1
|
He C, Mao Y, Wan H. In-depth understanding of the structure-based reactive metabolite formation of organic functional groups. Drug Metab Rev 2025:1-43. [PMID: 40008940 DOI: 10.1080/03602532.2025.2472076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025]
Abstract
Idiosyncratic drug-induced liver injury (DILI) is a leading cause of drug attrition and/or withdrawal. The formation of reactive metabolites is widely accepted as a key factor contributing to idiosyncratic DILI. Therefore, identifying reactive metabolites has become a critical focus during lead optimization, and a combination of GSH-/cyano-trapping and cytochrome P450 inactivation studies is recommended to identify compounds with the potential to generate reactive metabolites. Daily dose, clinical indication, detoxication pathways, administration route, and treatment duration are the most considerations when deprioritizing candidates that generate reactive metabolites. Removing the structural alerts is considered a pragmatic strategy for mitigating the risk associated with reactive metabolites, although this approach may sometimes exclude otherwise potent molecules. In this context, an in-depth insight into the structure-based reactive metabolite formation of organic functional groups can significantly aid in the rational design of drug candidates with improved safety profiles. The primary goal of this review is to delve into an analysis of the bioactivation mechanisms of organic functional groups and their potential detrimental effects with recent examples to assist medicinal chemists and metabolism scientists in designing safer drug candidates with a higher likelihood of success.
Collapse
Affiliation(s)
- Chunyong He
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co. Ltd., Shanghai, China
| | - Yuchang Mao
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co. Ltd., Shanghai, China
| | - Hong Wan
- WHDex Consulting AB, Mölndal, Sweden
| |
Collapse
|
2
|
Jung YH, Lee DC, Kwon YM, Jang E, Choi G, Kim YH, Kim TH, Kim JH. The Comparative Metabolism of a Novel Hepatocellular Carcinoma Therapeutic Agent, 2,3-Diamino- N-(4-(benzo[d]thiazol-2-yl)phenyl)propanamide, in Human and Animal Hepatocytes. Metabolites 2024; 14:425. [PMID: 39195521 DOI: 10.3390/metabo14080425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
[2,3-diamino-N-(4-(benzo[d]thiazol-2-yl)phenyl)propanamide], named as ETN101, is a novel therapeutic agent for hepatocellular carcinoma. In vitro studies examined ETN101 metabolites in human, mouse, rat, dog, and monkey hepatocytes and identified the drug-metabolizing enzymes involved using cDNA-expressed human recombinant cytochrome P450s (CYPs), carboxylesterases (CESs), N-acetyltransferase (NAT) 1, and human liver cytosol. ETN101 showed similar metabolic stability across hepatocytes from five species, with particularly comparable stability in humans, rats, and monkeys. Its half-life was 75.0 min in humans, 68.9 in rats, 73.1 in monkeys, 120.4 in mice, and 112.7 in dogs. Thirty-four ETN101 metabolites, including the major metabolite M1, were identified using liquid chromatography-high-resolution mass spectrometry. ETN101 was primarily metabolized to M1 and CYP1A2 is exclusively responsible for M1 metabolism. Both NAT1 and NAT2 were responsible for the N-acetylation of M1 to M2. ETN101 remained stable in human CESs. In conclusion, this study provides comprehensive insights into the metabolic characteristics of ETN101, valuable for its toxicological and clinical development.
Collapse
Affiliation(s)
- Young-Heun Jung
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dong-Cheol Lee
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Ye-Min Kwon
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Eunbee Jang
- Etnova Therapeutics, Suwon 16648, Republic of Korea
| | - Garam Choi
- Etnova Therapeutics, Suwon 16648, Republic of Korea
| | | | - Tae Hwan Kim
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Ju-Hyun Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
3
|
Rudolph B, Davis JA, Hainzl D, Walles M. A general perspective for the conduct of radiolabelled distribution, metabolism, and excretion studies for antibody-drug conjugates. Xenobiotica 2024; 54:521-532. [PMID: 39329287 DOI: 10.1080/00498254.2024.2336576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 09/28/2024]
Abstract
Antibody-drug conjugates (ADCs) are a class of biopharmaceuticals that combine the specificity of monoclonal antibodies (mAbs) with the cytotoxicity of small molecule drugs. 15 ADCs have been approved by regulatory authorities up to now, mainly for indications in oncology, however, this review paper will only focus on the 13 ADCs that have been approved by either the FDA or EMA.ADME (Absorption, Distribution, Metabolism, and Excretion) studies are essential for the development of small molecule drugs to evaluate their disposition properties. These studies help to select drug candidates, determine the optimal dosing regimen and help to identify potential safety concerns for the drug of interest in human. Tissue distribution studies are also important as they facilitate the understanding of the efficacy and safety for parent drug and its metabolites in preclinical and clinical studies.For biologics, ADME studies are usually not required. In this paper, we review the existing approval packages and literature for approved ADCs to determine the extent of ADME studies performed as part of ADC registration packages.We conclude that ADME studies are recommended for the development of ADCs if new linkers and payloads are used that have never been used in humans before as these studies provide valuable information on the pharmacokinetic properties, optimal dosing regimen, and potential safety concerns. However, for the development of ADCs with established linker payload combinations, radiolabelled ADME studies may not be necessary if the distribution, metabolism and excretion properties have been described before. Clinical radiolabelled ADME studies are not recommended where patients are treated for life threating diseases like for indications in oncology.
Collapse
Affiliation(s)
- Bettina Rudolph
- Pharmacokinetic Sciences, Biomedical Research, Novartis Pharma, Basel, Switzerland
| | - John A Davis
- Pharmacokinetic Sciences, Biomedical Research, Novartis Pharma, Cambridge, Massachusetts, USA
| | - Dominik Hainzl
- Pharmacokinetic Sciences, Biomedical Research, Novartis Pharma, Cambridge, Massachusetts, USA
| | - Markus Walles
- Pharmacokinetic Sciences, Biomedical Research, Novartis Pharma, Basel, Switzerland
| |
Collapse
|
4
|
Wei X, Liu J, Xu Z, Wang D, Zhu Q, Chen Q, Xu W. Research progress on the pharmacological mechanism, in vivo metabolism and structural modification of Erianin. Biomed Pharmacother 2024; 173:116295. [PMID: 38401517 DOI: 10.1016/j.biopha.2024.116295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/31/2024] [Accepted: 02/17/2024] [Indexed: 02/26/2024] Open
Abstract
Erianin is an important bibenzyl compound in dendrobium and has a wide spectrum of pharmacological properties. Since Erianin was discovered, abundant results have been achieved in the in vitro synthesis, structural modification, and pharmacological mechanism research. Researchers have developed a series of simple and efficient in vitro synthesis methods to improve the shortcomings of poor water solubility by replacing the chemical structure or coating it in nanomaterials. Erianin has a broad anti-tumor spectrum and significant anti-tumor effects. In addition, Erianin also has pharmacological actions like immune regulation, anti-inflammatory, and anti-angiogenesis. A comprehensive understanding of the synthesis, metabolism, structural modification, and pharmacological action pathways of Erianin is of great value for the utilization of Erianin. Therefore, this review conducts a relatively systematic look back at Erianin from the above four aspects, to give a reference for the evolvement and further appliance of Erianin.
Collapse
Affiliation(s)
- Xin Wei
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Jiajia Liu
- University of Science and Technology of China, Hefei 230026, PR China; Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, PR China
| | - Ziming Xu
- University of Science and Technology of China, Hefei 230026, PR China; Department of Ophthalmology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230001, PR China
| | - Dan Wang
- University of Science and Technology of China, Hefei 230026, PR China; Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, PR China
| | - Qizhi Zhu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Qi Chen
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Weiping Xu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China; Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, PR China; Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei 230001, PR China.
| |
Collapse
|
5
|
Zhang X, Zhao S, Wang T, Shu C, Ding L. Development and validation of an LC-MS/MS method for simultaneous determination of EVT201 and its five metabolites in human plasma: Application to a clinical study in Chinese healthy subjects. J Pharm Biomed Anal 2023; 235:115601. [PMID: 37523867 DOI: 10.1016/j.jpba.2023.115601] [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: 06/09/2023] [Revised: 07/16/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023]
Abstract
EVT201 is a partial GABAA receptor agonist, which inhibits nervous system to treat insomnia. EVT201 can form a variety of metabolites in vivo including Ro46-1927, Ro18-5528, Ro40-9970, Ro66-9196 and Ro66-5448. This study developed a simple method to realize the simultaneous determination of EVT201 and its five metabolites by HPLC-MS/MS with an electrospray ion source (ESI). The deuterium substitute of EVT201 was chosen as the internal standard and the multiple reaction monitoring (MRM) was used for the quantification. The separation of the six compounds was accomplished with an ACE Excel 3 AQ column (50 × 2.1 mm, 3 µm, ACE). The process of protein precipitating-transferring-nitrogen blowing-reconstituting was adopted for the sample pretreatment. This method was successfully validated according to the FDA guidance. Calibration curves were linear over the concentration range of 0.100-100 ng/mL for EVT201, 0.0300-30.0 ng/mL for Ro46-1927, 0.0600-6.00 ng/mL for Ro18-5528, 0.0200-4.00 ng/mL for Ro40-9970, 0.100-20.0 ng/mL for Ro66-9196 and 0.100-20.0 ng/mL for Ro66-5448. The intra-run and inter-run precisions and accuracies were all within 14.5%. This fully validated method was successfully applied to a clinical pharmacokinetic study of EVT201 and its five metabolites in Chinese healthy subjects after the single (2.5 mg and 5 mg, N = 12) and multiple dose (2.5 mg, N = 13) administration of EVT201 capsules. The test results of 2.5 mg dose group showed that for EVT201, Ro46-1927, Ro18-5528, Ro40-9970, Ro66-9196 and Ro66-5448, the Cmax values (ng/mL) were 39.2 ± 9.2, 10.3 ± 1.4, 0.218 ± 0.044, 0.128 ± 0.051, 7.01 ± 1.51, 8.73 ± 3.69, respectively; the AUC0-t values (h·ng/mL) were 231 ± 79, 143 ± 72, 10.9 ± 2.1, 1.84 ± 0.78, 55.9 ± 18.7, 135 ± 40 respectively. For EVT201, Ro46-1927, Ro66-5528, Ro66-9196 and Ro40-5448, the results of Cmax and AUC0-t proved that the five compounds showed linear pharmacokinetic profile over the dose ranges of 2.5 mg to 5 mg. Meanwhile, it is the first report to evaluate the pharmacokinetic characteristics of Ro40-9970, Ro66-9196 and Ro66-5448 in human plasma. It provided meaningful parameters for the safety and tolerability evaluation of EVT201 capsules in human.
