1
|
Lynn SG, Schultz IR, Matten SR, Patel PR, Watson SL, Yueh YL, Black SR, Wetmore BA. Cross-species comparisons of plasma binding and considerations for data evaluation. Toxicol In Vitro 2025; 106:106036. [PMID: 40023338 DOI: 10.1016/j.tiv.2025.106036] [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: 11/15/2024] [Revised: 02/12/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
The US Environmental Protection Agency is increasingly employing new approach methods (NAMs), including in vitro plasma binding and hepatocyte clearance experiments to collect chemical-species specific data. This paper presents data from plasma binding experiments using rapid equilibrium dialysis (RED) devices and plasma from humans, rats, and rainbow trout with a 4-h incubation time. A total of 54 chemicals, utilizing two concentrations, were tested across the three species resulting in 238 chemical-species specific datasets. Mass balance controls for chemical plasma stability and dialysis system recovery were used to evaluate the datasets and almost 40 % of the datasets (92/238 datasets) produced quantitative measurements. Cross-species comparisons and evaluations of the impact of physicochemical properties on chemical-assay performance were also evaluated. Comparisons of human-rat plasma binding revealed rat plasma generally demonstrated higher fup values for chemicals than human. While fup values in trout plasma were frequently lower than rat or human plasma. A comparison with literature data was performed and correlations between plasma binding, expressed as fraction unbound in plasma (fup), and log Kow across all three species indicate that the strongest relationship occurs at log Kow values between 1.5 and 4. The obtained datasets exhibited a wide range of behaviors, emphasizing the need for a robust approach to data quality assessment. The broader analysis of fup values indicates that chemicals with log Kow > 4.5 will be highly bound (fup ≤ 0.0001), difficult to measure, and have low reproducibility across laboratories, suggesting that use of different methods may be needed across different physicochemical properties.
Collapse
Affiliation(s)
- Scott G Lynn
- US Environmental Protection Agency, Office of Pesticide Programs, William Jefferson Clinton Building, 1200 Pennsylvania Avenue NW, Washington, DC 20460, USA.
| | - Irvin R Schultz
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, 2725 Montlake Boulevard East, Seattle, WA 98112, USA.
| | - Sharlene R Matten
- US Environmental Protection Agency, Office of Program Support, William Jefferson Clinton Building, 1200 Pennsylvania Avenue NW, Washington, DC 20460, USA.
| | - Purvi R Patel
- RTI International, Discovery Sciences, 3040 East Cornwallis Road, Research Triangle Park, NC 27709, USA.
| | - Scott L Watson
- RTI International, Discovery Sciences, 3040 East Cornwallis Road, Research Triangle Park, NC 27709, USA.
| | - Yun Lan Yueh
- RTI International, Discovery Sciences, 3040 East Cornwallis Road, Research Triangle Park, NC 27709, USA.
| | - Sherry R Black
- RTI International, Discovery Sciences, 3040 East Cornwallis Road, Research Triangle Park, NC 27709, USA.
| | - Barbara A Wetmore
- US Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, 109 T.W. Alexander Drive, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
2
|
Jung SM, Zhu HJ. Regulation of Human Hydrolases and Its Implications in Pharmacokinetics and Pharmacodynamics. Drug Metab Dispos 2024; 52:1139-1151. [PMID: 38777597 PMCID: PMC11495669 DOI: 10.1124/dmd.123.001609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Hydrolases represent an essential class of enzymes indispensable for the metabolism of various clinically essential medications. Individuals exhibit marked differences in the expression and activation of hydrolases, resulting in significant variability in the pharmacokinetics (PK) and pharmacodynamics (PD) of drugs metabolized by these enzymes. The regulation of hydrolase expression and activity involves both genetic polymorphisms and nongenetic factors. This review examines the current understanding of genetic and nongenetic regulators of six clinically significant hydrolases, including carboxylesterase (CES)-1 CES2, arylacetamide deacetylase (AADAC), paraoxonase (PON)-1 PON3, and cathepsin A (CTSA). We explore genetic variants linked to the expression and activity of the hydrolases and their effects on the PK and PD of their substrate drugs. Regarding nongenetic regulators, we focus on the inhibitors and inducers of these enzymes. Additionally, we examine the developmental expression patterns and gender differences in the hydrolases when pertinent information was available. Many genetic and nongenetic regulators were found to be associated with the expression and activity of the hydrolases and PK and PD. However, hydrolases remain generally understudied compared with other drug-metabolizing enzymes, such as cytochrome P450s. The clinical significance of genetic and nongenetic regulators has not yet been firmly established for the majority of hydrolases. Comprehending the mechanisms that underpin the regulation of these enzymes holds the potential to refine therapeutic regimens, thereby enhancing the efficacy and safety of drugs metabolized by the hydrolases. SIGNIFICANCE STATEMENT: Hydrolases play a crucial role in the metabolism of numerous clinically important medications. Genetic polymorphisms and nongenetic regulators can affect hydrolases' expression and activity, consequently influencing the exposure and clinical outcomes of hydrolase substrate drugs. A comprehensive understanding of hydrolase regulation can refine therapeutic regimens, ultimately enhancing the efficacy and safety of drugs metabolized by the enzymes.
