1
|
Review on Bortezomib Resistance in Multiple Myeloma and Potential Role of Emerging Technologies. Pharmaceuticals (Basel) 2023; 16:ph16010111. [PMID: 36678608 PMCID: PMC9864669 DOI: 10.3390/ph16010111] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Multiple myeloma is a hematological cancer type. For its treatment, Bortezomib has been widely used. However, drug resistance to this effective chemotherapeutic has been developed for various reasons. 2D cell cultures and animal models have failed to understand the MM disease and Bortezomib resistance. It is therefore essential to utilize new technologies to reveal a complete molecular profile of the disease. In this review, we in-depth examined the possible molecular mechanisms that cause Bortezomib resistance and specifically addressed MM and Bortezomib resistance. Moreover, we also included the use of nanoparticles, 3D culture methods, microfluidics, and organ-on-chip devices in multiple myeloma. We also discussed whether the emerging technology offers the necessary tools to understand and prevent Bortezomib resistance in multiple myeloma. Despite the ongoing research activities on MM, the related studies cannot provide a complete summary of MM. Nanoparticle and 3D culturing have been frequently used to understand MM disease and Bortezomib resistance. However, the number of microfluidic devices for this application is insufficient. By combining siRNA/miRNA technologies with microfluidic devices, a complete molecular genetic profile of MM disease could be revealed. Microfluidic chips should be used clinically in personal therapy and point-of-care applications. At least with Bortezomib microneedles, it could be ensured that MM patients can go through the treatment process more painlessly. This way, MM can be switched to the curable cancer type list, and Bortezomib can be targeted for its treatment with fewer side effects.
Collapse
|
2
|
Development and Challenges of Diclofenac-Based Novel Therapeutics: Targeting Cancer and Complex Diseases. Cancers (Basel) 2022; 14:cancers14184385. [PMID: 36139546 PMCID: PMC9496891 DOI: 10.3390/cancers14184385] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Diclofenac is a widely used drug for its anti-inflammatory and pain alleviating properties. This review summarizes the current understanding about the drug diclofenac. The potential applications of diclofenac beyond its well-known anti-inflammatory properties for other diseases such as cancer are discussed, along with existing limitations. Abstract Diclofenac is a highly prescribed non-steroidal anti-inflammatory drug (NSAID) that relieves inflammation, pain, fever, and aches, used at different doses depending on clinical conditions. This drug inhibits cyclooxygenase-1 and cyclooxygenase-2 enzymes, which are responsible for the generation of prostaglandin synthesis. To improve current diclofenac-based therapies, we require new molecular systematic therapeutic approaches to reduce complex multifactorial effects. However, the critical challenge that appears with diclofenac and other drugs of the same class is their side effects, such as signs of stomach injuries, kidney problems, cardiovascular issues, hepatic issues, and diarrhea. In this article, we discuss why defining diclofenac-based mechanisms, pharmacological features, and its medicinal properties are needed to direct future drug development against neurodegeneration and imperfect ageing and to improve cancer therapy. In addition, we describe various advance molecular mechanisms and fundamental aspects linked with diclofenac which can strengthen and enable the better designing of new derivatives of diclofenac to overcome critical challenges and improve their applications.
Collapse
|
3
|
Järvinen E, Deng F, Kiander W, Sinokki A, Kidron H, Sjöstedt N. The Role of Uptake and Efflux Transporters in the Disposition of Glucuronide and Sulfate Conjugates. Front Pharmacol 2022; 12:802539. [PMID: 35095509 PMCID: PMC8793843 DOI: 10.3389/fphar.2021.802539] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Glucuronidation and sulfation are the most typical phase II metabolic reactions of drugs. The resulting glucuronide and sulfate conjugates are generally considered inactive and safe. They may, however, be the most prominent drug-related material in the circulation and excreta of humans. The glucuronide and sulfate metabolites of drugs typically have limited cell membrane permeability and subsequently, their distribution and excretion from the human body requires transport proteins. Uptake transporters, such as organic anion transporters (OATs and OATPs), mediate the uptake of conjugates into the liver and kidney, while efflux transporters, such as multidrug resistance proteins (MRPs) and breast cancer resistance protein (BCRP), mediate expulsion of conjugates into bile, urine and the intestinal lumen. Understanding the active transport of conjugated drug metabolites is important for predicting the fate of a drug in the body and its safety and efficacy. The aim of this review is to compile the understanding of transporter-mediated disposition of phase II conjugates. We review the literature on hepatic, intestinal and renal uptake transporters participating in the transport of glucuronide and sulfate metabolites of drugs, other xenobiotics and endobiotics. In addition, we provide an update on the involvement of efflux transporters in the disposition of glucuronide and sulfate metabolites. Finally, we discuss the interplay between uptake and efflux transport in the intestine, liver and kidneys as well as the role of transporters in glucuronide and sulfate conjugate toxicity, drug interactions, pharmacogenetics and species differences.
Collapse
Affiliation(s)
- Erkka Järvinen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alli Sinokki
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Drug-drug-gene interactions as mediators of adverse drug reactions to diclofenac and statins: a case report and literature review. ACTA ACUST UNITED AC 2021; 72:114-128. [PMID: 34187111 PMCID: PMC8265195 DOI: 10.2478/aiht-2021-72-3549] [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: 04/01/2021] [Accepted: 05/01/2021] [Indexed: 01/29/2023]
Abstract
Concomitant treatment with drugs that inhibit drug metabolising enzymes and/or transporters, such as commonly prescribed statins and nonsteroidal anti-inflammatory drugs (NSAIDs), has been associated with prolonged drug exposure and increased risk of adverse drug reactions (ADRs) due to drug-drug interactions. The risk is further increased in patients with chronic diseases/comorbidities who are more susceptible because of their genetic setup or external factors. In that light, we present a case of a 46-year-old woman who had been experiencing acute renal and hepatic injury and myalgia over two years of concomitant treatment with diclofenac, atorvastatin, simvastatin/fenofibrate, and several other drugs, including pantoprazole and furosemide. Our pharmacogenomic findings supported the suspicion that ADRs, most notably the multi-organ toxicity experienced by our patient, may be owed to drug-drug-gene interactions and increased bioavailability of the prescribed drugs due to slower detoxification capacity and decreased hepatic and renal elimination. We also discuss the importance of CYP polymorphisms in the biotransformation of endogenous substrates such as arachidonic acid and their modulating role in pathophysiological processes. Yet even though the risks of ADRs related to the above mentioned drugs are substantially evidenced in literature, pre-emptive pharmacogenetic analysis has not yet found its way into common clinical practice.
Collapse
|
5
|
Nudischer R, Renggli K, Hierlemann A, Roth AB, Bertinetti-Lapatki C. Characterization of a long-term mouse primary liver 3D tissue model recapitulating innate-immune responses and drug-induced liver toxicity. PLoS One 2020; 15:e0235745. [PMID: 32645073 PMCID: PMC7347206 DOI: 10.1371/journal.pone.0235745] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional liver in vitro systems have recently attracted a lot of attention in drug development. These systems help to gain unprecedented insights into drug-induced liver injury (DILI), as they more closely reproduce liver biology, and as drug effects can be studied in isolated and controllable microenvironments. Many groups established human-based in vitro models but so far neglected the animal equivalent, although the availability of both models would be desirable. Animal in vitro models enable back- and forward translation of in vitro and in vivo findings, bridge the gap between rodent in vivo and human in vitro scenarios, and ultimately support the interpretation of data generated with preclinical species and humans. Since mice are often used in drug development and physiologically relevant in vitro systems are lacking, we established, for the first time, a mouse liver model that encompasses primary parenchymal and non-parenchymal cells with preserved viability and functionality over three weeks. Using our three-dimensional liver spheroids, we were able to predict the toxicity of known DILI compounds, demonstrated the interaction cascades between the different cell types and showed evidence of drug-induced steatosis and cholestasis. In summary, our mouse liver spheroids represent a valuable in vitro model that can be applied to study DILI findings, reported from mouse studies, and offers the potential to detect immune-mediated drug-induced liver toxicity.
Collapse
Affiliation(s)
- Ramona Nudischer
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
- * E-mail:
| | - Kasper Renggli
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Zurich, Switzerland
| | - Andreas Hierlemann
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Zurich, Switzerland
| | - Adrian B. Roth
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Cristina Bertinetti-Lapatki
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
6
|
Walles M, Brown AP, Zimmerlin A, End P. New Perspectives on Drug-Induced Liver Injury Risk Assessment of Acyl Glucuronides. Chem Res Toxicol 2020; 33:1551-1560. [PMID: 32525307 DOI: 10.1021/acs.chemrestox.0c00131] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Drug-induced liver injury (DILI) remains one of the key challenges in drug development due to the mechanisms of action being multifactorial in nature. This is particularly the case for idiosyncratic DILI which occurs in a very low frequency in humans (e.g., 1:10,000). Despite perceptions that acyl glucuronide metabolites are defacto risks for DILI, scientific evidence suggests that acyl glucuronide formation alone does not pose an increased risk compared to other drug metabolites. This applies in particular to those acyl glucuronides which are not reactive and do not form covalent adducts with proteins. The goal of this paper is to provide guidance on preclinical and clinical strategies to evaluate the potential for acyl glucuronide formation to contribute to DILI. A key element of our proposed safety assessment is to investigate whether a particular acyl glucuronide is reactive or not and whether systemic exposure in humans can be demonstrated in animal toxicology studies following administration of the parent drug. While standard animal toxicology studies can identify overtly hepatotoxic compounds, these studies are not predictive for drugs that produce idiosyncratic forms of DILI. In addition, we do not recommend conducting toxicology studies of administered individual acyl glucuronides due to differences in pharmacokinetic and dispositional properties from the endogenously produced metabolites. Once a drug candidate has entered clinical trials, the focus should be on clinical safety data and emerging risk-benefit analysis.
Collapse
Affiliation(s)
- Markus Walles
- PK Sciences, Novartis Institutes for Biomedical Research, Novartis Campus, 4052 Basel, Switzerland
| | - Alan P Brown
- Preclinical Safety, Novartis Institutes for Biomedical Research, 220 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Alfred Zimmerlin
- PK Sciences, Novartis Institutes for Biomedical Research, Novartis Campus, 4052 Basel, Switzerland
| | - Peter End
- PK Sciences, Novartis Institutes for Biomedical Research, Novartis Campus, 4052 Basel, Switzerland
| |
Collapse
|
7
|
Selvaraj S, Oh JH, Borlak J. An adverse outcome pathway for immune-mediated and allergic hepatitis: a case study with the NSAID diclofenac. Arch Toxicol 2020; 94:2733-2748. [PMID: 32372211 PMCID: PMC7395045 DOI: 10.1007/s00204-020-02767-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 04/22/2020] [Indexed: 12/26/2022]
Abstract
Many drugs have the potential to cause drug-induced liver injury (DILI); however, underlying mechanisms are diverse. The concept of adverse outcome pathways (AOPs) has become instrumental for risk assessment of drug class effects. We report AOPs specific for immune-mediated and drug hypersensitivity/allergic hepatitis by considering genomic, histo- and clinical pathology data of mice and dogs treated with diclofenac. The findings are relevant for other NSAIDs and drugs undergoing iminoquinone and quinone reactive metabolite formation. We define reactive metabolites catalyzed by CYP monooxygenase and myeloperoxidases of neutrophils and Kupffer cells as well as acyl glucuronides produced by uridine diphosphoglucuronosyl transferase as molecular initiating events (MIE). The reactive metabolites bind to proteins and act as neo-antigen and involve antigen-presenting cells to elicit B- and T-cell responses. Given the diverse immune systems between mice and dogs, six different key events (KEs) at the cellular and up to four KEs at the organ level are defined with mechanistic plausibility for the onset and progression of liver inflammation. With mice, cellular stress response, interferon gamma-, adipocytokine- and chemokine signaling provided a rationale for the AOP of immune-mediated hepatitis. With dogs, an erroneous programming of the innate and adaptive immune response resulted in mast cell activation; their infiltration into liver parenchyma and the shift to M2-polarized Kupffer cells signify allergic hepatitis and the occurrence of granulomas of the liver. Taken together, diclofenac induces divergent immune responses among two important preclinical animal species, and the injury pattern seen among clinical cases confirms the relevance of the developed AOP for immune-mediated hepatitis.
