. 345 kg, gestational age 31.20 ± 4.62 weeks) and 41 cases (30 males, 11 females, birth weight 1.220 ± 0.293 kg, gestational age 30.96 ± 4.17 weeks) of well-matched non-ROP controls. Metabolites were measured by ultra-performance liquid chromatography-tandem mass spectrometry. Standard multivariate and univariate analysis was performed to interpret metabolomic results. Results Glycine, glutamate, leucine, serine, piperidine, valine, tryptophan, citrulline, malonyl carnitine (C3DC), and homocysteine were identified as the top discriminant metabolites. In particular, discriminant concentrations of C3DC and glycine were also confirmed by univariate analysis as statistically significant different between ROP and non-ROP infants. Conclusions This study gained an insight into the metabolomic aspects of ROP development. We suggest that higher blood levels of C3DC and glycine can be promising biomarkers to predict the occurrence, but not the severity of ROP.
Collapse
|
16
|
Zhou ZM, Wang YK, Yan DM, Fang JH, Xiao XR, Zhang T, Cheng Y, Xu KP, Li F. Metabolic profiling of tyrosine kinase inhibitor nintedanib using metabolomics. J Pharm Biomed Anal 2019; 180:113045. [PMID: 31887668 DOI: 10.1016/j.jpba.2019.113045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/15/2019] [Accepted: 12/14/2019] [Indexed: 01/23/2023]
Abstract
Nintedanib is a promising tyrosine kinase inhibitor for clinically treating idiopathic pulmonary fibrosis (IPF). Some clinical cases reported that nintedanib treatment can cause hepatotoxicity and myocardial toxicity. U. S. FDA warns the potential drug-drug interaction when it is co-administrated with other drugs. In order to understand the potential toxicity of nintedanib and avoid drug-drug interaction, the metabolism of nintedanib was systematically investigated in human liver microsomes and mice using metabolomics approach, and the toxicity of metabolites was predicted by ADMET lab. Nineteen metabolites were detected in vivo and in vitro metabolism, and 8 of them were undescribed. Calculated partition coefficients (Clog P) were used to distinguish the isomers of nintedanib metabolites in this study. The major metabolic pathways of nintedanib majorly included hydroxylation, demethylation, glucuronidation, and acetylation reactions. The ADMET prediction indicated that nintedanib was a substrate of the cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (P-gp). And nintedanib and most of its metabolites might possess potential hepatotoxicity and cardiotoxicity. This study provided a global view of nintedanib metabolism, which could be used to understand the mechanism of adverse effects related to nintedanib and its potential drug-drug interaction.
Collapse
Affiliation(s)
- Zi-Meng Zhou
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Yi-Kun Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dong-Mei Yan
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Jian-He Fang
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Xue-Rong Xiao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Ting Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan Cheng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Kang-Ping Xu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Fei Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| |
Collapse
|
17
|
Wang YK, Yang XN, Zhu X, Xiao XR, Yang XW, Qin HB, Gonzalez FJ, Li F. Role of Metabolic Activation in Elemicin-Induced Cellular Toxicity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8243-8252. [PMID: 31271289 PMCID: PMC7385589 DOI: 10.1021/acs.jafc.9b02137] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Elemicin, an alkenylbenzene constituent of natural oils of several plant species, is widely distributed in food, dietary supplements, and medicinal plants. 1'-Hydroxylation is known to cause metabolic activation of alkenylbenzenes leading to their potential toxicity. The aim of this study was to explore the relationship between elemicin metabolism and its toxicity through comparing the metabolic maps between elemicin and 1'-hydroxyelemicin. Elemicin was transformed into a reactive metabolite of 1'-hydroxyelemicin, which was subsequently conjugated with cysteine (Cys) and N-acetylcysteine (NAC). Administration of NAC could significantly ameliorate the elemicin- and 1'-hydroxyelemicin-induced cytotoxicity of HepG2 cells, while depletion of Cys with diethyl maleate (DEM) increased cytotoxicity. Recombinant human CYP screening and CYP inhibition experiments revealed that multiple CYPs, notably CYP1A1, CYP1A2, and CYP3A4, were responsible for the metabolic activation of elemicin. This study revealed that metabolic activation plays a critical role in elemicin cytotoxicity.