Collapse
Affiliation(s)
- Sun Min Jung
- Departments of Pharmaceutical Sciences (S.M.J.) and Clinical Pharmacy (H.-J.Z.), University of Michigan, Ann Arbor, Michigan
| | - Hao-Jie Zhu
- Departments of Pharmaceutical Sciences (S.M.J.) and Clinical Pharmacy (H.-J.Z.), University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
3
|
El-Shorbagy HI, Mohamed MA, El-Gindy A, Hadad GM, Belal F. UPLC-PDA factorial design assisted method for simultaneous determination of oseltamivir, dexamethasone, and remdesivir in human plasma. Sci Rep 2024; 14:21758. [PMID: 39294224 PMCID: PMC11411088 DOI: 10.1038/s41598-024-71413-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 08/27/2024] [Indexed: 09/20/2024] Open
Abstract
A green and simple UPLC method was developed and optimized, adopting a factorial design for simultaneous determination of oseltamivir phosphate and remdesivir with dexamethasone as a co-administered drug in human plasma and using daclatasvir dihydrochloride as an internal standard within 5 min. The separation was established on UPLC column BEH C18 1.7 μm (2.1 × 100.0 mm) connected to UPLC pre-column BEH 1.7 μm (2.1 × 5.0 mm) at 50 °C with an injection volume of 10 μL. The photodiode array detector (PDA) was set at three wavelengths of 220, 315, and 245 nm for oseltamivir phosphate, the internal standard, and both dexamethasone and remdesivir, respectively. The mobile phase consisted of methanol and ammonium acetate solution (40 mM) adjusted to pH 4 in a ratio of 61.5:38.5 (v/v) with a flow rate of 0.25 mL min-1. The calibration curves were linear over 500.0-5000.0 ng mL-1 for oseltamivir phosphate, over 10.0-500.0 ng mL-1 and 500.0-5000.0 ng mL-1 for dexamethasone, and over 20.0-500 ng mL-1 and 500.0-5000.0 ng mL-1 for remdesivir. The Gibbs free energy and Van't Hoff plots were used to investigate the effect of column oven temperatures on retention times. Fluoride-EDTA anticoagulant showed inhibition activity on the esterase enzyme in plasma. The proposed method was validated according to the M10 ICH, FDA, and EMA's bioanalytical guidelines. According to Eco-score, GAPI, and AGREE criteria, the proposed method was considered acceptable green.
Collapse
Affiliation(s)
- Hanan I El-Shorbagy
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| | - Mona A Mohamed
- Pharmaceutical Chemistry Department, Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Alaa El-Gindy
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Ghada M Hadad
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Fathalla Belal
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
4
|
Zhang W, Oh JH, Zhang W, Aldrich CC, Sirianni RW, Elmquist WF. Pharmacokinetics of Panobinostat: Interspecies Difference in Metabolic Stability. J Pharmacol Exp Ther 2024; 389:96-105. [PMID: 38409112 PMCID: PMC10949161 DOI: 10.1124/jpet.123.002051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/13/2024] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
The deregulation of histone deacetylase (HDAC) expression is often seen in many cancers, and HDAC inhibitors have shown potency against a variety of cancer types. Panobinostat is a potent pan-HDAC inhibitor that has been tested in multiple studies for the treatment of brain tumors. There have been contrasting views surrounding its efficacy for the treatment of tumors in the central nervous system (CNS) following systemic administration when examined in different models or species. We conducted experiments using three different mouse strains or genotypes to have a more comprehensive understanding of the systemic as well as the CNS distributional kinetics of panobinostat. Our study found that panobinostat experienced rapid degradation in vitro in Friend leukemia virus strain B mouse matrices and a faster degradation rate was observed at 37°C compared with room temperature and 4°C, suggesting that the in vitro instability of panobinostat was due to enzymatic metabolism. Panobinostat also showed interstrain and interspecies differences in the in vitro plasma stability and was stable in human plasma. The objective of this study was to examine the in vitro metabolic stability of panobinostat in different matrices and assess the influence of that metabolic stability on the in vivo pharmacokinetics and CNS delivery of panobinostat. Importantly, the plasma stability in various mouse strains was not reflected in the in vivo systemic pharmacokinetic behavior of panobinostat. Several hypotheses arise from this finding, including: the binding of panobinostat to red blood cells, the existence of competing endogenous compounds to enzyme(s), the distribution into tissues with a lower level of enzymatic activity or the metabolism occurring in the plasma is a small fraction of the total metabolism in vivo. SIGNIFICANCE STATEMENT: Panobinostat showed different in vitro degradation in plasma from different mouse strains and genotypes. However, despite the differences surrounding in vitro plasma stability, panobinostat showed similar in vivo pharmacokinetic behavior in different mouse models. This suggests that the interstrain difference in enzymatic activity did not affect the in vivo pharmacokinetic behavior of panobinostat and its central nervous system distribution in mice. This lack of translation between in vitro metabolism assays and in vivo disposition can confound drug development.