Collapse
Affiliation(s)
- Xinrui Zhang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China; Nanjing Clinical Tech. Laboratories Inc., Nanjing, China
| | - Shunbo Zhao
- Nanjing Clinical Tech. Laboratories Inc., Nanjing, China
| | - Tao Wang
- Nanjing Clinical Tech. Laboratories Inc., Nanjing, China
| | - Chang Shu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China.
| | - Li Ding
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China; Nanjing Clinical Tech. Laboratories Inc., Nanjing, China.
| |
Collapse
|
6
|
He C, Mao Y, Wan H. Preclinical evaluation of chemically reactive metabolites and mitigation of bioactivation in drug discovery. Drug Discov Today 2023; 28:103621. [PMID: 37201781 DOI: 10.1016/j.drudis.2023.103621] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/25/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023]
Abstract
The formation of reactive metabolites (RMs) is thought to be one of the pathogeneses for some idiosyncratic adverse drug reactions (IADRs) which are considered one of the leading causes of some drug attritions and/or recalls. Minimizing or eliminating the formation of RMs via chemical modification is a useful tactic to reduce the risk of IADRs and time-dependent inhibition (TDI) of cytochrome P450 enzymes (CYPs). The RMs should be carefully handled before making a go-no-go decision. Herein, we highlight the role of RMs in the occurrence of IADRs and CYP TDI, the risk of structural alerts, the approaches of RM assessment at the discovery stage and strategies to minimize or eliminate RM liability. Finally, some considerations for developing a RM-positive drug candidate are suggested.
Collapse
Affiliation(s)
- Chunyong He
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical, No. 279 Wenjing Road, Shanghai 200245, China.
| | - Yuchang Mao
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical, No. 279 Wenjing Road, Shanghai 200245, China
| | - Hong Wan
- Department of DMPK/Bioanalysis, Shanghai Medicilon, No. 585 Chuanda Road, Shanghai 201299, China.
| |
Collapse
|
7
|
Hu A, Liu Q, Ouyang J. Identification and characterization of the metabolites of moscatilin in mouse, rat, dog, monkey and human hepatocytes by LC-Orbitrap-MS/MS combined with diagnostic fragment ions and accurate mass measurements. Biomed Chromatogr 2023; 37:e5573. [PMID: 36529812 DOI: 10.1002/bmc.5573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Moscatilin, a bibenzyl derivative from the stem of Dendrobium loddigesii, has been shown to have anticancer activity. The aim of this study was to identify and characterize the possible in vitro metabolites of moscatilin generated from hepatocytes. The metabolites generated in the hepatocytes of mouse, rat, dog, monkey and human were identified and characterized employing ultra-high-performance liquid chromatography coupled with quadrupole Orbitrap tandem mass spectrometry (LC-Orbitrap-MS/MS) based on diagnostic fragment ions and accurate mass measurements. A total of 18 metabolites were identified, among which seven were phase I and 11 were phase II metabolites. The plausible structures of the metabolites and the probable biotransformation pathways were proposed based on the diagnostic fragment ions, chemical formula and mass fragmentation pattern, as well as the accurate masses. The majority of phase I metabolites were generated by demethylation and hydroxylation, while phase II metabolites were mainly generated by glucuronidation, glutathione conjugation and sulfation. Our study first expounded the metabolites of moscatilin in mouse, rat, dog, monkey and human hepatocytes and provided a foundation for a further pharmacokinetic and toxicity study. More importantly, LC-Orbitrap-MS/MS combined with diagnostic fragment ions and accurate mass measurements has been proved to be an effective method for the rapid identification of bibenzyl derivatives and their metabolites.
Collapse
Affiliation(s)
- Aizhen Hu
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Qingwang Liu
- Institute of Heath and Medical Technology, Hefei Institute of Physical Science, Chinese Academy of Sciences, Hefei, Anhui Province, China
| | - Jing Ouyang
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
8
|
Wang W, Wan Q, Li M, Qu F, Liu H, Chen Y. Design, synthesis and biological evaluation of seco-DSP/DCK derivatives reversing P-glycoprotein-mediated paclitaxel resistance in A2780/T cells. Eur J Med Chem 2023; 250:115218. [PMID: 36871374 DOI: 10.1016/j.ejmech.2023.115218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023]
Abstract
P-glycoprotein transporter (P-gp, ABCB1) is a major contributor to multidrug resistance, making it a valuable target for the development of novel P-gp inhibitor to overcome multidrug resistance. In this study, forty-nine novel seco-DSPs and seco-DMDCK derivatives were synthesized and evaluated their chemo-sensitize abilities to paclitaxel in A2780/T cell lines. Most of them exhibited a comparable reversal multidrug-resistance activity than verapamil. Especially, compound 27f showed a remarkable chemo-sensitization with more than 425-fold reversal ratio in A2780/T cells. The study of preliminary pharmacological mechanism displayed that compound 27f was more effective to increase the accumulation of paclitaxel and Rhodamine 123 than verapamil via inhibiting P-gp for reversing multidrug-resistance. In addition, a higher than 40 μM IC50 values of hERG potassium channel inhibition concentration suggested that compound 27f hardly had relevant cardiac toxicity. These results indicated that compound 27f might be a potential candidate to further investigate for the development of chemosensitizer with MDR reversal activity.
Collapse
Affiliation(s)
- Weijie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Qi Wan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Mengru Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Feng Qu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Hongrui Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Ying Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
9
|
Yan M, Li W, Li WB, Huang Q, Li J, Cai HL, Gong H, Zhang BK, Wang YK. Metabolic activation of tyrosine kinase inhibitors: recent advance and further clinical practice. Drug Metab Rev 2023; 55:94-106. [PMID: 36453523 DOI: 10.1080/03602532.2022.2149775] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
At present, receptor tyrosine kinase signaling-related pathways have been successfully mediated to inhibit tumor proliferation and promote anti-angiogenesis effects for cancer therapy. Tyrosine kinase inhibitors (TKIs), a group of novel chemotherapeutic agents, have been applied to treat diverse malignant tumors effectively. However, the latent toxic and side effects of TKIs, such as hepatotoxicity and cardiotoxicity, limit their use in clinical practice. Metabolic activation has the potential to lead to toxic effects. Numerous TKIs have been demonstrated to be transformed into chemically reactive/potentially toxic metabolites following cytochrome P450-catalyzed activation, which causes severe adverse reactions, including hepatotoxicity, cardiotoxicity, skin toxicity, immune injury, mitochondria injury, and cytochrome P450 inactivation. However, the precise mechanisms of how these chemically reactive/potentially toxic species induce toxicity remain poorly understood. In addition, we present our viewpoints that regulating the production of reactive metabolites may decrease the toxicity of TKIs. Exploring this topic will improve understanding of metabolic activation and its underlying mechanisms, promoting the rational use of TKIs. This review summarizes the updated evidence concerning the reactive metabolites of TKIs and the associated toxicities. This paper provides novel insight into the safe use of TKIs and the prevention and treatment of multiple TKIs adverse effects in clinical practice.
Collapse
Affiliation(s)
- Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Wen-Bo Li
- Department of Plastic and Aesthetic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qi Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Hua-Lin Cai
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Hui Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, 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
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Yi-Kun Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| |
Collapse
|
10
|
Althaher AR. An Overview of Hormone-Sensitive Lipase (HSL). ScientificWorldJournal 2022; 2022:1964684. [PMID: 36530555 PMCID: PMC9754850 DOI: 10.1155/2022/1964684] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 07/30/2023] Open
Abstract
Hormone-sensitive lipase (HSL) is a pivotal enzyme that mediates triglyceride hydrolysis to provide free fatty acids and glycerol in adipocytes in a hormonally controlled lipolysis process. Elevated plasma-free fatty acids were accompanied by insulin resistance, type-2 diabetes, and obesity. Inhibition of lipolysis through HSL inhibition may provide a mechanism to prevent the accumulation of free fatty acids and to improve the affectability of insulin and blood glucose handling in type II diabetes. The published studies that examine the structure, regulation, and function of HSL and major inhibitors were reviewed in this paper.
Collapse
Affiliation(s)
- Arwa R. Althaher
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| |
Collapse
|
11
|
Zhang R, Wu Q, Gao H, Li Y, Zhang P. Rapid separation and characterization of the in vitro metabolites of moscatilin by ultra-high performance liquid chromatography coupled to hybrid quadrupole orbitrap tandem mass spectrometry. J Sep Sci 2022; 45:4167-4175. [PMID: 36168860 DOI: 10.1002/jssc.202200617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/10/2022] [Accepted: 09/22/2022] [Indexed: 12/13/2022]
Abstract
Moscatilin, a bioactive ingredient isolated from Dendrobium moscatum, has been demonstrated to have excellent anti-cancer activity. The goals of the present study were to investigate the metabolic profiles of moscatilin and to identify and characterize its metabolites. In vitro studies were performed by incubating moscatilin (10 μM) with rat, dog, monkey, and human liver microsomes (0.5 mg protein/ml) to generate the metabolites. An analytical method of liquid chromatography combined with hybrid quadrupole orbitrap high-resolution mass spectrometry in full mass/data-dependent tandem mass spectrometry scan was utilized to separate and identify the metabolites in accordance with their accurate masses, formulas, and tandem mass spectrometry fragment ions determination. A total of six phase I metabolites were detected and structurally characterized. The phase I metabolic pathways of moscatilin were hydroxylation, demethylation, and dehydrogenation. In glutathione-supplemented liver microsomes, nine glutathione conjugates were detected and identified. Our results demonstrated that moscatilin was susceptible to bioactivation with the result of ortho quinone and quinone-methide intermediates. The present study provided an overview of the in vitro metabolic profiles of moscatilin, which will aid in the understanding of the efficacy and safety of this active compound.
Collapse
Affiliation(s)
- Rui Zhang
- Anhui No. 2 Provincial People's Hospital, Hefei, P. R. China
| | - Qiguo Wu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China.,Department of Pharmacy, Anqing Medical College, Anqing, P. R. China
| | - Han Gao
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Yue Li
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Peiliang Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, P. R. China.,Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| |
Collapse
|
12
|
Evodiamine as an anticancer agent: a comprehensive review on its therapeutic application, pharmacokinetic, toxicity, and metabolism in various cancers. Cell Biol Toxicol 2022; 39:1-31. [PMID: 36138312 DOI: 10.1007/s10565-022-09772-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022]
Abstract
Evodiamine is a major alkaloid component found in the fruit of Evodia rutaecarpa. It shows the anti-proliferative potential against a wide range of cancers by suppressing cell growth, invasion, and metastasis and inducing apoptosis both in vitro and in vivo. Evodiamine shows its anticancer potential by modulating aberrant signaling pathways. Additionally, the review focuses on several therapeutic implications of evodiamine, such as epigenetic modification, cancer stem cells, and epithelial to mesenchymal transition. Moreover, combinatory drug therapeutics along with evodiamine enhances the anticancer efficacy of chemotherapeutic drugs in various cancers by overcoming the chemo resistance and radio resistance shown by cancer cells. It has been widely used in preclinical trials in animal models, exhibiting very negligible side effects against normal cells and effective against cancer cells. The pharmacokinetic and pharmacodynamics-based collaborations of evodiamine are also included. Due to its poor bioavailability, synthetic analogs of evodiamine and its nano capsule have been formulated to enhance its bioavailability and reduce toxicity. In addition, this review summarizes the ongoing research on the mechanisms behind the antitumor potential of evodiamine, which proposes an exciting future for such interests in cancer biology.