Collapse
Affiliation(s)
- Yi-Kun Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Nan Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plant, Nanning 530023, China
| | - Xu Zhu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue-Rong Xiao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Xiu-Wei Yang
- School of Pharmaceutical Sciences, Peking University Health Science Center, Peking University, Beijing 100191, China
| | - Hong-Bo Qin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Corresponding Authors. Tel: +86-871-65238010. Fax: +86-871-65238010. . Tel: +86-871-65216953. Fax: +86-871-65216953
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Fei Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Corresponding Authors. Tel: +86-871-65238010. Fax: +86-871-65238010. . Tel: +86-871-65216953. Fax: +86-871-65216953
| |
Collapse
|
18
|
Quilles Jr JC, Bernardi MD, Batista PH, Silva SC, Rocha CM, Montanari CA, Leitão A. Biological Activity and Physicochemical Properties of Dipeptidyl Nitrile Derivatives Against Pancreatic Ductal Adenocarcinoma Cells. Anticancer Agents Med Chem 2019; 19:112-120. [DOI: 10.2174/1871520618666181029141649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/09/2018] [Accepted: 10/17/2018] [Indexed: 01/26/2023]
Abstract
Background:
Pancreatic cancer is one of the most aggressive types with high mortality in patients. Therefore,
studies to discover new drugs based on cellular targets have been developed to treat this disease. Due to the
importance of Cysteine Protease (CP) to several cellular processes in cancer cells, CP inhibitors have been studied as
novel alternative approaches for pancreatic cancer therapy.
Objective:
The cytostatic potential of new CP inhibitors derived from dipeptidyl nitriles is analyzed in vitro using
pancreatic cancer (MIA PaCa-2) cells.
Methods:
The cytotoxic and cytostatic activities were studied using MTT colorimetric assay in 2D and 3D cultures.
Colony formation, migration in Boyden chamber and cell cycle analysis were applied to further study the cytostatic
activity. The inhibition of cysteine proteases was evaluated with Z-FR-MCA selective substrate, and ROS evaluation
was performed with DCFH-DA fluorophore. Permeability was investigated using HPLC-MS to obtain log kw. Combination
therapy was also evaluated using the best compound with gemcitabine.
Results:
The inhibition of intracellular CP activity by the compounds was confirmed, and the cytostatic effect was
established with cell cycle retention in the G1 phase. CP inhibitors were able to reduce cell proliferation by 50% in
the clonogenic assay, and the same result was achieved for the migration assay, without any cytotoxic effect. The
Neq0554 inhibitor was also efficient to increase the gemcitabine potency in the combination therapy. Physicochemical
properties using an artificial membrane model quantified 1.14 ≥ log Kw ≥ 0.75 for all inhibitors (also confirmed
using HPLC-MS analysis) along with the identification of intra and extracellular metabolites. Finally, these dipeptidyl
nitrile derivatives did not trigger the formation of reactive oxygen species, which is linked to genotoxicity.
Conclusion:
Altogether, these results provide a clear and favorable picture to develop CP inhibitors in pre-clinical
assays.