Collapse
Affiliation(s)
- Wenqiu Zhang
- Departments of Pharmaceutics, Brain Barriers Research Center (Wenq. Zhang, J.-H.O., Wenj. Zhang, W.F.E.) and Medicinal Chemistry (C.C.A.), University of Minnesota, Minneapolis, Minnesota; and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Ju-Hee Oh
- Departments of Pharmaceutics, Brain Barriers Research Center (Wenq. Zhang, J.-H.O., Wenj. Zhang, W.F.E.) and Medicinal Chemistry (C.C.A.), University of Minnesota, Minneapolis, Minnesota; and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Wenjuan Zhang
- Departments of Pharmaceutics, Brain Barriers Research Center (Wenq. Zhang, J.-H.O., Wenj. Zhang, W.F.E.) and Medicinal Chemistry (C.C.A.), University of Minnesota, Minneapolis, Minnesota; and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Courtney C Aldrich
- Departments of Pharmaceutics, Brain Barriers Research Center (Wenq. Zhang, J.-H.O., Wenj. Zhang, W.F.E.) and Medicinal Chemistry (C.C.A.), University of Minnesota, Minneapolis, Minnesota; and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Rachael W Sirianni
- Departments of Pharmaceutics, Brain Barriers Research Center (Wenq. Zhang, J.-H.O., Wenj. Zhang, W.F.E.) and Medicinal Chemistry (C.C.A.), University of Minnesota, Minneapolis, Minnesota; and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - William F Elmquist
- Departments of Pharmaceutics, Brain Barriers Research Center (Wenq. Zhang, J.-H.O., Wenj. Zhang, W.F.E.) and Medicinal Chemistry (C.C.A.), University of Minnesota, Minneapolis, Minnesota; and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| |
Collapse
|
5
|
Tang X, Zhang Y, Zhang YY, Zhang Z, Tian Y. Development and validation of a sensitive LC-MS/MS method for simultaneous analysis of clopidogrel and simvastatin and their main metabolites in beagles: Application to pharmacokinetic drug interactions. Biomed Chromatogr 2023; 37:e5714. [PMID: 37574765 DOI: 10.1002/bmc.5714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/13/2023] [Accepted: 04/18/2023] [Indexed: 08/15/2023]
Abstract
Clopidogrel (CLP) and simvastatin (SV) are commonly used in combination therapies as anti-cardiovascular drugs. However, the effect of coadministration on the absorption and metabolism of the two drugs in vivo is not clear. This study developed and validated an LC-MS/MS method for the simultaneous determination of CLP, clopidogrel carboxylic acid (CLPCA), 2-oxo-clopidogrel (2-O-CLP), SV, and simvastatin hydroxy acid (SVA) in beagle plasma. Chromatographic separation was achieved on an InfinityLab Poroshell 120 SB-C8 column (2.1 × 100 mm, 2.7 μm) using methanol and 0.1% formic acid in water as the mobile phase at a flow rate of 0.3 mL/min in gradient mode. The lower limits of quantification are 0.1, 0.8, 0.05, 0.05, and 0.05 ng/mL for CLP, CLPCA, 2-O-CLP, SV, and SVA, respectively. The selectivity, linearity, accuracy, precision, extraction recovery, matrix effect, and stability were validated within acceptable criteria. This method was successfully applied to the pharmacokinetic drug interaction study between CLP and SV, and the results revealed that combined administration affected the metabolic rate of CLP, SV, and their metabolites. This study is the first to detect CLP, CLPCA, 2-O-CLP, SV, and SVA simultaneously.
Collapse
Affiliation(s)
- Xiao Tang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, China
- Key Laboratory of Drug Consistency Evaluation, China Pharmaceutical University, Nanjing, China
| | - Yu Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, China
- Key Laboratory of Drug Consistency Evaluation, China Pharmaceutical University, Nanjing, China
| | - Yue Yi Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, China
- Key Laboratory of Drug Consistency Evaluation, China Pharmaceutical University, Nanjing, China
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, China
- Key Laboratory of Drug Consistency Evaluation, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yuan Tian
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, China
- Key Laboratory of Drug Consistency Evaluation, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
6
|
Simvastatin: In Vitro Metabolic Profiling of a Potent Competitive HMG-CoA Reductase Inhibitor. SEPARATIONS 2022. [DOI: 10.3390/separations9120400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Simvastatin (SV) is a semisynthetic derivative of lovastatin (LV), which is biosynthetically produced from the fungus Aspergillus terreus and has a high log p value (log p = 4.39)and thus high hepatic extraction and high efficacy in controlling cholesterol synthesis. The current study was undertaken to investigate the metabolic profile of SV using various mass spectrometry (MS) platforms. Metabolic profiling was studied in in vitro models, rat liver microsomes (RLMs), and isolated perfused rat liver hepatocytes (RLHs) using both ion trap and triple quadruple LC–MS/MS systems. A total of 29 metabolites were identified. Among them, three types of SV-related phase-I metabolites, namely exomethylene simvastatin acid (exomethylene SVA), monohydroxy SVA, and dihydrodiol SVA, were identified as new in RLMs. No phase-II metabolites were identified while incubating with RLHs.
