1
|
Ikuta H, Shimada H, Sakamoto K, Nakamura R, Kawase A, Iwaki M. Species differences in liver microsomal hydrolysis of acyl glucuronide in humans and rats. Xenobiotica 2022; 52:653-660. [PMID: 36190839 DOI: 10.1080/00498254.2022.2131484] [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: 12/13/2022]
Abstract
Acyl glucuronides (AGs) are known as one of the causes of idiosyncratic drug toxicity (IDT). Although AGs can be enzymatically hydrolysed by β-glucuronidase and esterase, much information on their characteristics and species differences is lacking. This study was aimed to clarify species differences in AG hydrolysis between human and rat liver microsomes (HLM and RLM).To evaluate the AG hydrolysis profile, and the contribution of β-glucuronidase and esterase towards AG hydrolysis in HLM and RLM, nonsteroidal anti-inflammatory drugs (NSAIDs) were used. AGs were incubated with 0.1 M Tris-HCl buffer (pH 7.4) and 0.3 mg/mL HLM or RLM in the absence or presence of β-glucuronidase inhibitor, D-saccharic acid 1,4-lactone (D-SL) and esterase inhibitor, phenylmethylsulfonyl fluoride (PMSF).AGs of mefenamic acid (MEF-AG) and etodolac (ETO-AG) showed significantly higher AG hydrolysis rates in RLM than in HLM. Esterases were found to serve as AG hydrolases dominantly in HLM, whereas both esterases and β-glucuronidase equally contribute to AG hydrolysis in RLM. However, MEF-AG and ETO-AG were hydrolysed only by β-glucuronidase.We demonstrated for the first time that the activity of AG hydrolases towards NSAID-AGs differs between humans and rats.
Collapse
Affiliation(s)
| | | | | | - Rena Nakamura
- Faculty of Pharmacy, Kindai University, Osaka, Japan
| | | | - Masahiro Iwaki
- Faculty of Pharmacy, Kindai University, Osaka, Japan.,Pharmaceutical Research and Technology Institute, Kindai University, Osaka, Japan.,Antiaging Center, Kindai University, Osaka, Japan
| |
Collapse
|
2
|
Hafez HM, Waz S, Rifaai RA, Mohamed MZ. Involvement of NOX-4/JAK/STAT pathway in the protective effect of aprepitant against diclofenac-induced renal toxicity. Life Sci 2022; 294:120381. [PMID: 35143828 DOI: 10.1016/j.lfs.2022.120381] [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: 12/21/2021] [Revised: 01/25/2022] [Accepted: 02/03/2022] [Indexed: 10/19/2022]
Abstract
AIMS Aprepitant, a neurokinin-1 (NK1) receptor antagonist, is a clinically approved anti-emetic drug. Recently, inhibition of the NK1 receptor has been reported as a potential nephroprotective strategy. We aimed to assess the pharmacological mechanisms of aprepitant against diclofenac (DIC)-induced renal toxicity. MAIN METHODS An in vivo study was conducted using twenty-four male Wistar rats, divided into 4 groups. Aprepitant was administered for 5 days (5 mg/kg/day) with or without DIC which was given on the 4th and 5th days (50 mg/kg, i.p.). At the end of the study, renal function biomarkers, renal oxidative parameters, prostaglandin E (PGE-2), and NADPH oxidase (NOX-4) were measured. Histopathological changes as well as expression of renal inflammatory and apoptotic markers (tumor necrosis factor alpha (TNF-α) and caspase-3) were investigated. KEY FINDINGS DIC caused significant renal damage, as evidenced by deterioration of renal functions, oxidative stress, inflammatory and apoptotic markers, and confirmed by histopathological findings. Pretreatment with aprepitant successfully ameliorated and improved all biochemical and molecular parameters induced by DIC. Moreover, aprepitant restored the decrease in renal PGE-2 concentration and inhibited DIC-activated Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling in renal tissues. SIGNIFICANCE The protective effect of aprepitant is possibly attributed to its anti-oxidant and anti-inflammatory roles via the NOX-4/JAK/STAT pathway.
