1
|
Hagenbuch B, Stieger B, Locher KP. Organic anion transporting polypeptides: Pharmacology, toxicology, structure, and transport mechanisms. Pharmacol Rev 2025; 77:100023. [PMID: 40148036 DOI: 10.1016/j.pharmr.2024.100023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/12/2024] [Indexed: 03/29/2025] Open
Abstract
Organic anion transporting polypeptides (OATPs) are membrane proteins that mediate the uptake of a wide range of substrates across the plasma membrane of various cells and tissues. They are classified into 6 subfamilies, OATP1 through OATP6. Humans contain 12 OATPs encoded by 11 solute carrier of organic anion transporting polypeptide (SLCO) genes: OATP1A2, OATP1B1, OATP1B3, the splice variant OATP1B3-1B7, OATP1C1, OATP2A1, OATP2B1, OATP3A1, OATP4A1, OATP4C1, OATP5A1, and OATP6A1. Most of these proteins are expressed in epithelial cells, where they mediate the uptake of structurally unrelated organic anions, cations, and even neutral compounds into the cytoplasm. The best-characterized members are OATP1B1 and OATP1B3, which have an important role in drug metabolism by mediating drug uptake into the liver and are involved in drug-drug interactions. In this review, we aimed to (1) provide a historical perspective on the identification of OATPs and their nomenclature and discuss their phylogenic relationships and molecular characteristics; (2) review the current knowledge of the broad substrate specificity and their role in drug disposition and drug-drug interactions, with a special emphasis on human hepatic OATPs; (3) summarize the different experimental systems that are used to study the function of OATPs and discuss their advantages and disadvantages; (4) review the available experimental 3-dimensional structures and examine how they can help elucidate the transport mechanisms of OATPs; and (5) finally, summarize the current knowledge of the regulation of OATP expression, discuss clinically important single-nucleotide polymorphisms, and outline challenges of physiologically based pharmacokinetic modeling and in vitro to in vivo extrapolation. SIGNIFICANCE STATEMENT: Organic anion transporting polypeptides (OATPs) are a family of 12 uptake transporters in the solute carrier superfamily. Several members, particularly the liver-expressed OATP1B1 and OATP1B3, are important drug transporters. They mediate the uptake of several endobiotics and xenobiotics, including statins and numerous other drugs, into hepatocytes, and their inhibition by other drugs or reduced expression due to single-nucleotide polymorphisms can lead to adverse drug effects. Their recently solved 3-dimensional structures should help to elucidate their transport mechanisms and broad substrate specificities.
Collapse
Affiliation(s)
- Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas.
| | - Bruno Stieger
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| | - Kaspar P Locher
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
2
|
Gu J, Guo Y, Wu B, He J. Liver injury associated with endothelin receptor antagonists: a pharmacovigilance study based on FDA adverse event reporting system data. Int J Clin Pharm 2024; 46:1307-1316. [PMID: 38902469 DOI: 10.1007/s11096-024-01757-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 05/17/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Endothelin receptor antagonists are commonly used in clinical practice, with concerns about their hepatotoxicity. AIM This study aimed to conduct a comprehensive pharmacovigilance study based on FDA adverse event reporting system data to evaluate the possible association between endothelin receptor antagonists and drug-induced liver injury. METHOD Adverse event reports from FDA adverse event reporting system between January 2004 and December 2022 were analyzed. Disproportionality algorithms, including reporting odds ratio and information component, were used to evaluate the association between endothelin receptor antagonists and liver injury. Sex- and age-stratified analyses of drug-induced liver injury events were also conducted in relation to endothelin receptor antagonists. RESULTS Significant associations between bosentan, macitentan, and liver injury were identified. Bosentan showed a strong link with liver injury, with reporting odds ratios for cholestatic injury at 7.59 (95% confidence interval: 6.90-8.35), hepatocellular injury at 5.63 (5.29-6.00), and serious drug-related hepatic disorders events at 1.33 (1.24-1.43). Drug-induced liver injury signals associated with bosentan were detected in all age groups. Macitentan was associated with liver injury, with reporting odds ratios for hepatic failure at 1.64 (1.39-1.94), cholestatic injury at 1.62 (1.43-1.83), and serious drug-related hepatic disorders events at 1.40 (1.29-1.51). No drug-induced liver injury signal was detected for ambrisentan, and no significant sex differences were observed in drug-induced liver injury events. CONCLUSION Both bosentan and macitentan are associated with liver injury. Routine monitoring of serum aminotransferase levels is recommended, especially in patients at higher risk of liver injury. Further research into drug-drug interactions involving endothelin receptor antagonists is warranted.
Collapse
Affiliation(s)
- Jinjian Gu
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Yuting Guo
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Wu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinhan He
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Hwang S, Lee Y, Jang Y, Cho JY, Yoon S, Chung JY. Comprehensive Evaluation of OATP- and BCRP-Mediated Drug-Drug Interactions of Methotrexate Using Physiologically-Based Pharmacokinetic Modeling. Clin Pharmacol Ther 2024; 116:1013-1022. [PMID: 38860384 DOI: 10.1002/cpt.3329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/16/2024] [Indexed: 06/12/2024]
Abstract
Methotrexate (MTX) is an antifolate agent widely used for treating conditions such as rheumatoid arthritis and hematologic cancer. This study aimed to quantitatively interpret the drug-drug interactions (DDIs) of MTX mediated by drug transporters using physiologically-based pharmacokinetic (PBPK) modeling. An open-label, randomized, 4-treatment, 6-sequence, 4-period crossover study was conducted to investigate the effects of rifampicin (RFP), an inhibitor of organic anionic transporting peptides (OATP) 1B1/3, and febuxostat (FBX), an inhibitor of breast cancer resistance protein (BCRP), on the pharmacokinetics of MTX in healthy volunteers. PBPK models of MTX, RFP, and FBX were developed based on in vitro and in vivo data, and the performance of the simulation results for final PBPK models was validated in a clinical study. In the clinical study, when MTX was co-administered with RFP or FBX, systemic exposure of MTX increased by 33% and 17%, respectively, compared with that when MTX was administered alone. When MTX was co-administered with RFP and FBX, systemic exposure increased by 52% compared with that when MTX was administered alone. The final PBPK model showed a good prediction performance for the observed clinical data. The PBPK model of MTX was well developed in this study and can be used as a potential mechanistic model to predict and evaluate drug transporter-mediated DDIs of MTX with other drugs.
Collapse
Affiliation(s)
- Sejung Hwang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University Medical Research Center, Seoul, Korea
| | - Yujin Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
| | - Yeonseo Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University Medical Research Center, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Seonghae Yoon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jae-Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Bundang Hospital, Seongnam, Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Korea
| |
Collapse
|
4
|
Marin JJG, Cives-Losada C, Macias RIR, Romero MR, Marijuan RP, Hortelano-Hernandez N, Delgado-Calvo K, Villar C, Gonzalez-Santiago JM, Monte MJ, Asensio M. Impact of liver diseases and pharmacological interactions on the transportome involved in hepatic drug disposition. Biochem Pharmacol 2024; 228:116166. [PMID: 38527556 DOI: 10.1016/j.bcp.2024.116166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
The liver plays a pivotal role in drug disposition owing to the expression of transporters accounting for the uptake at the sinusoidal membrane and the efflux across the basolateral and canalicular membranes of hepatocytes of many different compounds. Moreover, intracellular mechanisms of phases I and II biotransformation generate, in general, inactive compounds that are more polar and easier to eliminate into bile or refluxed back toward the blood for their elimination by the kidneys, which becomes crucial when the biliary route is hampered. The set of transporters expressed at a given time, i.e., the so-called transportome, is encoded by genes belonging to two gene superfamilies named Solute Carriers (SLC) and ATP-Binding Cassette (ABC), which account mainly, but not exclusively, for the uptake and efflux of endogenous substances and xenobiotics, which include many different drugs. Besides the existence of genetic variants, which determines a marked interindividual heterogeneity regarding liver drug disposition among patients, prevalent diseases, such as cirrhosis, non-alcoholic steatohepatitis, primary sclerosing cholangitis, primary biliary cirrhosis, viral hepatitis, hepatocellular carcinoma, cholangiocarcinoma, and several cholestatic liver diseases, can alter the transportome and hence affect the pharmacokinetics of drugs used to treat these patients. Moreover, hepatic drug transporters are involved in many drug-drug interactions (DDI) that challenge the safety of using a combination of agents handled by these proteins. Updated information on these questions has been organized in this article by superfamilies and families of members of the transportome involved in hepatic drug disposition.