Collapse
|
7
|
Solé M, Figueres E, Mañanós E, Rojo-Solís C, García-Párraga D. Characterisation of plasmatic B-esterases in bottlenose dolphins (Tursiops truncatus) and their potential as biomarkers of xenobiotic chemical exposures. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 313:120149. [PMID: 36115493 DOI: 10.1016/j.envpol.2022.120149] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/23/2022] [Accepted: 09/06/2022] [Indexed: 06/15/2023]
Abstract
A total of 164 blood samples from 16 clinically healthy bottlenose dolphins (Tursiops truncatus), were obtained from an aquarium in Spain between 2019 and 2020, as part of their preventive medicine protocol. In addition to conventional haematological and biochemical analyses, plasmatic B-esterase activities were characterised to determine the potential application of such analyses in wild counterparts. The hydrolysis rates for the substrates of acetylcholinesterase (AChE), butyrylcholinesterase (BuChE) and carboxylesterase (CE) activity in plasma were measured, the last using two commercial substrates, p-nitrophenyl acetate (pNPA) and p-nitrophenyl butyrate (pNPB). Activity rates (mean ± SEM in nmol/min/mL plasma) were (in descending order): AChE (125.6 ± 3.8), pNPB-CE (65.0 ± 2.2), pNPA-CE (49.7 ± 1.1) and BuChE (12.8 ± 1.3). These values for dolphins are reported in here for the first time in this species. Additionally, the in vitro sensitivity of two B-esterases (AChE and pNPB-CE) to chemicals of environmental concern was determined, and the protective role of plasmatic albumin assessed. Out of the B-esterases measured in plasma of dolphin, AChE activity was more responsive in vitro to pesticides, while CEs had a low response to plastic additives, likely due to the protective presence of albumin. However, the clear in vitro interaction of these environmental chemicals with purified AChE from electric eels and recombinant human hCEs (hCE1 and hCE2) and albumin, predicts their impact in other tissues that require in vivo validation. A relationship between esterase-like activities and health parameters in terrestrial mammals has already been established. Thus, B-esterase measures could be easily included in marine mammal health assessment protocols for dolphins as well, once the relationship between these measures and the animal's fitness has been established.
Collapse
Affiliation(s)
- M Solé
- Institut de Ciències del Mar, CSIC, Psg. Marítim de La Barceloneta 37-49, 08003, Barcelona, Spain.
| | - E Figueres
- Institut de Ciències del Mar, CSIC, Psg. Marítim de La Barceloneta 37-49, 08003, Barcelona, Spain
| | - E Mañanós
- Institute of Aquaculture Torre La Sal (IATS),-CSIC, 12595, Cabanes, Castellón, Spain
| | - C Rojo-Solís
- Veterinary Services, Oceanogràfic, Ciudad de Las Artes y Las Ciencias, C/Eduardo Primo Yúfera (Científic) 1B, 46013, València, Spain
| | - D García-Párraga
- Veterinary Services, Oceanogràfic, Ciudad de Las Artes y Las Ciencias, C/Eduardo Primo Yúfera (Científic) 1B, 46013, València, Spain
| |
Collapse
|
8
|
Lamichhane S, Mohammed CJ, Haller ST, Kennedy DJ, Isailovic D. Quantification of Cardiotonic Steroids Potentially Regulated by Paraoxonase 3 in a Rat Model of Chronic Kidney Disease Using UHPLC-Orbitrap-MS. Int J Mol Sci 2022; 23:ijms232113565. [PMID: 36362352 PMCID: PMC9654389 DOI: 10.3390/ijms232113565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Endogenous cardiotonic steroids (CTSs), such as telocinobufagin (TCB) and marinobufagin (MBG) contain a lactone moiety critical to their binding and signaling through the Na+/K+-ATPase. Their concentrations elevate in response to sodium intake and under volume-expanded conditions. Paraoxonase 3 (PON3) is an enzyme that can hydrolyze lactone substrates. Here, we examine the role of PON3 in regulating CTS levels in a rat model of chronic kidney diseases (CKD). TCB and MBG were extracted from rat urine samples, and the analyses were carried out using ultra-high pressure liquid chromatography−Orbitrap-mass spectrometry (UHPLC-Orbitrap-MS). Ten-week-old Dahl salt-sensitive wild type (SS-WT) and Dahl salt-sensitive PON3 knockout (SS-PON3 KO) rats were maintained on a high-salt diet (8% NaCl) for 8 weeks to initiate salt-sensitive hypertensive renal disease characteristic of this model. CTS extraction recovery from urine >80% was achieved. For animals maintained on a normal chow diet, the baseline amount of TCB excreted in 24 h urine of SS-PON3 KO rats (6.08 ± 1.47 ng/24 h; or 15.09 ± 3.25 pmol) was significantly higher than for SS-WT rats (1.48 ± 0.69 ng/24 h; or 3.67 ± 1.54 pmol, p < 0.05). Similarly, for the same animals, the amount of excreted MBG was higher in the urine of SS-PON3 KO rats (4.74 ± 1.30 ng/24 h versus 1.03 ± 0.25 ng/24 h in SS-WT; or 11.83 ± 2.91 pmol versus 2.57 ± 0.56 pmol in SS-WT, p < 0.05). For animals on a high-salt diet, the SS-PON3 KO rats had significantly increased levels of TCB (714.52 ± 79.46 ng/24 h; or 1774.85 ± 175.55 pmol) compared to SS-WT control (343.84 ± 157.54 ng/24 h; or 854.09 ± 350.02 pmol, p < 0.05), and comparatively higher levels of MBG were measured for SS-PON3 KO (225.55 ± 82.61 ng/24 h; or 563.19 ± 184.5 pmol) versus SS-WT (157.56 ± 85.53 ng/24 h; or 393.43 ± 191.01 pmol, p > 0.05) rats. These findings suggest that the presence and absence of PON3 dramatically affect the level of endogenous CTSs, indicating its potential role in CTS regulation.