Collapse
Affiliation(s)
- José C. Quilles Jr
- Medicinal Chemistry Group (NEQUIMED), Sao Carlos Institute of Chemistry (IQSC), University of Sao Paulo (USP) - Av. Trabalhador Sao-carlense, 400, Sao Carlos, SP, Brazil
| | - Murillo D.L. Bernardi
- Medicinal Chemistry Group (NEQUIMED), Sao Carlos Institute of Chemistry (IQSC), University of Sao Paulo (USP) - Av. Trabalhador Sao-carlense, 400, Sao Carlos, SP, Brazil
| | - Pedro H.J. Batista
- Medicinal Chemistry Group (NEQUIMED), Sao Carlos Institute of Chemistry (IQSC), University of Sao Paulo (USP) - Av. Trabalhador Sao-carlense, 400, Sao Carlos, SP, Brazil
| | - Samelyn C.M. Silva
- Medicinal Chemistry Group (NEQUIMED), Sao Carlos Institute of Chemistry (IQSC), University of Sao Paulo (USP) - Av. Trabalhador Sao-carlense, 400, Sao Carlos, SP, Brazil
| | - Camila M.R. Rocha
- Medicinal Chemistry Group (NEQUIMED), Sao Carlos Institute of Chemistry (IQSC), University of Sao Paulo (USP) - Av. Trabalhador Sao-carlense, 400, Sao Carlos, SP, Brazil
| | - Carlos A. Montanari
- Medicinal Chemistry Group (NEQUIMED), Sao Carlos Institute of Chemistry (IQSC), University of Sao Paulo (USP) - Av. Trabalhador Sao-carlense, 400, Sao Carlos, SP, Brazil
| | - Andrei Leitão
- Medicinal Chemistry Group (NEQUIMED), Sao Carlos Institute of Chemistry (IQSC), University of Sao Paulo (USP) - Av. Trabalhador Sao-carlense, 400, Sao Carlos, SP, Brazil
| |
Collapse
|
19
|
Kadi AA, Yin W, Rahman AFMM. In-vitro metabolic profiling study of potential topoisomerase inhibitors 'pyrazolines' in RLMs by mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1114-1115:125-133. [PMID: 30953840 DOI: 10.1016/j.jchromb.2019.03.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/24/2019] [Accepted: 03/19/2019] [Indexed: 01/25/2023]
Abstract
Taking into consideration of the cytotoxicity and topo-IIα inhibitory activity of pyrazoline derivatives (1-3) against HCT15 cells, and known topo-IIα inhibitor, etoposide, respectively, the compounds were biotransformed in rat liver microsomes. LC-MS/MS and MALDI mass spectrometric techniques has been used for analysis. All three compounds were biotransformed into demethylated metabolites. Among three compounds, compounds 1 and 2 were biotransformed into mono-hydroxylated metabolites and compound 3 biotransformed into reduced and epoxidized metabolites. Reduced and reduced along with demethylation metabolites were identified from MALDI Orbitrap spectrometric analysis. Without NADPH or microsomes no compounds (1-3) were generated metabolites, it shows CYP450 enzymes involvement in the presence of NADPH in the metabolisms.
Collapse
Affiliation(s)
- Adnan A Kadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wencui Yin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - A F M Motiur Rahman
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
20
|
Chen C, Fan Z, Xu H, Tan X, Zhu M. Metabolomics-based parallel discovery of xenobiotics and induced endogenous metabolic dysregulation in clinical toxicology. Biomed Chromatogr 2018; 33:e4413. [PMID: 30357883 DOI: 10.1002/bmc.4413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/07/2018] [Accepted: 10/17/2018] [Indexed: 12/23/2022]
Abstract
Intoxication by xenobiotics triggers the perturbation of metabolic fingerprints in biofluids, including the accumulation of xenobiotic compounds and the dysregulation of endogenous metabolites. In this work, an untargeted metabolomics workflow was developed to simultaneously profile both xenobiotic and endogenous metabolites for the identification of the xenobiotic origin and an in-depth understanding of the intoxication mechanism. This workflow was demonstrated in a real-world clinical case. Plasma samples were collected from four intoxicated children and another three healthy children. Untargeted metabolomics analysis was performed using ultraperformance liquid chromatography (UPLC) coupled to a high-resolution mass spectrometer (HRMS) with data-independent MSE acquisition. LC-MSE data was processed using an untargeted metabolomics data interpretation workflow, in which the identities of xenobiotics and altered endogenous metabolic features were determined via database searching. Five xenobiotic chemicals and 19 endogenous metabolites were found to be dysregulated. Combined with the clinical evidence, penfluridol was confirmed as the xenobiotic toxin. Furthermore, a mechanistic hypothesis was developed to explain the dysregulation of the four endogenous acyl-carnitines. This workflow can be readily applied to a wide range of clinical toxicology cases, offering a powerful and convenient means of simultaneous discovery of intoxication source and the understanding of intoxication mechanisms.