Collapse
Affiliation(s)
- Saravanakumar Selvaraj
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625, Hannover, Germany
| | - Jung-Hwa Oh
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625, Hannover, Germany.,Department of Predictive Toxicology, Korea Institute of Toxicology, Gajeong-ro, Yuseong, Daejeon, 34114, Republic of Korea
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
8
|
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: 23] [Impact Index Per Article: 4.6] [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
|
9
|
Garcia-Lino AM, Blanco-Paniagua E, Astorga-Simon EN, Alvarez-Fernandez L, Garcia-Mateos D, Alvarez-Fernandez I, Alvarez AI, Merino G. Abcg2 transporter affects plasma, milk and tissue levels of meloxicam. Biochem Pharmacol 2020; 175:113924. [PMID: 32217099 DOI: 10.1016/j.bcp.2020.113924] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/19/2020] [Indexed: 12/15/2022]
Abstract
ATP-binding cassette (ABCG2) is an efflux transporter that extrudes xenotoxins from cells in liver, intestine, mammary gland, brain and other organs, affecting the pharmacokinetics, brain accumulation and secretion into milk of several compounds, including antitumoral, antimicrobial and anti-inflammatory drugs. The aim of this study was to investigate whether the widely used anti-inflammatory drug meloxicam is an Abcg2 sustrate, and how this transporter affects its systemic distribution. Using polarized ABCG2-transduced cell lines, we found that meloxicam is efficiently transported by murine Abcg2 and human ABCG2. After oral administration of meloxicam, the area under the plasma concentration-time curve in Abcg2-/- mice was 2-fold higher than in wild type mice (146.06 ± 10.57 µg·h/ml versus 73.80 ± 10.00 µg·h/ml). Differences in meloxicam distribution were reported for several tissues after oral and intravenous administration, with a 20-fold higher concentration in the brain of Abcg2-/- after oral administration. Meloxicam secretion into milk was also affected by the transporter, with a 2-fold higher milk-to-plasma ratio in wild-type compared with Abcg2-/- lactating female mice after oral and intravenous administration. We conclude that Abcg2 is an important determinant of the plasma and brain distribution of meloxicam and is clearly involved in its secretion into milk.
Collapse
Affiliation(s)
- Alba M Garcia-Lino
- Department of Biomedical Sciences-Physiology, Veterinary Faculty, Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, Campus de Vegazana, León, Spain
| | - Esther Blanco-Paniagua
- Department of Biomedical Sciences-Physiology, Veterinary Faculty, Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, Campus de Vegazana, León, Spain
| | - Elsa N Astorga-Simon
- Department of Biomedical Sciences-Physiology, Veterinary Faculty, Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, Campus de Vegazana, León, Spain
| | - Laura Alvarez-Fernandez
- Department of Biomedical Sciences-Physiology, Veterinary Faculty, Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, Campus de Vegazana, León, Spain
| | - Dafne Garcia-Mateos
- Department of Biomedical Sciences-Physiology, Veterinary Faculty, Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, Campus de Vegazana, León, Spain
| | - Indira Alvarez-Fernandez
- Department of Biomedical Sciences-Physiology, Veterinary Faculty, Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, Campus de Vegazana, León, Spain
| | - Ana I Alvarez
- Department of Biomedical Sciences-Physiology, Veterinary Faculty, Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, Campus de Vegazana, León, Spain
| | - Gracia Merino
- Department of Biomedical Sciences-Physiology, Veterinary Faculty, Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, Campus de Vegazana, León, Spain.
| |
Collapse
|
10
|
Scialis RJ, Aleksunes LM, Csanaky IL, Klaassen CD, Manautou JE. Identification and Characterization of Efflux Transporters That Modulate the Subtoxic Disposition of Diclofenac and Its Metabolites. Drug Metab Dispos 2019; 47:1080-1092. [PMID: 31399506 PMCID: PMC6750190 DOI: 10.1124/dmd.119.086603] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/12/2019] [Indexed: 11/22/2022] Open
Abstract
In the present work, in vivo transporter knockout (KO) mouse models were used to characterize the disposition of diclofenac (DCF) and its primary metabolites following a single subtoxic dose in mice lacking breast cancer resistance protein (Bcrp) or multidrug resistance-associated protein (Mrp)3. The results indicate that Bcrp acts as a canalicular efflux mediator for DCF, as wild-type (WT) mice had biliary excretion values that were 2.2- to 2.6-fold greater than Bcrp KO mice, although DCF plasma levels were not affected. The loss of Bcrp resulted in a 1.8- to 3.2-fold increase of diclofenac acyl glucuronide (DCF-AG) plasma concentrations in KO animals compared with WT mice, while the biliary excretion of DCF-AG increased 1.4-fold in WT versus KO mice. Furthermore, Mrp3 was found to mediate the basolateral transport of DCF-AG, but not DCF or 4'-hydroxy diclofenac. WT mice had DCF-AG plasma concentrations 7.0- to 8.6-fold higher than Mrp3 KO animals; however, there were no changes in biliary excretion of DCF-AG. Vesicular transport experiments with human MRP3 demonstrated that MRP3 is able to transport DCF-AG via low- and high-affinity binding sites. The low-affinity MRP3 transport had a V max and K m of 170 pmol/min/mg and 98.2 µM, respectively, while the high-affinity V max and K m parameters were estimated to be 71.9 pmol/min/mg and 1.78 µM, respectively. In summary, we offer evidence that the disposition of DCF-AG can be affected by both Bcrp and Mrp3, and these findings may be applicable to humans.
Collapse
Affiliation(s)
- Renato J Scialis
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (R.J.S., J.E.M.), Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (L.M.A., I.L.C., C.D.K.) and Department of Internal Medicine, University of Kansas Medical Center, Kansas City (C.D.K.)
| | - Lauren M Aleksunes
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (R.J.S., J.E.M.), Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (L.M.A., I.L.C., C.D.K.) and Department of Internal Medicine, University of Kansas Medical Center, Kansas City (C.D.K.)
| | - Iván L Csanaky
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (R.J.S., J.E.M.), Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (L.M.A., I.L.C., C.D.K.) and Department of Internal Medicine, University of Kansas Medical Center, Kansas City (C.D.K.)
| | - Curtis D Klaassen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (R.J.S., J.E.M.), Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (L.M.A., I.L.C., C.D.K.) and Department of Internal Medicine, University of Kansas Medical Center, Kansas City (C.D.K.)
| | - José E Manautou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (R.J.S., J.E.M.), Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (L.M.A., I.L.C., C.D.K.) and Department of Internal Medicine, University of Kansas Medical Center, Kansas City (C.D.K.)
| |
Collapse
|
11
|
Ghanem CI, Manautou JE. Modulation of Hepatic MRP3/ABCC3 by Xenobiotics and Pathophysiological Conditions: Role in Drug Pharmacokinetics. Curr Med Chem 2019; 26:1185-1223. [PMID: 29473496 DOI: 10.2174/0929867325666180221142315] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/17/2018] [Accepted: 02/05/2018] [Indexed: 12/13/2022]
Abstract
Liver transporters play an important role in the pharmacokinetics and disposition of pharmaceuticals, environmental contaminants, and endogenous compounds. Among them, the family of ATP-Binding Cassette (ABC) transporters is the most important due to its role in the transport of endo- and xenobiotics. The ABCC sub-family is the largest one, consisting of 13 members that include the cystic fibrosis conductance regulator (CFTR/ABCC7); the sulfonylurea receptors (SUR1/ABCC8 and SUR2/ABCC9) and the multidrug resistanceassociated proteins (MRPs). The MRP-related proteins can collectively confer resistance to natural, synthetic drugs and their conjugated metabolites, including platinum-containing compounds, folate anti-metabolites, nucleoside and nucleotide analogs, among others. MRPs can be also catalogued into "long" (MRP1/ABCC1, -2/C2, -3/C3, -6/C6, and -7/C10) and "short" (MRP4/C4, -5/C5, -8/C11, -9/C12, and -10/C13) categories. While MRP2/ABCC2 is expressed in the canalicular pole of hepatocytes, all others are located in the basolateral membrane. In this review, we summarize information from studies examining the changes in expression and regulation of the basolateral hepatic transporter MPR3/ABCC3 by xenobiotics and during various pathophysiological conditions. We also focus, primarily, on the consequences of such changes in the pharmacokinetic, pharmacodynamic and/or toxicity of different drugs of clinical use transported by MRP3.
Collapse
Affiliation(s)
- Carolina I Ghanem
- Instituto de Investigaciones Farmacologicas (ININFA), Facultad de Farmacia y Bioquimica. CONICET. Universidad de Buenos Aires, Buenos Aires, Argentina.,Catedra de Fisiopatologia. Facultad de Farmacia y Bioquimica. Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jose E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
12
|
Pomes LM, Guglielmetti M, Bertamino E, Simmaco M, Borro M, Martelletti P. Optimising migraine treatment: from drug-drug interactions to personalized medicine. J Headache Pain 2019; 20:56. [PMID: 31101004 PMCID: PMC6734220 DOI: 10.1186/s10194-019-1010-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/05/2019] [Indexed: 11/16/2022] Open
Abstract
Migraine is the most disabling and expensive chronic disorders, the etiology of which is still not fully known. The neuronal systems, (glutammatergic, dopaminergic, serotoninergic and GABA-ergic) whose functionality is partly attributable to genetically determined factors, has been suggested to play an important role. The treatment of acute attacks and the prophylactic management of chronic forms include the use of different category of drugs, and it is demonstrated that not each subject has the same clinical answer to them. The reason of this is to be searched in different functional capacity and quantity of phase I enzymes (such as different isoforms of CYP P450), phase II enzymes (such as UDP-glucuronosyltransferases), receptors (such as OPRM1 for opioids) and transporters (such as ABCB1) involved in the metabolic destiny of each drug, all of these dictated by DNA and RNA variations. The general picture is further exacerbated by the need for polytherapies, often also to treat comorbidities, which may interfere with the pharmacological action of anti-migraine drugs. Personalized medicine has the objective of setting the optimal therapies in the light of the functional biochemical asset and of the comorbidities of the individual patient, in order to obtain the best clinical response. Novel therapeutic perspectives in migraine includes biotechnological drugs directed against molecules (such as CGRP and its receptor) that cause vasodilatation at the peripheral level of the meningeal blood vessels and reflex stimulation of the parasympathetic system. Drug-drug interactions and the possible competitive metabolic destiny should be studied by the application of pharmacogenomics in large scale. Drug-drug interactions and their possible competitive metabolic destiny should be studied by the application of pharmacogenomics in large scale.
Collapse
Affiliation(s)
- Leda Marina Pomes
- Residency Program in Laboratory Medicine, Gabriele d'Annunzio University, Chieti, Italy
| | - Martina Guglielmetti
- Regional Referral Headache Centre, Sant'Andrea Hospital, Rome, Italy.,Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Enrico Bertamino
- Residency Program in Hygiene and Preventive Medicine, Sapienza University of Rome, Rome, Italy
| | - Maurizio Simmaco
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Rome, Italy.,Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marina Borro
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Rome, Italy.,Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Paolo Martelletti
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy. .,Internal Medicine and Emergency Medicine Unit, Sant'Andrea Hospital, Rome, Italy.
| |
Collapse
|
13
|
Overview: Role of Drug Transporters in Drug Disposition and Its Clinical Significance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:1-12. [PMID: 31571163 DOI: 10.1007/978-981-13-7647-4_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Absorption, distribution, and excretion of drugs are involved in drug transport across plasma membrane, most of which are mediated by drug transporters. These drug transporters are generally divided into solute carrier (SLC) family and ATP-binding cassette (ABC) family. These transporters not only mediate transport of therapeutic drugs across membrane but also transport various kinds of endogenous compounds. Thus besides being participated in disposal of drug and its clinical efficacy/toxicity, these transporters also play vital roles in maintaining cell homeostasis via regulating transport of endogenous compounds. This chapter will outline classification of drug transporters, their roles in drug disposal/drug response, and remote communication between tissues/organs.