Collapse
Affiliation(s)
- Heba M Hafez
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt.
| | - Shaimaa Waz
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia 61511, Egypt
| | - Rehab Ahmed Rifaai
- Department of Histology and Cell biology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt
| | - Mervat Z Mohamed
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt
| |
Collapse
|
3
|
Description and Analysis of Glycosidic Residues in the Largest Open Natural Products Database. Biomolecules 2021; 11:biom11040486. [PMID: 33804966 PMCID: PMC8063959 DOI: 10.3390/biom11040486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/20/2022] Open
Abstract
Natural products (NPs), biomolecules produced by living organisms, inspire the pharmaceutical industry and research due to their structural characteristics and the substituents from which they derive their activities. Glycosidic residues are frequently present in NP structures and have particular pharmacokinetic and pharmacodynamic importance as they improve their solubility and are often involved in molecular transport, target specificity, ligand–target interactions, and receptor binding. The COlleCtion of Open Natural prodUcTs (COCONUT) is currently the largest open database of NPs, and therefore a suitable starting point for the detection and analysis of the diversity of glycosidic residues in NPs. In this work, we report and describe the presence of circular, linear, terminal, and non-terminal glycosidic units in NPs, together with their importance in drug discovery.
Collapse
|
4
|
Scialis RJ, Ghanem CI, Manautou JE. The modulation of transcriptional expression and inhibition of multidrug resistance associated protein 4 (MRP4) by analgesics and their primary metabolites. Curr Res Toxicol 2020; 1:34-41. [PMID: 34345835 PMCID: PMC8320619 DOI: 10.1016/j.crtox.2020.04.002] [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: 12/20/2019] [Revised: 04/02/2020] [Accepted: 04/24/2020] [Indexed: 11/17/2022] Open
Abstract
During the course of a toxic challenge, changes in gene expression can manifest such as induction of metabolizing enzymes as a compensatory detoxification response. We currently report that a single 400 mg/kg acetaminophen (APAP) dose to C57BL/6J mice led to an increase in multidrug resistance-associated (Mrp) 4 (Abcc4) mRNA 12 h after administration. Alanine aminotransferase, as a marker of liver injury, was also elevated indicating hepatotoxicity had occurred. Therefore, induction of Mrp4 mRNA was likely attributable to APAP-induced liver injury. Mrp4 has been shown to be upregulated during oxidative stress, and it is well-established that APAP overdose causes oxidative stress due to depletion of glutathione. Given the importance of Mrp4 upregulation as an adaptive response during cholestatic and oxidative liver injury, we next investigated the extent by which human MRP4 can be inhibited by the analgesics, APAP, diclofenac (DCF), and their metabolites. Using an in vitro assay with inside out human MRP4 vesicles, we determined that APAP-cysteine inhibited MRP4-mediated transport of leukotriene C4 with an apparent IC50 of 125 μM. APAP-glutathione also attenuated MRP4 activity though it achieved only 28% inhibition at 300 μM. Diclofenac acyl glucuronide (DCF-AG) inhibited MRP4 transport by 34% at 300 μM. The MRP4 in vitro inhibition occurs at APAP-cysteine and DCF-AG concentrations seen in vivo after toxic doses of APAP or DCF in mice, hence the findings are important given the role that Mrp4 serves as a compensatory response during oxidative stress following toxic challenge. Following 400 mg/kg APAP in mice, mean ALT 12 hours post-dose was 1,140 U/L A statistically significant increase in Mrp4 mRNA was observed 12 hours post-dose APAP-CYS inhibited human MRP4 transport of LTC4 with an IC50 = 125 μM (Ki = 122 μM) APAP-GSH decreased MRP4 transport by 29% inhibition at 300 μM APAP, APAP-GLU, APAP-NAC, and APAP-SUL did not exhibit significant MRP4 inhibition
Collapse
Key Words
- ALT, alanine aminotransferase
- AMP, adenosine monophosphate
- APAP, acetaminophen
- APAP-CYS, acetaminophen cysteine
- APAP-GLU, acetaminophen glucuronide
- APAP-NAC, acetaminophen N-acetylcysteine
- APAP-SUL, acetaminophen sulfate
- ATP, adenosine triphosphate
- Acetaminophen
- DCF, diclofenac
- DCF-AG, diclofenac acyl glucuronide
- Diclofenac
- Fmo, flavin containing monooxygenase
- IS, internal standard
- Inhibition
- LTC4, leukotriene C4
- MRP, multidrug resistance-associated protein
- MRP4
- Metabolite
- OH-DCF, 4′-hydroxy diclofenac
- PGE2, prostaglandin E2
Collapse
Affiliation(s)
| | | | - José E. Manautou
- Corresponding author at: University of Connecticut, Dept. of Pharmaceutical Sciences, 69 North Eagleville Road, Storrs, CT 06269-3092, USA.