Collapse
Affiliation(s)
- Sabitri Lamichhane
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Chrysan J. Mohammed
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Steven T. Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - David J. Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
- Correspondence: (D.J.K.); (D.I.); Tel.: +1+419-383-6822 (D.J.K.); +1+419-530-5523 (D.I.)
| | - Dragan Isailovic
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
- Correspondence: (D.J.K.); (D.I.); Tel.: +1+419-383-6822 (D.J.K.); +1+419-530-5523 (D.I.)
| |
Collapse
|
9
|
Prieto Garcia L, Lundahl A, Ahlström C, Vildhede A, Lennernäs H, Sjögren E. Does the choice of applied physiologically‐based pharmacokinetics platform matter? A case study on simvastatin disposition and drug–drug interaction. CPT Pharmacometrics Syst Pharmacol 2022; 11:1194-1209. [PMID: 35722750 PMCID: PMC9469690 DOI: 10.1002/psp4.12837] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022] Open
Abstract
Physiologically‐based pharmacokinetic (PBPK) models have an important role in drug discovery/development and decision making in regulatory submissions. This is facilitated by predefined PBPK platforms with user‐friendly graphical interface, such as Simcyp and PK‐Sim. However, evaluations of platform differences and the potential implications for disposition‐related applications are still lacking. The aim of this study was to assess how PBPK model development, input parameters, and model output are affected by the selection of PBPK platform. This is exemplified via the establishment of simvastatin PBPK models (workflow, final models, and output) in PK‐Sim and Simcyp as representatives of established whole‐body PBPK platforms. The major finding was that the choice of PBPK platform influenced the model development strategy and the final model input parameters, however, the predictive performance of the simvastatin models was still comparable between the platforms. The main differences between the structure and implementation of Simcyp and PK‐Sim were found in the absorption and distribution models. Both platforms predicted equally well the observed simvastatin (lactone and acid) pharmacokinetics (20–80 mg), BCRP and OATP1B1 drug–gene interactions (DGIs), and drug–drug interactions (DDIs) when co‐administered with CYP3A4 and OATP1B1 inhibitors/inducers. This study illustrates that in‐depth knowledge of established PBPK platforms is needed to enable an assessment of the consequences of PBPK platform selection. Specifically, this work provides insights on software differences and potential implications when bridging PBPK knowledge between Simcyp and PK‐Sim users. Finally, it provides a simvastatin model implemented in both platforms for risk assessment of metabolism‐ and transporter‐mediated DGIs and DDIs.
Collapse
Affiliation(s)
- Luna Prieto Garcia
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and Development Uppsala University Uppsala Sweden
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Anna Lundahl
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Christine Ahlström
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Anna Vildhede
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Hans Lennernäs
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and Development Uppsala University Uppsala Sweden
| | - Erik Sjögren
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and Development Uppsala University Uppsala Sweden
| |
Collapse
|
10
|
Fei B, Cai X, Huang G. Status and strategies of college English teaching using adaptive deep learning from the perspective of multiculturalism. Front Psychol 2022; 13:910667. [PMID: 35967666 PMCID: PMC9366170 DOI: 10.3389/fpsyg.2022.910667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/07/2022] [Indexed: 11/26/2022] Open
Abstract
The aim is to deeply understand the current situation of College English Teaching (CET). By consulting the theories of Deep Learning (DL) and Edge Computing (EC), this work designs a Questionnaire Survey (QS) to understand the current situation of college English learning and teaching. Then, Adaptive Deep Learning (ADL) and EC are introduced into CET. Finally, the corresponding conclusions and suggestions are drawn. Specifically, the purpose and time of college students’ vocabulary learning are investigated. The results suggest that students’ English vocabulary learning is shallow. They have not really mastered the meaning and usage of vocabulary. Simultaneously, teachers’ vocabulary teaching methods are analyzed from three aspects: root affixes, vocabulary collocation, or repeated reading and memory. The teaching method is excellent from the teacher’s perspective but far from perfect from the students’ perspective. Introducing ADL and EC into CET enhances students’ class concentration time, homework submission efficiency, and academic performance. More than half of the students believe that the new teaching method introducing ADL and EC can make them more confident and motivated in English learning. Therefore, it has great reference significance for applying ADL and EC in CET.
Collapse
Affiliation(s)
- Bi Fei
- Institute of Education, Sichuan Normal University, Chengdu, China
- College of Foreign Languages and Cultures, Chengdu University of Technology, Chengdu, China
| | - Xia Cai
- Sports Institute, Chengdu University of Technology, Chengdu, China
- *Correspondence: Xia Cai,
| | - Guo Huang
- Sichuan Province University Key Laboratory of Internet Natural Language Intelligent Processing, Leshan Normal University, Leshan, China
| |
Collapse
|
11
|
A UHPLC-QTOF-MS/MS method with a superimposed multiple product ion strategy and esterase inhibitor improved sensitivity for the determination of xylocarpin H in rat plasma. J Pharm Biomed Anal 2022; 216:114803. [DOI: 10.1016/j.jpba.2022.114803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/11/2022] [Accepted: 04/23/2022] [Indexed: 11/23/2022]
|
12
|
Mohammed CJ, Lamichhane S, Connolly JA, Soehnlen SM, Khalaf FK, Malhotra D, Haller ST, Isailovic D, Kennedy DJ. A PON for All Seasons: Comparing Paraoxonase Enzyme Substrates, Activity and Action including the Role of PON3 in Health and Disease. Antioxidants (Basel) 2022; 11:antiox11030590. [PMID: 35326240 PMCID: PMC8945423 DOI: 10.3390/antiox11030590] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/17/2022] Open
Abstract
Paraoxonases (PONs) are a family of hydrolytic enzymes consisting of three members, PON1, PON2, and PON3, located on human chromosome 7. Identifying the physiological substrates of these enzymes is necessary for the elucidation of their biological roles and to establish their applications in the biomedical field. PON substrates are classified as organophosphates, aryl esters, and lactones based on their structure. While the established native physiological activity of PONs is its lactonase activity, the enzymes’ exact physiological substrates continue to be elucidated. All three PONs have antioxidant potential and play an important anti-atherosclerotic role in several diseases including cardiovascular diseases. PON3 is the last member of the family to be discovered and is also the least studied of the three genes. Unlike the other isoforms that have been reviewed extensively, there is a paucity of knowledge regarding PON3. Thus, the current review focuses on PON3 and summarizes the PON substrates, specific activities, kinetic parameters, and their association with cardiovascular as well as other diseases such as HIV and cancer.