Collapse
Affiliation(s)
- Chang Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Ziquan Fan
- Waters Technology (Shanghai) Co. Ltd, Shanghai, China
| | - Hui Xu
- PICU, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Xiaojie Tan
- Waters Technology (Shanghai) Co. Ltd, Shanghai, China
| | | |
Collapse
|
21
|
Shestakova K, Brito A, Mesonzhnik NV, Moskaleva NE, Kurynina KO, Grestskaya NM, Serkov IV, Lyubimov II, Bezuglov VV, Appolonova SA. Rabbit plasma metabolomic analysis of Nitroproston®: a multi target natural prostaglandin based-drug. Metabolomics 2018; 14:112. [PMID: 30830378 DOI: 10.1007/s11306-018-1413-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/12/2018] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Nitroproston® is a novel multi-target drug bearing natural prostaglandin E2 (PGE2) and nitric oxide (NO)-donating fragments for treatment of inflammatory and obstructive diseases (i.e., asthma and obstructive bronchitis). OBJECTIVES To investigate the effects of Nitroproston® administration on plasma metabolomics in vivo. METHODS Experimental in vivo study randomly assigning the target drug (treatment group) or a saline solution without the drug (vehicle control group) to 12 rabbits (n = 6 in each group). Untargeted (5880 initial features; 1869 negative-4011 positive ion peaks; UPLC-IT-TOF/MS) and 84 targeted moieties (Nitroproston® related metabolites, prostaglandins, steroids, purines, pyrimidines and amino acids; HPLC-QQQ-MS/MS) were measured from plasma at 0, 2, 4, 6, 8, 12, 18, 24, 32 and 60 min after administration. RESULTS PGE2, 13,14-dihydro-15-keto-PGE2, PGB2, 1,3-GDN and 15-keto-PGE2 increased in the treatment group. Steroids (i.e., cortisone, progesterone), organic acids, 3-oxododecanoic acid, nicotinate D-ribonucleoside, thymidine, the amino acids serine and aspartate, and derivatives pyridinoline, aminoadipic acid and uric acid increased (p < 0.05 AUCROC curve > 0.75) after treatment. Purines (i.e., xanthine, guanine, guanosine), bile acids, acylcarnitines and the amino acids L-tryptophan and L-phenylalanine were decreased. Nitroproston® impacted steroidogenesis, purine metabolism and ammonia recycling pathways, among others. CONCLUSION Nitroproston®, a multi action novel drug based on natural prostaglandins, altered metabolites (i.e., guanine, adenine, cortisol, cortisone and aspartate) involved in purine metabolism, urea and ammonia biological cycles, steroidogenesis, among other pathways. Suggested mechanisms of action, metabolic pathway interconnections and useful information to further understand the metabolic effects of prostaglandin administration are presented.
Collapse
Affiliation(s)
- Ksenia Shestakova
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya St., Moscow, Russia, 119991
- PhD Program in Nanoscience and Advanced Technology, Department of Diagnostics and Public Health, University of Verona, Policlinico G.B. Rossi - P.le L.A. Scuro 10, 37134, Verona, Italy
| | - Alex Brito
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya St., Moscow, Russia, 119991
| | - Natalia V Mesonzhnik
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya St., Moscow, Russia, 119991
| | - Natalia E Moskaleva
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya St., Moscow, Russia, 119991
| | - Ksenia O Kurynina
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya St., Moscow, Russia, 119991
| | - Natalia M Grestskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Ulitsa Miklukho-Maklaya, 16/10, Moscow, Russia, 117997
| | - Igor V Serkov
- Institute of Physiologically Active Compounds RAS, Severniy pr., 1, Chernogolovka, Russia, 142432
| | - Igor I Lyubimov
- LLC "Gurus BioPharm", Territory of Skolkovo Innovation Center, Bolshoy Boulevard, 42 Building 1, Moscow, Russia, 143026
| | - Vladimir V Bezuglov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Ulitsa Miklukho-Maklaya, 16/10, Moscow, Russia, 117997
| | - Svetlana A Appolonova
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya St., Moscow, Russia, 119991.