| |
Collapse
|
5
|
Huo X, Meng Q, Wang C, Wu J, Wang C, Zhu Y, Ma X, Sun H, Liu K. Protective effect of cilastatin against diclofenac-induced nephrotoxicity through interaction with diclofenac acyl glucuronide via organic anion transporters. Br J Pharmacol 2020; 177:1933-1948. [PMID: 32000294 PMCID: PMC7161545 DOI: 10.1111/bph.14957] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/21/2019] [Accepted: 12/02/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Diclofenac is a widely used nonsteroidal anti-inflammatory drug. However, adverse effects in the kidney limit its clinical application. The present study was aimed to evaluate the potential effect of cilastatin on diclofenac-induced acute kidney injury and to clarify the potential roles of renal organic anion transporters (OATs) in the drug-drug interaction between cilastatin and diclofenac. EXPERIMENTAL APPROACH The effect of cilastatin was evaluated in diclofenac-induced acute kidney injury in mice. Human OAT1/3-transfected HEK293 cells and renal primary proximal tubule cells (RPTCs) were used to investigate OAT1/3-mediated transport and the cytotoxicity of diclofenac. KEY RESULTS Cilastatin treatment decreased the pathological changes, renal dysfunction and elevated renal levels of oxidation products, cytokine production and apoptosis induced by diclofenac in mice. Moreover, cilastatin increased the plasma concentration and decreased the renal distribution of diclofenac and its glucuronide metabolite, diclofenac acyl glucuronide (DLF-AG). Similarly, cilastatin inhibited cytotoxicity and mitochondrial damage in RPTCs but did not change the intracellular accumulation of diclofenac. DLF-AG but not diclofenac exhibited OAT-dependent cytotoxicity and was identified as an OAT1/3 substrate. Cilastatin inhibited the intracellular accumulation and decreased the cytotoxicity of DLF-AG in RPTCs. CONCLUSION AND IMPLICATIONS Cilastatin alleviated diclofenac-induced acute kidney injury in mice by restoring the redox balance, suppressing inflammation, and reducing apoptosis. Cilastatin inhibited OATs and decreased the renal distribution of diclofenac and DLF-AG, which further ameliorated the diclofenac-induced nephrotoxicity in mice. Cilastatin can be potentially used in the clinic as a therapeutic agent to alleviate the adverse renal reaction to diclofenac.
Collapse
Affiliation(s)
- Xiaokui Huo
- Department of Clinical Pharmacology, College of PharmacyDalian Medical UniversityDalianChina
- College (Institute) of Integrative MedicineDalian Medical UniversityDalianChina
- Provincial Key Laboratory for Pharmacokinetics and Transport, LiaoningDalian Medical UniversityDalianChina
| | - Qiang Meng
- Department of Clinical Pharmacology, College of PharmacyDalian Medical UniversityDalianChina
- College (Institute) of Integrative MedicineDalian Medical UniversityDalianChina
- Provincial Key Laboratory for Pharmacokinetics and Transport, LiaoningDalian Medical UniversityDalianChina
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of PharmacyDalian Medical UniversityDalianChina
- College (Institute) of Integrative MedicineDalian Medical UniversityDalianChina
- Provincial Key Laboratory for Pharmacokinetics and Transport, LiaoningDalian Medical UniversityDalianChina
| | - Jingjing Wu
- Department of Clinical Pharmacology, College of PharmacyDalian Medical UniversityDalianChina
- College (Institute) of Integrative MedicineDalian Medical UniversityDalianChina
- Provincial Key Laboratory for Pharmacokinetics and Transport, LiaoningDalian Medical UniversityDalianChina
| | - Chong Wang