Collapse
Affiliation(s)
- Jose J G Marin
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain.
| | - Candela Cives-Losada
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Rocio I R Macias
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Marta R Romero
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Rebeca P Marijuan
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain
| | | | - Kevin Delgado-Calvo
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain
| | - Carmen Villar
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain; Department of Gastroenterology and Hepatology, University Hospital of Salamanca, Salamanca, Spain
| | - Jesus M Gonzalez-Santiago
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain; Department of Gastroenterology and Hepatology, University Hospital of Salamanca, Salamanca, Spain
| | - Maria J Monte
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Maitane Asensio
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| |
Collapse
|
5
|
Kang MJ, Kim MJ, Seol Y, Chang JE, Lee KR, Chae YJ. Interactions of bosutinib with drug transporters: In vitro and In vivo inhibition of organic cation transporter 2, multidrug and toxin extrusion protein 1, and breast cancer resistance protein by bosutinib. Biomed Pharmacother 2024; 178:117114. [PMID: 39053425 DOI: 10.1016/j.biopha.2024.117114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/04/2024] [Accepted: 07/07/2024] [Indexed: 07/27/2024] Open
Abstract
Bosutinib has been approved for use in patients with chronic myeloid leukemia. Information regarding the effects of bosutinib on clinically important drug transporters is limited, particularly regarding its inhibitory potency on transporters and in vivo effects. Therefore, we conducted a study investigating the in vitro and in vivo effects of bosutinib on drug transporters. Bosutinib showed moderate or strong inhibitory effects on organic cation transporter 2, multidrug and toxin extrusion protein 1, and breast cancer resistance protein with IC50 values of 0.0894, 0.598, and 10.8 µM, respectively. In vivo experiments in rats showed that bosutinib significantly inhibited organic cation transporter 2 and multidrug and toxin extrusion protein 1, leading to a marked reduction in the renal clearance of metformin and an increase in systemic exposure to metformin. Bosutinib increased systemic exposure to sulfasalazine, a probe substrate of breast cancer resistance protein, by 75 % in rats, highlighting its potential to significantly affect intestinal drug efflux. These quantitative changes suggest that bosutinib may alter the in vivo pharmacokinetics of drugs that are substrates of these transporters, potentially leading to increased drug exposure and enhanced or unexpected pharmacological effects.
Collapse
Affiliation(s)
- Min-Ji Kang
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea
| | - Min Ju Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Yunjin Seol
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea
| | - Ji-Eun Chang
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Kyeong-Ryoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Bioscience, University of Science and Technology, Daejeon 34113, Republic of Korea.
| | - Yoon-Jee Chae
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea; Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Republic of Korea.
| |
Collapse
|
6
|
de Bruijn VMP, Rietjens IMCM. From hazard to risk prioritization: a case study to predict drug-induced cholestasis using physiologically based kinetic modeling. Arch Toxicol 2024; 98:3077-3095. [PMID: 38755481 PMCID: PMC11324677 DOI: 10.1007/s00204-024-03775-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024]
Abstract
Cholestasis is characterized by hepatic accumulation of bile acids. Clinical manifestation of cholestasis only occurs in a small proportion of exposed individuals. The present study aims to develop a new approach methodology (NAM) to predict drug-induced cholestasis as a result of drug-induced hepatic bile acid efflux inhibition and the resulting bile acid accumulation. To this end, hepatic concentrations of a panel of drugs were predicted by a generic physiologically based kinetic (PBK) drug model. Their effects on hepatic bile acid efflux were incorporated in a PBK model for bile acids. The predicted bile acid accumulation was used as a measure for a drug's cholestatic potency. The selected drugs were known to inhibit hepatic bile acid efflux in an assay with primary suspension-cultured hepatocytes and classified as common, rare, or no for cholestasis incidence. Common cholestasis drugs included were atorvastatin, chlorpromazine, cyclosporine, glimepiride, ketoconazole, and ritonavir. The cholestasis incidence of the drugs appeared not to be adequately predicted by their Ki for inhibition of hepatic bile acid efflux, but rather by the AUC of the PBK model predicted internal hepatic drug concentration at therapeutic dose level above this Ki. People with slower drug clearance, a larger bile acid pool, reduced bile salt export pump (BSEP) abundance, or given higher than therapeutic dose levels were predicted to be at higher risk to develop drug-induced cholestasis. The results provide a proof-of-principle of using a PBK-based NAM for cholestasis risk prioritization as a result of transporter inhibition and identification of individual risk factors.
Collapse
Affiliation(s)
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands.
| |
Collapse
|
7
|
Kang MJ, Kim MJ, Kim A, Koo TS, Lee KR, Chae YJ. Pharmacokinetic interactions of niclosamide in rats: Involvement of organic anion transporters 1 and 3 and organic cation transporter 2. Chem Biol Interact 2024; 390:110886. [PMID: 38280639 DOI: 10.1016/j.cbi.2024.110886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/30/2023] [Accepted: 01/19/2024] [Indexed: 01/29/2024]
Abstract
Niclosamide is an anthelmintic drug with a long history of use and is generally safe and well tolerated in humans. As the conventional dose of niclosamide results in a low but certain level in systemic circulation, drug interactions with concomitant drugs should be considered. We aimed to investigate the interaction between niclosamide and drug transporters, as such information is currently limited. Niclosamide inhibited the transport activity of OATP1B1, OATP1B3, OAT1, OAT3, and OCT2 in vitro. Among them, the inhibitory effects on OAT1, OAT3, and OCT2 were strong, with IC50 values of less than 1 μM. When 3 mg/kg of niclosamide was co-administered to rats, systemic exposure to furosemide (a substrate of OAT1/3) and metformin (a substrate of OCT2) increased, and the renal clearance (CLr) of the drugs significantly decreased. These results suggest that niclosamide inhibits renal transporters, OAT1/3 and OCT2, not only in vitro but also in vivo, resulting in increased systemic exposure to the substrates of the transporters by strongly blocking the urinary elimination pathway in rats. The findings of this study will support a meticulous understanding of the transporter-mediated drug interactions of niclosamide and consequently aid in effective and safe use of niclosamide.
Collapse
Affiliation(s)
- Min-Ji Kang
- College of Pharmacy, Woosuk University, Wanju, 55338, Republic of Korea
| | - Min Ju Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Aeran Kim
- College of Pharmacy, Woosuk University, Wanju, 55338, Republic of Korea
| | - Tae-Sung Koo
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Kyeong-Ryoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea; Department of Bioscience, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Yoon-Jee Chae
- College of Pharmacy, Woosuk University, Wanju, 55338, Republic of Korea; Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju, 55338, Republic of Korea.
| |
Collapse
|
8
|
Parvez MM, Sadighi A, Ahn Y, Keller SF, Enoru JO. Uptake Transporters at the Blood-Brain Barrier and Their Role in Brain Drug Disposition. Pharmaceutics 2023; 15:2473. [PMID: 37896233 PMCID: PMC10610385 DOI: 10.3390/pharmaceutics15102473] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Uptake drug transporters play a significant role in the pharmacokinetic of drugs within the brain, facilitating their entry into the central nervous system (CNS). Understanding brain drug disposition is always challenging, especially with respect to preclinical to clinical translation. These transporters are members of the solute carrier (SLC) superfamily, which includes organic anion transporter polypeptides (OATPs), organic anion transporters (OATs), organic cation transporters (OCTs), and amino acid transporters. In this systematic review, we provide an overview of the current knowledge of uptake drug transporters in the brain and their contribution to drug disposition. Here, we also assemble currently available proteomics-based expression levels of uptake transporters in the human brain and their application in translational drug development. Proteomics data suggest that in association with efflux transporters, uptake drug transporters present at the BBB play a significant role in brain drug disposition. It is noteworthy that a significant level of species differences in uptake drug transporters activity exists, and this may contribute toward a disconnect in inter-species scaling. Taken together, uptake drug transporters at the BBB could play a significant role in pharmacokinetics (PK) and pharmacodynamics (PD). Continuous research is crucial for advancing our understanding of active uptake across the BBB.