Collapse
|
13
|
Jung YH, Lee DC, Kim JO, Kim JH. Untargeted metabolomics-assisted comparative cytochrome P450-dependent metabolism of fenbendazole in human and dog liver microsomes. Xenobiotica 2022; 52:986-996. [PMID: 36533905 DOI: 10.1080/00498254.2022.2160676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Fenbendazole (FBZ), a benzimidazole carbamate anthelmintic, has attracted attention for its antitumor activity. This study examined the metabolic characteristics of FBZ in humans compared with those in dogs. The phase I metabolites were identified in liver microsomal incubates using liquid chromatography-mass spectrometry (MS)-based untargeted metabolomics approaches. Seven metabolites of FBZ were identified by principal component analysis and orthogonal partial least square-discriminant analysis based on the global ion variables of the FBZ incubation groups. The chemical structure of the FBZ metabolites was suggested by examining the MS/MS spectrum and isotope distribution pattern. Cytochrome P450 (CYP) 1A1, CYP2D6, and CYP2J2 were the major isozymes responsible for the FBZ metabolism. No differences in the types of metabolites produced by the two species were noted. Multivariate analysis of human and dog incubation groups showed that five metabolites were relatively abundant in humans and the other two were not. In summary, the phase I metabolic profile of FBZ and the comparative metabolism between humans and dogs were examined using an untargeted metabolomics approach. This study suggests a successful investigation of FBZ metabolism in humans for conducting safety assessments regarding drug repositioning.
Collapse
Affiliation(s)
| | - Dong-Cheol Lee
- College of Pharmacy, Yeungnam University, Gyeongsan, Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Korea
| | - Ju-Hyun Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Korea
| |
Collapse
|
14
|
Li X, Yin D, Sun Y. Identification of the metabolite of ophiopogonanone A by liquid chromatography/quadrupole time-of-flight mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2022; 36:e9311. [PMID: 35557016 DOI: 10.1002/rcm.9311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 06/15/2023]
Abstract
RATIONALE Ophiopogonanone A (OPA) is one of the representative homoisoflavonoids isolated from Ophiopogonis Radix. The aim of this study was to identify and characterize the metabolites of OPA generated in the liver microsomes and hepatocytes of rats and humans. METHODS The metabolites were generated by incubating OPA (5 μM) with liver microsomes or hepatocytes at 37°C. To trap the reactive metabolites, glutathione (GSH, 5mM) was added into microsomal incubations. The metabolite identification and profiling were performed using ultra-high-performance liquid chromatography combined with photo-diode array detector and quadrupole time-of-flight tandem mass spectrometry (LC-Q/TOF-MS). The acquired mass data were processed by MetaboLynx software. The structures of the metabolites were tentatively characterized in terms of their accurate masses, product ions, and retention times. RESULTS Under the present conditions, a total of nine metabolites were detected and their structures were tentatively identified. Among these metabolites, M8 (OPA catechol) was the most abundant metabolite both in rat and human liver microsomes. M7 (glucuronidation product of M8) was the major metabolite both in rat and human hepatocytes. The metabolic pathways of OPA include demethylenation, dehydrogenation, hydroxylation, methylation and glucuronidation and GSH conjugation. CONCLUSION Our results provided valuable information regarding the in vitro metabolism of OPA, which would help us understand the mechanism of the elimination of OPA and in turn the effectiveness and potential toxicity.
Collapse
Affiliation(s)
- Xiao Li
- Department of Pharmacy, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Dengyang Yin
- Department of Clinical Pharmacy, Jingjiang People's Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu Province, China
| | - Ying Sun
- Department of Pharmacy, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| |
Collapse
|
15
|
Tu D, Ning J, Zou L, Wang P, Zhang Y, Tian X, Zhang F, Zheng J, Ge G. Unique Oxidative Metabolism of Bufalin Generates Two Reactive Metabolites That Strongly Inactivate Human Cytochrome P450 3A. J Med Chem 2022; 65:4018-4029. [DOI: 10.1021/acs.jmedchem.1c01875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Dongzhu Tu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Ning
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Liwei Zou
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yani Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiangge Tian
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550025, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
16
|
Wu L, Xie CL, Yang XW, Chen G. Pharmacokinetics and Metabolism Study of Deep-Sea-Derived Butyrolactone I in Rats by UHPLC-MS/MS and UHPLC-Q-TOF-MS. Mar Drugs 2021; 20:md20010011. [PMID: 35049869 PMCID: PMC8780701 DOI: 10.3390/md20010011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
Butyrolactone I (BTL-I) is a butanolide isolated from the deep-sea-derived fungus, Aspergillus sp. It provides a potential new target for the prevention and treatment of food allergies. This study aimed to investigate the metabolic and pharmacokinetic profile of BTL-I in rats. The metabolic profiles were obtained by UHPLC–Q-TOF-MS. As a result, eleven metabolites were structurally identified, and the proposed metabolic pathways of BTL-I were characterized. The main metabolites were the oxidative and glucuronidative metabolites. In addition, a sensitive UHPLC–MS/MS method was established for the quantitation of BTL-I in rat plasma (LOQ = 2 ng/mL). The method was fully validated and successfully applied to the pharmacokinetic study of BTL-I in rats after oral administration or intravenous administration. The oral bioavailability was calculated as 6.29%, and the maximum plasma concentrations were 9.85 ± 1.54 ng/mL and 17.97 ± 1.36 ng/mL for intravenous and intragastric dosing groups, respectively.
Collapse
Affiliation(s)
- Liang Wu
- Department of Pharmaceutical Analysis, School of Pharmacy, Fudan University, Shanghai 201203, China;
| | - Chun-Lan Xie
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China;
| | - Xian-Wen Yang
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China;
- Correspondence: (X.-W.Y.); (G.C.); Tel.: +86-592-219-5319 (X.-W.Y.); +86-21-51980168 (G.C.)
| | - Gang Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fudan University, Shanghai 201203, China;
- Correspondence: (X.-W.Y.); (G.C.); Tel.: +86-592-219-5319 (X.-W.Y.); +86-21-51980168 (G.C.)
| |
Collapse
|
17
|
Zhang KX, Dong N, Guo L. Characterization of the metabolites of dichotomitin in rat, monkey and human by ultra-high-performance liquid chromatography/high-resolution mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e9182. [PMID: 34418186 DOI: 10.1002/rcm.9182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 06/13/2023]
Abstract
RATIONALE Dichotomitin is one of the bioactive constituents isolated from Belamcanda chinensis. The goal of this study was to identify the metabolites of dichotomitin produced by liver microsomes and hepatocytes. METHODS Using ultra-high-performance liquid chromatography and high-resolution mass spectrometry (UPLC/HRMS), the metabolites were profiled and identified. The exact masses, elemental compositions and product ions of the metabolites were used to characterize their structures. RESULTS A total of ten metabolites were found and identified. The main metabolites identified in the incubation samples were M6 (3',5,6,7-tetrahydroxy-4',5'-dimethoxyisoflavone) and M8 (8-hydroxydichotomitin). Opening of 1,3-benzodioxole, demethylation, hydroxylation, glucuronidation and sulfation were among the metabolic modifications for dichotomitin. A human recombinant cytochrome P450 enzyme study revealed that CYP 1A2, 2C19, and 2D6 facilitated the formation of M6, whereas CYP 1A2 catalyzed the formation of M8 exclusively. CONCLUSIONS For the first time, data on the in vitro metabolic fates of dichotomitin were revealed in this work which would be helpful for us to understand the disposition of this bioactive constituent.
Collapse
Affiliation(s)
- Kai-Xuan Zhang
- Department of Clinical Laboratory, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Na Dong
- Department of Pharmacy, Hebei Women's Vocational College, Shijiazhuang, Hebei Province, China
| | - Le Guo
- Department of Pharmacy, Hebei Foreign Studies University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
18
|
Su G, Qin L, Su X, Tao C. Pyrotinib in vitro metabolite profiling via rat, dog and human hepatocytes using liquid chromatography-quadrupole/orbitrap mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e9195. [PMID: 34491599 DOI: 10.1002/rcm.9195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/04/2021] [Accepted: 09/05/2021] [Indexed: 02/05/2023]
Abstract
RATIONALE Pyrotinib is an irreversible EGFR/HER2 inhibitor that has shown antitumor activity and tolerance in the treatment of breast cancer. Studies focused on its metabolic pathways and major metabolites are insufficient. In the evaluation of drug safety and therapeutic use, metabolite characterization is critical. The metabolism of pyrotinib in vitro was studied utilizing rat, dog and human hepatocytes in this study. METHODS Pyrotinib (10 μM) was incubated with hepatocytes in Williams' E medium. The metabolites were examined and profiled using ultrahigh-performance liquid chromatography coupled with quadrupole/orbitrap high-resolution mass spectrometry. The metabolite structures were deduced by comparing their precise molecular weights, fragment ions and retention times with those of the parent drug. RESULTS A total of 16 metabolites, including 6 novel ones, were discovered and structurally described under the present conditions. Oxidation, demethylation, dehydrogenation, O-dealkylation and glutathione (GSH) conjugation were all involved in the metabolism of pyrotinib in hepatocytes. The most predominant metabolic route was identified as GSH conjugation (M5). CONCLUSIONS This study generated valuable metabolite profiles of pyrotinib in several species, which will aid in the understanding of the drug's disposition in various species and in evaluating the contribution of metabolites to overall effectiveness and toxicity of pyrotinib.