- Department of Clinical Pharmacology, College of PharmacyDalian Medical UniversityDalianChina
- College (Institute) of Integrative MedicineDalian Medical UniversityDalianChina
- Provincial Key Laboratory for Pharmacokinetics and Transport, LiaoningDalian Medical UniversityDalianChina
| | - Yanna Zhu
- Department of Clinical Pharmacology, College of PharmacyDalian Medical UniversityDalianChina
- College (Institute) of Integrative MedicineDalian Medical UniversityDalianChina
- Provincial Key Laboratory for Pharmacokinetics and Transport, LiaoningDalian Medical UniversityDalianChina
| | - Xiaodong Ma
- Department of Clinical Pharmacology, College of PharmacyDalian Medical UniversityDalianChina
| | - Huijun Sun
- Department of Clinical Pharmacology, College of PharmacyDalian Medical UniversityDalianChina
- College (Institute) of Integrative MedicineDalian Medical UniversityDalianChina
- Provincial Key Laboratory for Pharmacokinetics and Transport, LiaoningDalian Medical UniversityDalianChina
| | - Kexin Liu
- Department of Clinical Pharmacology, College of PharmacyDalian Medical UniversityDalianChina
- College (Institute) of Integrative MedicineDalian Medical UniversityDalianChina
- Provincial Key Laboratory for Pharmacokinetics and Transport, LiaoningDalian Medical UniversityDalianChina
| |
Collapse
|
6
|
Lang SQ, Lang WH, Yu HY, Wang L. Metabolic activation of TM5441 in vitro and in vivo: Formation of reactive metabolites and human enzymes involved. Eur J Pharm Sci 2019; 143:105195. [PMID: 31852629 DOI: 10.1016/j.ejps.2019.105195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 12/13/2019] [Accepted: 12/14/2019] [Indexed: 11/19/2022]
Abstract
TM5441, a furan-containing drug, is an inhibitor of plasminogen activator inhibitor-1 (PAI-1), which can induce intrinsic apoptosis of human cancer cell lines. The aim of this study was to identify the reactive metabolites of TM5441 and to reveal the bioactivation pathways that are associated with its hepatotoxicity. The reactive metabolites were trapped by using glutathione (GSH) or N-acetyl-lysine (NAL) in rat, dog, and human liver microsomal incubation system after exposure to TM5441. Two metabolic activation pathways were disclosed. The first bioactivation pathway was dominated by Cytochrome P450 enzymes (CYP450s); TM5441 was metabolized into cis-2-butene-1,4-dial derivative dependent on NADPH, which can be trapped in the liver microsomal incubations fortified with GSH or NAL as trapping agents. Five metabolites (M1, M2, M9, M12 and M13) associated with GSH and three metabolites (M4, M7 and M14) associated with NAL were identified by liquid chromatography-high resolution mass spectrometry. The second bioactivation pathway was catalyzed by UDP-glucuronosyltransferases (UGTs); TM5441 was conjugated with glucuronide to form acyl-glucuronide (M10), which further reacted with GSH, resulting in the identification of a TM5441-S-acyl-GSH adduct (M11) in liver microsomal incubations fortified with uridine-5'-diphosphoglucuronidc acid (UDPGA) and GSH. M9, M10, M11, M12 and M13 were also detected in bile samples of rats given TM5441. Compared with rat, dog would display closer bioactivation profiles to human. The CYP450 enzyme responsible for the bioactivation of TM5441 was mainly identified as CYP3A4, using human recombinant CYP450 enzymes and specific inhibitory studies. The UGT enzymes responsible for the bioactivation of TM5441 mainly involved UGT2B7, 1A1 and 1A4. These results facilitate the understanding of the bioactivation of TM5441 and potential toxicological implications.