Collapse
Affiliation(s)
- Md Masud Parvez
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| | - Armin Sadighi
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| | - Yeseul Ahn
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S Coulter St., Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Steve F. Keller
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| | - Julius O. Enoru
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| |
Collapse
|
9
|
Coons JC, Empey PE. Pharmacogenomics in the Management of Pulmonary Arterial Hypertension: Current Perspectives. Pharmgenomics Pers Med 2023; 16:729-737. [PMID: 37457231 PMCID: PMC10349598 DOI: 10.2147/pgpm.s361222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease with heterogeneous causes that can lead to right ventricular (RV) failure and death if left untreated. There are currently 10 medications representative of five unique pharmacologic classes that are approved for treatment. These have led to significant improvements in overall clinical outcome. However, substantial variability in dosing requirements and treatment response is evident, leading to suboptimal outcome for many patients. Furthermore, dosing is empiric and iterative and can lead to delays in meeting treatment goals and burdensome adverse effects. Pharmacogenomic (PGx) associations have been reported with certain PAH medications, such as treprostinil and bosentan, and can explain some of the variability in response. Relevant genes associated with treprostinil include CYP2C8, CYP2C9, CAMK2D, and PFAS. CYP2C8 and CYP2C9 are the genes encoding the major metabolizing liver enzymes for treprostinil, and reduced function variants (*2, *3) with CYP2C9 were associated with lower treatment persistence. Additionally, a higher CYP2C9 activity score was associated with a significantly less risk of treatment discontinuation. Other genes of interest that have been explored with treprostinil include CAMK2D, which is associated with right ventricular dysfunction and significantly higher dose requirements. Similarly, PFAS is associated with lower concentrations of cyclic adenosine monophosphate and significantly higher dose requirements. Genes of interest with the endothelin receptor antagonist (ERA) class include GNG2 and CYP2C9. A genetic variant in GNG2 (rs11157866) was linked to a significantly increased rate of clinical improvement with ERAs. The *2 variant with CYP2C9 (encoding for the major metabolizing enzyme for bosentan) was significantly associated with a higher risk for elevations in hepatic aminotransferases and liver injury. In summary, this article reviews the relevant pharmacogenes that have been associated to date with dosing and outcome among patients who received PAH medications.
Collapse
Affiliation(s)
- James C Coons
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
- Department of Pharmacy, UPMC Presbyterian-Shadyside Hospital, Pittsburgh, PA, USA
| | - Philip E Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| |
Collapse
|
10
|
Marie S, Frost KL, Hau RK, Martinez-Guerrero L, Izu JM, Myers CM, Wright SH, Cherrington NJ. Predicting disruptions to drug pharmacokinetics and the risk of adverse drug reactions in non-alcoholic steatohepatitis patients. Acta Pharm Sin B 2023; 13:1-28. [PMID: 36815037 PMCID: PMC9939324 DOI: 10.1016/j.apsb.2022.08.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 12/18/2022] Open
Abstract
The liver plays a central role in the pharmacokinetics of drugs through drug metabolizing enzymes and transporters. Non-alcoholic steatohepatitis (NASH) causes disease-specific alterations to the absorption, distribution, metabolism, and excretion (ADME) processes, including a decrease in protein expression of basolateral uptake transporters, an increase in efflux transporters, and modifications to enzyme activity. This can result in increased drug exposure and adverse drug reactions (ADRs). Our goal was to predict drugs that pose increased risks for ADRs in NASH patients. Bibliographic research identified 71 drugs with reported ADRs in patients with liver disease, mainly non-alcoholic fatty liver disease (NAFLD), 54 of which are known substrates of transporters and/or metabolizing enzymes. Since NASH is the progressive form of NAFLD but is most frequently undiagnosed, we identified other drugs at risk based on NASH-specific alterations to ADME processes. Here, we present another list of 71 drugs at risk of pharmacokinetic disruption in NASH, based on their transport and/or metabolism processes. It encompasses drugs from various pharmacological classes for which ADRs may occur when used in NASH patients, especially when eliminated through multiple pathways altered by the disease. Therefore, these results may inform clinicians regarding the selection of drugs for use in NASH patients.
Collapse
Affiliation(s)
- Solène Marie
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Kayla L. Frost
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Raymond K. Hau
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Lucy Martinez-Guerrero
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Jailyn M. Izu
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Cassandra M. Myers
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Stephen H. Wright
- College of Medicine, Department of Physiology, University of Arizona, Tucson, AZ 85724, USA
| | - Nathan J. Cherrington
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA,Corresponding author. Tel.: +1 520 6260219; fax: +1 520 6266944.
| |
Collapse
|
11
|
Božina T, Ganoci L, Karačić E, Šimičević L, Vrkić-Kirhmajer M, Klarica-Domjanović I, Križ T, Sertić Z, Božina N. ABCG2 and SLCO1B1 gene polymorphisms in the Croatian population. Ann Hum Biol 2022; 49:323-331. [PMID: 36382878 DOI: 10.1080/03014460.2022.2140826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Organic anion-transporting polypeptide 1B1 (OATP1B1) and the ATP-binding cassette subfamily G member 2, ABCG2, are important transporters involved in the transport of endogenous substrates and xenobiotics, including drugs. Genetic polymorphisms of these transporters have effect on transporter activity. There is significant interethnic variability in the frequency of allele variants. AIM To determined allele and genotype frequencies of ABCG2 and SLCO1B1 genes in Croatian populations of European descent. SUBJECTS AND METHODS A total of 905 subjects (482 women) were included. Genotyping for ABCG2 c.421C > A (rs2231142) and for SLCO1B1 c.521T > C (rs4149056), was performed by real-time polymerase chain reaction (PCR) using TaqMan® DME Genotyping Assays. RESULTS For ABCG2 c.421C > A, the frequency of CC, CA and AA genotypes was 81.4%, 17.8% and 0.8% respectively. The frequency of variant ABCG2 421 A allele was 9.7%. For SLCO1B1 c.521T > C, the frequency of TT, TC and CC genotypes was 61.7%, 34.8% and 3.5% respectively. The frequency of variant SLCO1B1 521 C allele was 20.9%. CONCLUSION The frequency of the ABCG2 and SLCO1B1 allelic variants and genotypes in the Croatian population is in accordance with other European populations. Pharmacogenetic analysis can serve to individualise drug therapy and minimise the risk of developing adverse drug reactions.
Collapse
Affiliation(s)
- Tamara Božina
- Department of Medical Chemistry, Biochemistry, and Clinical Chemistry, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lana Ganoci
- Division of Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ena Karačić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Livija Šimičević
- Division of Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Majda Vrkić-Kirhmajer
- Department of Cardiovascular Diseases Zagreb, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Croatia
| | | | - Tena Križ
- Department of Ophthalmology, University Hospital Centre "Sestre milosrdnice", Zagreb, Croatia
| | - Zrinka Sertić
- Department of Emergency Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nada Božina
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
12
|
Nies AT, Schaeffeler E, Schwab M. Hepatic solute carrier transporters and drug therapy: Regulation of expression and impact of genetic variation. Pharmacol Ther 2022; 238:108268. [DOI: 10.1016/j.pharmthera.2022.108268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
|
13
|
Dashti M, Al-Matrouk A, Channanath A, Al-Mulla F, Thanaraj TA. Frequency of functional exonic single-nucleotide polymorphisms and haplotype distribution in the SLCO1B1 gene across genetic ancestry groups in the Qatari population. Sci Rep 2022; 12:14858. [PMID: 36050458 PMCID: PMC9437070 DOI: 10.1038/s41598-022-19318-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 11/09/2022] Open
Abstract
Organic anion transporting polypeptides (OATP), which are encoded by SLCO genes, participate in the hepatic elimination of drugs and xenobiotics. SLCO1B1 is an important pharmacogenomic gene (encoding OATP1B1) associated with response to the uptake of endogenous compounds, such as statin and bilirubin. Ethnicity of the patient modulates the response to these drugs; the frequency and haplotype data for SLCO1B1 genetic variants in the Arab population is lacking. Therefore, we determined the frequencies of two well-characterized SLCO1B1 single nucleotide polymorphisms (SNP) and haplotypes that affect the OATP1B1 drugs transportation activity in Qatari population. Genotyping data for two SLCO1B1 SNPs (c.388A > G, c.521 T > C) were extracted from whole exome data of 1050 Qatari individuals, who were divided into three ancestry groups, namely Bedouins, Persians/South Asians, and Africans. By way of using Fisher's exact and Chi-square tests, we evaluated the differences in minor allele frequency (MAF) of the two functional SNPs and haplotype frequencies (HF) among the three ancestry groups. The OATP1B1 phenotypes were assigned according to their function by following the guidelines from the Clinical Pharmacogenetics Implementation Consortium for SLCO1B1 and Simvastatin-Induced Myopathy.The MAF of SLCO1B1:c.388A > G was higher compared to that of SLCO1B1:c.521 T > C in the study cohort. It was significantly high in the African ancestry group compared with the other two groups, whereas SLCO1B1:c.521 T > C was significantly low in the African ancestry group compared with the other two groups. The SLCO1B1 *15 haplotype had the highest HF, followed by *1b, *1a, and *5. Only the SLCO1B1 *5 haplotype showed no significant difference in frequency across the three ancestry groups. Furthermore, we observed that the OATP1B1 normal function phenotype accounted for 58% of the Qatari individuals, the intermediate function phenotype accounted for 35% with significant differences across the ancestry groups, and the low function phenotype accounted for 6% of the total Qatari individuals with a higher trend observed in the Bedouin group.The results indicate that the phenotype frequencies of the OATP1B1 intermediate and low function in the Qatari population appear at the higher end of the frequency range seen worldwide. Thus, a pharmacogenetic screening program for SLCO1B1 variants may be necessary for the Qatari population.