Collapse
|
14
|
Kawase A, Kaneto A, Ishibashi M, Kobayashi A, Shimada H, Iwaki M. Involvement of diclofenac acyl-β-d-glucuronide in diclofenac-induced cytotoxicity in glutathione-depleted isolated murine hepatocytes co-cultured with peritoneal macrophages. Toxicol Mech Methods 2018; 29:203-210. [PMID: 30489186 DOI: 10.1080/15376516.2018.1544384] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Direct hepatotoxic effects of drugs can occur when a parent drug and/or its reactive metabolites induces the formation of reactive oxygen species. Reactive metabolites of diclofenac (DIC) such as DIC acyl-β-d-glucuronide (DIC-AG) bind covalently to proteins, potentially decreasing protein function or inducing an immune response. However, it is unclear whether the macrophages and GSH depletion participate in DIC-induced cytotoxicity. Mouse hepatocytes (Hep) co-cultured with peritoneal macrophages (PMs) were used to clarify the effects of presence of PM with GSH depletion on DIC-induced cytotoxicity in Hep. DIC-AG but not hydroxy-DIC concentrations in medium were significantly increased in Hep co-cultured with PM with GSH depletion. Depletion of GSH resulted in significantly higher LDH leakage. Interestingly, LDH leakage in Hep/PM (1:0.4) with GSH depletion was significantly higher than in Hep/PM (1:0 and 1:0.1) with BSO. It is likely that macrophages with GSH depletion could facilitate DIC-induced cytotoxicity.
Collapse
Affiliation(s)
- Atsushi Kawase
- a Department of Pharmacy, Faculty of Pharmacy , Kindai University , Osaka , Japan
| | - Ayaka Kaneto
- a Department of Pharmacy, Faculty of Pharmacy , Kindai University , Osaka , Japan
| | - Mao Ishibashi
- a Department of Pharmacy, Faculty of Pharmacy , Kindai University , Osaka , Japan
| | - Akihiro Kobayashi
- a Department of Pharmacy, Faculty of Pharmacy , Kindai University , Osaka , Japan
| | - Hiroaki Shimada
- a Department of Pharmacy, Faculty of Pharmacy , Kindai University , Osaka , Japan
| | - Masahiro Iwaki
- a Department of Pharmacy, Faculty of Pharmacy , Kindai University , Osaka , Japan
| |
Collapse
|
15
|
Ryder TF, Calabrese MF, Walker GS, Cameron KO, Reyes AR, Borzilleri KA, Delmore J, Miller R, Kurumbail RG, Ward J, Kung DW, Brown JA, Edmonds DJ, Eng H, Wolford AC, Kalgutkar AS. Acyl Glucuronide Metabolites of 6-Chloro-5-[4-(1-hydroxycyclobutyl)phenyl]-1 H-indole-3-carboxylic Acid (PF-06409577) and Related Indole-3-carboxylic Acid Derivatives are Direct Activators of Adenosine Monophosphate-Activated Protein Kinase (AMPK). J Med Chem 2018; 61:7273-7288. [PMID: 30036059 DOI: 10.1021/acs.jmedchem.8b00807] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Studies on indole-3-carboxylic acid derivatives as direct activators of human adenosine monophosphate-activated protein kinase (AMPK) α1β1γ1 isoform have culminated in the identification of PF-06409577 (1), PF-06885249 (2), and PF-06679142 (3) as potential clinical candidates. Compounds 1-3 are primarily cleared in animals and humans via glucuronidation. Herein, we describe the biosynthetic preparation, purification, and structural characterization of the glucuronide conjugates of 1-3. Spectral characterization of the purified glucuronides M1, M2, and M3 indicated that they were acyl glucuronide derivatives. In vitro pharmacological evaluation revealed that all three acyl glucuronides retained selective activation of β1-containing AMPK isoforms. Inhibition of de novo lipogenesis with representative parent carboxylic acids and their respective acyl glucuronide conjugates in human hepatocytes demonstrated their propensity to activate cellular AMPK. Cocrystallization of the AMPK α1β1γ1 isoform with 1-3 and M1-M3 provided molecular insights into the structural basis for AMPK activation by the glucuronide conjugates.
Collapse
Affiliation(s)
- Tim F Ryder
- Medicine Design , Pfizer Worldwide Research & Development , Groton , Connecticut 06340 , United States
| | - Matthew F Calabrese
- Medicine Design , Pfizer Worldwide Research & Development , Groton , Connecticut 06340 , United States
| | - Gregory S Walker
- Medicine Design , Pfizer Worldwide Research & Development , Groton , Connecticut 06340 , United States
| | | | | | - Kris A Borzilleri
- Medicine Design , Pfizer Worldwide Research & Development , Groton , Connecticut 06340 , United States
| | | | | | - Ravi G Kurumbail
- Medicine Design , Pfizer Worldwide Research & Development , Groton , Connecticut 06340 , United States
| | | | - Daniel W Kung
- Medicine Design , Pfizer Worldwide Research & Development , Groton , Connecticut 06340 , United States
| | - Janice A Brown
- Medicine Design , Pfizer Worldwide Research & Development , Groton , Connecticut 06340 , United States
| | | | - Heather Eng
- Medicine Design , Pfizer Worldwide Research & Development , Groton , Connecticut 06340 , United States
| | - Angela C Wolford
- Medicine Design , Pfizer Worldwide Research & Development , Groton , Connecticut 06340 , United States
| | | |
Collapse
|
16
|
Lee EH, Oh JH, Selvaraj S, Park SM, Choi MS, Spanel R, Yoon S, Borlak J. Immunogenomics reveal molecular circuits of diclofenac induced liver injury in mice. Oncotarget 2017; 7:14983-5017. [PMID: 26934552 PMCID: PMC4924767 DOI: 10.18632/oncotarget.7698] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/25/2015] [Indexed: 12/12/2022] Open
Abstract
Diclofenac is a non-steroidal anti-inflammatory drug and its use can be associated with severe adverse reactions, notably myocardial infarction, stroke and drug-induced liver injury (DILI). In pursue of immune-mediated DILI mechanisms an immunogenomic study was carried out. Diclofenac treatment of mice at 30 mg/kg for 3 days caused significant serum ALT and AST elevations, hepatomegaly and degenerative changes including hepatic glycogen depletion, hydropic swelling, cholesterolosis and eosinophilic hepatocytes with one animal presenting subsegmental infarction due to portal vein thrombosis. Furthermore, portal/periportal induction of the rate limiting enzyme in ammonia detoxification, i.e. carbamoyl phosphate synthetase 1 was observed. The performed microarray studies informed on > 600 differential expressed genes of which 35, 37 and 50 coded for inflammation, 51, 44 and 61 for immune and 116, 129 and 169 for stress response, respectively after single and repeated dosing for 3 and 14 days. Bioinformatic analysis defined molecular circuits of hepatic inflammation with the growth hormone (Ghr)− and leptin receptor, the protein-tyrosine-phosphatase, selectin and the suppressor-of-cytokine-signaling (Socs) to function as key nodes in gene regulatory networks. Western blotting confirmed induction of fibronectin and M-CSF to hallmark tissue repair and differentiation of monocytes and macrophages. Transcript expression of the macrophage receptor with collagenous structure increased > 7-fold and immunohistochemistry of CD68 evidenced activation of tissue-resident macrophages. Importantly, diclofenac treatment prompted strong expression of phosphorylated Stat3 amongst individual animals and the associated 8- and 4-fold Soc3 and Il-6 induction reinforced Ghr degradation as evidenced by immunoblotting. Moreover, immunohistochemistry confirmed regulation of master regulatory proteins of diclofenac treated mice to suggest complex pro-and anti-inflammatory reactions in immune-mediated hepatic injury. The findings encourage translational research.
Collapse
Affiliation(s)
- Eun-Hee Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 305-343, Republic of Korea
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 305-343, Republic of Korea.,Department of Human and Environmental Toxicology, School of Engineering, Korea University of Science and Technology, Daejeon, 305-343, Republic of Korea
| | - Saravanakumar Selvaraj
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625 Hannover, Germany
| | - Se-Myo Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 305-343, Republic of Korea
| | - Mi-Sun Choi
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 305-343, Republic of Korea
| | - Reinhard Spanel
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625 Hannover, Germany.,Institute for Clinical Pathology, 41747 Viersen, Germany
| | - Seokjoo Yoon
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 305-343, Republic of Korea.,Department of Human and Environmental Toxicology, School of Engineering, Korea University of Science and Technology, Daejeon, 305-343, Republic of Korea
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
17
|
Utzeri E, Usai P. Role of non-steroidal anti-inflammatory drugs on intestinal permeability and nonalcoholic fatty liver disease. World J Gastroenterol 2017; 23:3954-3963. [PMID: 28652650 PMCID: PMC5473116 DOI: 10.3748/wjg.v23.i22.3954] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/19/2017] [Accepted: 02/08/2017] [Indexed: 02/06/2023] Open
Abstract
The use of non-steroidal anti-inflammatory drugs (NSAIDs) is widespread worldwide thanks to their analgesic, anti-inflammatory and antipyretic effects. However, even more attention is placed upon the recurrence of digestive system complications in the course of their use. Recent data suggests that the complications of the lower gastro-intestinal tract may be as frequent and severe as those of the upper tract. NSAIDs enteropathy is due to enterohepatic recycling of the drugs resulting in a prolonged and repeated exposure of the intestinal mucosa to the compound and its metabolites. Thus leading to so-called topical effects, which, in turn, lead to an impairment of the intestinal barrier. This process determines bacterial translocation and toxic substances of intestinal origin in the portal circulation, leading to an endotoxaemia. This condition could determine a liver inflammatory response and might promote the development of non-alcoholic steatohepatitis, mostly in patients with risk factors such as obesity, metabolic syndrome and a high fat diet, which may induce a small intestinal bacterial overgrowth and dysbiosis. This alteration of gut microbiota may contribute to nonalcoholic fatty liver disease and its related disorders in two ways: firstly causing a malfunction of the tight junctions that play a critical role in the increase of intestinal permeability, and then secondly leading to the development of insulin resistance, body weight gain, lipogenesis, fibrogenesis and hepatic oxidative stress.
Collapse
|
18
|
Yang G, Ge S, Singh R, Basu S, Shatzer K, Zen M, Liu J, Tu Y, Zhang C, Wei J, Shi J, Zhu L, Liu Z, Wang Y, Gao S, Hu M. Glucuronidation: driving factors and their impact on glucuronide disposition. Drug Metab Rev 2017; 49:105-138. [PMID: 28266877 DOI: 10.1080/03602532.2017.1293682] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Glucuronidation is a well-recognized phase II metabolic pathway for a variety of chemicals including drugs and endogenous substances. Although it is usually the secondary metabolic pathway for a compound preceded by phase I hydroxylation, glucuronidation alone could serve as the dominant metabolic pathway for many compounds, including some with high aqueous solubility. Glucuronidation involves the metabolism of parent compound by UDP-glucuronosyltransferases (UGTs) into hydrophilic and negatively charged glucuronides that cannot exit the cell without the aid of efflux transporters. Therefore, elimination of parent compound via glucuronidation in a metabolic active cell is controlled by two driving forces: the formation of glucuronides by UGT enzymes and the (polarized) excretion of these glucuronides by efflux transporters located on the cell surfaces in various drug disposition organs. Contrary to the common assumption that the glucuronides reaching the systemic circulation were destined for urinary excretion, recent evidences suggest that hepatocytes are capable of highly efficient biliary clearance of the gut-generated glucuronides. Furthermore, the biliary- and enteric-eliminated glucuronides participate into recycling schemes involving intestinal microbes, which often prolong their local and systemic exposure, albeit at low systemic concentrations. Taken together, these recent research advances indicate that although UGT determines the rate and extent of glucuronide generation, the efflux and uptake transporters determine the distribution of these glucuronides into blood and then to various organs for elimination. Recycling schemes impact the apparent plasma half-life of parent compounds and their glucuronides that reach intestinal lumen, in addition to prolonging their gut and colon exposure.