Collapse
Affiliation(s)
- Guosheng Su
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,Department of Laboratory Medicine, People's Hospital of Guangxi-ASEAN Economic and Technological Development Zone, Nanning Tenth People's Hospital, Nanning, Guangxi Province, China
| | - Lihua Qin
- Graduate School Master Arts of Nursing, University of Perpetual Help System DALTA, Las Piñas City, Philippines.,Department of Internal Medicine, People's Hospital of Guangxi-ASEAN Economic and Technological Development Zone, Nanning Tenth People's Hospital, Nanning, Guangxi Province, China
| | - Xiaoye Su
- Graduate School Master Arts of Nursing, University of Perpetual Help System DALTA, Las Piñas City, Philippines
| | - Chuanmin Tao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
19
|
Li J, Zhou M, Lai X, Wang Y, Zou Y, Li K, Li W, Zheng J. Toxicokinetic and bioavailability studies on retrorsine in mice, and ketoconazole-induced alteration in toxicokinetic properties. Biomed Chromatogr 2021; 36:e5270. [PMID: 34727371 DOI: 10.1002/bmc.5270] [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: 07/27/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 11/11/2022]
Abstract
Retrorsine (RTS) is a toxic retronecine-type pyrrolizidine alkaloid, which is widely distributed. The purpose of this study was to develop a high-performance liquid chromatography-tandem mass spectrometric (LC-MS/MS) method for serum RTS determination in mice. Serum samples were deproteinated by acetonitrile, separated on a C18 -PFP column and delivered at 0.8 ml/min with an eluting system composed of water containing 0.1% (v/v) formic acid and acetonitrile containing 0.1% (v/v) formic acid as mobile phases. RTS and the internal standard S-hexylglutathione (H-GSH) were quantitatively monitored with precursor-to-product transitions of m/z 352.1 → 120.1 and m/z 392.2 → 246.3, respectively. The method showed excellent linearity over the concentration range 0.05-50 μg/ml, with correlation coefficient r2 = 0.9992. The extraction recovery was >86.34%, and the matrix effect was not significant. Inter- and intra-day precisions (RSD) were <4.99%. The validated LC-MS/MS method was successfully applied to study the toxicokinetic profiles of serum RTS in mice after intravenous, oral administration and co-treated with ketoconazole, which showed that RTS displayed a long half-life (~11.05 h) and good bioavailability (81.80%). Co-administration of ketoconazole (KTZ) increased the peak serum concentration and area under the concentration-time curve and decreased the clearance and mean residence time. Summing up, a new standardized method was established for quantitative determination of RTS in sera.
Collapse
Affiliation(s)
- Jing Li
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Mengyue Zhou
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Xiaoqiong Lai
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Yang Wang
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Ying Zou
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Kunna Li
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Weiwei Li
- National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Jiang Zheng
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China.,Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China
| |
Collapse
|
20
|
Jiao W, Qin N, Wang K, Wu D, Yu H, Du L, Wu G, Wu H, Zhao X. LC-MS/MS for determination of aesculetin in rat plasma and its application to a pharmacokinetic study. Biomed Chromatogr 2021; 36:e5233. [PMID: 34519055 DOI: 10.1002/bmc.5233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 01/11/2023]
Abstract
Aesculetin, a coumarin compound present in the sancho tree and chicory, exhibits excellent antioxidant and anti-inflammatory activities in the vascular and immune system. In this study, a rapid and sensitive ultra-high performance liquid chromatography electrospray ionization-tandem mass spectrometry (UHPLC-ESI-MS/MS) method was established and validated for the determination of aesculetin in rat plasma. Plasma samples were prepared by protein precipitation with acetonitrile. Chromatographic separation was performed on an Acquity UPLC HSS T3 C18 column (2.1 × 100 mm, 1.8 μm) with gradient elution at a flow rate of 0.3 ml/min, using mobile phase consisting of 0.1% formic acid (A) and acetonitrile (B). Aesculetin and puerarin (internal standard) were detected by multiple reaction monitoring in negative ion mode. The method was fully validated according to the US Food and Drug Administration guidelines. The calibration curve was linear over the range of 2-1,000 ng/ml with correlation coefficient >0.9980. The carry-over, matrix effect, extraction recovery, dilution effect, intra- and inter-day precision and the accuracy were within acceptable limits. The method was then applied to a pharmacokinetic study of aesculetin in rats. After oral administration at doses of 5, 10 and 20 mg/kg, the plasma concentration reached peaks of 95.7, 219.9, 388.6 ng/ml at times of 1.22-1.78 h. The oral bioavailability was calculated as 15.6-20.3% in rat plasma. The result provided pre-clinical information for further application of aesculetin.
Collapse
Affiliation(s)
- Weijie Jiao
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine Zhengzhou, Henan, China
| | - Nan Qin
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Kun Wang
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine Zhengzhou, Henan, China
| | - Dongmei Wu
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine Zhengzhou, Henan, China
| | - Hongyan Yu
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine Zhengzhou, Henan, China
| | - Lei Du
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine Zhengzhou, Henan, China
| | - Guiyue Wu
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine Zhengzhou, Henan, China
| | - Hong Wu
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xu Zhao
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine Zhengzhou, Henan, China
| |
Collapse
|
21
|
Metabolite identification of iridin in rats by using UHPLC-MS/MS and pharmacokinetic study of its metabolite irigenin. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1181:122914. [PMID: 34492510 DOI: 10.1016/j.jchromb.2021.122914] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 11/22/2022]
Abstract
Iridin, one of the main bioactive components isolated from Belamcanda chinensis (L.) DC, exerts various pharmacological activities, such as anti-inflammation, antioxidant, and antitumor. However, the metabolism and pharmacokinetics of iridin are still unknown. After 100 mg/kg administration of iridin orally, the plasma, urine, and fecal bio-samples from Sprague-Dawley (SD) rats were collected and detected by ultrahigh-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). The pharmacokinetics of the major metabolite irigenin (aglycon of iridin) and a total of thirteen metabolites of iridin were identified, including five metabolites in plasma, ten metabolites in urine, and six metabolites in feces. The most principal metabolic pathway of iridin was glucuronidation after demethylation and was mediated by UDP-glucuronosyltransferases (UGTs) 1A7, 1A8, 1A9 and 1A10. This study highlights the first-time investigation of the metabolism of iridin in vivo, and the pharmacokinetics of irigenin (the major metabolite of iridin) in rats. These results provide robust evidence for further research and clinical application of iridin.
Collapse
|
22
|
In Vitro Metabolism of Donepezil in Liver Microsomes Using Non-Targeted Metabolomics. Pharmaceutics 2021; 13:pharmaceutics13070936. [PMID: 34201744 PMCID: PMC8309179 DOI: 10.3390/pharmaceutics13070936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 11/17/2022] Open
Abstract
Donepezil is a reversible acetylcholinesterase inhibitor that is currently the most commonly prescribed drug for the treatment of Alzheimer’s disease. In general, donepezil is known as a safe and well-tolerated drug, and it was not associated with liver abnormalities in several clinical trials. However, rare cases of drug-related liver toxicity have been reported since it has become commercially available. Few studies have investigated the metabolic profile of donepezil, and the mechanism of liver damage caused by donepezil has not been elucidated. In this study, the in vitro metabolism of donepezil was investigated using liquid chromatography–tandem mass spectrometry based on a non-targeted metabolomics approach. To identify metabolites, the data were subjected to multivariate data analysis and molecular networking. A total of 21 donepezil metabolites (17 in human liver microsomes, 21 in mice liver microsomes, and 17 in rat liver microsomes) were detected including 14 newly identified metabolites. One potential reactive metabolite was identified in rat liver microsomal incubation samples. Metabolites were formed through four major metabolic pathways: (1) O-demethylation, (2) hydroxylation, (3) N-oxidation, and (4) N-debenzylation. This study indicates that a non-targeted metabolomics approach combined with molecular networking is a reliable tool to identify and detect unknown drug metabolites.
Collapse
|
23
|
Yadav J, El Hassani M, Sodhi J, Lauschke VM, Hartman JH, Russell LE. Recent developments in in vitro and in vivo models for improved translation of preclinical pharmacokinetics and pharmacodynamics data. Drug Metab Rev 2021; 53:207-233. [PMID: 33989099 DOI: 10.1080/03602532.2021.1922435] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Improved pharmacokinetics/pharmacodynamics (PK/PD) prediction in the early stages of drug development is essential to inform lead optimization strategies and reduce attrition rates. Recently, there have been significant advancements in the development of new in vitro and in vivo strategies to better characterize pharmacokinetic properties and efficacy of drug leads. Herein, we review advances in experimental and mathematical models for clearance predictions, advancements in developing novel tools to capture slowly metabolized drugs, in vivo model developments to capture human etiology for supporting drug development, limitations and gaps in these efforts, and a perspective on the future in the field.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Inc., Boston, MA, USA
| | | | - Jasleen Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jessica H Hartman
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
24
|
Advancements in practical and scientific bioanalytical approaches to metabolism studies in drug development. Bioanalysis 2021; 13:913-930. [PMID: 33961500 DOI: 10.4155/bio-2021-0050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Advancement in metabolism profiling approaches and bioanalytical techniques has been revolutionized over the last two decades. Different in vitro and in vivo approaches along with advanced bioanalytical techniques are enabling the accurate qualitative and quantitative analysis of metabolites. This review summarizes various modern in vitro and in vivo approaches for executing metabolism studies with special emphasis on the recent advancement in the field. Advanced bioanalytical techniques, which can be employed in metabolism studies, have been discussed suggesting their particular application based on specific study objectives. This article can efficiently guide the researchers to scientifically plan metabolism studies and their bioanalysis during drug development programs taking advantage of a detailed understanding of instances of failure in the past.
Collapse
|
25
|
Zhang X, Xu M, Wu Z, Liu G, Tang Y, Li W. Assessment of CYP2C9 Structural Models for Site of Metabolism Prediction. ChemMedChem 2021; 16:1754-1763. [PMID: 33600055 DOI: 10.1002/cmdc.202000964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/07/2021] [Indexed: 11/07/2022]
Abstract
Structure-based prediction of a compound's potential sites of metabolism (SOMs) mediated by cytochromes P450 (CYPs) is highly advantageous in the early stage of drug discovery. However, the accuracy of the SOMs prediction can be influenced by several factors. CYP2C9 is one of the major drug-metabolizing enzymes in humans and is responsible for the metabolism of ∼13 % of clinically used drugs. In this study, we systematically evaluated the effects of protein crystal structure models, scoring functions, heme forms, conserved active-site water molecules, and protein flexibility on SOMs prediction of CYP2C9 substrates. Our results demonstrated that, on average, ChemScore and GlideScore outperformed four other scoring functions: Vina, GoldScore, ChemPLP, and ASP. The performance of the crystal structure models with pentacoordinated heme was generally superior to that of the hexacoordinated iron-oxo heme (referred to as Compound I) models. Inclusion of the conserved active-site water molecule improved the prediction accuracy of GlideScore, but reduced the accuracy of ChemScore. In addition, the effect of the conserved water on SOMs prediction was found to be dependent on the receptor model and the substrate. We further found that one of snapshots from molecular dynamics simulations on the apo form can improve the prediction accuracy when compared to the crystal structural model.