Collapse
Affiliation(s)
- Mohammed Dashti
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Abdullah Al-Matrouk
- Narcotic and Psychotropic Department, Ministry of Interior, Farwaniya, Kuwait
| | - Arshad Channanath
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait.
| | | |
Collapse
|
14
|
Wang Z, Li Y, Villanueva CE, Peng T, Han W, Bo Z, Zhang H, Hagenbuch B, Gui C. The Importance of Val386 in Transmembrane Domain 8 for the Activation of OATP1B3 by Epigallocatechin Gallate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:6552-6560. [PMID: 35603894 PMCID: PMC9438777 DOI: 10.1021/acs.jafc.2c02692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Estrone-3-sulfate (E3S) uptake mediated by organic anion transporting polypeptide 1B3 (OATP1B3) can be activated by epigallocatechin gallate (EGCG). In this study, by using chimeric transporters and site-directed mutagenesis, we found that Val386 in transmembrane domain 8 (TM8) is essential for OATP1B3's activation by EGCG. Kinetic studies showed that the loss of activation of 1B3-TM8 and 1B3-V386F in the presence of EGCG is due to their decreased substrate binding affinity and reduced maximal transport rate. The overall transport efficiencies of OATP1B3, 1B3-TM8, and 1B3-V386F in the absence and presence of EGCG are 8.6 ± 0.7 vs 15.9 ± 1.4 (p < 0.05), 11.2 ± 2.1 vs 2.7 ± 0.3 (p < 0.05), and 10.2 ± 1.0 vs 2.5 ± 0.3 (p < 0.05), respectively. While 1B3-V386F cannot be activated by EGCG, its transport activity for EGCG is also diminished. OATP1B3's activation by EGCG is substrate-dependent as EGCG inhibits OATP1B3-mediated pravastatin uptake. Furthermore, the activation of OATP1B3-mediated E3S uptake by quercetin 3-O-α-l-arabinopyranosyl(1 → 2)-α-l-rhamnopyranoside is not affected by TM8 and V386F. Taken together, the activation of OATP1B3 by small molecules is substrate- and modulator-dependent, and V386 in TM8 plays a critical role in the activation of OATP1B3-mediated E3S uptake by EGCG.
Collapse
Affiliation(s)
- Zhongmin Wang
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Ying Li
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Cecilia E. Villanueva
- Department of Pharmacology, Toxicology and Therapeutics, the University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, Kansas 66160, United States
| | - Taotao Peng
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Wanjun Han
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Zheyue Bo
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Hongjian Zhang
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, the University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, Kansas 66160, United States
| | - Chunshan Gui
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, People’s Republic of China
| |
Collapse
|
15
|
Oorts M, Van Brantegem P, Deferm N, Chatterjee S, Dreesen E, Cooreman A, Vinken M, Richert L, Annaert P. Bosentan Alters Endo- and Exogenous Bile Salt Disposition in Sandwich-Cultured Human Hepatocytes. J Pharmacol Exp Ther 2021; 379:20-32. [PMID: 34349015 DOI: 10.1124/jpet.121.000695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/28/2021] [Indexed: 01/29/2023] Open
Abstract
Bosentan, a well-known cholestatic agent, was not identified as cholestatic at concentrations up to 200 µM based on the drug-induced cholestasis (DIC) index value, determined in a sandwich-cultured human hepatocyte (SCHH)-based DIC assay. To obtain further quantitative insights into the effects of bosentan on cellular bile salt handling by human hepatocytes, the present study determined the effect of 2.5-25 µM bosentan on endogenous bile salt levels and on the disposition of 10 µM chenodeoxycholic acid (CDCA) added to the medium in SCHHs. Bosentan reduced intracellular as well as extracellular concentrations of both endogenous glycochenodeoxycholic acid (GCDCA) and glycocholic acid in a concentration-dependent manner. When exposed to 10 µM CDCA, bosentan caused a shift from canalicular efflux to sinusoidal efflux of GCDCA. CDCA levels were not affected. Our mechanistic model confirmed the inhibitory effect of bosentan on canalicular GCDCA clearance. Moreover, our results in SCHHs also indicated reduced GCDCA formation. We confirmed the direct inhibitory effect of bosentan on CDCA conjugation with glycine in incubations with liver S9 fraction. SIGNIFICANCE STATEMENT: Bosentan was evaluated at therapeutically relevant concentrations (2.5-25 µM) in sandwich-cultured human hepatocytes. It altered bile salt disposition and inhibited canalicular secretion of glycochenodeoxycholic acid (GCDCA). Within 24 hours, bosentan caused a shift from canalicular to sinusoidal efflux of GCDCA. These results also indicated reduced GCDCA formation. This study confirmed a direct effect of bosentan on chenodeoxycholic acid conjugation with glycine in liver S9 fraction.
Collapse
Affiliation(s)
- Marlies Oorts
- Drug Delivery and Disposition (M.O., P.V.B., N.D., P.A.) and Clinical Pharmacology and Pharmacotherapy (E.D.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (A.C.); Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb Research Center, Syngene International, Bangalore, India (S.C.); Uppsala Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden (E.D.); Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium (M.V.); KaLy-Cell, Plobsheim, France (L.R.); and BioNotus, Niel, Belgium (P.A.)
| | - Pieter Van Brantegem
- Drug Delivery and Disposition (M.O., P.V.B., N.D., P.A.) and Clinical Pharmacology and Pharmacotherapy (E.D.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (A.C.); Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb Research Center, Syngene International, Bangalore, India (S.C.); Uppsala Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden (E.D.); Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium (M.V.); KaLy-Cell, Plobsheim, France (L.R.); and BioNotus, Niel, Belgium (P.A.)
| | - Neel Deferm
- Drug Delivery and Disposition (M.O., P.V.B., N.D., P.A.) and Clinical Pharmacology and Pharmacotherapy (E.D.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (A.C.); Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb Research Center, Syngene International, Bangalore, India (S.C.); Uppsala Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden (E.D.); Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium (M.V.); KaLy-Cell, Plobsheim, France (L.R.); and BioNotus, Niel, Belgium (P.A.)
| | - Sagnik Chatterjee
- Drug Delivery and Disposition (M.O., P.V.B., N.D., P.A.) and Clinical Pharmacology and Pharmacotherapy (E.D.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (A.C.); Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb Research Center, Syngene International, Bangalore, India (S.C.); Uppsala Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden (E.D.); Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium (M.V.); KaLy-Cell, Plobsheim, France (L.R.); and BioNotus, Niel, Belgium (P.A.)
| | - Erwin Dreesen
- Drug Delivery and Disposition (M.O., P.V.B., N.D., P.A.) and Clinical Pharmacology and Pharmacotherapy (E.D.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (A.C.); Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb Research Center, Syngene International, Bangalore, India (S.C.); Uppsala Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden (E.D.); Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium (M.V.); KaLy-Cell, Plobsheim, France (L.R.); and BioNotus, Niel, Belgium (P.A.)
| | - Axelle Cooreman
- Drug Delivery and Disposition (M.O., P.V.B., N.D., P.A.) and Clinical Pharmacology and Pharmacotherapy (E.D.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (A.C.); Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb Research Center, Syngene International, Bangalore, India (S.C.); Uppsala Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden (E.D.); Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium (M.V.); KaLy-Cell, Plobsheim, France (L.R.); and BioNotus, Niel, Belgium (P.A.)
| | - Mathieu Vinken
- Drug Delivery and Disposition (M.O., P.V.B., N.D., P.A.) and Clinical Pharmacology and Pharmacotherapy (E.D.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (A.C.); Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb Research Center, Syngene International, Bangalore, India (S.C.); Uppsala Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden (E.D.); Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium (M.V.); KaLy-Cell, Plobsheim, France (L.R.); and BioNotus, Niel, Belgium (P.A.)
| | - Lysiane Richert
- Drug Delivery and Disposition (M.O., P.V.B., N.D., P.A.) and Clinical Pharmacology and Pharmacotherapy (E.D.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (A.C.); Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb Research Center, Syngene International, Bangalore, India (S.C.); Uppsala Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden (E.D.); Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium (M.V.); KaLy-Cell, Plobsheim, France (L.R.); and BioNotus, Niel, Belgium (P.A.)
| | - Pieter Annaert
- Drug Delivery and Disposition (M.O., P.V.B., N.D., P.A.) and Clinical Pharmacology and Pharmacotherapy (E.D.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (A.C.); Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb Research Center, Syngene International, Bangalore, India (S.C.); Uppsala Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden (E.D.); Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium (M.V.); KaLy-Cell, Plobsheim, France (L.R.); and BioNotus, Niel, Belgium (P.A.)