| |
Collapse
|
22
|
Johnsi Rani P, Vishnuvardhan C, Nimbalkar RD, Garg P, Satheeshkumar N. Metabolite characterization of ambrisentan, in in vitro and in vivo matrices by UHPLC/QTOF/MS/MS: Detection of glutathione conjugate of epoxide metabolite evidenced by in vitro GSH trapping assay. J Pharm Biomed Anal 2018; 155:320-328. [DOI: 10.1016/j.jpba.2018.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 01/11/2023]
|
23
|
Li F, Zhang N, Gorantla S, Gilbertson SR, Pati D. The Metabolism of Separase Inhibitor Sepin-1 in Human, Mouse, and Rat Liver Microsomes. Front Pharmacol 2018; 9:313. [PMID: 29867452 PMCID: PMC5949348 DOI: 10.3389/fphar.2018.00313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/19/2018] [Indexed: 01/02/2023] Open
Abstract
Separase, a known oncogene, is widely overexpressed in numerous human tumors of breast, bone, brain, blood, and prostate. Separase is an emerging target for cancer therapy, and separase enzymatic inhibitors such as sepin-1 are currently being developed to treat separase-overexpressed tumors. Drug metabolism plays a critical role in the efficacy and safety of drug development, as well as possible drug–drug interactions. In this study, we investigated the in vitro metabolism of sepin-1 in human, mouse, and rat liver microsomes (RLM) using metabolomic approaches. In human liver microsomes (HLM), we identified seven metabolites including one cysteine–sepin-1 adduct and one glutathione–sepin-1 adduct. All the sepin-1 metabolites in HLM were also found in both mouse and RLM. Using recombinant CYP450 isoenzymes, we demonstrated that multiple enzymes contributed to the metabolism of sepin-1, including CYP2D6 and CYP3A4 as the major metabolizing enzymes. Inhibitory effects of sepin-1 on seven major CYP450s were also evaluated using the corresponding substrates recommended by the US Food and Drug Administration. Our studies indicated that sepin-1 moderately inhibits CYP1A2, CYP2C19, and CYP3A4 with IC50 < 10 μM but weakly inhibits CYP2B6, CYP2C8/9, and CYP2D6 with IC50 > 10 μM. This information can be used to optimize the structures of sepin-1 for more suitable pharmacological properties and to predict the possible sepin-1 interactions with other chemotherapeutic drugs.
Collapse
Affiliation(s)
- Feng Li
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.,Advance Technology Core, Baylor College of Medicine, Houston, TX, United States
| | - Nenggang Zhang
- Texas Children's Cancer Center, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Siddharth Gorantla
- Texas Children's Cancer Center, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Scott R Gilbertson
- Department of Chemistry, University of Houston, Houston, TX, United States
| | - Debananda Pati
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Cancer Center, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
24
|
Metabolomics-assisted metabolite profiling of itraconazole in human liver preparations. J Chromatogr B Analyt Technol Biomed Life Sci 2018. [PMID: 29524695 DOI: 10.1016/j.jchromb.2018.02.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Itraconazole (ITZ) is a first-generation triazole-containing antifungal agent that effectively treats various fungal infections. As ITZ has a better safety profile than that of ketoconazole (KCZ), ITZ has been used worldwide for over 25 years. However, few reports have explored the metabolic profile of ITZ, and the underlying mechanism of ITZ-induced liver injury is not clearly understood. In the present study, we revisited ITZ metabolism in humans, using a non-targeted metabolomics approach, and identified several novel metabolic pathways including O-dearylation, piperazine oxidation, and piperazine-N,N'-deethylation. Furthermore, we explored the formation of reactive ITZ metabolites using trapping agents as surrogates, to assess the possibility of metabolism-mediated toxicity. We found that ITZ and its metabolites did not form any adducts with nucleophiles including glutathione, potassium cyanide, and semicarbazide. The present study expands our knowledge of ITZ metabolism and supports the suggestion that ITZ has a better safety profile than that of KCZ in terms of metabolism-mediated toxicity.