Collapse
Affiliation(s)
- Xiaoxiao Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Minjie Xu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| |
Collapse
|
26
|
Dong J, Li S, Liu G. Binimetinib Is a Potent Reversible and Time-Dependent Inhibitor of Cytochrome P450 1A2. Chem Res Toxicol 2021; 34:1169-1174. [PMID: 33728909 DOI: 10.1021/acs.chemrestox.1c00036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Binimetinib is a selective MEK1/2 inhibitor, which is indicative of melanoma. We aimed to investigate the inhibitory effect of binimetinib on cytochrome P450 using human liver microsomes. Binimetinib was demonstrated to display reversible and time-dependent inhibitory effects on human CYP1A2. Binimetinib can inhibit the activity of phenacetin deethylation with IC50 of 5.6 μM. A 30 min preincubation of binimetinib with NADPH-supplemented human liver microsomes raised a significant left IC50 shift (6.5-fold), from 5.69-0.88 μM. The inactivation parameters Kinact and KI were 0.063 min-1 and 15.47 μM, and the half-life of inactivation was 11 min. Glutathione (GSH) and catalase/superoxide exhibited minor or no protective effect on binimetinib-induced enzyme inactivation. Trapping experiment by GSH induced a detection of GSH adduct, of which the formation was believed to be through the oxidation of electron-rich 1,4-benzenediamine to reactive 1,4-diiminoquinone species. Cytochrome P450 3A4, 2C9, and 2D6 were involved in the bioactivation of binimetinib. In conclusion, binimetinib was proven to display reversible and time-dependent inhibitory effect on CYP1A2, which may have implications for the toxicity of binimetinib.
Collapse
Affiliation(s)
- Jiangnan Dong
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Su Li
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Guangxuan Liu
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| |
Collapse
|
27
|
Ji QG, Ma MH, Hu XM, Zhang YJ, Xu XH, Nian H. Detection and structural characterization of the metabolites of dihydroresveratrol in rats by liquid chromatography coupled to high-resolution tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e8991. [PMID: 33125777 DOI: 10.1002/rcm.8991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/12/2020] [Accepted: 10/21/2020] [Indexed: 06/11/2023]
Abstract
RATIONALE Dihydroresveratrol has been demonstrated to possess a wide spectrum of bioactivities, such as anti-oxidant and anti-inflammatory effects. The aim of the present study was to investigate the metabolic profiles of dihydroresveratrol in rats. METHODS The in vitro metabolism was elucidated by incubating dihydroresveratrol with rat hepatocytes for 2 h at 37°C. For in vivo metabolism, dihydroresveratrol was orally administered to rats at a single dose of 50 mg/kg and the resulting biliary and urinary samples were collected. All the samples were analyzed by liquid chromatography combined with electrospray ionization high-resolution mass spectrometry. The structures of the metabolites were proposed based on their accurate masses and their MS/MS product ions. RESULTS A total of 16 metabolites including three phase I metabolites and 13 phase II metabolites were detected and structurally proposed. Among these metabolites, M6 and M14 were unambiguously identified as 3'-hydroxylresveratrol and resveratrol, respectively, using reference standards. Dihydroresveratrol was mainly metabolized into resveratrol (M14) and a glucuronide conjugate (M12), which were excreted into urine and bile as the major metabolites. CONCLUSIONS The metabolic pathways of dihydroresveratrol involved hydroxylation, dehydrogenation, glucuronidation, glutathione (GSH) conjugation and methylation. The present study provided useful information with regard to the metabolic profiles of dihydroresveratrol in rats.
Collapse
Affiliation(s)
- Qiang-Guo Ji
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Ming-Hua Ma
- Department of Pharmacy, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China
| | - Xue-Mei Hu
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yi-Jun Zhang
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiao-Hong Xu
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Hua Nian
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| |
Collapse
|
28
|
Chen X, Li Y. Identification of the stable and reactive metabolites of tetrahydropiperine using ultrahigh-performance liquid chromatography combined with diode-array detection and high-resolution mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e8975. [PMID: 33049799 DOI: 10.1002/rcm.8975] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/22/2020] [Accepted: 10/08/2020] [Indexed: 06/11/2023]
Abstract
RATIONALE Tetrahydropiperine is one of the natural arylpentanamide compounds isolated from Piper nigrum L., which has been demonstrated to have insecticidal activity. The aim of this study was to investigate the metabolic profiles of tetrahydropiperine in mouse, rat, dog, monkey and human hepatocytes. METHODS The in vitro metabolism of tetrahydropiperine was elucidated via incubation with hepatocytes for 2 h at 37°C. The samples were analyzed using ultrahigh-performance liquid chromatography combined with diode-array detection and high-resolution tandem mass spectrometry operated in positive electrospray ionization mode. The structures of the metabolites were characterized using their retention times and their tandem mass spectrometric product ions. RESULTS A total of 20 metabolites were detected and their structures were proposed. These metabolites were formed mainly through the following pathways: (1) 1,3-benzodioxole ring opening to form a catechol derivative (M12), which was prone to glucuronidation (M6 and M8), methylation (M17) and glutathione (GSH)-derived conjugation through an ortho-quinone intermediate (M4) or via an aldehyde intermediate (M7); (2) dehydrogenation to form a piperanine (M15), which was subsequently subject to hydroxylation (M2 and M5) and GSH conjugation (M10 and M11) via Michael addition; (3) hydroxylation (M13, M14, M16, M18 and M19); and (4) direct GSH conjugation through an aldehyde intermediate (M3). CONCLUSIONS The major metabolic pathways of tetrahydropiperine were hydroxylation, dehydrogenation, methylation, GSH conjugation and glucuronidation. Tetrahydropiperine was bioactivated through ortho-quinone, Michael receptor and aldehyde intermediates.
Collapse
Affiliation(s)
- Xiaoling Chen
- Department of Laboratory, Jinmen First People's Hospital, 67 Xiangshan Avenue, Jinmen, Hubei Province, 448000, China
| | - Yanghua Li
- Department of Pharmacy, Jinmen First People's Hospital, 67 Xiangshan Avenue, Jinmen, Hubei Province, 448000, China
| |
Collapse
|
29
|
Dan Z, Min H, Chengbo X, Chengjuan C, Jiangong S, Tiantai Z, Pengmei L. Pharmacokinetic Characterization of ZT55, A Novel Indole Derivative Isolated from Radix Isatidis, using Liquid Chromatography/Tandem Mass and Q-TOF/Tandem Mass Spectrometry. CURR PHARM ANAL 2021. [DOI: 10.2174/1573412915666191007090906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
ZT55 is a novel natural product isolated from Radix isatidis. It is a highlyselective
tyrosine kinase inhibitor against myeloproliferative neoplasms. Although earlier research has
described the pharmacodynamic properties of ZT55 in vivo and in vitro, the quantitative determination
and pharmacokinetic profile in vivo have not been thoroughly studied.
Methods:
A novel liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed
and validated for the quantification of ZT55 in rat plasma. A Waters symmetry C18 column was
used for chromatographic separation; 0.1% formic acid in acetonitrile and 0.1% formic aqueous solution
was used as the mobile phase. Detection was performed by Multiple Reaction Monitoring (MRM)
mode using electrospray ionization in the positive ion mode. UPLC-QTOF-MS was used for the identification
of metabolites.
Results:
The method was linear (R2=0.9988) over the concentration range of 1-2500 ng/mL. The lower
limit of quantification was 1 ng/mL. The intra-day and inter-day precision of ZT55 showed a relative
standard deviation within 8.47%, whereas the accuracy (RE) ranged from -4.84% to 4.45%. The recoveries
ranged from 92.89% to 97.21%. ZT55 reached the highest plasma concentration at 0.5h. The peak
concentrations with three dosages were 103.59±10.11, 185.23±29.56, and 355.98±28.86 ng/mL. The
AUC0-24 of three dosages were 874.70±72.33, 433.80±49.33, and 231.65±19.41 ng•h/ml respectively.
Five metabolites of ZT55 from plasma were confirmed. The main pathways of ZT55 in vivo were hydrolysis,
N-dealkylation, glycosylation, and sulfonation.
Conclusions:
LC-MS/MS method was successfully applied to the pharmacokinetic study of ZT55 after
oral administration and intravenous. ZT55 exhibited rapid oral absorption, high elimination, and low
absolute bioavailability. This study provides important pharmacokinetic and metabolism information
for further pharmacological and toxicological research on ZT55.
Collapse
Affiliation(s)
- Zhang Dan
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Hu Min
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xu Chengbo
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Chen Chengjuan
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shi Jiangong
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zhang Tiantai
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Li Pengmei
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
30
|
Identification of the New In Vivo Metabolites of Ilaprazole in Rat Plasma after Oral Administration by LC-MS: In Silico Prediction of the H +/K +-ATPase Inhibitor. Molecules 2021; 26:molecules26020459. [PMID: 33467211 PMCID: PMC7829900 DOI: 10.3390/molecules26020459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 11/22/2022] Open
Abstract
Ilaprazole is a proton pump inhibitor used to treat digestive diseases. In this study, blood samples were collected after oral administration of ilaprazole and prepared by liquid–liquid extraction. The metabolites of ilaprazole were detected by liquid chromatography–high-resolution mass spectrometry (LC-HRMS) and LC-MSn. A total of twelve in vivo metabolites were detected in rat plasma and six new metabolites of ilaprazole, including one reductive metabolite with sulfide (M3), two hydroxylated metabolites with sulfoxide (M7 and M8), and three oxidative metabolites with sulfone (M9, M11, and M12), were identified. The possible metabolic pathways of ilaprazole and the fragmentation behaviors of its metabolites were elucidated. The result of the in silico prediction indicates that all the new metabolites showed the potential ability to inhibit H+/K+-ATPase activity.
Collapse
|
31
|
Ju Z, Tang X, Liao Q, Guan H, Yang L, Wang Z. Pharmacokinetic, bioavailability, and metabolism studies of lusianthridin, a dihydrophenanthrene compound, in rats by liquid chromatography/electrospray ionization tandem mass spectrometry. J Pharm Biomed Anal 2020; 195:113836. [PMID: 33358433 DOI: 10.1016/j.jpba.2020.113836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 11/26/2022]
Abstract
Lusianthridin was reported to possess many biological properties such as anti-oxidant and anti-cancer activities. However, its metabolic profiles and pharmacokinetics in vivo remain unknown. This study was carried out to investigate the metabolic profiles and pharmacokinetics of lusianthridin in rats. The metabolic profiles were obtained by an ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF-MS). A total of eighteen metabolites involved three phase I metabolites and fifteen phase II metabolites were detected and identified. The major metabolic pathways of lusianthridin were demethylation, oxidation, sulfation, glucuronidation and glutathione conjugation. In addition, a simple and sensitive ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method was established for determination of lusianthridin in rat plasma. After extracted by protein precipitation, lusianthridin was quantitated in positive ion mode. The method was linear over the range of 0.5-500 ng/mL (r ≥ 0.995) with the LLOQ of 0.5 ng/mL. The intra- and inter- precision and accuracy, extraction recovery, matrix effect and stability were within the acceptable limits. The validated method was applied to the pre-clinical pharmacokinetic study of lusianthridin in rats. After oral administration, lusianthridin was quickly absorbed into plasma and reached the max concentration of 236.22 ng/mL at 22.00 min. The elimination half life of lusianthridin from plasma was approximately 83.05-104.47 min and the oral absolute bioavailability was calculated as 30.93 %.