Collapse
Affiliation(s)
- Su-Qin Lang
- Department of Outpatient, Weifang People's Hospital, Weifang 261041, Shandong Province, China
| | - Wen-Hua Lang
- Department of Spine surgery, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Hai-Yan Yu
- Department of Radiology, Weifang People's Hospital, Weifang 261041, Shandong Province, China
| | - Li Wang
- Department of Pharmacy, Jining No.1 People's Hospital, Jining 272011, Shandong Province, China.
| |
Collapse
|
7
|
Protective effects of Ajwa date extract against tissue damage induced by acute diclofenac toxicity. J Taibah Univ Med Sci 2019; 14:553-559. [PMID: 31908644 PMCID: PMC6940670 DOI: 10.1016/j.jtumed.2019.10.002] [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: 06/28/2019] [Revised: 09/28/2019] [Accepted: 10/05/2019] [Indexed: 11/23/2022] Open
Abstract
Objectives To investigate the tissue-protective effects of Ajwa date fruits (a Prophetic medicinal remedy) against acute diclofenac toxicity. Methods Albino Sprague–Dawley rats were allocated to four experimental groups: a negative control group, an Ajwa-only group that received 2 g/kg of Ajwa date extract (ADE) orally, an acute diclofenac toxicity group that received 200 mg diclofenac once intraperitoneally, and a treatment group that received diclofenac and ADE after 4 h. Histological examinations of rat lung and liver tissues were performed. Results Acute diclofenac toxicity caused marked hepatic derangements, such as congested central veins, congested blood sinusoids, hyaline degeneration, and hepatocyte necrosis. Toxic diclofenac overdose resulted in markedly congested alveolar capillaries and alveolar haemorrhages, thick edematous alveolar walls, and edema fluid exudates in the alveoli. Upon treatment with ADE, significant reduction in diclofenac-induced hepatic and pulmonary derangements were observed. Conclusion ADE is a safe, tissue-protective nutritional agent that alleviates cellular and tissue-damaging effects due to acute diclofenac toxicity. ADE relieved hepatic and pulmonary changes induced by acute diclofenac toxicity. The use of ADE is recommended for the treatment of acute diclofenac toxicity.
Collapse
|
8
|
Nie Y, Yang J, Liu S, Sun R, Chen H, Long N, Jiang R, Gui C. Genetic polymorphisms of human hepatic OATPs: functional consequences and effect on drug pharmacokinetics. Xenobiotica 2019; 50:297-317. [DOI: 10.1080/00498254.2019.1629043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Yingmin Nie
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jingjie Yang
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Shuai Liu
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Ruiqi Sun
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Huihui Chen
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Nan Long
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Rui Jiang
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chunshan Gui
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
9
|
Baba A, Yamada K, Satoh T, Watanabe K, Yoshioka T. Chemo-Enzymatic Synthesis, Structural and Stereochemical Characterization, and Intrinsic Degradation Kinetics of Diastereomers of 1-β- O-Acyl Glucuronides Derived from Racemic 2-{4-[(2-Methylprop-2-en-1-yl)amino]phenyl}propanoic Acid. ACS OMEGA 2018; 3:4932-4940. [PMID: 31458709 PMCID: PMC6641924 DOI: 10.1021/acsomega.8b00443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/24/2018] [Indexed: 06/10/2023]
Abstract
Alminoprofen, (RS)-2-{4-[(2-methylprop-2-en-1-yl)amino]phenyl}propanoic acid (ALP) 1, is a racemic drug categorized as a 2-arylpropanoic acid-class nonsteroidal anti-inflammatory drug. Pharmacokinetic studies of 1 in patients have revealed that the corresponding acyl glucuronide 5 is a major urinary metabolite, but little is known about the structure and stereochemistry of 5. The present work describes the synthesis of a diastereomeric mixture of 1-β-O-acyl glucuronides (2RS)-5 from 1 and methyl 2,3,4-tri-O-acetyl-1-bromo-1-deoxy-α-d-glucopyranuronate 2 using our chemo-enzymatic method that has complete specificity for the β-configuration. The structure of (2RS)-5 was characterized by 1H and 13C NMR spectroscopy and high-resolution mass spectrometry as well as by complete hydrolysis by β-glucuronidase. The absolute stereochemistry of (2RS)-5 was determined by comparison with (2R)-5 synthesized alternatively from (2R)-1 and 2. Compound (2R)-1 was prepared in two steps starting from chiral (R)-2-(4-nitrophenyl)propanoic acid (2R)-6. Chiral resolution of (2RS)-1 was achieved using a chiral high-performance liquid chromatography column, and its stereochemistry was determined by comparison with (2R)-1. The intrinsic degradation rate constant of (2R)-5 was 0.405 ± 0.002 h-1, which is approximately twice that of (2S)-5 (the k value was 0.226 ± 0.002 h-1) under physiological conditions (pH 7.40, 37 °C).