| |
Collapse
|
16
|
Leuenberger M, Häusler S, Höhn V, Euler A, Stieger B, Lochner M. Characterization of Novel Fluorescent Bile Salt Derivatives for Studying Human Bile Salt and Organic Anion Transporters. J Pharmacol Exp Ther 2021; 377:346-357. [PMID: 33782042 DOI: 10.1124/jpet.120.000449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/23/2021] [Indexed: 11/22/2022] Open
Abstract
Bile salts, such as cholate, glycocholate, taurocholate, and glycochenodeoxycholate, are taken up from the portal blood into hepatocytes via transporters, such as the Na+-taurocholate-cotransporting polypeptide (NTCP) and organic anion-transporting polypeptides (OATPs). These bile salts are later secreted into bile across the canalicular membrane, which is facilitated by the bile salt export pump (BSEP). Apart from bile salt transport, some of these proteins (e.g., OATPs) are also key transporters for drug uptake into hepatocytes. In vivo studies of transporter function in patients by using tracer compounds have emerged as an important diagnostic tool to complement classic liver parameter measurements by determining dynamic liver function both for diagnosis and monitoring progression or improvement of liver diseases. Such approaches include use of radioactively labeled bile salts (e.g., for positron emission tomography) and fluorescent bile salt derivatives or dyes (e.g., indocyanine green). To expand the list of liver function markers, we synthesized fluorescent derivatives of cholic and chenodeoxycholic acid by conjugating small organic dyes to the bile acid side chain. These novel fluorescent probes were able to block substrate transport in a concentration-dependent manner of NTCP, OATP1B1, OATP1B3, OATP2B1, BSEP, and intestinal apical sodium-dependent bile salt transporter (ASBT). Whereas the fluorescent bile acid derivatives themselves were transported across the membrane by OATP1B1, OATP1B3, and OATP2B1, they were not transport substrates for NTCP, ASBT, BSEP, and multidrug resistance-related protein 2. Accordingly, these novel fluorescent bile acid probes can potentially be used as imaging agents to monitor the function of OATPs. SIGNIFICANCE STATEMENT: Synthetic modification of common bile acids by attachment of small organic fluorescent dyes to the bile acid side chain resulted in bright, fluorescent probes that interact with hepatic and intestinal organic anion [organic anion-transporting polypeptide (OATP) 1B1, OATP1B3, OATP2B1], bile salt uptake (Na+-taurocholate-cotransporting polypeptide, apical sodium-dependent bile salt transporter), and bile salt efflux (bile salt export pump, multidrug resistance-related protein 2) transporters. Although the fluorescent bile salt derivatives are taken up into cells via the OATPs, the efflux transporters do not transport any of them but one.
Collapse
Affiliation(s)
- Michele Leuenberger
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland (M.Le., M.Lo.); Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland (S.H., V.H., A.E., B.S.); and Swiss National Center of Competence in Research, NCCR TransCure, Bern, Switzerland (M.Le., S.H., A.E., B.S., M.Lo.)
| | - Stephanie Häusler
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland (M.Le., M.Lo.); Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland (S.H., V.H., A.E., B.S.); and Swiss National Center of Competence in Research, NCCR TransCure, Bern, Switzerland (M.Le., S.H., A.E., B.S., M.Lo.)
| | - Vera Höhn
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland (M.Le., M.Lo.); Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland (S.H., V.H., A.E., B.S.); and Swiss National Center of Competence in Research, NCCR TransCure, Bern, Switzerland (M.Le., S.H., A.E., B.S., M.Lo.)
| | - Adriana Euler
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland (M.Le., M.Lo.); Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland (S.H., V.H., A.E., B.S.); and Swiss National Center of Competence in Research, NCCR TransCure, Bern, Switzerland (M.Le., S.H., A.E., B.S., M.Lo.)
| | - Bruno Stieger
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland (M.Le., M.Lo.); Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland (S.H., V.H., A.E., B.S.); and Swiss National Center of Competence in Research, NCCR TransCure, Bern, Switzerland (M.Le., S.H., A.E., B.S., M.Lo.)
| | - Martin Lochner
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland (M.Le., M.Lo.); Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland (S.H., V.H., A.E., B.S.); and Swiss National Center of Competence in Research, NCCR TransCure, Bern, Switzerland (M.Le., S.H., A.E., B.S., M.Lo.)
| |
Collapse
|
17
|
Ouranidis A, Tsiaxerli A, Vardaka E, Markopoulou CK, Zacharis CK, Nicolaou I, Hatzichristou D, Haidich AB, Kostomitsopoulos N, Kachrimanis K. Sildenafil 4.0-Integrated Synthetic Chemistry, Formulation and Analytical Strategies Effecting Immense Therapeutic and Societal Impact in the Fourth Industrial Era. Pharmaceuticals (Basel) 2021; 14:365. [PMID: 33920975 PMCID: PMC8071249 DOI: 10.3390/ph14040365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022] Open
Abstract
Sildenafil is a potent selective, reversible inhibitor of phosphodiesterase type 5 (PDE5) approved for the treatment of erectile dysfunction and pulmonary arterial hypertension. Whilst twenty years have passed since its original approval by the US Food and Drug Administration (USFDA), sildenafil enters the fourth industrial era catalyzing the treatment advances against erectile dysfunction and pulmonary hypertension. The plethora of detailed clinical data accumulated and the two sildenafil analogues marketed, namely tadalafil and vardenafil, signify the relevant therapeutic and commercial achievements. The pharmacokinetic and pharmacodynamic behavior of the drug appears complex, interdependent and of critical importance whereas the treatment of special population cohorts is considered. The diversity of the available formulation strategies and their compatible administration routes, extend from tablets to bolus suspensions and from per os to intravenous, respectively, inheriting the associated strengths and weaknesses. In this comprehensive review, we attempt to elucidate the multi-disciplinary elements spanning the knowledge fields of chemical synthesis, physicochemical properties, pharmacology, clinical applications, biopharmaceutical profile, formulation approaches for different routes of administration and analytical strategies, currently employed to guide the development of sildenafil-based compositions.
Collapse
Affiliation(s)
- Andreas Ouranidis
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (E.V.)
- Department of Chemical Engineering, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Anastasia Tsiaxerli
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (E.V.)
| | - Elisavet Vardaka
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (E.V.)
| | - Catherine K. Markopoulou
- Laboratory of Pharmaceutical Analysis, Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (C.K.M.); (C.K.Z.)
| | - Constantinos K. Zacharis
- Laboratory of Pharmaceutical Analysis, Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (C.K.M.); (C.K.Z.)
| | - Ioannis Nicolaou
- Laboratory of Pharmaceutical Chemistry, Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Dimitris Hatzichristou
- Department of Urology, Medical School, Aristotle University of Thessaloniki, 54635 Thessaloniki, Greece;
| | - Anna-Bettina Haidich
- Department of Hygiene, Social-Preventive Medicine and Medical Statistics, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Nikolaos Kostomitsopoulos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Kyriakos Kachrimanis
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (E.V.)
| |
Collapse
|
18
|
Koyama S, Toshimoto K, Lee W, Aoki Y, Sugiyama Y. Revisiting Nonlinear Bosentan Pharmacokinetics by Physiologically Based Pharmacokinetic Modeling: Target Binding, Albeit Not a Major Contributor to Nonlinearity, Can Offer Prediction of Target Occupancy. Drug Metab Dispos 2021; 49:298-304. [PMID: 33558262 DOI: 10.1124/dmd.120.000023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 01/15/2021] [Indexed: 11/22/2022] Open
Abstract
Bosentan is a high-affinity antagonist of endothelin receptors and one of the earliest examples for target-mediated drug disposition [a type of nonlinear pharmacokinetics (PKs) caused by saturable target binding]. The previous physiologically based PK (PBPK) modeling indicated that the nonlinear PKs of bosentan was explainable by considering saturable hepatic uptake. However, it remained unexamined to what extent the saturable target binding contributes to the nonlinear PKs of bosentan. Here, we developed a PBPK model incorporating saturable target binding and hepatic uptake and analyzed the clinical bosentan PK data using the cluster Gauss-Newton method (CGNM). The PBPK model without target binding fell short in capturing the bosentan concentrations below 100 nM, based on the PK profiles and the goodness-of-fit plot. Both global and local identifiability analyses (using the CGNM and Fisher information matrix, respectively) informed that the target binding parameters were identifiable only if the observations from the lowest dose (10 mg) were included. By analyzing blood PK profiles alone, the PBPK model with target binding yielded practically identifiable target binding parameters and predicted the maximum target occupancies of 0.6-0.8 at clinical bosentan doses. Our results indicate that target binding, albeit not a major contributor to the nonlinear bosentan PKs, may offer a prediction of target occupancy from blood PK profiles alone and potential guidance on achieving optimal efficacy outcomes, under the condition when the high-affinity drug target is responsible for the efficacy of interest and when the dose ranges cover varying degrees of target binding. SIGNIFICANCE STATEMENT: By incorporating saturable target binding, our physiologically based pharmacokinetic (PBPK) model predicted in vivo target occupancy of bosentan based only on the blood concentration-time profiles obtained from a wide range of doses. Our analysis highlights the potential utility of PBPK models that incorporate target binding in predicting target occupancy in vivo.