Collapse
Affiliation(s)
- Guangyi Yang
- a Department of Pharmacy , Institute of Wudang Herbal Medicine Research, Taihe Hospital, Hubei University of Medicine , Shiyan , Hubei , China.,b Hubei Provincial Technology and Research Center for Comprehensive Development of Medicinal Herbs, Hubei University of Medicine , Shiyan , Hubei , China
| | - Shufan Ge
- c Department of Pharmacological and Pharmaceutical Sciences , College of Pharmacy, University of Houston , Houston , TX , USA
| | - Rashim Singh
- c Department of Pharmacological and Pharmaceutical Sciences , College of Pharmacy, University of Houston , Houston , TX , USA
| | - Sumit Basu
- c Department of Pharmacological and Pharmaceutical Sciences , College of Pharmacy, University of Houston , Houston , TX , USA
| | - Katherine Shatzer
- c Department of Pharmacological and Pharmaceutical Sciences , College of Pharmacy, University of Houston , Houston , TX , USA
| | - Ming Zen
- d Department of Thoracic and Cardiomacrovascular Surgery , Taihe Hospital, Hubei University of Medicine , Shiyan , Hubei , China
| | - Jiong Liu
- e Department of Digestive Diseases Surgery , Taihe Hospital, Hubei University of Medicine , Shiyan , Hubei , China
| | - Yifan Tu
- c Department of Pharmacological and Pharmaceutical Sciences , College of Pharmacy, University of Houston , Houston , TX , USA
| | - Chenning Zhang
- a Department of Pharmacy , Institute of Wudang Herbal Medicine Research, Taihe Hospital, Hubei University of Medicine , Shiyan , Hubei , China
| | - Jinbao Wei
- a Department of Pharmacy , Institute of Wudang Herbal Medicine Research, Taihe Hospital, Hubei University of Medicine , Shiyan , Hubei , China
| | - Jian Shi
- f Department of Pharmacy , Institute of Translational Chinese Medicine, Guangzhou University of Chinese Medicine , Guangzhou , Guangdong , China
| | - Lijun Zhu
- f Department of Pharmacy , Institute of Translational Chinese Medicine, Guangzhou University of Chinese Medicine , Guangzhou , Guangdong , China
| | - Zhongqiu Liu
- f Department of Pharmacy , Institute of Translational Chinese Medicine, Guangzhou University of Chinese Medicine , Guangzhou , Guangdong , China
| | - Yuan Wang
- g Department of Pharmacy , College of Pharmacy, Hubei University of Medicine , Shiyan , Hubei , China
| | - Song Gao
- c Department of Pharmacological and Pharmaceutical Sciences , College of Pharmacy, University of Houston , Houston , TX , USA.,g Department of Pharmacy , College of Pharmacy, Hubei University of Medicine , Shiyan , Hubei , China
| | - Ming Hu
- c Department of Pharmacological and Pharmaceutical Sciences , College of Pharmacy, University of Houston , Houston , TX , USA.,g Department of Pharmacy , College of Pharmacy, Hubei University of Medicine , Shiyan , Hubei , China
| |
Collapse
|
19
|
Kawase A, Hashimoto R, Shibata M, Shimada H, Iwaki M. Involvement of Reactive Metabolites of Diclofenac in Cytotoxicity in Sandwich-Cultured Rat Hepatocytes. Int J Toxicol 2017; 36:260-267. [DOI: 10.1177/1091581817700584] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background and Objectives: Diclofenac (DIC) is metabolized to reactive metabolites such as diclofenac acyl-β-d-glucuronide (DIC-AG). It is possible that such reactive metabolites could cause tissue damage by formation of covalent protein adducts and other modification of cellular proteins or by induction of immune responses against its covalent protein adducts. However, the detailed mechanisms of idiosyncratic drug-induced liver injury (DILI) have been unclear. The objective is to clarify the involvement of DIC-AG and 4′hydroxydiclofenac (4′OH-DIC) in acute DILI. Methods: We examined the effects of inhibiting DIC-AG and 4′OH-DIC production on covalent protein adduct formation and lactate dehydrogenase leakage using sandwich-cultured rat hepatocytes (SCRHs). Results: After pretreatment of SCRH with (−)-borneol (BOR, a uridine diphosphate (UDP)-glucuronosyltransferase inhibitor) or sulfaphenazole (SUL, a cytochrome P450 2C9 inhibitor) for 30 minutes, intracellular concentrations of DIC, DIC-AG, and 4′OH-DIC were determined after further treating cells with 300 μM DIC for 3 hours. The decreased levels of reactive metabolites caused by BOR or SUL pretreatment resulted in decreased lactate dehydrogenase leakage from SCRH, although the formation of covalent protein adducts was not affected. Conclusion: These results suggested that both DIC-AG and 4′OH-DIC may be involved in acute cytotoxicity by DIC.
Collapse
Affiliation(s)
- Atsushi Kawase
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Higashi-osaka, Osaka, Japan
| | - Ryota Hashimoto
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Higashi-osaka, Osaka, Japan
| | - Mai Shibata
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Higashi-osaka, Osaka, Japan
| | - Hiroaki Shimada
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Higashi-osaka, Osaka, Japan
| | - Masahiro Iwaki
- Department of Pharmacy, Faculty of Pharmacy, Kindai University, Higashi-osaka, Osaka, Japan
| |
Collapse
|
20
|
Ge S, Wei Y, Yin T, Xu B, Gao S, Hu M. Transport–Glucuronidation Classification System and PBPK Modeling: New Approach To Predict the Impact of Transporters on Disposition of Glucuronides. Mol Pharm 2017; 14:2884-2898. [DOI: 10.1021/acs.molpharmaceut.6b00941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shufan Ge
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| | - Yingjie Wei
- Key
Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, 100 Shizi Street, Nanjing 210028, China
| | - Taijun Yin
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| | - Beibei Xu
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| | - Song Gao
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| | - Ming Hu
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| |
Collapse
|
21
|
Wang L, Chen Q, Zhu L, Li Q, Zeng X, Lu L, Hu M, Wang X, Liu Z. Metabolic Disposition of Luteolin Is Mediated by the Interplay of UDP-Glucuronosyltransferases and Catechol-O-Methyltransferases in Rats. Drug Metab Dispos 2017; 45:306-315. [PMID: 28031430 PMCID: PMC6041822 DOI: 10.1124/dmd.116.073619] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/27/2016] [Indexed: 11/22/2022] Open
Abstract
Luteolin partially exerts its biologic effects via its metabolites catalyzed by UDP-glucuronosyltransferases (UGTs) and catechol-O-methyltransferases (COMTs). However, the interplay of UGTs and COMTs in mediating luteolin disposition has not been well clarified. In this study, we investigated the glucuronidation and methylation pathways of luteolin mediated by the interplay of UGTs and COMTs in vivo and in vitro. A total of nine luteolin metabolites was detected in rat plasma and bile by liquid chromatography-tandem mass spectrometry, namely, three glucuronides, two methylated metabolites, and four methylated glucuronides. Luteolin-3'-glucuronide (Lut-3'-G) exhibited the highest systemic exposure among these metabolites. Kinetics studies in rat liver S9 fractions suggested two pathways, as follows: 1) Luteolin was glucuronidated to luteolin-7-glucuronide, luteolin-4'-glucuronide, and Lut-3'-G by UGTs, and then Lut-7-G was methylated to chrysoeriol-7-glucuronide and diosmetin-7-glucuronide by COMTs. 2) Alternatively, luteolin was methylated to chrysoeriol and diosmetin by COMTs, and then chrysoeriol and diosmetin were glucuronidated by UGTs to their respective glucuronides. The methylation rate of luteolin was significantly increased by the absence of glucuronidation, whereas the glucuronidation rate was increased by the absence of methylation, but to a lesser extent. In conclusion, two pathways mediated by the interplay of UGTs and COMTs are probably involved in the metabolic disposition of luteolin. The glucuronidation and methylation of luteolin compensate for each other, although glucuronidation is the predominant pathway.
Collapse
Affiliation(s)
- Liping Wang
- First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, PR China (L.W., Q.C., X.Z., X.W.); International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (L.W., L.Z., L.L., M.H., Z.L.); Department of Pharmacy, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China (Q.L.); and College of Pharmacy, University of Houston, Houston, Texas (M.H.)
| | - Qingwei Chen
- First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, PR China (L.W., Q.C., X.Z., X.W.); International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (L.W., L.Z., L.L., M.H., Z.L.); Department of Pharmacy, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China (Q.L.); and College of Pharmacy, University of Houston, Houston, Texas (M.H.)
| | - Lijun Zhu
- First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, PR China (L.W., Q.C., X.Z., X.W.); International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (L.W., L.Z., L.L., M.H., Z.L.); Department of Pharmacy, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China (Q.L.); and College of Pharmacy, University of Houston, Houston, Texas (M.H.)
| | - Qiang Li
- First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, PR China (L.W., Q.C., X.Z., X.W.); International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (L.W., L.Z., L.L., M.H., Z.L.); Department of Pharmacy, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China (Q.L.); and College of Pharmacy, University of Houston, Houston, Texas (M.H.)
| | - Xuejun Zeng
- First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, PR China (L.W., Q.C., X.Z., X.W.); International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (L.W., L.Z., L.L., M.H., Z.L.); Department of Pharmacy, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China (Q.L.); and College of Pharmacy, University of Houston, Houston, Texas (M.H.)
| | - Linlin Lu
- First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, PR China (L.W., Q.C., X.Z., X.W.); International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (L.W., L.Z., L.L., M.H., Z.L.); Department of Pharmacy, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China (Q.L.); and College of Pharmacy, University of Houston, Houston, Texas (M.H.)
| | - Ming Hu
- First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, PR China (L.W., Q.C., X.Z., X.W.); International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (L.W., L.Z., L.L., M.H., Z.L.); Department of Pharmacy, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China (Q.L.); and College of Pharmacy, University of Houston, Houston, Texas (M.H.)
| | - Xinchun Wang
- First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, PR China (L.W., Q.C., X.Z., X.W.); International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (L.W., L.Z., L.L., M.H., Z.L.); Department of Pharmacy, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China (Q.L.); and College of Pharmacy, University of Houston, Houston, Texas (M.H.)
| | - Zhongqiu Liu
- First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, PR China (L.W., Q.C., X.Z., X.W.); International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (L.W., L.Z., L.L., M.H., Z.L.); Department of Pharmacy, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China (Q.L.); and College of Pharmacy, University of Houston, Houston, Texas (M.H.)
| |
Collapse
|
22
|
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]
|
23
|
Daly AK. Are Polymorphisms in Genes Relevant to Drug Disposition Predictors of Susceptibility to Drug-Induced Liver Injury? Pharm Res 2016; 34:1564-1569. [PMID: 28028769 PMCID: PMC5498650 DOI: 10.1007/s11095-016-2091-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/20/2016] [Indexed: 01/06/2023]
Abstract
Despite considerable progress in identifying specific HLA alleles as genetic risk factors for some forms of drug-induced liver injury, progress in understanding whether genetic polymorphisms relevant to drug disposition also contribute to risk for developing this serious toxicity has been more limited. Evidence from both candidate-gene case control studies and genome-wide association studies is now discussed. In the case of genes relevant to drug metabolism, polymorphisms in cytochromes P450, UDP-glucuronosyltransferases, N-acetyltransferases and glutathione S-transferases as risk factors for DILI are assessed. The relevance of ABC transporters to drug-induced liver injury is also considered, together with data showing associations of particular ABCB11, ABCB1 and ABCC2 polymorphisms with some forms of drug-induced liver injury. Very few of the associations with genes relevant to drug disposition that have been reported have been well replicated. Even apparently well-studied associations such as that between isoniazid liver injury and N-acetyltransferase 2 slow acetylators remain problematic, though it seems likely that polymorphisms in drug metabolism genes do contribute to risk for some specific drugs. A better understanding of genetic risk factors for drug-induced liver injury will require further genome-wide association studies with larger numbers of cases, especially for forms of drug-induced liver injury where HLA genotype does not appear to be a risk factor.