Collapse
|
25
|
Nikles S, Monschein M, Zou H, Liu Y, He X, Fan D, Lu A, Yu K, Isaac G, Bauer R. Metabolic profiling of the traditional Chinese medicine formulation Yu Ping Feng San for the identification of constituents relevant for effects on expression of TNF-α, IFN-γ, IL-1β and IL-4 in U937 cells. J Pharm Biomed Anal 2017; 145:219-229. [PMID: 28667937 DOI: 10.1016/j.jpba.2017.03.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/22/2017] [Accepted: 03/24/2017] [Indexed: 01/13/2023]
Abstract
Yu Ping Feng San (YPFS) is a classical TCM formulation which has been traditionally used for treatment of immune system related diseases such as chronic bronchitis, allergic rhinitis and asthma. The formula is a mixture of Radix Saposhnikoviae (Fangfeng), Radix Astragali (Huangqi), and Rhizoma Atractylodis macrocephalae (Baizhu). TLC- and LC-DAD-ESI-MS/MS methods have been developed for the analysis of the metabolic profiles of the single herbs and of the formula. Decoctions and ASE extracts were analyzed in order to trace components of the individual herbs in YPFS. Nine constituents of Radix Saposhnikoviae, ten constituents of Radix Astragali and five constituents of Rhizoma Atractylodis macrocephalae have been assigned in the chemical profiles of the formula, which now allow the standardisation of YPFS. The pharmacological testing showed that all extracts significantly inhibited expression of TNF-α, IFN-γ, and IL-1β in U937 cells, while the inhibition of IL-4 was consistently low. Compared to conventional analyses which are focused on a limited set of compounds, metabolomics approaches, together with novel data processing tools, enable a more holistic comparison of the herbal extracts. In order to identify the constituents which are relevant for the immunomodulatory effects of the formula, metabolomics studies (PCA, OPLS-DA) have been performed using UPLC/QTOF MS data.
Collapse
Affiliation(s)
- Stefanie Nikles
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Universitätsplatz 4/I, 8010 Graz, Austria
| | - Marlene Monschein
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Universitätsplatz 4/I, 8010 Graz, Austria
| | - Huiqin Zou
- School of Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Liu
- School of Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojuan He
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Danping Fan
- China Academy of Chinese Medical Sciences, Institute of Basic Research in Clinical Medicine, Dongzhimennei Nanxiaojie 16, 100700 Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China; China Academy of Chinese Medical Sciences, Institute of Basic Research in Clinical Medicine, Dongzhimennei Nanxiaojie 16, 100700 Beijing, China
| | - Kate Yu
- Waters Corporation, 5 Technology Drive, 01757 Milford, USA
| | - Giorgis Isaac
- Waters Corporation, 5 Technology Drive, 01757 Milford, USA
| | - Rudolf Bauer
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Universitätsplatz 4/I, 8010 Graz, Austria.
| |
Collapse
|
26
|
Revisiting the Metabolism and Bioactivation of Ketoconazole in Human and Mouse Using Liquid Chromatography-Mass Spectrometry-Based Metabolomics. Int J Mol Sci 2017; 18:ijms18030621. [PMID: 28335386 PMCID: PMC5372636 DOI: 10.3390/ijms18030621] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 01/01/2023] Open
Abstract
Although ketoconazole (KCZ) has been used worldwide for 30 years, its metabolic characteristics are poorly described. Moreover, the hepatotoxicity of KCZ limits its therapeutic use. In this study, we used liquid chromatography–mass spectrometry-based metabolomics to evaluate the metabolic profile of KCZ in mouse and human and identify the mechanisms underlying its hepatotoxicity. A total of 28 metabolites of KCZ, 11 of which were novel, were identified in this study. Newly identified metabolites were classified into three categories according to the metabolic positions of a piperazine ring, imidazole ring, and N-acetyl moiety. The metabolic characteristics of KCZ in human were comparable to those in mouse. Moreover, three cyanide adducts of KCZ were identified in mouse and human liver microsomal incubates as “flags” to trigger additional toxicity study. The oxidation of piperazine into iminium ion is suggested as a biotransformation responsible for bioactivation. In summary, the metabolic characteristics of KCZ, including reactive metabolites, were comprehensively understood using a metabolomics approach.