Collapse
Affiliation(s)
- Satoshi Koyama
- Sugiyama Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Kanagawa, Japan (S.K., K.T., Y.S.); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-gu, Seoul, Korea (W.L.); and National Institute of Informatics, Chiyoda-ku, Tokyo, Japan (Y.A.)
| | - Kota Toshimoto
- Sugiyama Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Kanagawa, Japan (S.K., K.T., Y.S.); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-gu, Seoul, Korea (W.L.); and National Institute of Informatics, Chiyoda-ku, Tokyo, Japan (Y.A.)
| | - Wooin Lee
- Sugiyama Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Kanagawa, Japan (S.K., K.T., Y.S.); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-gu, Seoul, Korea (W.L.); and National Institute of Informatics, Chiyoda-ku, Tokyo, Japan (Y.A.)
| | - Yasunori Aoki
- Sugiyama Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Kanagawa, Japan (S.K., K.T., Y.S.); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-gu, Seoul, Korea (W.L.); and National Institute of Informatics, Chiyoda-ku, Tokyo, Japan (Y.A.)
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Kanagawa, Japan (S.K., K.T., Y.S.); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-gu, Seoul, Korea (W.L.); and National Institute of Informatics, Chiyoda-ku, Tokyo, Japan (Y.A.)
| |
Collapse
|
19
|
Ma R, Li G, Wang X, Bi Y, Zhang Y. Inhibitory effect of sixteen pharmaceutical excipients on six major organic cation and anion uptake transporters. Xenobiotica 2020; 51:95-104. [PMID: 32544367 DOI: 10.1080/00498254.2020.1783720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To date, relatively little is known about the interactions of pharmaceutical excipients with hepatic and renal drug uptake transporters. The present study was designed to systematically evaluate the effects of 16 commonly consumed excipients on human organic cation transporter 1 and 2 (hOCT1 and hOCT2), human organic anion transporter 1 and 3 (hOAT1 and hOAT3) and human organic anion transporting polypeptide 1B1 and 1B3 (hOATP1B1 and hOATP1B3). The inhibitory effects and mechanisms of excipients on transporters were investigated using in vitro uptake studies, cell viability assays, concentration-dependent studies, and the Lineweaver-Burk plot method. Triton X-100 is a non-competitive inhibitor for all six transporters. Tween 20 inhibits hOCT2, hOAT1, hOAT3, and hOATP1B3 in a mixed way, whereas it competitively inhibits hOATP1B1. The inhibition of Tween 80 is competitive for hOCT2, non-competitive for hOATP1B1 and hOATP1B3, and mixed for hOAT1 and hOAT3. Concentration-dependent studies identify Triton X-100 as a strong inhibitor of hOCT1 and hOCT2 with IC50 values of 20.1 and 4.54 μg/mL, respectively. Additionally, Triton X-100, Tween 20, and Tween 80 strongly inhibit hOAT3 with IC50 values ≤31.0 μg/mL. The present study is significant in understanding the excipient-drug interactions and provides valuable information for excipient selection in drug development.
Collapse
Affiliation(s)
- Ruicong Ma
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Gentao Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xue Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Yajuan Bi
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
20
|
Yorifuji K, Uemura Y, Horibata S, Tsuji G, Suzuki Y, Nakayama K, Hatae T, Kumagai S, Emoto N. Predictive model of bosentan-induced liver toxicity in Japanese patients with pulmonary arterial hypertension. Can J Physiol Pharmacol 2020; 98:625-628. [PMID: 32433892 DOI: 10.1139/cjpp-2019-0656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Bosentan, an endothelin receptor antagonist, has been widely used as a first-line medication for the treatment of pulmonary arterial hypertension (PAH). It has been shown to improve symptoms of hypertension, exercise capacity, and hemodynamics and prolong time to clinical worsening. However, liver dysfunction is a major side effect of bosentan treatment that could hamper the optimal management of patients with PAH. Previously, we demonstrated, using drug metabolism enzymes and transporters analysis, that the carbohydrate sulfotransferase 3 (CHST3) and CHST13 alleles are significantly more frequent in patients with elevated aminotransferases during therapy with bosentan than they are in patients without liver toxicity. In addition, we constructed a pharmacogenomics model to predict bosentan-induced liver injury in patients with PAH using two single-nucleotide polymorphisms and two nongenetic factors. The purpose of the present study was to externally validate the predictive model of bosentan-induced liver toxicity in Japanese patients. We evaluated five cases of patients treated with bosentan, and one presented with liver dysfunction. We applied mutation alleles of CHST3 and CHST13, serum creatinine, and age to our model to predict liver dysfunction. The sensitivity and specificity were calculated as 100% and 50%, respectively. Considering that PAH is a rare disease, multicenter collaboration would be necessary to validate our model.
Collapse
Affiliation(s)
- Kennosuke Yorifuji
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama-kitamachi, Higashinada, Kobe 658-8558, Japan
- The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
- Department of Pharmacy, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Yuko Uemura
- The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Shinji Horibata
- The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
- Department of Pharmacy, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Goh Tsuji
- The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
- Center for Rheumatic Diseases, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Yoko Suzuki
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama-kitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 650-0017, Japan
| | - Kazuhiko Nakayama
- Department of Cardiovascular Medicine, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Takashi Hatae
- Education and Research Center for Clinical Pharmacy, Kobe Pharmaceutical University, 4-19-1 Motoyama-kitamachi, Higashinada, Kobe 658-8558, Japan
| | - Shunichi Kumagai
- The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
- Center for Rheumatic Diseases, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama-kitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 650-0017, Japan
| |
Collapse
|
21
|
Treiber A, Delahaye S, Seeland S, Gnerre C. The endothelin receptor antagonist macitentan for the treatment of pulmonary arterial hypertension: A cross-species comparison of its cytochrome P450 induction pattern. Pharmacol Res Perspect 2020; 8:e00619. [PMID: 32613761 PMCID: PMC7330163 DOI: 10.1002/prp2.619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 11/18/2022] Open
Abstract
The dual endothelin receptor antagonist macitentan was approved in 2013 for the treatment of pulmonary arterial hypertension. Macitentan is an inducer of cytochrome P450 expression in vivo in animal species but not in man. In rat and dog, changes in P450 expression manifest as autoinduction upon repeat dosing. The induction pattern, however, significantly differed between both species, and between male and female rats. While macitentan exposure steadily declined with dose in the dog, P450 induction was saturable in the rat reaching levels of 40%-60% and 60%-80% at steady-state in male and female animals, respectively. The nature and number of P450 enzymes involved in macitentan clearance were identified as a major reason for the observed species differences. In the dog, macitentan was metabolized by a single P450 enzyme, that is, Cyp3a12, whereas several members of the Cyp2c and Cyp3a families were involved in the rat. Macitentan selectively upregulated Cyp3a expression in rat, whereas the expression of the Cyp2c enzymes involved in macitentan metabolism remained mostly unchanged, eventually leading to a higher contribution of Cyp3a upon induction. Macitentan also induced CYP3A4 expression in human hepatocytes via initial activation of the human pregnane X receptor. No such induction was evident in humans at the therapeutic macitentan dose of 10 mg as shown in a clinical drug-drug interaction study with the CYP3A4 substrate sildenafil.