Collapse
Affiliation(s)
- Ann K Daly
- Institute of Cellular Medicine, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
24
|
Ge S, Tu Y, Hu M. Challenges and Opportunities with Predicting in Vivo Phase II Metabolism via Glucuronidation from in Vitro Data. ACTA ACUST UNITED AC 2016; 2:326-338. [PMID: 28966903 DOI: 10.1007/s40495-016-0076-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Glucuronidation is the most important phase II metabolic pathway which is responsible for the clearance of many endogenous and exogenous compounds. To better understand the elimination process for compounds undergoing glucuronidation and identify compounds with desirable in vivo pharmacokinetic properties, many efforts have been made to predict in vivo glucuronidation using in vitro data. In this article, we reviewed typical approaches used in previous predictions. The problems and challenges in prediction of glucuronidation were discussed. Besides that different incubation conditions can affect the prediction accuracy, other factors including efflux / uptake transporters, enterohepatic recycling, and deglucuronidation reactions also contribute to the disposition of glucuronides and make the prediction more difficult. PBPK modeling, which can describe more complicated process in vivo, is a promising prediction strategy which may greatly improve the prediction of glucuronidation and potential DDIs involving glucuronidation. Based on previous studies, we proposed a transport-glucuronidation classification system, which was built based on the kinetics of both glucuronidation and transport of the glucuronide. This system could be a very useful tool to achieve better in vivo predictions.
Collapse
Affiliation(s)
- Shufan Ge
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, TX, 77030, USA
| | - Yifan Tu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, TX, 77030, USA
| | - Ming Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, TX, 77030, USA
| |
Collapse
|
25
|
Zhang Y, Han YH, Putluru SP, Matta MK, Kole P, Mandlekar S, Furlong MT, Liu T, Iyer RA, Marathe P, Yang Z, Lai Y, Rodrigues AD. Diclofenac and Its Acyl Glucuronide: Determination of In Vivo Exposure in Human Subjects and Characterization as Human Drug Transporter Substrates In Vitro. Drug Metab Dispos 2016; 44:320-8. [PMID: 26714763 DOI: 10.1124/dmd.115.066944] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/28/2015] [Indexed: 01/05/2023] Open
Abstract
Although the metabolism and disposition of diclofenac (DF) has been studied extensively, information regarding the plasma levels of its acyl-β-d-glucuronide (DF-AG), a major metabolite, in human subjects is limited. Therefore, DF-AG concentrations were determined in plasma (acidified blood derived) of six healthy volunteers following a single oral DF dose (50 mg). Levels of DF-AG in plasma were high, as reflected by a DF-AG/DF ratio of 0.62 ± 0.21 (Cmax mean ± S.D.) and 0.84 ± 0.21 (area under the concentration-time curve mean ± S.D.). Both DF and DF-AG were also studied as substrates of different human drug transporters in vitro. DF was identified as a substrate of organic anion transporter (OAT) 2 only (Km = 46.8 µM). In contrast, DF-AG was identified as a substrate of numerous OATs (Km = 8.6, 60.2, 103.9, and 112 µM for OAT2, OAT1, OAT4, and OAT3, respectively), two organic anion-transporting polypeptides (OATP1B1, Km = 34 µM; OATP2B1, Km = 105 µM), breast cancer resistance protein (Km = 152 µM), and two multidrug resistance proteins (MRP2, Km = 145 µM; MRP3, Km = 196 µM). It is concluded that the disposition of DF-AG, once formed, can be mediated by various candidate transporters known to be expressed in the kidney (basolateral, OAT1, OAT2, and OAT3; apical, MRP2, BCRP, and OAT4) and liver (canalicular, MRP2 and BCRP; basolateral, OATP1B1, OATP2B1, OAT2, and MRP3). DF-AG is unstable in plasma and undergoes conversion to parent DF. Therefore, caution is warranted when assessing renal and hepatic transporter-mediated drug-drug interactions with DF and DF-AG.
Collapse
Affiliation(s)
- Yueping Zhang
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Yong-Hae Han
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Siva Prasad Putluru
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Murali Krishna Matta
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Prashant Kole
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Sandhya Mandlekar
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Michael T Furlong
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Tongtong Liu
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Ramaswamy A Iyer
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Punit Marathe
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Zheng Yang
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Yurong Lai
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - A David Rodrigues
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| |
Collapse
|
26
|
Iwamura A, Ito M, Mitsui H, Hasegawa J, Kosaka K, Kino I, Tsuda M, Nakajima M, Yokoi T, Kume T. Toxicological evaluation of acyl glucuronides utilizing half-lives, peptide adducts, and immunostimulation assays. Toxicol In Vitro 2015; 30:241-9. [DOI: 10.1016/j.tiv.2015.10.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 10/06/2015] [Accepted: 10/30/2015] [Indexed: 11/28/2022]
|
27
|
Niu X, de Graaf IAM, van de Vegte D, Langelaar-Makkinje M, Sekine S, Groothuis GMM. Consequences of Mrp2 deficiency for diclofenac toxicity in the rat intestine ex vivo. Toxicol In Vitro 2015; 29:168-75. [PMID: 25450747 DOI: 10.1016/j.tiv.2014.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 10/01/2014] [Accepted: 10/06/2014] [Indexed: 02/07/2023]
Abstract
The non-steroidal anti-inflammatory drug diclofenac (DCF) has a high prevalence of intestinal side effects in humans and rats. It has been reported that Mrp2 transporter deficient rats (Mrp2) are more resistant to DCF induced intestinal toxicity. This was explained in vivo by impaired Mrp2-dependent biliary transport of DCF-acylglucuronide (DAG), leading to decreased intestinal exposure to DAG and DCF. However, it is not known to what extent adaptive changes in the Mrp2 intestine itself influence its sensitivity to DCF toxicity without the influence of liver metabolites. To investigate this, DCF toxicity and disposition were studied ex vivo by precision-cut intestinal slices and Ussing chamber using intestines from wild type(WT) and Mrp2 rats. The results show that adaptive changes due to Mrp2 deficiency concerning Mrp2, Mrp3 and BCRP gene expression, GSH content and DAG formation were different between liver and intestine. Furthermore, Mrp2 intestine was intrinsically more resistant to DCF toxicity than its WT counterpart ex vivo. This can at least partly be explained by a reduced DCF uptake by the Mrp2 intestine, but isnot related to the other adaptive changes in the intestine. The extrapolation of this data to humans with MRP2 deficiency is uncertain due to species differences in activity and regulation of transporters.
Collapse
Affiliation(s)
- Xiaoyu Niu
- Division of Pharmacokinetics, Toxicology and Targeting, Department of Pharmacy, University of Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
28
|
Scialis RJ, Csanaky IL, Goedken MJ, Manautou JE. Multidrug Resistance-Associated Protein 3 Plays an Important Role in Protection against Acute Toxicity of Diclofenac. Drug Metab Dispos 2015; 43:944-950. [PMID: 25897176 PMCID: PMC4468432 DOI: 10.1124/dmd.114.061705] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 04/20/2015] [Indexed: 11/22/2022] Open
Abstract
Diclofenac (DCF) is a nonsteroidal anti-inflammatory drug commonly prescribed to reduce pain in acute and chronic inflammatory diseases. One of the main DCF metabolites is a reactive diclofenac acyl glucuronide (DCF-AG) that covalently binds to biologic targets and may contribute to adverse drug reactions arising from DCF use. Cellular efflux of DCF-AG is partially mediated by multidrug resistance-associated proteins (Mrp). The importance of Mrp2 during DCF-induced toxicity has been established, yet the role of Mrp3 remains largely unexplored. In the present work, Mrp3-null (KO) mice were used to study the toxicokinetics and toxicodynamics of DCF and its metabolites. DCF-AG plasma concentrations were 90% lower in KO mice than in wild-type (WT) mice, indicating that Mrp3 mediates DCF-AG basolateral efflux. In contrast, there were no differences in DCF-AG biliary excretion between WT and KO, suggesting that only DCF-AG basolateral efflux is compromised by Mrp3 deletion. Susceptibility to toxicity was also evaluated after a single high DCF dose. No signs of injury were detected in livers and kidneys; however, ulcers were found in the small intestines. Furthermore, the observed intestinal injuries were consistently more severe in KO compared with WT. DCF covalent adducts were observed in liver and small intestines; however, staining intensity did not correlate with the severity of injuries, implying that tissues respond differently to covalent modification. Overall, the data provide strong evidence that (1) in vivo Mrp3 plays an important role in DCF-AG disposition and (2) compromised Mrp3 function can enhance injury in the gastrointestinal tract after DCF treatment.
Collapse
Affiliation(s)
- Renato J Scialis
- University of Connecticut, School of Pharmacy, Department of Pharmaceutical Sciences, Storrs, Connecticut (R.J.S., J.E.M.); University of Kansas Medical Center, Department of Internal Medicine, Kansas City, Kansas (I.L.C.); and Office of Translational Science, Rutgers University, Piscataway, New Jersey (M.J.G.)
| | - Iván L Csanaky
- University of Connecticut, School of Pharmacy, Department of Pharmaceutical Sciences, Storrs, Connecticut (R.J.S., J.E.M.); University of Kansas Medical Center, Department of Internal Medicine, Kansas City, Kansas (I.L.C.); and Office of Translational Science, Rutgers University, Piscataway, New Jersey (M.J.G.)
| | - Michael J Goedken
- University of Connecticut, School of Pharmacy, Department of Pharmaceutical Sciences, Storrs, Connecticut (R.J.S., J.E.M.); University of Kansas Medical Center, Department of Internal Medicine, Kansas City, Kansas (I.L.C.); and Office of Translational Science, Rutgers University, Piscataway, New Jersey (M.J.G.)
| | - José E Manautou
- University of Connecticut, School of Pharmacy, Department of Pharmaceutical Sciences, Storrs, Connecticut (R.J.S., J.E.M.); University of Kansas Medical Center, Department of Internal Medicine, Kansas City, Kansas (I.L.C.); and Office of Translational Science, Rutgers University, Piscataway, New Jersey (M.J.G.)
| |
Collapse
|
29
|
Sharma R, Litchfield J, Bergman A, Atkinson K, Kazierad D, Gustavson SM, Di L, Pfefferkorn JA, Kalgutkar AS. Comparison of the circulating metabolite profile of PF-04991532, a hepatoselective glucokinase activator, across preclinical species and humans: potential implications in metabolites in safety testing assessment. Drug Metab Dispos 2015; 43:190-8. [PMID: 25384899 DOI: 10.1124/dmd.114.061218] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A previous report from our laboratory disclosed the identification of PF-04991532 [(S)-6-(3-cyclopentyl-2-(4-trifluoromethyl)-1H-imidazol-1-yl)propanamido)nicotinic acid] as a hepatoselective glucokinase activator for the treatment of type 2 diabetes mellitus. Lack of in vitro metabolic turnover in microsomes and hepatocytes from preclinical species and humans suggested that metabolism would be inconsequential as a clearance mechanism of PF-04991532 in vivo. Qualitative examination of human circulating metabolites using plasma samples from a 14-day multiple ascending dose clinical study, however, revealed a glucuronide (M1) and monohydroxylation products (M2a and M2b/M2c) whose abundances (based on UV integration) were greater than 10% of the total drug-related material. Based on this preliminary observation, mass balance/excretion studies were triggered in animals, which revealed that the majority of circulating radioactivity following the oral administration of [¹⁴C]PF-04991532 was attributed to an unchanged parent (>70% in rats and dogs). In contrast with the human circulatory metabolite profile, the monohydroxylated metabolites were not detected in circulation in either rats or dogs. Available mass spectral evidence suggested that M2a and M2b/M2c were diastereomers derived from cyclopentyl ring oxidation in PF-04991532. Because cyclopentyl ring hydroxylation on the C-2 and C-3 positions can generate eight possible diastereomers, it was possible that additional diastereomers may have also formed and would need to be resolved from the M2a and M2b/M2c peaks observed in the current chromatography conditions. In conclusion, the human metabolite scouting study in tandem with the animal mass balance study allowed early identification of PF-04991532 oxidative metabolites, which were not predicted by in vitro methods and may require additional scrutiny in the development phase of PF-04991532.