Collapse
Affiliation(s)
- Zhengcai Ju
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaowen Tang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qi Liao
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huida Guan
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Yang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai R&D Centre for Standardization of Chinese Medicines, Shanghai, 201203, China.
| | - Zhengtao Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai R&D Centre for Standardization of Chinese Medicines, Shanghai, 201203, China.
| |
Collapse
|
32
|
Zhang H, Liu C, Wang M, Sui Y. Metabolic profiling of senkyunolide A and identification of its metabolites in hepatocytes by ultra-high-performance liquid chromatography combined with diode-array detector and high-resolution mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34:e8894. [PMID: 32663340 DOI: 10.1002/rcm.8894] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 06/11/2023]
Abstract
RATIONALE Senkyunolide A is one of the bioactive constituents originally isolated from Ligusticum chuanxiong Hort. To better understand the action of this constituent, it is necessary to study the metabolic profiles in different species. METHODS For the metabolic stability study, senkyunolide A at a concentration of 0.5 μM was individually incubated with hepatocytes of mouse, rat, dog, monkey and human at 37°C for 2 h. For metabolite profiling and identification, senkyunolide A (10 μM) was incubated with hepatocytes and the incubation samples were analyzed by ultra-high-performance liquid chromatography combined with diode-array detector and high-resolution mass spectrometry (UHPLC/DAD-HRMS). The identities of the metabolites were characterized by accurate masses, product ions and retention times. RESULTS Senkyunolide A was metabolically unstable in hepatocytes. The in vitro half-lives were 136.2, 60.6, 33.65, 55.96 and 138 min in mouse, rat, dog, monkey and human hepatocytes, respectively. Furthermore, a total of 14 metabolites were detected. M1 and M9 were the most abundant metabolites in all species. The metabolic pathways of senkyunolide A involved the following pathways: (1) hydroxylation to form 10- and 11-hydroxysenkyunolide A, which further underwent epoxidation followed by GSH conjugation; (2) epoxidation followed by epoxide hydrolysis or GSH conjugation; and (3) aromatization to form 3-butylphthalide followed by hydroxylation. CONCLUSIONS Hydroxylation, epoxidation, aromatization and GSH conjugation were the main metabolic pathways of senkyunolide A. This study provides an overview of the metabolic profiles of senkyunolide A, which is helpful for a better understanding of the action of this compound.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pharmacy, Maternal and Child Health Care Hospital of Zaozhuang, No.25 East Cultural Road, Shandong Province, Zaozhuang, 277100, China
| | - Chunjuan Liu
- Department of Pharmacy, Maternal and Child Health Care Hospital of Zaozhuang, No.25 East Cultural Road, Shandong Province, Zaozhuang, 277100, China
| | - Minghua Wang
- Department of Pharmacy, Maternal and Child Health Care Hospital of Zaozhuang, No.25 East Cultural Road, Shandong Province, Zaozhuang, 277100, China
| | - Yong Sui
- Department of Pharmacy, Maternal and Child Health Care Hospital of Zaozhuang, No.25 East Cultural Road, Shandong Province, Zaozhuang, 277100, China
| |
Collapse
|
33
|
Ju Z, Liao Q, Yang Y, Guan H, Ma C, Tang X, Yang L, Wang Z. Identification of lusianthridin metabolites in rat liver microsomes by liquid chromatography combined with electrospray ionization time-of-flight mass spectrometry. Biomed Chromatogr 2020; 35:e5001. [PMID: 33063881 DOI: 10.1002/bmc.5001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/01/2020] [Accepted: 10/06/2020] [Indexed: 11/07/2022]
Abstract
Lusianthridin, a bioactive component isolated from Dendrobium venustum, has been demonstrated to have many biological properties such as antioxidant and anticancer activities. However, the metabolic profiles remain unknown. This study was carried out to investigate the metabolic profiles of lusianthridin in liver microsomes. Lusianthridin was co-incubated with liver microsomes in the presence of nicotinamide adenine dinucleotide phosphate and UDP-glucuronic acid or glutathione at 37°C for 1 h. The incubation samples were analyzed by liquid chromatography combined with electrospray ionization high-resolution mass spectrometry. The data were acquired and processed. The structures of the metabolites were proposed by comparing their accurate mass and MS2 spectra with those of the parent compound. A total of 15 metabolites were detected in vitro, including two phase I and 13 phase II metabolites. The phase I metabolic pathways were oxidation, demethylation and dehydrogenation. The phase II metabolic pathways referred to glucuronidation and glutathione conjugation. The present study provides an overview pertaining to the metabolic profiles of lusianthridin in vitro, which is indispensable for understanding the efficacy and safety of lusianthridin, as well as the herbal medicine D. venustum.
Collapse
Affiliation(s)
- Zhengcai Ju
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Liao
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuangui Yang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huida Guan
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Ma
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaowen Tang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Yang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Centre for Standardization of Chinese Medicines, Shanghai, China
| | - Zhengtao Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Centre for Standardization of Chinese Medicines, Shanghai, China
| |
Collapse
|
34
|
Tao S, Li H, Liu J. Metabolic profiling of ligustilide and identification of the metabolite in rat and human hepatocytes by liquid chromatography combined with high-resolution mass spectrometry. J Sep Sci 2020; 43:4405-4413. [PMID: 33098237 DOI: 10.1002/jssc.202000951] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 01/03/2023]
Abstract
Ligustilide is one of the most abundant bioactive ingredients in Rhizoma Chuanxiong that has been widely prescribed for medicinal purposes in China. To better understand the disposition and action of ligustilide, it is necessary to investigate the metabolic profiles. The in vitro metabolism was elucidated through incubating ligustilide with human and rat hepatocytes at 37°C. The incubation samples were collected at predefined time points to determine the metabolic stability. Upon metabolite identification and profiling, the incubation samples were analyzed by ultra-high-performance liquid chromatography combined with diode array detector and high-resolution mass spectrometry. The structures of the metabolites were characterized based on their mass spectrometry spectra, tandem mass spectrometry spectra, and fragmentation patterns. Ligustilide showed fast metabolism with high intrinsic clearance both in rat and human hepatocyte incubations. The half-lives of ligustilide in rat and human hepatocyte incubations were 8.0 and 15.0 min, respectively. Most of the parent (>90%) was biotransformed into the metabolites. Among these metabolites, M1 (senkyunolide I) was the major metabolite both in rat and human hepatocytes with the percentage of 42 and 70%, respectively. The metabolic pathways of ligustilide included epoxidation, epoxide hydrolysis, aromatization, hydroxylation, and glutathionylation. This work provided an overview of the metabolic profiles of ligustilide, which would be helpful for us to understand the action of this compound.
Collapse
Affiliation(s)
- Simei Tao
- Department of Pharmacy, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China
| | - Huidi Li
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China
| | - Jie Liu
- Department of Pharmacy, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China
| |
Collapse
|
35
|
Computer-Aided Estimation of Biological Activity Profiles of Drug-Like Compounds Taking into Account Their Metabolism in Human Body. Int J Mol Sci 2020; 21:ijms21207492. [PMID: 33050610 PMCID: PMC7593915 DOI: 10.3390/ijms21207492] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/25/2022] Open
Abstract
Most pharmaceutical substances interact with several or even many molecular targets in the organism, determining the complex profiles of their biological activity. Moreover, due to biotransformation in the human body, they form one or several metabolites with different biological activity profiles. Therefore, the development and rational use of novel drugs requires the analysis of their biological activity profiles, taking into account metabolism in the human body. In silico methods are currently widely used for estimating new drug-like compounds' interactions with pharmacological targets and predicting their metabolic transformations. In this study, we consider the estimation of the biological activity profiles of organic compounds, taking into account the action of both the parent molecule and its metabolites in the human body. We used an external dataset that consists of 864 parent compounds with known metabolites. It is shown that the complex assessment of active pharmaceutical ingredients' interactions with the human organism increases the quality of computer-aided estimates. The toxic and adverse effects showed the most significant difference: reaching 0.16 for recall and 0.14 for precision.
Collapse
|
36
|
Zhang Z, Nong L, Chen M, Liu M, Cheng W. A validated LC–MS/MS method for the quantification of capivasertib in dog plasma: Application to its pharmacokinetics study. Biomed Chromatogr 2020; 34:e4920. [DOI: 10.1002/bmc.4920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 11/10/2022]
Affiliation(s)
- Zhe Zhang
- Department of Integrated Therapy Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Li Nong
- Cancer Center, Union Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Meng‐Lei Chen
- Department of Integrated Therapy Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Ming‐Hui Liu
- Department of Integrated Therapy Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Wen‐Wu Cheng
- Department of Integrated Therapy Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| |
Collapse
|
37
|
Liu Y, Ma C, Jiao W, Yang Y, Zhang H, Du L, Ma M, Sun P, Li X, Chen J. Pharmacokinetics and bioavailability of ipatasertib in dog plasma using LC/MS/MS. Biomed Chromatogr 2020; 34:e4923. [PMID: 32558944 DOI: 10.1002/bmc.4923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/19/2020] [Accepted: 06/15/2020] [Indexed: 11/11/2022]
Abstract
A rapid, sensitive, and reliable liquid chromatography-tandem mass spectrometric method was developed to quantify ipatasertib in dog plasma. The dog plasma sample was deproteinated by using acetonitrile with ulixertinib as an internal standard followed by separation on a Spursil C18 -EP column with a gradient mobile phase comprising 2 mM ammonium acetate containing 0.1% formic acid and acetonitrile. Positive ion electrospray was used, and multiple reaction monitoring transitions were m/z 458.2 > 387.2 for ipatasertib and m/z 433.1 > 262.1 for the internal standard. The developed method was validated with a linear range of 0.3-1500 ng/mL, and with correlation coefficient greater than 0.9989. The lower limit of quantification was 0.3 ng/mL. The intra- and inter-day precision ranged from 3.58 to 14.32%, whereas the intra- and inter-day accuracy was in the range of -2.50-13.25%. No carry-over and matrix effects were observed under the current conditions. The extraction recovery was demonstrated to be greater than 85.43%. Ipatasertib was stable during the storage, processing, and determination. The validated assay was further successfully applied to a pharmacokinetic study of ipatasertib in dogs after oral and intravenous administrations. The bioavailability of ipatasertib was determined to be 19.3%.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Chunzheng Ma
- Department of Oncology, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Weijie Jiao
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yanhua Yang
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Huifang Zhang
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Lei Du
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ming Ma
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Pin Sun
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xiaokun Li
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Jianshe Chen
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
38
|
Agnew-Francis KA, Williams CM. Squaramides as Bioisosteres in Contemporary Drug Design. Chem Rev 2020; 120:11616-11650. [DOI: 10.1021/acs.chemrev.0c00416] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kylie A. Agnew-Francis
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Craig M. Williams
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
39
|
Li R, Ren M, Lu W, Yuan Y, Li J, Zhong W. A validated LC-MS/MS method for the determination of RAF inhibitor LXH254: Application to pharmacokinetic study in rat. Biomed Chromatogr 2020; 35:e4968. [PMID: 32881002 DOI: 10.1002/bmc.4968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 11/06/2022]
Abstract
In this study, a simple and sensitive UHPLC-ESI-MS/MS method was established for the determination of LXH254 in rat plasma. The developed method was validated according to the Food and Drug administration guidelines. After extraction using ethyl acetate, the sample was separated on an ACQUITY BEH C18 column. The mobile phase consisted of 2 mM ammonium acetate containing 0.1% formic acid and acetonitrile as the mobile phase with gradient elution. The flow rate was 0.3 mL/min. A TSQ triple quadrupole mass spectrometer operated in positive-ion mode was used for mass detection, with multiple reaction monitoring transitions of m/z 503.3 > 459.1 and m/z 435.3 > 367.1 for LXH254 and olaparib (internal standard), respectively. An excellent linearity was achieved in the concentration range of 0.1-1000 ng/mL, with correlation coefficient >0.998. The mean recovery was more than 78.55%. Inter- and intra-day precision (percentage of relative standard deviation) did not exceed 12.87%, and accuracy was in the range of -2.50 to 13.50%. LXH254 was demonstrated to be stable under the tested storage conditions. The validated UHPLC-MS/MS method was further applied to the pharmacokinetic study of LXH254 in rat plasma after oral (2, 5, and 15 mg/kg) and intravenous (2 mg/kg) administrations. The pharmacokinetic study revealed that LXH254 showed low clearance, moderate bioavailability (~30%), and linear pharmacokinetic profile over the oral dose range of 2-15 mg/kg. To the best of our knowledge, this is the first report on the method development and validation of the determination of LXH254 and its application to pharmacokinetic study.