Collapse
Affiliation(s)
- Chrysan J. Mohammed
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (C.J.M.); (J.A.C.); (S.M.S.); (F.K.K.); (D.M.); (S.T.H.)
| | - Sabitri Lamichhane
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA; (S.L.); (D.I.)
| | - Jacob A. Connolly
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (C.J.M.); (J.A.C.); (S.M.S.); (F.K.K.); (D.M.); (S.T.H.)
| | - Sophia M. Soehnlen
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (C.J.M.); (J.A.C.); (S.M.S.); (F.K.K.); (D.M.); (S.T.H.)
| | - Fatimah K. Khalaf
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (C.J.M.); (J.A.C.); (S.M.S.); (F.K.K.); (D.M.); (S.T.H.)
- Department of Clinical Pharmacy, College of Pharmacy, University of Alkafeel, Najaf 61001, Iraq
| | - Deepak Malhotra
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (C.J.M.); (J.A.C.); (S.M.S.); (F.K.K.); (D.M.); (S.T.H.)
| | - Steven T. Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (C.J.M.); (J.A.C.); (S.M.S.); (F.K.K.); (D.M.); (S.T.H.)
| | - Dragan Isailovic
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA; (S.L.); (D.I.)
| | - David J. Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (C.J.M.); (J.A.C.); (S.M.S.); (F.K.K.); (D.M.); (S.T.H.)
- Correspondence: ; Tel.: +1-419-383-6822
| |
Collapse
|
13
|
Kumar D, Gautam N, Alnouti Y. Analyte recovery in LC-MS/MS bioanalysis: An old issue revisited. Anal Chim Acta 2022; 1198:339512. [PMID: 35190119 PMCID: PMC8864627 DOI: 10.1016/j.aca.2022.339512] [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: 11/17/2021] [Revised: 01/04/2022] [Accepted: 01/15/2022] [Indexed: 11/24/2022]
Abstract
There are several challenges associated with LC-MS/MS bioanalytical method development and validation. Low and variable recovery of some analytes, especially the more hydrophobic ones, is often challenging. Analytes can be lost to various extents throughout the process of sample collection, storage, before, during, and/or after sample preparation and analysis. The calculation of overall extraction recovery can detect problems of low recovery during sample preparation but does not identify the source(s) of analyte losses. Low overall analyte recovery is the net result of losses that can happen for multiple reasons at all steps of sample preparation and analysis. Therefore, identifying the source(s) of analyte loss during sample preparation can help guide the optimization the bioanalysis conditions to minimize these losses. In this article we propose a practical protocol to systematically identify and quantify the sources of low analyte recovery. This allows the proper choice of strategies to optimize the relevant bioanalytical conditions to minimize analyte losses and improve overall recovery.
Collapse
Affiliation(s)
- Devendra Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Nagsen Gautam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
14
|
Filppula AM, Hirvensalo P, Parviainen H, Ivaska VE, Lönnberg KI, Deng F, Viinamäki J, Kurkela M, Neuvonen M, Niemi M. Comparative Hepatic and Intestinal Metabolism and Pharmacodynamics of Statins. Drug Metab Dispos 2021; 49:658-667. [PMID: 34045219 DOI: 10.1124/dmd.121.000406] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
This study aimed to comprehensively investigate the in vitro metabolism of statins. The metabolism of clinically relevant concentrations of atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin, and their metabolites were investigated using human liver microsomes (HLMs), human intestine microsomes (HIMs), liver cytosol, and recombinant cytochrome P450 enzymes. We also determined the inhibitory effects of statin acids on their pharmacological target, 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase. In HLMs, statin lactones were metabolized to a much higher extent than their acid forms. Atorvastatin lactone and simvastatin (lactone) showed extensive metabolism [intrinsic clearance (CLint) values of 3700 and 7400 µl/min per milligram], whereas the metabolism of the lactones of 2-hydroxyatorvastatin, 4-hydroxyatorvastatin, and pitavastatin was slower (CLint 20-840 µl/min per milligram). The acids had CLint values in the range <0.1-80 µl/min per milligram. In HIMs, only atorvastatin lactone and simvastatin (lactone) exhibited notable metabolism, with CLint values corresponding to 20% of those observed in HLMs. CYP3A4/5 and CYP2C9 were the main statin-metabolizing enzymes. The majority of the acids inhibited HMG-CoA reductase, with 50% inhibitory concentrations of 4-20 nM. The present comparison of the metabolism and pharmacodynamics of the various statins using identical methods provides a strong basis for further application, e.g., comparative systems pharmacology modeling. SIGNIFICANCE STATEMENT: The present comparison of the in vitro metabolic and pharmacodynamic properties of atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin and their metabolites using unified methodology provides a strong basis for further application. Together with in vitro drug transporter and clinical data, the present findings are applicable for use in comparative systems pharmacology modeling to predict the pharmacokinetics and pharmacological effects of statins at different dosages.