Collapse
Affiliation(s)
- Raman Sharma
- Pfizer Inc., Groton, Connecticut (R.S., A.B., K.A., S.M.G., L.D.); and Pfizer Inc., Cambridge, Massachusetts (J.L., D.K., J.A.P., A.S.K.)
| | - John Litchfield
- Pfizer Inc., Groton, Connecticut (R.S., A.B., K.A., S.M.G., L.D.); and Pfizer Inc., Cambridge, Massachusetts (J.L., D.K., J.A.P., A.S.K.)
| | - Arthur Bergman
- Pfizer Inc., Groton, Connecticut (R.S., A.B., K.A., S.M.G., L.D.); and Pfizer Inc., Cambridge, Massachusetts (J.L., D.K., J.A.P., A.S.K.)
| | - Karen Atkinson
- Pfizer Inc., Groton, Connecticut (R.S., A.B., K.A., S.M.G., L.D.); and Pfizer Inc., Cambridge, Massachusetts (J.L., D.K., J.A.P., A.S.K.)
| | - David Kazierad
- Pfizer Inc., Groton, Connecticut (R.S., A.B., K.A., S.M.G., L.D.); and Pfizer Inc., Cambridge, Massachusetts (J.L., D.K., J.A.P., A.S.K.)
| | - Stephanie M Gustavson
- Pfizer Inc., Groton, Connecticut (R.S., A.B., K.A., S.M.G., L.D.); and Pfizer Inc., Cambridge, Massachusetts (J.L., D.K., J.A.P., A.S.K.)
| | - Li Di
- Pfizer Inc., Groton, Connecticut (R.S., A.B., K.A., S.M.G., L.D.); and Pfizer Inc., Cambridge, Massachusetts (J.L., D.K., J.A.P., A.S.K.)
| | - Jeffrey A Pfefferkorn
- Pfizer Inc., Groton, Connecticut (R.S., A.B., K.A., S.M.G., L.D.); and Pfizer Inc., Cambridge, Massachusetts (J.L., D.K., J.A.P., A.S.K.)
| | - Amit S Kalgutkar
- Pfizer Inc., Groton, Connecticut (R.S., A.B., K.A., S.M.G., L.D.); and Pfizer Inc., Cambridge, Massachusetts (J.L., D.K., J.A.P., A.S.K.)
| |
Collapse
|
30
|
Breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (P-GP/ABCB1) restrict oral availability and brain accumulation of the PARP inhibitor rucaparib (AG-014699). Pharm Res 2014; 32:37-46. [PMID: 24962512 DOI: 10.1007/s11095-014-1442-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 06/10/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND Rucaparib is a potent, orally available, small-molecule inhibitor of poly ADP-ribose polymerase (PARP) 1 and 2. Ongoing clinical trials are assessing the efficacy of rucaparib alone or in combination with other cytotoxic drugs, mainly in breast and ovarian cancer patients with mutations in the breast cancer associated (BRCA) genes. PURPOSE We aimed to establish whether the multidrug efflux transporters ABCG2 (BCRP) and ABCB1 (P-gp, MDR1) affect the oral availability and brain penetration of rucaparib in mice. RESULTS In vitro, rucaparib was efficiently transported by both human ABCB1 and ABCG2, and very efficiently by mouse Abcg2. Transport could be inhibited by the small-molecule ABCB1 and ABCG2 inhibitors zosuquidar and Ko143, respectively. In vivo, oral availability (plasma AUC0-1 and AUC0-24) and brain levels of rucaparib at 1 and 24 h were increased by the absence of both Abcg2 and Abcb1a/1b after oral administration of rucaparib at 10 mg/kg. CONCLUSIONS Our data show to our knowledge for the first time that oral availability and brain accumulation of a PARP inhibitor are markedly and additively restricted by Abcg2 and Abcb1a/1b. This may have clinical relevance for improvement of rucaparib therapy in PARP inhibitor-resistant tumors with ABCB1 and/or ABCG2 expression and in patients with brain (micro)metastases positioned behind a functional blood-brain barrier.
Collapse
|
31
|
Bateman TJ, Reddy VGB, Kakuni M, Morikawa Y, Kumar S. Application of chimeric mice with humanized liver for study of human-specific drug metabolism. Drug Metab Dispos 2014; 42:1055-65. [PMID: 24700822 DOI: 10.1124/dmd.114.056978] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Human-specific or disproportionately abundant human metabolites of drug candidates that are not adequately formed and qualified in preclinical safety assessment species pose an important drug development challenge. Furthermore, the overall metabolic profile of drug candidates in humans is an important determinant of their drug-drug interaction susceptibility. These risks can be effectively assessed and/or mitigated if human metabolic profile of the drug candidate could reliably be determined in early development. However, currently available in vitro human models (e.g., liver microsomes, hepatocytes) are often inadequate in this regard. Furthermore, the conduct of definitive radiolabeled human ADME studies is an expensive and time-consuming endeavor that is more suited for later in development when the risk of failure has been reduced. We evaluated a recently developed chimeric mouse model with humanized liver on uPA/SCID background for its ability to predict human disposition of four model drugs (lamotrigine, diclofenac, MRK-A, and propafenone) that are known to exhibit human-specific metabolism. The results from these studies demonstrate that chimeric mice were able to reproduce the human-specific metabolite profile for lamotrigine, diclofenac, and MRK-A. In the case of propafenone, however, the human-specific metabolism was not detected as a predominant pathway, and the metabolite profiles in native and humanized mice were similar; this was attributed to the presence of residual highly active propafenone-metabolizing mouse enzymes in chimeric mice. Overall, the data indicate that the chimeric mice with humanized liver have the potential to be a useful tool for the prediction of human-specific metabolism of xenobiotics and warrant further investigation.
Collapse
Affiliation(s)
- Thomas J Bateman
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck Research Laboratories, Rahway, New Jersey (T.J.B, V.G.B.R., S.K.); and PhoenixBio Corporation Limited, Higashi-Hiroshima, Japan (M.K., Y.M.)
| | | | | | | | | |
Collapse
|
32
|
Structure and function of BCRP, a broad specificity transporter of xenobiotics and endobiotics. Arch Toxicol 2014; 88:1205-48. [PMID: 24777822 DOI: 10.1007/s00204-014-1224-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/06/2014] [Indexed: 12/20/2022]
|
33
|
Vredenburg G, Elias NS, Venkataraman H, Hendriks DFG, Vermeulen NPE, Commandeur JNM, Vos JC. Human NAD(P)H:quinone Oxidoreductase 1 (NQO1)-Mediated Inactivation of Reactive Quinoneimine Metabolites of Diclofenac and Mefenamic Acid. Chem Res Toxicol 2014; 27:576-86. [DOI: 10.1021/tx400431k] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Galvin Vredenburg
- Division
of Molecular Toxicology,
Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Faculty
of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Naura S. Elias
- Division
of Molecular Toxicology,
Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Faculty
of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Harini Venkataraman
- Division
of Molecular Toxicology,
Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Faculty
of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Delilah F. G. Hendriks
- Division
of Molecular Toxicology,
Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Faculty
of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Nico P. E. Vermeulen
- Division
of Molecular Toxicology,
Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Faculty
of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Jan N. M. Commandeur
- Division
of Molecular Toxicology,
Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Faculty
of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - J. Chris Vos
- Division
of Molecular Toxicology,
Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Faculty
of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
34
|
HPLC–MS Profiling and Structural Identification of [14C]-Diclofenac Metabolites in Mouse Bile. Chromatographia 2013. [DOI: 10.1007/s10337-013-2594-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
35
|
Ramm S, Mally A. Role of drug-independent stress factors in liver injury associated with diclofenac intake. Toxicology 2013; 312:83-96. [PMID: 23939143 DOI: 10.1016/j.tox.2013.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 07/26/2013] [Accepted: 08/01/2013] [Indexed: 01/08/2023]
Abstract
Although a basic understanding of the chemical and biological events leading to idiosyncratic drug toxicity is still lacking, it appears that drug-independent risk factors that increase reactive metabolite formation or alter cellular stress and immune response may be critical determinants in the response to an otherwise non-toxic drug. Thus, we were interested to determine the impact of various drug-independent stress factors - lipopolysaccharide (LPS), poly I:C (PIC) or glutathione depletion via buthionine sulfoximine (BSO) - on the toxicity of diclofenac (Dcl), a model drug associated with rare but significant cases of serious hepatotoxicity, and to understand if enhanced toxicity occurs through alterations of drug metabolism and/or modulation of stress response pathways. Co-treatment of rats repeatedly given therapeutic doses of Dcl for 7 days with a single dose of LPS 2h before the last Dcl dose resulted in severe liver toxicity. Neither LPS nor diclofenac alone or in combination with PIC or BSO had such an effect. While it is thought that bioactivation to reactive Dcl acyl glucuronides (AG) and subsequent protein adduct formation contribute to Dcl induced liver injury, LC-MS/MS analyses did not reveal increased formation of 4'- and 5-hydroxy-Dcl, Dcl-AG or Dcl-AG dependent protein adducts in animals treated with LPS/Dcl. Hepatic gene expression analysis suggested enhanced activation of NFκB and MAPK pathways and up-regulation of co-stimulatory molecules (IL-1β, TNF-α, CINC-1) by LPS/Dcl and PIC/Dcl, while protective factors (HSPs, SOD2) were down-regulated. LPS/Dcl led to extensive release of pro-inflammatory cytokines (IL-1β, IL-6, IFN-γ, TNF-α) and factors thought to constitute danger signals (HMGB1, CINC-1) into plasma. Taken together, our results show that Dcl enhanced the inflammatory response induced by LPS - and to a lesser extent by PIC - through up-regulation of pro-inflammatory molecules and down-regulation of protective factors. This suggests sensitization of cells to cellular stress mediated by non-drug-related risk factors by therapeutic doses of Dcl, rather than potentiation of Dcl toxicity by the stress factors.
Collapse
Affiliation(s)
- Susanne Ramm
- Department of Toxicology, University of Würzburg, Germany
| | | |
Collapse
|
36
|
Pharmacogenetics of chronic pain and its treatment. Mediators Inflamm 2013; 2013:864319. [PMID: 23766564 PMCID: PMC3671679 DOI: 10.1155/2013/864319] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 04/12/2013] [Indexed: 12/20/2022] Open
Abstract
This paper reviews the impact of genetic variability of drug metabolizing enzymes, transporters, receptors, and pathways involved in chronic pain perception on the efficacy and safety of analgesics and other drugs used for chronic pain treatment. Several candidate genes have been identified in the literature, while there is usually only limited clinical evidence substantiating for the penetration of the testing for these candidate biomarkers into the clinical practice. Further, the pain-perception regulation and modulation are still not fully understood, and thus more complex knowledge of genetic and epigenetic background for analgesia will be needed prior to the clinical use of the candidate genetic biomarkers.
Collapse
|
37
|
More VR, Cheng Q, Donepudi AC, Buckley DB, Lu ZJ, Cherrington NJ, Slitt AL. Alcohol cirrhosis alters nuclear receptor and drug transporter expression in human liver. Drug Metab Dispos 2013; 41:1148-55. [PMID: 23462698 PMCID: PMC3629807 DOI: 10.1124/dmd.112.049676] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 03/05/2013] [Indexed: 12/15/2022] Open
Abstract
Unsafe use of alcohol results in approximately 2.5 million deaths worldwide, with cirrhosis contributing to 16.6% of reported deaths. Serum insulin levels are often elevated in alcoholism and may result in diabetes, which is why alcoholic liver disease and diabetes often are present together. Because there is a sizable population with these diseases alone or in combination, the purpose of this study was to determine whether transporter expression in human liver is affected by alcoholic cirrhosis, diabetes, and alcoholic cirrhosis coexisting with diabetes. Transporters aid in hepatobiliary excretion of many drugs and toxic chemicals and can be determinants of drug-induced liver injury. Drug transporter expression and transcription factor-relative mRNA and protein expression in normal, diabetic, cirrhotic, and cirrhosis with diabetes human livers were quantified. Cirrhosis significantly increased ABCC4, 5, ABCG2, and solute carrier organic anion (SLCO) 2B1 mRNA expression and decreased SLCO1B3 mRNA expression in the liver. ABCC1, 3-5, and ABCG2 protein expression was also upregulated by alcoholic cirrhosis. ABCC3-5 and ABCG2 protein expression was also upregulated in diabetic cirrhosis. Cirrhosis increased nuclear factor E2-related factor 2 mRNA expression, whereas it decreased pregnane-X-receptor and farnesoid-X-receptor mRNA expression in comparison with normal livers. Hierarchical cluster analysis indicated that expressions of ABCC2, 3, and 6; SLCO1B1 and 1B3; and ABCC4 and 5 were more closely related in the livers from this cohort. Overall, alcoholic cirrhosis altered transporter expression in human liver.