Collapse
Affiliation(s)
- Akiko Baba
- Department
of Medicinal Chemistry and Department of Pharmaceutics, Faculty
of Pharmaceutical Sciences, Hokkaido University
of Science, 7-15-4-1
Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Koki Yamada
- Department
of Medicinal Chemistry and Department of Pharmaceutics, Faculty
of Pharmaceutical Sciences, Hokkaido University
of Science, 7-15-4-1
Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Takashi Satoh
- Department
of Medicinal Chemistry and Department of Pharmaceutics, Faculty
of Pharmaceutical Sciences, Hokkaido University
of Science, 7-15-4-1
Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Kazuhiro Watanabe
- Department
of Medicinal Chemistry and Department of Pharmaceutics, Faculty
of Pharmaceutical Sciences, Hokkaido University
of Science, 7-15-4-1
Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Tadao Yoshioka
- Department
of Medicinal Chemistry and Department of Pharmaceutics, Faculty
of Pharmaceutical Sciences, Hokkaido University
of Science, 7-15-4-1
Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| |
Collapse
|
10
|
Wilson CE, Dickie AP, Schreiter K, Wehr R, Wilson EM, Bial J, Scheer N, Wilson ID, Riley RJ. The pharmacokinetics and metabolism of diclofenac in chimeric humanized and murinized FRG mice. Arch Toxicol 2018; 92:1953-1967. [PMID: 29721588 DOI: 10.1007/s00204-018-2212-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/25/2018] [Indexed: 11/30/2022]
Abstract
The pharmacokinetics of diclofenac were investigated following single oral doses of 10 mg/kg to chimeric liver humanized and murinized FRG and C57BL/6 mice. In addition, the metabolism and excretion were investigated in chimeric liver humanized and murinized FRG mice. Diclofenac reached maximum blood concentrations of 2.43 ± 0.9 µg/mL (n = 3) at 0.25 h post-dose with an AUCinf of 3.67 µg h/mL and an effective half-life of 0.86 h (n = 2). In the murinized animals, maximum blood concentrations were determined as 3.86 ± 2.31 µg/mL at 0.25 h post-dose with an AUCinf of 4.94 ± 2.93 µg h/mL and a half-life of 0.52 ± 0.03 h (n = 3). In C57BL/6J mice, mean peak blood concentrations of 2.31 ± 0.53 µg/mL were seen 0.25 h post-dose with a mean AUCinf of 2.10 ± 0.49 µg h/mL and a half-life of 0.51 ± 0.49 h (n = 3). Analysis of blood indicated only trace quantities of drug-related material in chimeric humanized and murinized FRG mice. Metabolic profiling of urine, bile and faecal extracts revealed a complex pattern of metabolites for both humanized and murinized animals with, in addition to unchanged parent drug, a variety of hydroxylated and conjugated metabolites detected. The profiles in humanized mice were different to those of both murinized and wild-type animals, e.g., a higher proportion of the dose was detected in the form of acyl glucuronide metabolites and much reduced amounts as taurine conjugates. Comparison of the metabolic profiles obtained from the present study with previously published data from C57BL/6J mice and humans revealed a greater, though not complete, match between chimeric humanized mice and humans, such that the liver humanized FRG model may represent a model for assessing the biotransformation of such compounds in humans.
Collapse
Affiliation(s)
- C E Wilson
- Nestlé Skin Health R&D, Les Templiers, Route des Colles, BP 87, 06902, Sophia-Antipolis, France.