Collapse
Affiliation(s)
- Anne M Filppula
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Päivi Hirvensalo
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Heli Parviainen
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Vilma E Ivaska
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - K Ivar Lönnberg
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Feng Deng
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Jenni Viinamäki
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Mika Kurkela
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Mikko Neuvonen
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Mikko Niemi
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| |
Collapse
|
15
|
Li Z, Lyu Y, Zhao J, Li D, Lin Z, To KKW, Yan X, Zuo Z. Disease Status-Dependent Drug-Herb Interactions: NASH Lowered the Risk of Hepatotoxicity in Rats Coadministered With Simvastatin and Gardenia jasminoides J. Ellis. Front Pharmacol 2021; 12:622040. [PMID: 33967756 PMCID: PMC8103205 DOI: 10.3389/fphar.2021.622040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/18/2021] [Indexed: 11/13/2022] Open
Abstract
Concurrent use of simvastatin (SV) and Gardenia jasminoides J. Ellis (GJ) was adopted in patients with multi-morbidity, such as stroke rehabilitation patients with NASH. Although hepatotoxicity has been reported in both of them and NASH could alter the pharmacokinetics of drugs/herbs, the interaction between SV and GJ and the related hepatotoxicity remained uninvestigated under neither healthy nor NASH condition. The current study aimed to evaluate the potential hepatotoxicity resulted from the interactions between SV and GJ in both healthy and NASH rats. Both healthy and NASH rats received two-week SV (p. o., 8.66 mg/kg, once daily) and/or GJ (p.o., 325 mg/kg, twice daily). Pharmacokinetic profiles of SV, simvastatin acid (SVA, active metabolite of SV), and geniposide (major component in GJ); hepatic Cyp2c11/Oatp1b2/P-gp expression; and biomarker levels of liver function, lipid levels, and liver histology were compared to demonstrate the interactions in rats. To explore the mechanism of the interaction-mediated hepatotoxicity, hepatic genipin-protein adduct content and iNOS/COX-1/COX-2 expressions from related groups were compared. Moreover, liver histology of healthy/NASH rats at 90 days after discontinuation of two-week GJ in the absence and presence of SV was evaluated to estimate the long-term impact of the interactions. GJ reduced the systemic exposures of SV and SVA by up-regulating the hepatic P-gp expression in healthy but not NASH rats. Meanwhile, SV increased the systemic exposure of geniposide via inhibiting the activity of P-gp in both healthy and NASH rats. Although neither SV nor GJ induced hepatotoxicity in healthy rats, their co-treatment elevated serum ALT and AST levels, which may attribute to the aggravated genipin-protein adduct formation, inflammation infiltration, and iNOS/COX-1 expressions in the liver. In NASH rats, SV and/or GJ reduced serum ALT, AST, LDL/vLDL, and TC levels via alleviating hepatic inflammation infiltration and iNOS/COX-1 expressions. Moreover, in comparison to NASH rats, more severe fibrosis was observed in the livers of healthy rats at 90 days after discontinuation of two-week SV and GJ coadministration. Although interactions between SV and GJ induced short-term and long-term liver injuries in healthy rats, NASH condition in rats could lower such risk.
Collapse
Affiliation(s)
- Ziwei Li
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuanfeng Lyu
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jiajia Zhao
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Dan Li
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zhixiu Lin
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xiaoyu Yan
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zhong Zuo
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
16
|
Hu SX, Ernst K, Benner CP, Feenstra KL. Stability of Ketoprofen Methylester in Plasma of Different Species. Curr Drug Metab 2021; 22:215-223. [PMID: 33334282 DOI: 10.2174/1389200221666201217141025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/30/2020] [Accepted: 11/04/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Pharmacokinetic and pharmacodynamic assessment of ester-containing drugs can be impacted by hydrolysis of the drugs in plasma samples post blood collection. The impact is different in the plasma of different species. OBJECTIVE This study evaluated the stability of a prodrug, ketoprofen methylester (KME), in commercially purchased and freshly collected plasma of mouse, rat, dog, cat, pig, sheep, cattle and horse. METHODS KME hydrolysis was determined following its incubation in commercially purchased and freshly collected plasma of those species. Different esterase inhibitors were evaluated for prevention of the hydrolysis in rat, dog and pig plasma. RESULTS KME was rapidly hydrolyzed in both commercially purchased and freshly collected plasma of mouse, rat, and horse. The hydrolysis was initially quick and then limited in cat plasma. KME hydrolysis was minimum in commercially purchased plasma of dog, pig, sheep and cattle but substantial in freshly collected plasma of those species. Different esterase inhibitors showed different effects on the stability of KME in rat, dog and pig plasma. CONCLUSION These results indicate that plasma of different species has different hydrolytic activities to estercontaining drugs. The activities in commercially purchased and freshly collected plasma may be different and species-dependent. Esterase inhibitors have different effects on preventing hydrolysis of the ester-containing drugs in the plasma of different species.