Collapse
Affiliation(s)
- Alexander Treiber
- Department of Non‐Clinical Drug Metabolism and PharmacokineticsIdorsia Pharmaceuticals LtdAllschwilSwitzerland
| | - Stephane Delahaye
- Department of Non‐Clinical Drug Metabolism and PharmacokineticsIdorsia Pharmaceuticals LtdAllschwilSwitzerland
| | - Swen Seeland
- Department of Non‐Clinical Drug Metabolism and PharmacokineticsIdorsia Pharmaceuticals LtdAllschwilSwitzerland
| | - Carmela Gnerre
- Department of Non‐Clinical Drug Metabolism and PharmacokineticsIdorsia Pharmaceuticals LtdAllschwilSwitzerland
| |
Collapse
|
22
|
Ogawa SI, Shimizu M, Kamiya Y, Uehara S, Suemizu H, Yamazaki H. Increased plasma concentrations of an antidyslipidemic drug pemafibrate co-administered with rifampicin or cyclosporine A in cynomolgus monkeys genotyped for the organic anion transporting polypeptide 1B1. Drug Metab Pharmacokinet 2020; 35:354-360. [DOI: 10.1016/j.dmpk.2020.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/02/2020] [Accepted: 03/24/2020] [Indexed: 12/21/2022]
|
23
|
Influence of Single Nucleotide Polymorphisms on Rifampin Pharmacokinetics in Tuberculosis Patients. Antibiotics (Basel) 2020; 9:antibiotics9060307. [PMID: 32521634 PMCID: PMC7344705 DOI: 10.3390/antibiotics9060307] [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: 05/06/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/03/2022] Open
Abstract
Rifampin (RF) is metabolized in the liver into an active metabolite 25-desacetylrifampin and excreted almost equally via biliary and renal routes. Various influx and efflux transporters influence RF disposition during hepatic uptake and biliary excretion. Evidence has also shown that Vitamin D deficiency (VDD) and Vitamin D receptor (VDR) polymorphisms are associated with tuberculosis (TB). Hence, genetic polymorphisms of metabolizing enzymes, drug transporters and/or their transcriptional regulators and VDR and its pathway regulators may affect the pharmacokinetics of RF. In this narrative review, we aim to identify literature that has explored the influence of single nucleotide polymorphisms (SNPs) of genes encoding drug transporters and their transcriptional regulators (SLCO1B1, ABCB1, PXR and CAR), metabolizing enzymes (CES1, CES2 and AADAC) and VDR and its pathway regulators (VDR, CYP27B1 and CYP24A1) on plasma RF concentrations in TB patients on antitubercular therapy. Available reports to date have shown that there is a lack of any association of ABCB1, PXR, CAR, CES1 and AADAC genetic variants with plasma concentrations of RF. Further evidence is required from a more comprehensive exploration of the association of SLCO1B1, CES2 and Vitamin D pathway gene variants with RF pharmacokinetics in distinct ethnic groups and a larger population to reach conclusive information.
Collapse
|
24
|
Ben Salem C, Sahnoun D, Slim R, Ben Fradj F, Aouani C. Atorvastatin and sildenafil interaction-induced rhabdomyolysis. Ann Pharmacother 2020; 54:1047-1048. [PMID: 32418442 DOI: 10.1177/1060028020919933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
25
|
Investigation of the interactions between flavonoids and human organic anion transporting polypeptide 1B1 using fluorescent substrate and 3D-QSAR analysis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183210. [PMID: 32006472 DOI: 10.1016/j.bbamem.2020.183210] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/10/2020] [Accepted: 01/27/2020] [Indexed: 01/31/2023]
Abstract
Organic anion transporting polypeptide 1B1 (OATP1B1) is a key hepatic uptake transporter whose inhibition could lead to adverse drug-drug and drug-food interactions. Flavonoids are widely distributed in food and beverages and thus our bodies are frequently exposed to them. Therefore, investigation of the interactions between OATP1B1 and flavonoids could be of great significance. In the present study, 25 common flavonoids were investigated for their interactions with OATP1B1 using the fluorescent substrate 2',7'-dichlorofluorescein (DCF) and three-dimensional quantitative structure-activity relationship (3D-QSAR) analysis. Kinetic study showed that OATP1B1-mediated DCF uptake exhibited a monophasic saturation kinetics with a Km value of 9.7 ± 2.4 μM. Inhibition assay for flavonoids on OATP1B1-mediated DCF uptake was performed and their IC50 values were determined upon which reliable and predictive CoMFA (q2 = 0.604, r2 = 0.841) and CoMSIA (q2 = 0.534, r2 = 0.807) models were developed. Our experimental and computational results showed that flavonoid aglycones interacted with OATP1B1 much stronger than their glycosides such as 3-O- and 7-O-glycosides as bulky hydrophilic and hydrogen-bond forming substituents at C-3 and C-7 positions on rings A and C were unfavorable for their binding. On the other hand, the presence of hydrogen-bond forming groups on ring B was beneficial as long as the number of hydroxyl groups was not >2. Our results also indicated that flavones usually interacted with OATP1B1 much stronger than their 3-hydroxyflavone counterparts (flavonols). The obtained information and 3D-QSAR models could be useful for elucidating and predicting the interactions between flavonoids and human OATP1B1.
Collapse
|
26
|
Fan X, Bai J, Hu M, Xu Y, Zhao S, Sun Y, Wang B, Hu J, Li Y. Drug interaction study of flavonoids toward OATP1B1 and their 3D structure activity relationship analysis for predicting hepatoprotective effects. Toxicology 2020; 437:152445. [PMID: 32259555 DOI: 10.1016/j.tox.2020.152445] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/14/2020] [Accepted: 03/25/2020] [Indexed: 12/22/2022]
Abstract
Organic anion transporting polypeptide 1B1 (OATP1B1), a liver-specific uptake transporter, was associated with drug induced liver injury (DILI). Screening and identifying potent OATP1B1 inhibitors with little toxicity is of great value in reducing OATP1B1-mediated DILI. Flavonoids are a group of polyphenols ubiquitously present in vegetables, fruits and herbal products, some of them were reported to produce transporter-mediated DDI. Our objective was to investigate potential inhibitors of OATP1B1 from 99 flavonoids, and to assess the hepatoprotective effects on bosentan induced liver injury. Eight flavonoids, including biochanin A, hispidulin, isoliquiritigenin, isosinensetin, kaempferol, licochalcone A, luteolin and sinensetin exhibited significant inhibition (>50 %) on OATP1B1 in OATP1B1-HEK293 cells, which reduced the OATP1B1-mediated influx of methotrexate, accordingly decreased its cytotoxicity in OATP1B1-HEK293 cells and increased its AUC0-t in different extents in rats, from 28.27%-82.71 %. In bosentan-induced rat liver injury models, 8 flavonoids reduced the levels of serum total bile acid (TBA) and the liver concentration of bosentan in different degrees. Among them, kaempferol decreased the concentration most significantly, by 54.17 %, which indicated that flavonoids may alleviate bosentan-induced liver injury by inhibiting OATP1B1-mediated bosentan uptake. Furthermore, the pharmacophore model indicated the hydrogen bond acceptors and hydrogen bond donors may play critical role in the potency of flavonoids inhibition on OATP1B1. Taken together, our findings would provide helpful information for predicting the potential risks of flavonoid-containing food/herb-drug interactions in humans and alleviating bosentan -induced liver injury by OATP1B1 regulation.
Collapse
Affiliation(s)
- Xiaoqing Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jie Bai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Minwan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanxia Xu
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Shengyu Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanhong Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Baolian Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jinping Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Yan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
27
|
Zhi H, Yuan Y, Zhang C, Jiang Y, Zhang H, Wang C, Ruan J. Importance of OATP1B1 and 1B3 in the Liver Uptake of Luteolin and Its Consequent Glucuronidation Metabolites. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:2063-2070. [PMID: 32009392 DOI: 10.1021/acs.jafc.9b06954] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Luteolin is a typical flavonoid and broadly distributed in the plants. Oral bioavailability of luteolin is low owing to extensive metabolism. Regioselective glucuronidation by UDP-glucuronosyltransferases (UGTs) and liver uptake by organic anion transporting polypeptides (OATPs) of luteolin and consequent glucuronidation metabolites were studied. Luteolin-3'-O-glucuronide (L-3'-G) and luteolin-7-O-glucuronide (L-7-G) were the major metabolites in human liver microsomes. Further study demonstrated that UGT1A9 played a predominant role in the glucuronidation of luteolin. Transporter study showed that OATP1B1- and 1B3-transfected cells selectively uptake L-3'-G into cells but not luteolin or L-7-G. After intravenous administration of luteolin to mice, the area under the curve of L-3'-G in the plasma was the highest among luteolin, L-3'-G, and L-7-G. In the liver, the concentration of L-3'-G was significantly greater than L-7-G. In conclusion, OATP1B1 and OATP1B3 play an important role in the liver disposition of luteolin and its glucuronidation metabolites.