Collapse
|
27
|
Mumtaz MW, Hamid AA, Akhtar MT, Anwar F, Rashid U, AL-Zuaidy MH. An overview of recent developments in metabolomics and proteomics – phytotherapic research perspectives. FRONTIERS IN LIFE SCIENCE 2017. [DOI: 10.1080/21553769.2017.1279573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Muhammad Waseem Mumtaz
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Malaysia
- Department of Chemistry, Faculty of Science, University of Gujrat, Gujrat, Pakistan
| | - Azizah Abdul Hamid
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Malaysia
| | - Muhammad Tayyab Akhtar
- Institute of Bioscience, Laboratory of Natural Products, Universiti Putra Malaysia, Serdang, Malaysia
| | - Farooq Anwar
- Department of Chemistry, University of Sargodha, Sargodha, Pakistan
| | - Umer Rashid
- Institute of Advanced Technology, Universiti Putra Malaysia, Serdang, Malaysia
| | - Mizher Hezam AL-Zuaidy
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
28
|
|
29
|
Genovino J, Sames D, Hamann LG, Touré BB. Die Erschließung von Wirkstoffmetaboliten durch übergangsmetallkatalysierte C-H-Oxidation: die Leber als Inspiration für die Synthese. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201602644] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Julien Genovino
- Pfizer Inc.; Worldwide Medicinal Chemistry, Cardiovascular, Metabolic, and Endocrine Diseases (CVMED); 558 Eastern Point Road Groton CT 06340 USA
| | - Dalibor Sames
- Columbia University; Department of Chemistry and Neurotechnology Center; 3000 Broadway MC3101 New York NY 10027 USA
| | - Lawrence G. Hamann
- Novartis Institutes for Biomedical Sciences (NIBR), Global Discovery Chemistry (GDC); 181 Massachusetts Avenue Cambridge MA 02139 USA
| | - B. Barry Touré
- Novartis Institutes for Biomedical Sciences (NIBR), Global Discovery Chemistry (GDC); 100 Technology Square Cambridge MA 02139 USA
| |
Collapse
|
30
|
Genovino J, Sames D, Hamann LG, Touré BB. Accessing Drug Metabolites via Transition-Metal Catalyzed C-H Oxidation: The Liver as Synthetic Inspiration. Angew Chem Int Ed Engl 2016; 55:14218-14238. [PMID: 27723189 DOI: 10.1002/anie.201602644] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/08/2016] [Indexed: 11/07/2022]
Abstract
Can classical and modern chemical C-H oxidation reactions complement biotransformation in the synthesis of drug metabolites? We have surveyed the literature in an effort to try to answer this important question of major practical significance in the pharmaceutical industry. Drug metabolites are required throughout all phases of the drug discovery and development process; however, their synthesis is still an unsolved problem. This Review, not intended to be comprehensive or historical, highlights relevant applications of chemical C-H oxidation reactions, electrochemistry and microfluidic technologies to drug templates in order to access drug metabolites, and also highlights promising reactions to this end. Where possible or appropriate, the contrast with biotransformation is drawn. In doing so, we have tried to identify gaps where they exist in the hope to spur further activity in this very important research area.
Collapse
Affiliation(s)
- Julien Genovino
- Pfizer Inc., Worldwide Medicinal Chemistry, Cardiovascular, Metabolic, and Endocrine Diseases (CVMED), 558 Eastern Point Road, Groton, CT, 06340, USA
| | - Dalibor Sames
- Columbia University, Department of Chemistry and Neurotechnology Center, 3000 Broadway MC3101, New York, NY, 10027, USA
| | - Lawrence G Hamann
- Novartis Institutes for Biomedical Sciences (NIBR), Global Discovery Chemistry (GDC), 181 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - B Barry Touré
- Novartis Institutes for Biomedical Sciences (NIBR), Global Discovery Chemistry (GDC), 100 Technology Square, Cambridge, MA, 02139, USA.