| | - A P Dickie
- Evotec (UK) Ltd, 114 Innovation Drive, Abingdon, Oxfordshire, OX14 4RZ, UK
| | - K Schreiter
- Evotec International GmbH, Manfred Eigen Campus, Essener Bogen 7, Hamburg, Germany
| | - R Wehr
- Evotec International GmbH, Manfred Eigen Campus, Essener Bogen 7, Hamburg, Germany
| | - E M Wilson
- Yecuris Corporation, PO Box 4645, Tualatin, OR, 97062, USA
| | - J Bial
- Yecuris Corporation, PO Box 4645, Tualatin, OR, 97062, USA
| | - N Scheer
- CEVEC Pharmaceuticals GmbH, Gottfried-Hagen-Str. 60-62, 51105, Cologne, Germany
| | - I D Wilson
- Department of Surgery and Cancer, Imperial College, Exhibition Rd, South Kensington, London, SW7 2AZ, UK
| | - R J Riley
- Evotec (UK) Ltd, Alderley Park, Nether Alderley, Cheshire, SK10 4TG, UK
| |
Collapse
|
11
|
Van Vleet TR, Liu H, Lee A, Blomme EAG. Acyl glucuronide metabolites: Implications for drug safety assessment. Toxicol Lett 2017; 272:1-7. [PMID: 28286018 DOI: 10.1016/j.toxlet.2017.03.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/17/2017] [Accepted: 03/05/2017] [Indexed: 12/23/2022]
Abstract
Acyl glucuronides are important metabolites of compounds with carboxylic acid moieties and have unique properties that distinguish them from other phase 2 metabolites. In particular, in addition to being often unstable, acyl glucuronide metabolites can be chemically reactive leading to covalent binding with macromolecules and toxicity. While there is circumstantial evidence that drugs forming acyl glucuronide metabolites can be associated with rare, but severe idiosyncratic toxic reactions, many widely prescribed drugs with good safety records are also metabolized through acyl glucuronidation. Therefore, there is a need to understand the various factors that can affect the safety of acyl glucuronide-producing drugs including the rate of acyl glucuronide formation, the relative reactivity of the acyl glucuronide metabolite formed, the rate of elimination, potential proteins being targeted, and the rate of aglucuronidation. In this review, these factors are discussed and various approaches to de-risk the safety liabilities of acyl glucuronide metabolites are evaluated.
Collapse
Affiliation(s)
- Terry R Van Vleet
- Abbvie, Development Sciences, Department of Preclinical Safety, United States.
| | - Hong Liu
- Abbvie, Development Sciences, Biomeasure and Metabolism, United States
| | - Anthony Lee
- Abbvie, Development Sciences, Biomeasure and Metabolism, United States
| | - Eric A G Blomme
- Abbvie, Development Sciences, Department of Preclinical Safety, United States
| |
Collapse
|
12
|
Toxicological potential of acyl glucuronides and its assessment. Drug Metab Pharmacokinet 2017; 32:2-11. [DOI: 10.1016/j.dmpk.2016.11.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 12/22/2022]
|
13
|
Kovacsics D, Patik I, Özvegy-Laczka C. The role of organic anion transporting polypeptides in drug absorption, distribution, excretion and drug-drug interactions. Expert Opin Drug Metab Toxicol 2016; 13:409-424. [PMID: 27783531 DOI: 10.1080/17425255.2017.1253679] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION The in vivo fate and effectiveness of a drug depends highly on its absorption, distribution, metabolism, excretion and toxicity (ADME-Tox). Organic anion transporting polypeptides (OATPs) are membrane proteins involved in the cellular uptake of various organic compounds, including clinically used drugs. Since OATPs are significant players in drug absorption and distribution, modulation of OATP function via pharmacotherapy with OATP substrates/inhibitors, or modulation of their expression, affects drug pharmacokinetics. Given their cancer-specific expression, OATPs may also be considered anticancer drug targets. Areas covered: We describe the human OATP family, discussing clinically relevant consequences of altered OATP function. We offer a critical analysis of published data on the role of OATPs in ADME and in drug-drug interactions, especially focusing on OATP1A2, 1B1, 1B3 and 2B1. Expert opinion: Four members of the OATP family, 1A2, 1B1, 1B3 and 2B1, have been characterized in detail. As biochemical and pharmacological knowledge on the other OATPs is lacking, it seems timely to direct research efforts towards developing the experimental framework needed to investigate the transport mechanism and substrate specificity of the poorly described OATPs. In addition, elucidating the role of OATPs in tumor development and therapy response are critical avenues for further research.
Collapse
Affiliation(s)
- Daniella Kovacsics
- a Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - Izabel Patik
- a Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - Csilla Özvegy-Laczka
- a Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| |
Collapse
|