Collapse
Affiliation(s)
- Rong Li
- Department of Pharmacy, Luzhou People's Hospital, Luzhou, China
| | - Meiping Ren
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wei Lu
- Department of Internal Medicine, Luzhou People's Hospital, Luzhou, China
| | - Yunhua Yuan
- Department of Neurology, Luzhou People's Hospital, Luzhou, China
| | - Jian Li
- Department of Urology, Luzhou People's Hospital, Luzhou, China
| | - Wu Zhong
- Department of Vascular Surgery, Luzhou People's Hospital, Luzhou, China
| |
Collapse
|
40
|
Wang J, Liu Y, Liu C, Shi Q. Characterization of the metabolites of gigantol in rat, dog, monkey, and human hepatocytes using ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34:e8810. [PMID: 32267985 DOI: 10.1002/rcm.8810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/16/2020] [Accepted: 04/03/2020] [Indexed: 06/11/2023]
Abstract
RATIONALE Gigantol (3',4-dihydroxy-3,5'-dimethoxybibenzyl) is a bibenzyl compound isolated from Dendrobii Caulis that has been widely used as a medicinal herb in China. To fully understand the mechanism of action of gigantol, it is necessary to determine its metabolic profile. METHODS Gigantol at a concentration of 20 μM was incubated with hepatocytes (rat, dog, monkey, and human) at 37°C. After 120 min incubation, the samples were analyzed using liquid chromatography coupled with electrospray ionization tandem mass spectrometry. The structures of the metabolites were characterized by their molecular masses, product ions, and retention times. RESULTS A total of 17 metabolites were detected and structurally identified. The metabolism involved the following pathways: (a) oxidation to form quinone-methide species and subsequently conjugation with glutathione (GSH); (b) demethylation to form demethylated gigantol, which was further conjugated with GSH; (c) hydroxylation to yield hydroxyl-gigantol followed by glucuronidation or GSH conjugation; and (d) glucuronidation to form glucuronide conjugates. Glucuronidation was the primary metabolic pathway in all tested species. CONCLUSIONS Hydroxylation, demethylation, glucuronidation, and GSH conjugation were the major metabolic pathways of gigantol. This study provides new information on the metabolic profiles of gigantol and helps us understand the disposition of the compound.
Collapse
Affiliation(s)
- Jingying Wang
- Department of Clinical Laboratory, The Third Hospital of Jilin University, Changchun, 130033, Jilin Province, China
| | - Yang Liu
- Department of Radiological, Second Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Chunyan Liu
- Department of Radiological, Second Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Qinghong Shi
- Department of Clinical Laboratory, The Third Hospital of Jilin University, Changchun, 130033, Jilin Province, China
| |
Collapse
|
41
|
Huang Y, Lin H, Chen Y, Huang X. Pharmacokinetic and bioavailability study of kurarinone in dog plasma by UHPLC-MS/MS. Biomed Chromatogr 2020; 34:e4945. [PMID: 32656774 DOI: 10.1002/bmc.4945] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/22/2020] [Accepted: 07/06/2020] [Indexed: 11/06/2022]
Abstract
Kurarinone, a natural prenylated flavonone isolated from Sophora flavescens, has been exhibited various activities. This study aimed to establish a simple and sensitive ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method for determining kurarinone in dog plasma. Acetonitrile-mediated precipitation was applied for sample pretreatment. Chromatographic separation was achieved on a Waters ACQUITY HSS T3 (100 × 2.1 mm, i. d., 1.8 μm) column with gradient elution using water containing 0.1% formic acid and acetonitrile as mobile phase. Quantitation was performed using an electrospray ionization source in negative multiple reaction monitoring mode. The linearity of this method was over the concentration range 0.1-500 ng/mL with the lowest limit of quantification (LLOQ) of 0.1 ng/mL. The intra- and inter-day precision was less than 10.51% and the accuracy ranged from 94.85% to 97.72%, respectively. The extraction recovery of kurarinone in dog plasma was more than 82.37% and no significant matrix effect was observed. The analyte was stable under tested storage conditions. The validated method was further successfully applied to a preclinical pharmacokinetic study of kurarinone in dog after a single intravenous (2 mg/kg) and oral (20 mg/kg) administration. The results revealed that kurarinone was rapidly absorbed into plasma with good bioavailability (38.19%) and low clearance.
Collapse
Affiliation(s)
- Yiqian Huang
- Department of Pharmacy, School of Medicine, The Fourth Affiliated Hospital of Zhejiang University, Yiwu, China
| | - Huashan Lin
- Department of Common Surgery, The First Affiliated Hospital, School of Medicine, Nanchang University, Nanchang, China
| | - Yaping Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiaosong Huang
- Department of Pharmacy, School of Medicine, The Fourth Affiliated Hospital of Zhejiang University, Yiwu, China
| |
Collapse
|
42
|
Shi Q, Liu Y, Liu C, Wang J. Characterization of the metabolites of TUG-891 in rat, dog, and human hepatocytes using ultra-high-performance liquid chromatography tandem mass spectrometry and nuclear magnetic resonance spectroscopy. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34:e8766. [PMID: 32108961 DOI: 10.1002/rcm.8766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 06/10/2023]
Abstract
RATIONALE TUG-891 is a potent and selective agonist of the long chain free fatty acid receptor 4. However, its metabolic profiles have not been revealed. The aim of this study was to investigate the in vitro metabolism of TUG-891 in hepatocytes. METHODS TUG-891 at a concentration of 20 μM was incubated with rat, dog, and human hepatocytes at 37°C for 120 min. The samples were analyzed using ultra-high-performance liquid chromatography combined with electrospray ionization tandem mass spectrometry. The structures of the metabolites were proposed according to their MS/MS product ions. Furthermore, M4 and M5 were biosynthesized using human liver microsomes, and their structures were characterized using 13 C-NMR spectroscopy. RESULTS Under the current conditions, eight metabolites were detected and structurally identified using high-resolution LC/MS and MS/MS spectra. The metabolites M4 and M5 were unambiguously confirmed to be TUG-891 alcohol and TUG-891 acid, respectively, using 13 C-NMR spectroscopy. Our results revealed that hydroxylation of methyl group at C-21 position to form TUG-891 alcohol (M5) followed by oxidation to yield TUG-891 aldehyde (M7) and carboxylic acid (M4) were the major metabolism processes. Phase II metabolism processes included glucuronidation and sulphation. CONCLUSIONS Hydroxylation at the C-21 position was the primary metabolic site of TUG-891. This study provided an overview of the metabolic profiles of TUG-891 in hepatocytes.
Collapse
Affiliation(s)
- Qinghong Shi
- Department of Clinical Laboratory, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yang Liu
- Department of Radiological, Second Hospital of Jilin University, Changchun, Jilin, China
| | - Chunyan Liu
- Department of Radiological, Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jingying Wang
- Department of Clinical Laboratory, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
43
|
Liu N, Wang X, Liu H, Zhang C. Identification of the metabolites of erianin in rat and human by liquid chromatography/electrospray ionization tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34:e8661. [PMID: 31732995 DOI: 10.1002/rcm.8661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/09/2019] [Accepted: 11/13/2019] [Indexed: 06/10/2023]
Abstract
RATIONALE Erianin, a bioactive component isolated from Dctidrobium chrysotoxum Lindl, was demonstrated to have many biological properties relevant to cancer prevention and therapy. However, the metabolic profiles of erianin remain unknown. This study was carried out to investigate the metabolic profiles of erianin in rats and humans. METHODS Erianin was orally administered to rats at a single dose of 50 mg/kg. Urine and bile samples were collected. For in vitro metabolism, erianin was co-incubated with rat or human hepatocytes at 37°C for 2 h. The samples from incubations and rat were analyzed by liquid chromatography combined with electrospray ionization high-resolution mass spectrometry. The data were processed by MetWorks software. The structures of the metabolites were proposed by comparing the mass spectra with that of the parent compound. RESULTS A total of twenty-four metabolites were detected in vitro and in vivo, including seven phase I and eighteen phase II metabolites. The phase I metabolic pathways of erianin were hydroxylation, demethylation and dehydrogenation. Erianin undergoes metabolic activation to form reactive metabolites quinoid intermediates, which were further trapped by glutathione (GSH) or N-acetylcysteine. The phase II metabolic pathways were glucuronidation, glutathione and N-acetylcysteine conjugation. CONCLUSIONS The present study provides an overview pertaining to the in vitro and in vivo metabolic profiles of erianin, which is indispensable for us to understand the efficacy and safety of erianin, as well as the herbal medicine D. chrysotoxum.