| |
Collapse
|
31
|
Liu X, Lu YF, Guan X, Zhao M, Wang J, Li F. Characterizing novel metabolic pathways of melatonin receptor agonist agomelatine using metabolomic approaches. Biochem Pharmacol 2016; 109:70-82. [PMID: 27021842 DOI: 10.1016/j.bcp.2016.03.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/24/2016] [Indexed: 12/19/2022]
Abstract
Agomelatine (AGM), an analog of melatonin, is a potential agonist at melatonin receptors 1/2 and a selective antagonist at 5-hydroxytryptamine 2C receptors. AGM is widely used for the treatment of major depressive episodes in adults. However, multiple adverse effects associated with AGM have been reported in clinical practice. It is little known about AGM metabolism in vitro and in vivo, although metabolism plays a pivotal role in its efficacy and safety. To elucidate metabolic pathways of AGM, we systemically investigated AGM metabolism and its bioactivation in human liver microsomes (HLM) and mice using metabolomic approaches. We identified thirty-eight AGM metabolites and adducts, among which thirty-two are novel. In HLM, we uncovered five GSH-trapped adducts and two semicarbazide-trapped aldehydes. Moreover, we characterized three N-acetyl cysteine conjugated-AGM adducts in mouse urine and feces, which were formed from the degradation of AGM_GSH adducts. Using recombinant CYP450 isoenzymes and chemical inhibitors, we demonstrated that CYP1A2 and CYP3A4 are primary enzymes contributing to the formation of AGM_GSH adducts and AGM_hydrazones. This study provided a global view of AGM metabolism and identified the novel pathways of AGM bioactivation, which could be utilized for further understanding the mechanism of adverse effects related to AGM and possible drug-drug interactions.
Collapse
Affiliation(s)
- Xing Liu
- Department of Molecular and Cellular Biology, Alkek Center for Molecular Discovery, Advanced Technology Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuan-Fu Lu
- Key Lab for Basic Pharmacology of Ministry of Education, Zunyi Medical College, Zunyi, Guizhou 563000, China
| | - Xinfu Guan
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mingkun Zhao
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Wang
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Feng Li
- Department of Molecular and Cellular Biology, Alkek Center for Molecular Discovery, Advanced Technology Core, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Le H, Jerums M, Goudar CT. Characterization of intrinsic variability in time-series metabolomic data of cultured mammalian cells. Biotechnol Bioeng 2015; 112:2276-83. [DOI: 10.1002/bit.25646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/19/2015] [Accepted: 05/07/2015] [Indexed: 01/10/2023]
Affiliation(s)
- Huong Le
- Drug Substance Technologies, Process Development; Amgen, Inc.; One Amgen Center Drive, Thousand Oaks, California, 91320
| | - Matthew Jerums
- Drug Substance Technologies, Process Development; Amgen, Inc.; One Amgen Center Drive, Thousand Oaks, California, 91320
| | - Chetan T. Goudar
- Drug Substance Technologies, Process Development; Amgen, Inc.; One Amgen Center Drive, Thousand Oaks, California, 91320
| |
Collapse
|
33
|
Chemical dereplication of marine actinomycetes by liquid chromatography-high resolution mass spectrometry profiling and statistical analysis. Anal Chim Acta 2013; 805:70-9. [PMID: 24296145 DOI: 10.1016/j.aca.2013.10.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/02/2013] [Accepted: 10/11/2013] [Indexed: 12/15/2022]
Abstract
Discovery of novel bioactive metabolites from marine bacteria is becoming increasingly challenging, and the development of novel approaches to improve the efficiency of early steps in the microbial drug discovery process is therefore of interest. For example, current protocols for the taxonomic dereplication of microbial strains generally use molecular tools which do not take into consideration the ability of these selected bacteria to produce secondary metabolites. As the identification of novel chemical entities is one of the key elements driving drug discovery programs, this study reports a novel methodology to dereplicate microbial strains by a metabolomics approach using liquid chromatography-high resolution mass spectrometry (LC-HRMS). In order to process large and complex three dimensional LC-HRMS datasets, the reported method uses a bucketing and presence-absence standardization strategy in addition to statistical analysis tools including principal component analysis (PCA) and cluster analysis. From a closely related group of Streptomyces isolated from geographically varied environments, we demonstrated that grouping bacteria according to the chemical diversity of produced metabolites is reproducible and provides greatly improved resolution for the discrimination of microbial strains compared to current molecular dereplication techniques. Importantly, this method provides the ability to identify putative novel chemical entities as natural product discovery leads.
Collapse
|
34
|
Forcisi S, Moritz F, Kanawati B, Tziotis D, Lehmann R, Schmitt-Kopplin P. Liquid chromatography–mass spectrometry in metabolomics research: Mass analyzers in ultra high pressure liquid chromatography coupling. J Chromatogr A 2013; 1292:51-65. [DOI: 10.1016/j.chroma.2013.04.017] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 03/29/2013] [Accepted: 04/10/2013] [Indexed: 12/27/2022]
|