Collapse
Affiliation(s)
- Vijay R More
- Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Tan JR, Chakravarthi S, Judson JP, Haleagrahara N, Segarra I. Potential protective effect of sunitinib after administration of diclofenac: biochemical and histopathological drug-drug interaction assessment in a mouse model. Naunyn Schmiedebergs Arch Pharmacol 2013; 386:619-33. [PMID: 23552887 DOI: 10.1007/s00210-013-0861-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 03/22/2013] [Indexed: 01/27/2023]
Abstract
Sunitinib is a tyrosine kinase inhibitor for GIST and advanced renal cell carcinoma. Diclofenac is used in cancer pain management. Coadministration may mediate P450 toxicity. We evaluate their interaction, assessing biomarkers ALT, AST, BUN, creatinine, and histopathological changes in the liver, kidney, heart, brain, and spleen. ICR mice (male, n = 6 per group/dose) were administered saline (group A) or 30 mg/kg diclofenac ip (group B), or sunitinib po at 25, 50, 80, 100, 140 mg/kg (group C) or combination of diclofenac (30 mg/kg, ip) and sunitinib (25, 50, 80, 100, 140 mg/kg po). Diclofenac was administered 15 min before sunitinib, mice were euthanized 4 h post-sunitinib dose, and biomarkers and tissue histopathology were assessed. AST was 92.2 ± 8.0 U/L in group A and 159.7 ± 14.6 U/L in group B (p < 0.05); in group C, it the range was 105.1-152.6 U/L, and in group D, it was 156.0-209.5 U/L (p < 0.05). ALT was 48.9 ± 1.6 U/L (group A), 95.1 ± 4.5 U/L (p < 0.05) in group B, and 50.5-77.5 U/L in group C and 82.3-115.6 U/L after coadministration (p < 0.05). Renal function biomarker BUN was 16.3 ± 0.6 mg/dl (group A) and increased to 29.9 ± 2.6 mg/dl in group B (p < 0.05) and it the range was 19.1-33.3 mg/dl (p < 0.05) and 26.9-40.8 mg/dl in groups C and D, respectively. Creatinine was 5.9 pmol/ml in group A; 6.2 pmol/ml in group B (p < 0.01), and the range was 6.0-6.2 and 6.2-6.4 pmol/ml in groups C and D, respectively (p < 0.05 for D). Histopathological assessment (vascular and inflammation damages) showed toxicity in group B (p < 0.05) and mild toxicity in group C. Damage was significantly lesser in group D than group B (p < 0.05). Spleen only showed toxicity after coadministration. These results suggest vascular and inflammation protective effects of sunitinib, not shown after biomarker analysis.
Collapse
Affiliation(s)
- Jian Ren Tan
- Department of Human Biology, School of Medicine, International Medical University, Jalan 19/155B, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | | | | | | | | |
Collapse
|
39
|
Zamek-Gliszczynski MJ, Abraham TL, Alberts JJ, Kulanthaivel P, Jackson KA, Chow KH, McCann DJ, Hu H, Anderson S, Furr NA, Barbuch RJ, Cassidy KC. Pharmacokinetics, metabolism, and excretion of the glycogen synthase kinase-3 inhibitor LY2090314 in rats, dogs, and humans: a case study in rapid clearance by extensive metabolism with low circulating metabolite exposure. Drug Metab Dispos 2013; 41:714-26. [PMID: 23305709 DOI: 10.1124/dmd.112.048488] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
LY2090314 (3-[9-fluoro-2-(piperidin-1-ylcarbonyl)-1,2,3,4-tetrahydro[1,4]diazepino[6,7,1-hi]indol-7-yl]-4-imidazo[1,2-a]pyridin-3-yl-1H-pyrrole-2,5-dione) is an intravenous glycogen synthase kinase-3 inhibitor in oncology trials. Drug disposition was characterized after intravenous infusion of [(14)C]LY2090314 to rats and dogs, and was related to available clinical data. LY2090314 exhibited high clearance (approximating hepatic blood flow) and a moderate volume of distribution (∼1-2 l/kg) resulting in rapid elimination (half-life ∼0.4, 0.7, and 1.8-3.4 hours in rats, dogs, and humans, respectively). Scaled clearance from liver microsomes accurately predicted perfusion-limited clearance across species. LY2090314 was cleared by extensive metabolism, and the numerous metabolites were rapidly excreted into feces via bile (69-97% of dose; 62-93% within 0-24 hours); urinary recovery of drug-related material was low (≤3% of dose). Despite extensive metabolism, in rats and humans the parent compound was the sole identifiable drug-related moiety in plasma. Even in Mdr1a-, Bcrp-, and Mrp2-knockout rats, LY2090314 metabolites did not appear in circulation, and their urinary excretion was not enhanced, because the hypothesized impaired biliary excretion of metabolites in the absence of these canalicular transporters was not observed. Canine metabolite disposition was generally similar, with the notable exception of dog-unique LY2090314 glucuronide. This conjugate was formed in the dog liver and was preferentially excreted into the blood, where it accounted for the majority of circulating radioactivity at later times, and was predominantly recovered in urine (16% of dose). In conclusion, LY2090314 was rapidly cleared by extensive metabolism with negligible circulating metabolite exposures due to biliary excretion of metabolites into feces with no apparent intestinal reabsorption.
Collapse
|
40
|
Stachulski AV, Baillie TA, Kevin Park B, Scott Obach R, Dalvie DK, Williams DP, Srivastava A, Regan SL, Antoine DJ, Goldring CEP, Chia AJL, Kitteringham NR, Randle LE, Callan H, Castrejon JL, Farrell J, Naisbitt DJ, Lennard MS. The Generation, Detection, and Effects of Reactive Drug Metabolites. Med Res Rev 2012; 33:985-1080. [DOI: 10.1002/med.21273] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Andrew V. Stachulski
- Department of Chemistry, Robert Robinson Laboratories; University of Liverpool; Liverpool; L69 7ZD; UK
| | - Thomas A. Baillie
- School of Pharmacy; University of Washington; Box 357631; Seattle; Washington; 98195-7631
| | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - R. Scott Obach
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; Groton; Connecticut 06340
| | - Deepak K. Dalvie
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; La Jolla; California 94121
| | - Dominic P. Williams
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Abhishek Srivastava
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Sophie L. Regan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Daniel J. Antoine
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Christopher E. P. Goldring
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Alvin J. L. Chia
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Neil R. Kitteringham
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Laura E. Randle
- School of Pharmacy and Biomolecular Sciences, Faculty of Science; Liverpool John Moores University; James Parsons Building, Byrom Street; Liverpool L3 3AF; UK
| | - Hayley Callan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - J. Luis Castrejon
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - John Farrell
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Martin S. Lennard
- Academic Unit of Medical Education; University of Sheffield; 85 Wilkinson Street; Sheffield S10 2GJ; UK
| |
Collapse
|
41
|
Boelsterli UA, Redinbo MR, Saitta KS. Multiple NSAID-induced hits injure the small intestine: underlying mechanisms and novel strategies. Toxicol Sci 2012; 131:654-67. [PMID: 23091168 DOI: 10.1093/toxsci/kfs310] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) can cause serious gastrointestinal (GI) injury including jejunal/ileal mucosal ulceration, bleeding, and even perforation in susceptible patients. The underlying mechanisms are largely unknown, but they are distinct from those related to gastric injury. Based on recent insights from experimental models, including genetics and pharmacology in rodents typically exposed to diclofenac, indomethacin, or naproxen, we propose a multiple-hit pathogenesis of NSAID enteropathy. The multiple hits start with an initial pharmacokinetic determinant caused by vectorial hepatobiliary excretion and delivery of glucuronidated NSAID or oxidative metabolite conjugates to the distal small intestinal lumen, where bacterial β-glucuronidase produces critical aglycones. The released aglycones are then taken up by enterocytes and further metabolized by intestinal cytochrome P450s to potentially reactive intermediates. The "first hit" is caused by the NSAID and/or oxidative metabolites that induce severe endoplasmic reticulum stress or mitochondrial stress and lead to cell death. The "second hit" is created by the significant subsequent inflammatory response that would follow such a first-hit injury. Based on these putative mechanisms, strategies have been developed to protect the enterocytes from being exposed to the parent NSAID and/or oxidative metabolites. Among these, a novel strategy already demonstrated in a murine model is the selective disruption of bacteria-specific β-glucuronidases with a novel small molecule inhibitor that does not harm the bacteria and that alleviates NSAID-induced enteropathy. Such mechanism-based strategies require further investigation but provide potential avenues for the alleviation of the GI toxicity caused by multiple NSAID hits.
Collapse
Affiliation(s)
- Urs A Boelsterli
- Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, Connecticut 06269, USA.
| | | | | |
Collapse
|
42
|
Darnell M, Ulvestad M, Ellis E, Weidolf L, Andersson TB. In vitro evaluation of major in vivo drug metabolic pathways using primary human hepatocytes and HepaRG cells in suspension and a dynamic three-dimensional bioreactor system. J Pharmacol Exp Ther 2012; 343:134-44. [PMID: 22776955 DOI: 10.1124/jpet.112.195834] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Major human specific metabolites, not detected during in vivo and in vitro preclinical studies, may cause unexpected drug interactions and toxicity in human and delays in clinical programs. Thus, reliable preclinical tools for the detection of major human metabolites are of high importance. The aim of this study was to compare major drug metabolic pathways in HepaRG cells, a human hepatoma cell line, to fresh human hepatocytes, cryopreserved human hepatocytes, and human in vivo data. Furthermore, the maintenance of cytochrome P450 (P450) and UDP-glucuronosyltransferase (UGT) activities in a dynamic three-dimensional (3D) bioreactor were evaluated over time by using HepaRG cells and human hepatocytes. (14)C-diclofenac and a candidate from AstraZeneca's drug development program, (14)C-AZD6610, which are metabolized by P450 and UGT in vivo, were used as model substrates. The proportion of relevant biotransformation pathways of the investigated drug was clearly different in the various cell systems. The hydroxylation route was favored in primary human hepatocytes, whereas the glucuronidation route was favored in HepaRG cells. The human in vivo metabolite profile of AZD6610 was best represented by human hepatocytes, whereas all major diclofenac metabolites were detected in HepaRG cells. Moreover, the metabolite profiles in cryopreserved and fresh human hepatocytes were essentially the same. The liver bioreactor using both fresh human hepatocytes and HepaRG cells retained biotransformation capacity over 1 week. Thus, the incubation time can be increased from a few hours in suspension to several days in 3D cultures, which opens up for detection of metabolites from slowly metabolized drugs.