Collapse
Affiliation(s)
- Na Liu
- Department of Pharmacy, Jining No. 1 People's Hospital, Jining, 272011, Shandong Province, China
- Jining Medical University, Jining, 272011, Shandong Province, China
| | - Xige Wang
- Department of Pharmacy, Jining Cancer Hospital, Jining, 272007, Shandong Province, China
| | - Hongqiang Liu
- Department of Pharmacy, Jining No. 1 People's Hospital, Jining, 272011, Shandong Province, China
- Jining Medical University, Jining, 272011, Shandong Province, China
| | - Changpo Zhang
- Department of Pharmacy, Jining Hospital of TCM, Jining, 272037, Shandong Province, China
| |
Collapse
|
44
|
Yu B, Duan J, Ning H, Huang Y, Ling B, Lin F. Pharmacokinetics and metabolism of ulixertinib in rat by liquid chromatography combined with electrospray ionization tandem mass spectrometry. J Sep Sci 2020; 43:1275-1283. [PMID: 31970927 DOI: 10.1002/jssc.201901139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/14/2020] [Accepted: 01/19/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Bin Yu
- Department of PharmacyMianyang Central Hospital Mianyang P. R. China
| | - Jie Duan
- Department of Clinical PharmacyPidu District People's Hospital Chengdu P. R. China
| | - Hong Ning
- Department of PharmacyMianyang Central Hospital Mianyang P. R. China
| | - Yi‐Lan Huang
- Department of PharmacyThe Affiliated Hospital of Southwest Medical University Luzhou P. R. China
| | - Bao‐Dong Ling
- Sichuan Province College Key Laboratory of Structure‐Specific Small Molecule DrugsSchool of Pharmacy, Chengdu Medical College Chengdu P. R. China
| | - Fei Lin
- Department of PharmacyThe First Affiliated Hospital of Chengdu Medical College Chengdu P. R. China
| |
Collapse
|
45
|
Su J, Jia F, Lu J, Chen W, Sun H, Liu T, Wu X. Characterization of the metabolites of rosmarinic acid in human liver microsomes using liquid chromatography combined with electrospray ionization tandem mass spectrometry. Biomed Chromatogr 2020; 34:e4806. [PMID: 32012312 DOI: 10.1002/bmc.4806] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/22/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022]
Abstract
Rosmarinic acid (RA) is a phenolic acid originally isolated from the herb medicine Rosmarinus officinalis. The purpose of this study was to identify the metabolites of RA. RA was incubated with human liver microsomes in the presence of β-nicotinamide adenine dinucleotide phosphate tetrasodium salt and/or uridine diphosphate glucuronic acid using glutathione (GSH) as a trapping agent. After 60-min incubation, the samples were analyzed using high-resolution liquid chromatography tandem mass spectrometry. Under the current conditions, 14 metabolites were detected and identified. Our data revealed that RA was metabolized through the following pathways: the first pathway is the oxidation of catechol to form ortho-quinone intermediates, which react with GSH to form mono-GSH adducts (M1, M2, and M3) and bis-GSH adducts (M4 and M5); the second pathway is conjugation with glucuronide to yield acylglucuronide (M7), which further reacts with GSH to form RA-S-acyl-GSH adduct (M9); the third pathway is hydroxylation to form M10, M11, and M12, which further react with GSH to form mono-GSH adducts (M13 and M14); the fourth pathway is conjugation with GSH through Michael addition (M6); the fifth pathway is conjugation with glucuronidation, forming M8, which is the major metabolic pathway of RA.
Collapse
Affiliation(s)
- Jing Su
- Department of Gastroenterology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu Province, China
| | - Fangyuan Jia
- Department of Gastroenterology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu Province, China
| | - Junjie Lu
- Department of Neurology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu Province, China
| | - Weixu Chen
- Department of Gastroenterology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu Province, China
| | - Han Sun
- Department of Gastroenterology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu Province, China
| | - Tong Liu
- Department of Gastroenterology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu Province, China
| | - Xia Wu
- Department of Gastroenterology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu Province, China
| |
Collapse
|
46
|
Wang L, Wang M. In vitro metabolism of olaparib in liver microsomes by liquid chromatography/electrospray ionization high-resolution mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34:e8575. [PMID: 31499579 DOI: 10.1002/rcm.8575] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/31/2019] [Accepted: 09/02/2019] [Indexed: 06/10/2023]
Abstract
RATIONALES Olaparib is a Poly (ADP-ribose) Polymerase (PARP) inhibitor which has been developed as an anti-cancer agent. The purpose of this study was to characterize the metabolites of olaparib from liver microsomes and to reveal the interspecies differences between animals and humans. METHODS Olaparib (20 μM) was incubated with different species of liver microsomes at 37°C for 1 h in the presence of NADPH. The incubation samples were analyzed by liquid chromatography/electrospray ionization high-resolution mass spectrometry (LC/ESI-HRMS) operated in positive ion mode. The metabolites were characterized by accurate masses, MS2 spectra and retention times. RESULTS A total of 12 metabolites were detected and the structures of the metabolites were characterized based on their accurate masses, fragment ions and retention times. Four metabolites, i.e., M1, M10, M11 and M12, were unambiguously identified by using reference standards. The metabolic pathways of olaparib included hydroxylation, bis-hydroxylation, hydrolysis, dealkylation, dehydrogenation, and alcohol oxidation. CONCLUSIONS Compared with animal species, no human-specific metabolite was found in HLM. Dog also had a closer metabolic profile to humans. This study will be helpful for a better understanding of the species difference in pharmacokinetics/pharmacodynamics.
Collapse
Affiliation(s)
- Li Wang
- Department of Pharmacy, Jining No. 1 People's Hospital, Jining, 272011, Shandong Province, China
| | - Meixia Wang
- Department of Pharmacy, Affiliated Hospital of Jining Medical University, No. 89 Guhuai Road, Jining, 272000, Shandong Province, China
| |
Collapse
|
47
|
Sun Z, Yang J, Liu L, Xu Y, Zhou L, Jia Q, Shi Y, Du X, Kang J, Zuo L. Pharmacokinetics and Metabolite Profiling of Trepibutone in Rats Using Ultra-High Performance Liquid Chromatography Combined With Hybrid Quadrupole-Orbitrap and Triple Quadrupole Mass Spectrometers. Front Pharmacol 2019; 10:1266. [PMID: 31749700 PMCID: PMC6843799 DOI: 10.3389/fphar.2019.01266] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/30/2019] [Indexed: 12/24/2022] Open
Abstract
Trepibutone was widely used for cholelithiasis, cholecystitis, biliary tract dyskinesia, cholecystectomy syndrome, and chronic pancreatitis in clinic. However, few investigations on trepibutone have been conducted. In this study, an accurate, sensitive, and selective analytical method was developed and successfully applied to assess the pharmacokinetic behavior of trepibutone in rats. Trepibutone and carbamazepine (internal standard, IS) were quantified using multiple reaction monitoring (MRM) mode with the transitions of m/z 311.09→265.08 and m/z 237.06→194.08, respectively. The linearity, precision, accuracy, extraction recovery, matrix effect, and stability of the established method were all excellent within acceptable range. A total of 30 metabolites were identified in plasma and urine by Q-Exactive high resolution mass spectrometry, and several common metabolic pathways were observed such as dealkylation, oxidation, reduction, glucuronidation, and so on. This research provides more information on trepibutone in pharmacodynamics and toxicology and will assist the usage of trepibutone in clinical.
Collapse
Affiliation(s)
- Zhi Sun
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, China
| | - Jie Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, China
| | - Liwei Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, China
| | - Yanyan Xu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, China
| | - Lin Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, China
| | - Qingquan Jia
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yingying Shi
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, China
| | - Xiangyu Du
- Department of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Jian Kang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, China
| | - Lihua Zuo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, China
| |
Collapse
|
48
|
Xi W, Zou Q, Ouyang P. Pharmacokinetics of cligosiban in dog plasma after oral administration by liquid chromatography electrospray ionization tandem mass spectrometry. Biomed Chromatogr 2019; 33:e4611. [DOI: 10.1002/bmc.4611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/19/2019] [Accepted: 05/24/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Wang Xi
- School of Pharmaceutical SciencesNanjing Tech University Nanjing China
| | - Qiaogen Zou
- School of Pharmaceutical SciencesNanjing Tech University Nanjing China
| | - Pingkai Ouyang
- College of Biotechnology and Pharmaceutical EngineeringNanjing Tech University Nanjing China
| |
Collapse
|
49
|
He F, Zheng W, Chen Y, Mo C, Chen Y. Development and validation of a simple and sensitive high‐resolution LC/MS method for determination of PF‐04620110 in dog plasma: Application to a pharmacokinetic study. Biomed Chromatogr 2019; 33:e4562. [DOI: 10.1002/bmc.4562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/21/2019] [Accepted: 04/17/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Fan He
- Guangzhou Women and Children's Medical Center Guangzhou China
| | - Wei Zheng
- Guangzhou Women and Children's Medical Center Guangzhou China
| | - Yongzhuang Chen
- Guangzhou Women and Children's Medical Center Guangzhou China
| | - Chengke Mo
- Guangzhou Women and Children's Medical Center Guangzhou China
| | - Yilu Chen
- Guangzhou Women and Children's Medical Center Guangzhou China
| |
Collapse
|
50
|
Yang J, Yu H, Tang Q. Simultaneous determination of evobrutinib and its metabolite evobrutinib-diol in dog plasma by liquid chromatography combined with electrospray ionization tandem mass spectrometry. Biomed Chromatogr 2019; 33:e4575. [PMID: 31069837 DOI: 10.1002/bmc.4575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/18/2019] [Accepted: 05/03/2019] [Indexed: 11/06/2022]
Abstract
A rapid and sensitive liquid chromatography hyphenated with electrospray ionization tandem mass spectrometric method (LC-ESI-MS/MS) was developed and validated for simultaneous determination of evobrutinib and evobrutinib-diol in dog plasma. The plasma sample was processed using acetonitrile and chromatographic separation was carried out on a Waters Acquity BEH C18 column (50 × 2.1 mm, 1.7 μm). The mobile phase was composed of 0.1% formic acid and acetonitrile, with an optimized gradient elution at a flow rate of 0.4 mL/min. Detection was accomplished in selective reaction monitoring mode via electrospray ionization interface operated in positive ion mode. The precursor-to-product transitions for quantification were m/z 430.2 → 98.1 for evobrutinib, m/z 464.2 → 98.1 for evobrutinib-diol and m/z 441.2 → 138.1 for ibrutinib (internal standard). The developed assay was linear over the tested concentration ranges with correlation coefficient >0.995. The LLOQ was 0.1 ng/mL for both analytes. The inter- and intra-day precisions were <9.65% and the accuracy ranged from -3.94 to 6.37%. The extraction recovery was >85.41% and no significant matrix effect was observed. The developed assay was successfully applied to the pharmacokinetic study of evobrutinib and evobrutinib-diol in dogs after oral administration of evobrutinib at a single dose of 5 mg/kg.
Collapse
Affiliation(s)
- Jie Yang
- Department of Pharmacy, Shandong Provincial Jining No.1 People's Hospital, Jining, Shandong Province, China
| | - Haiyang Yu
- Department of Pharmacy, Shandong Provincial Jining No.1 People's Hospital, Jining, Shandong Province, China
| | - Qingmeng Tang
- Department of Pharmacy, Shandong Provincial Jining No.1 People's Hospital, Jining, Shandong Province, China
| |
Collapse
|