Collapse
Affiliation(s)
- Steven X Hu
- Zoetis, Inc., Veterinary Medicine Research and Development, Kalamazoo, MI 49007, United States
| | - Kelsey Ernst
- Zoetis, Inc., Veterinary Medicine Research and Development, Kalamazoo, MI 49007, United States
| | - Charles P Benner
- Zoetis, Inc., Veterinary Medicine Research and Development, Kalamazoo, MI 49007, United States
| | - Kenneth L Feenstra
- Zoetis, Inc., Veterinary Medicine Research and Development, Kalamazoo, MI 49007, United States
| |
Collapse
|
17
|
Li Z, Zhang J, Zhang Y, Zhou L, Zhao J, Lyu Y, Poon LH, Lin Z, To KKW, Yan X, Zuo Z. Intestinal absorption and hepatic elimination of drugs in high-fat high-cholesterol diet-induced non-alcoholic steatohepatitis rats: exemplified by simvastatin. Br J Pharmacol 2020; 178:582-599. [PMID: 33119943 DOI: 10.1111/bph.15298] [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: 03/18/2020] [Revised: 10/12/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Altered drug pharmacokinetics is a significant concern in non-alcoholic steatohepatitis (NASH) patients. Although high-fat high-cholesterol (HFHC) diet-induced NASH (HFHC-NASH) rats could simulate the typical dysregulation of cholesterol in NASH patients, experimental investigation on the altered drug pharmacokinetics in this model are limited. Thus, the present study comprehensive investigates the nature of such altered pharmacokinetics using simvastatin as the model drug. EXPERIMENTAL APPROACH Pharmacokinetic profiles of simvastatin and its active metabolite simvastatin acid together with compartmental pharmacokinetic modelling were used to identify the key factors involved in the altered pharmacokinetics of simvastatin in HFHC-NASH rats. Experimental investigations via in situ single-pass intestinal perfusion and intrahepatic injection of simvastatin were carried out. Histology, Ces1 activities and mRNA/protein levels of Oatp1b2/CYP2c11/P-gp in the small intestine/liver of healthy and HFHC-NASH rats were compared. KEY RESULTS Reduced intestinal absorption and more extensive hepatic elimination in HFHC-NASH rats resulted in less systemic exposures of simvastatin/simvastatin acid. In the small intestine of HFHC-NASH rats, thicker intestinal wall with more collagen fibres, increased Ces1 activity and up-regulated P-gp protein decreased the permeability of simvastatin, accelerated the hydrolysis of simvastatin and promoted the efflux of simvastatin acid respectively. In the liver of HFHC-NASH rats, higher hepatic P-gp expression accelerated the hepatic elimination of simvastatin. CONCLUSION AND IMPLICATIONS Altered histology, Ces1 activity and P-gp expression in the small intestine/liver were identified to be the major contributing factors leading to less systemic exposure of drugs in HFHC-NASH rats, which may be applicable to NASH patients.
Collapse
Affiliation(s)
- Ziwei Li
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jun Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yufeng Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Limin Zhou
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Jiajia Zhao
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuanfeng Lyu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Long Hin Poon
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zhixiu Lin
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kenneth Kin Wah To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xiaoyu Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zhong Zuo
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
18
|
Shi CC, Song YQ, He RJ, Guan XQ, Song LL, Chen ST, Sun MR, Ge GB, Zhang LR. Rapalogues as hCES2A Inhibitors: In Vitro and In Silico Investigations. Eur J Drug Metab Pharmacokinet 2020; 46:129-139. [PMID: 33140264 DOI: 10.1007/s13318-020-00659-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND AND OBJECTIVE Rapamycin and its semi-synthetic analogues (rapalogues) are frequently used in combination with other prescribed medications in clinical settings. Although the inhibitory effects of rapalogues on cytochrome P450 enzymes (CYPs) have been well examined, the inhibition potentials of rapalogues on human esterases have not been investigated. Herein, the inhibition potentials and inhibitory mechanisms of six marketed rapalogues on human esterases are investigated. METHODS The inhibitory effects of six marketed rapalogues (rapamycin, zotarolimus, temsirolimus, everolimus, pimecrolimus and tacrolimus) on three major esterases, including human carboxylesterases 1 (hCES1A), human carboxylesterases 2 (hCES2A) and butyrylcholinesterase (BuChE), were assayed using isozyme-specific substrates. Inhibition kinetic analyses and docking simulations were performed to investigate the inhibitory mechanisms of the rapalogues with strong hCES2A inhibition potency. RESULTS Zotarolimus and pimecrolimus displayed strong inhibition of human hCES2A but these agents did not inhibit hCES1A or BuChE. Further investigation demonstrated that zotarolimus could strongly inhibit intracellular hCES2A in living HepG2 cells, with an estimated IC50 value of 4.09 µM. Inhibition kinetic analyses revealed that zotarolimus inhibited hCES2A-catalyzed fluorescein diacetate hydrolysis in a mixed manner, with the Ki value of 1.61 µM. Docking simulations showed that zotarolimus could tightly bind on hCES2A at two district ligand-binding sites, consistent with its mixed inhibition mode. CONCLUSION Our findings demonstrate that several marketed rapalogues are potent and specific hCES2A inhibitors, and these agents can serve as leading compounds for the development of more efficacious hCES2A inhibitors to modulate the pharmacokinetic profiles and toxicity of hCES2A-substrate drugs (such as the anticancer agent irinotecan).
Collapse
Affiliation(s)
- Cheng-Cheng Shi
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China. .,Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
| | - Yun-Qing Song
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Rong-Jing He
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Xiao-Qing Guan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Li-Lin Song
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Shi-Tong Chen
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Meng-Ru Sun
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China.
| | - Li-Rong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
| |
Collapse
|