Collapse
Affiliation(s)
- Malin Darnell
- DMPK Innovative Medicines, AstraZeneca R&D Mölndal, Mölndal, Sweden
| | | | | | | | | |
Collapse
|
43
|
LoGuidice A, Wallace BD, Bendel L, Redinbo MR, Boelsterli UA. Pharmacologic targeting of bacterial β-glucuronidase alleviates nonsteroidal anti-inflammatory drug-induced enteropathy in mice. J Pharmacol Exp Ther 2012; 341:447-54. [PMID: 22328575 PMCID: PMC3336811 DOI: 10.1124/jpet.111.191122] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 02/09/2012] [Indexed: 01/05/2023] Open
Abstract
Small intestinal mucosal injury is a frequent adverse effect caused by nonsteroidal anti-inflammatory drugs (NSAIDs). The underlying mechanisms are not completely understood, but topical (luminal) effects have been implicated. Many carboxylic acid-containing NSAIDs, including diclofenac (DCF), are metabolized to acyl glucuronides (AGs), and/or ether glucuronides after ring hydroxylation, and exported into the biliary tree. In the gut, these conjugates are cleaved by bacterial β-glucuronidase, releasing the potentially harmful aglycone. We first confirmed that DCF-AG was an excellent substrate for purified Escherichia coli β-D-glucuronidase. Using a previously characterized novel bacteria-specific β-glucuronidase inhibitor (Inhibitor-1), we then found that the enzymatic hydrolysis of DCF-AG in vitro was inhibited concentration dependently (IC₅₀ ∼164 nM). We next hypothesized that pharmacologic inhibition of bacterial β-glucuronidase would reduce exposure of enterocytes to the aglycone and, as a result, alleviate enteropathy. C57BL/6J mice were administered an ulcerogenic dose of DCF (60 mg/kg i.p.) with or without oral pretreatment with Inhibitor-1 (10 μg per mouse, b.i.d.). Whereas DCF alone caused the formation of numerous large ulcers in the distal parts of the small intestine and increased (2-fold) the intestinal permeability to fluorescein isothiocyanate-dextran, Inhibitor-1 cotreatment significantly alleviated mucosal injury and reduced all parameters of enteropathy. Pharmacokinetic profiling of DCF plasma levels in mice revealed that Inhibitor-1 coadministration did not significantly alter the C(max), half-life, or area under the plasma concentration versus time curve of DCF. Thus, highly selective pharmacologic targeting of luminal bacterial β-D-glucuronidase by a novel class of small-molecule inhibitors protects against DCF-induced enteropathy without altering systemic drug exposure.
Collapse
Affiliation(s)
- Amanda LoGuidice
- Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, Connecticut, USA
| | | | | | | | | |
Collapse
|
44
|
Amacher DE. The primary role of hepatic metabolism in idiosyncratic drug-induced liver injury. Expert Opin Drug Metab Toxicol 2012; 8:335-47. [DOI: 10.1517/17425255.2012.658041] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
45
|
|
46
|
Wang Z, Zhou Q, Kruh GD, Gallo JM. Dose-dependent disposition of methotrexate in Abcc2 and Abcc3 gene knockout murine models. Drug Metab Dispos 2011; 39:2155-61. [PMID: 21841039 PMCID: PMC3198900 DOI: 10.1124/dmd.111.041228] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 08/08/2011] [Indexed: 11/22/2022] Open
Abstract
Methotrexate (MTX) is a substrate for numerous human ATP-binding cassette (ABC) efflux transporters, yet the impact of these transporters on MTX pharmacokinetics (PK) over a large dose range has not been examined. To investigate the effects of two transporters-ABC subfamily C member 2 (Abcc2; multidrug resistance protein 2) and ABC subfamily C member 3 (Abcc3; multidrug resistance protein 3)-involved in MTX hepatobiliary disposition in vivo, MTX plasma, urine, and feces concentrations were analyzed after 10, 50, and 200 mg/kg i.v. doses to groups of wild type (WT), Abcc2(-/-), and Abcc3(-/-) mice. The absence of Abcc2 caused a decrease in total clearance of MTX relative to WT mice at all dose levels yet was accompanied by compensatory increases in renal excretion and metabolism to 7-hydroxymethotrexate (7OH-MTX). In Abcc3(-/-) mice, total clearance was elevated at the two lower dose levels and was attributed to stimulation of biliary excretion and confirmed by elevated fecal excretion; however, at the high 200 mg/kg dose, clearance was severely retarded and could be attributed to hepatotoxicity because conversion to 7OH-MTX was diminished. The findings confirmed that both Abcc2 and Abcc3 significantly influenced the PK properties of MTX, and depending on the MTX dose and strain, alternate elimination pathways were elicited and saturable.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Pharmaceutical Sciences, Temple University, Philadelphia, Pennsylvania, USA
| | | | | | | |
Collapse
|
47
|
Sarda S, Page C, Pickup K, Schulz-Utermoehl T, Wilson I. Diclofenac metabolism in the mouse: novel in vivo metabolites identified by high performance liquid chromatography coupled to linear ion trap mass spectrometry. Xenobiotica 2011; 42:179-94. [PMID: 21955289 DOI: 10.3109/00498254.2011.607865] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The metabolism of [(14)C]-diclofenac in mice was investigated following a single oral dose of 10 mg/kg. The majority of the drug-related material was excreted in the urine within 24 h of administration (49.7 %). Liquid chromatographic analyses of urine and faecal extracts revealed extensive metabolism to at least 37 components, with little unchanged diclofenac excreted. Metabolites were identified using a hybrid linear ion-trap mass spectrometer via exact mass determinations of molecular ions and subsequent multi-stage fragmentation. The major routes of metabolism identified included: 1) conjugation with taurine; and 2) hydroxylation (probably at the 4'-and 5-arene positions) followed by conjugation to taurine, glucuronic acid or glucose. Ether, rather than acyl glucuronidation, predominated. There was no evidence for p-benzoquinone-imine formation (i.e. no glutathione or mercapturic acid conjugates were detected). A myriad of novel minor drug-related metabolites were also detected, including ribose, glucose, sulfate and glucuronide ether-linked conjugates of hydroxylated diclofenac derivatives. Combinations of these hydroxylated derivatives with acyl conjugates (glucose, glucuronide and taurine) or N-linked sulfation or glucosidation were also observed. Acyl- or amide-linked-conjugates of benzoic acid metabolites and several indolinone derivatives with further hydroxylated and conjugated moieties were also evident. The mechanisms involved in the generation of benzoic acid and indolinone products indicate the formation reactive intermediates in vivo that may possibly contribute to hepatotoxicity.
Collapse
Affiliation(s)
- Sunil Sarda
- AstraZeneca, DMPK IM , Alderley Park, Macclesfield, United Kingdom.
| | | | | | | | | |
Collapse
|
48
|
Regan SL, Maggs JL, Hammond TG, Lambert C, Williams DP, Park BK. Acyl glucuronides: the good, the bad and the ugly. Biopharm Drug Dispos 2011; 31:367-95. [PMID: 20830700 DOI: 10.1002/bdd.720] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acyl glucuronidation is the major metabolic conjugation reaction of most carboxylic acid drugs in mammals. The physiological consequences of this biotransformation have been investigated incompletely but include effects on drug metabolism, protein binding, distribution and clearance that impact upon pharmacological and toxicological outcomes. In marked contrast, the exceptional but widely disparate chemical reactivity of acyl glucuronides has attracted far greater attention. Specifically, the complex transacylation and glycation reactions with proteins have provoked much inconclusive debate over the safety of drugs metabolised to acyl glucuronides. It has been hypothesised that these covalent modifications could initiate idiosyncratic adverse drug reactions. However, despite a large body of in vitro data on the reactions of acyl glucuronides with protein, evidence for adduct formation from acyl glucuronides in vivo is limited and potentially ambiguous. The causal connection of protein adduction to adverse drug reactions remains uncertain. This review has assessed the intrinsic reactivity, metabolic stability and pharmacokinetic properties of acyl glucuronides in the context of physiological, pharmacological and toxicological perspectives. Although numerous experiments have characterised the reactions of acyl glucuronides with proteins, these might be attenuated substantially in vivo by rapid clearance of the conjugates. Consequently, to delineate a relationship between acyl glucuronide formation and toxicological phenomena, detailed pharmacokinetic analysis of systemic exposure to the acyl glucuronide should be undertaken adjacent to determining protein adduct concentrations in vivo. Further investigation is required to ascertain whether acyl glucuronide clearance is sufficient to prevent covalent modification of endogenous proteins and consequentially a potential immunological response.
Collapse
Affiliation(s)
- Sophie L Regan
- MRC Centre for Drug Safety Science, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3GE, UK.
| | | | | | | | | | | |
Collapse
|
49
|
Koga T, Fujiwara R, Nakajima M, Yokoi T. Toxicological evaluation of acyl glucuronides of nonsteroidal anti-inflammatory drugs using human embryonic kidney 293 cells stably expressing human UDP-glucuronosyltransferase and human hepatocytes. Drug Metab Dispos 2011; 39:54-60. [PMID: 20926620 DOI: 10.1124/dmd.110.035600] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The chemical reactivity of acyl glucuronide (AG) has been thought to be associated with the toxic properties of drugs containing carboxylic acid moieties, but there has been no direct evidence that AG formation was related to the toxicity. In the present study, the cytotoxicity and genotoxicity of AGs were investigated. Human embryonic kidney (HEK) 293 cells stably expressing UDP-glucuronosyltransferase (UGT) 1A3 (HEK/UGT1A3) were constructed to assess the cytotoxicity of AGs, and HEK/UGT1A4 cells were also used as a negative reference. After exposure to nonsteroidal anti-inflammatory drugs (NSAIDs) such as naproxen (1 mM), diclofenac (0.1 mM), ketoprofen (1 mM), or ibuprofen (1 mM) for 24 h, HEK/UGT1A3 cells produced AG in a time-dependent manner. However, HEK/UGT1A4 cells hardly produced AG. The cytotoxicity of HEK/UGT1A3 cells was not increased compared with that of HEK/UGT1A4 cells. In addition, the AG formed in NSAID-treated human hepatocytes was decreased from one-third to one-ninth by treatment with (-)-borneol, an inhibitor of acyl glucuronidation, but the cytotoxicity was increased. These results indicated that AG formation reflected the detoxification process in human hepatocytes. Furthermore, the possibility of genotoxicity from the AG formed in NSAID-treated HEK/UGT cells was investigated by the comet assay, and DNA damage was not detected in any HEK/UGT cell lines. In conclusion, the in vitro cytotoxic and genotoxic effects of the AGs of NSAIDs were investigated and AG was not found to be a causal factor in the toxicity.
Collapse
Affiliation(s)
- Toshihisa Koga
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | | | | | | |
Collapse
|
50
|
van Midwoud PM, Janssen J, Merema MT, de Graaf IAM, Groothuis GMM, Verpoorte E. On-line HPLC analysis system for metabolism and inhibition studies in precision-cut liver slices. Anal Chem 2010; 83:84-91. [PMID: 21128611 DOI: 10.1021/ac1018638] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A novel approach for on-line monitoring of drug metabolism in continuously perifused, precision-cut liver slices (PCLS) in a microfluidic system has been developed using high-performance liquid chromatography with UV detection (HPLC-UV). In this approach, PCLS are incubated in a microfluidic device made of poly(dimethylsiloxane) (PDMS) by continuous, single-pass perifusion with fresh medium. Two syringe pumps are incorporated into the system to infuse substrates or inhibitors at varying concentrations into the perfusion medium just before the chip entrance. The medium containing the metabolites produced by the PCLS is directed toward an injection loop. Once filled, the content of this injection loop is automatically injected onto an HPLC for analysis. The on-line analysis of metabolites was tested by using the substrate, 7-hydroxycoumarin (7-HC). Rapid switching between substrate and solvent control was possible, and a direct metabolic response of the liver slice to perifusion with substrate was detected. Very stable phase II metabolism over a period of 24 h was observed. The inhibitory effect of phloxine B on the formation of 7-hydroxycoumarin glucuronide (phase II product of 7-HC) was also investigated. Phloxine B was injected into the incubation medium in increasing concentrations varying from 0 to 200 μM. The results showed a concentration-dependent inhibition of 7-HC glucuronide formation and allowed the calculation of an IC50 value (concentration in which 50% of the enzyme is inhibited) of ∼85 μM using one single liver slice. On-line detection was also shown to be advantageous for the detection of unstable metabolites. This was demonstrated by determination of the metabolites of the drug diclofenac. The reactive metabolite, acyl glucuronide, was detected at relatively high concentrations which remained very constant over a period of 4 h. In contrast, only low and decreasing amounts of diclofenac acyl glucuronide could be measured in the conventional well-plate incubation system. The advantages of this novel on-line analysis system for PCLS include the capability to obtain direct information about tissue function, assess the concentration dependence of drug-drug interactions in one single slice, and detect unstable metabolites. The system also enables fast analysis without the need to store samples, thus eliminating the associated freeze-thaw problems, and allows the simultaneous analysis of multiple metabolites.
Collapse
Affiliation(s)
- Paul M van Midwoud
- Pharmaceutical Analysis, Department of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|