Collapse
Affiliation(s)
- Hui Zhi
- College of Pharmaceutical Sciences , Soochow University , Suzhou 215123 , China
- Clinical Pharmacy Lab, Department of Pharmacy , The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University , Suzhou 215123 , China
| | - Yuan Yuan
- Department of Pharmacy , The Affiliated Wuxi Matemity and Child Health Care Hospital of Nanjing Medical University , Wuxi 214000 , China
| | - Chunzhen Zhang
- College of Pharmaceutical Sciences , Soochow University , Suzhou 215123 , China
| | - Yiguo Jiang
- Clinical Pharmacy Lab, Department of Pharmacy , The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University , Suzhou 215123 , China
| | - Hongjian Zhang
- Clinical Pharmacy Lab, Department of Pharmacy , The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University , Suzhou 215123 , China
| | - Cheng Wang
- Clinical Pharmacy Lab, Department of Pharmacy , The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University , Suzhou 215123 , China
| | - Jianqing Ruan
- College of Pharmaceutical Sciences , Soochow University , Suzhou 215123 , China
| |
Collapse
|
28
|
Han LW, Gao C, Zhang Y, Wang J, Mao Q. Transport of Bupropion and its Metabolites by the Model CHO and HEK293 Cell Lines. Drug Metab Lett 2020; 13:25-36. [PMID: 30488806 DOI: 10.2174/1872312813666181129101507] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/19/2018] [Accepted: 11/07/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Bupropion (BUP) is widely used as an antidepressant and smoking cessation aid. There are three major pharmacologically active metabolites of BUP, Erythrohydrobupropion (EB), Hydroxybupropion (OHB) and Threohydrobupropion (TB). At present, the mechanisms underlying the overall disposition and systemic clearance of BUP and its metabolites have not been well understood, and the role of transporters has not been studied. OBJECTIVE The goal of this study was to investigate whether BUP and its active metabolites are substrates of the major hepatic uptake and efflux transporters. METHOD CHO or HEK293 cell lines or plasma membrane vesicles that overexpress OATP1B1, OATP1B3, OATP2B1, OATP4A1, OCT1, BCRP, MRP2 or P-gp were used in cellular or vesicle uptake and inhibition assays. Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) was used to quantify transport activity. RESULTS BUP and its major active metabolites were actively transported into the CHO or HEK293 cells overexpressing OATP1B1, OATP1B3 or OATP2B1; however, such cellular active uptake could not be inhibited at all by prototypical inhibitors of any of the OATP transporters. These compounds were not transported by OCT1, BCRP, MRP2 or P-gp either. These results suggest that the major known hepatic transporters likely play a minor role in the overall disposition and systemic clearance of BUP and its active metabolites in humans. We also demonstrated that BUP and its metabolites were not transported by OATP4A1, an uptake transporter on the apical membrane of placental syncytiotrophoblasts, suggesting that OATP4A1 is not responsible for the transfer of BUP and its metabolites from the maternal blood to the fetal compartment across the placental barrier in pregnant women. CONCLUSION BUP and metabolites are not substrates of the major hepatic transporters tested and thus these hepatic transporters likely do not play a role in the overall disposition of the drug. Our results also suggest that caution should be taken when using the model CHO and HEK293 cell lines to evaluate potential roles of transporters in drug disposition.
Collapse
Affiliation(s)
- Lyrialle W Han
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Chunying Gao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Yuchen Zhang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Joanne Wang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
29
|
Liu X. Transporter-Mediated Drug-Drug Interactions and Their Significance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:241-291. [PMID: 31571167 DOI: 10.1007/978-981-13-7647-4_5] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Drug transporters are considered to be determinants of drug disposition and effects/toxicities by affecting the absorption, distribution, and excretion of drugs. Drug transporters are generally divided into solute carrier (SLC) family and ATP binding cassette (ABC) family. Widely studied ABC family transporters include P-glycoprotein (P-GP), breast cancer resistance protein (BCRP), and multidrug resistance proteins (MRPs). SLC family transporters related to drug transport mainly include organic anion-transporting polypeptides (OATPs), organic anion transporters (OATs), organic cation transporters (OCTs), organic cation/carnitine transporters (OCTNs), peptide transporters (PEPTs), and multidrug/toxin extrusions (MATEs). These transporters are often expressed in tissues related to drug disposition, such as the small intestine, liver, and kidney, implicating intestinal absorption of drugs, uptake of drugs into hepatocytes, and renal/bile excretion of drugs. Most of therapeutic drugs are their substrates or inhibitors. When they are comedicated, serious drug-drug interactions (DDIs) may occur due to alterations in intestinal absorption, hepatic uptake, or renal/bile secretion of drugs, leading to enhancement of their activities or toxicities or therapeutic failure. This chapter will illustrate transporter-mediated DDIs (including food drug interaction) in human and their clinical significances.
Collapse
Affiliation(s)
- Xiaodong Liu
- China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
30
|
Pan G. Roles of Hepatic Drug Transporters in Drug Disposition and Liver Toxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:293-340. [PMID: 31571168 DOI: 10.1007/978-981-13-7647-4_6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatic drug transporters are mainly distributed in parenchymal liver cells (hepatocytes), contributing to drug's liver disposition and elimination. According to their functions, hepatic transporters can be roughly divided into influx and efflux transporters, translocating specific molecules from blood into hepatic cytosol and mediating the excretion of drugs and metabolites from hepatic cytosol to blood or bile, respectively. The function of hepatic transport systems can be affected by interspecies differences and inter-individual variability (polymorphism). In addition, some drugs and disease can redistribute transporters from the cell surface to the intracellular compartments, leading to the changes in the expression and function of transporters. Hepatic drug transporters have been associated with the hepatic toxicity of drugs. Gene polymorphism of transporters and altered transporter expressions and functions due to diseases are found to be susceptible factors for drug-induced liver injury (DILI). In this chapter, the localization of hepatic drug transporters, their regulatory factors, physiological roles, and their roles in drug's liver disposition and DILI are reviewed.
Collapse
Affiliation(s)
- Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, Shanghai, China.
| |
Collapse
|
31
|
Yue M, Yang J, Jin M, Steiert B, Xiang Y, Zhang H, Hagenbuch B, Gui C. Gly45 and Phe555 in Transmembrane Domains 1 and 10 Are Critical for the Activation of Organic Anion Transporting Polypeptide 1B3 by Epigallocatechin Gallate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9079-9087. [PMID: 31353905 PMCID: PMC6892160 DOI: 10.1021/acs.jafc.9b03812] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Organic anion transporting polypeptides (OATPs) 1B1 and 1B3 are two highly homologous transporters expressed in the human liver. However, epigallocatechin gallate (EGCG), which is the most predominant catechin in green tea, has opposite effects on the function of OATP1B1 and OATP1B3. In the present study, the critical structural domains and amino acid residues for the activation of OATP1B3 by EGCG have been determined by characterizing the function of a series of OATP1B3-derived chimeric transporters, site-directed mutagenesis, and kinetic studies. Our results showed that G45 and F555 in transmembrane domains 1 and 10 are the most important amino acid residues for OATP1B3 activation. Kinetic studies showed that the activation of OATP1B3 by EGCG at a low substrate concentration was due to its increased substrate binding affinity. However, EGCG caused increased Km and decreased Vmax for 1B3-G45A and 1B3-F555H. The flexibility at position 45 and aromaticity at position 555 might be important for OATP1B3 activation. While 1B3-G45A and 1B3-F555H could not be activated by EGCG, their transport activity for EGCG was comparable to that of wild-type OATP1B3. In conclusion, the present study elucidated the molecular mechanism for OATP1B3 activation by EGCG.
Collapse
Affiliation(s)
- Mei Yue
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jingjie Yang
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Meng Jin
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Brianna Steiert
- Department of Pharmacology, Toxicology and Therapeutics, the University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Yiqun Xiang
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Hongjian Zhang
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, the University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Chunshan Gui
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
- Corresponding author: Chunshan Gui, Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou 215123, China. Tel.: +86-512-65882089; Fax: +86-512-65882089
| |
Collapse
|
32
|
Nie Y, Yang J, Liu S, Sun R, Chen H, Long N, Jiang R, Gui C. Genetic polymorphisms of human hepatic OATPs: functional consequences and effect on drug pharmacokinetics. Xenobiotica 2019; 50:297-317. [DOI: 10.1080/00498254.2019.1629043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Yingmin Nie
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jingjie Yang
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Shuai Liu
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Ruiqi Sun
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Huihui Chen
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Nan Long
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Rui Jiang
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chunshan Gui
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
33
|
Crowe A, Zheng W, Miller J, Pahwa S, Alam K, Fung KM, Rubin E, Yin F, Ding K, Yue W. Characterization of Plasma Membrane Localization and Phosphorylation Status of Organic Anion Transporting Polypeptide (OATP) 1B1 c.521 T>C Nonsynonymous Single-Nucleotide Polymorphism. Pharm Res 2019; 36:101. [PMID: 31093828 PMCID: PMC8456979 DOI: 10.1007/s11095-019-2634-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 04/27/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Membrane transport protein organic anion transporting polypeptide (OATP) 1B1 mediates hepatic uptake of many drugs (e.g. statins). The OATP1B1 c.521 T > C (p. V174A) polymorphism has reduced transport activity. Conflicting in vitro results exist regarding whether V174A-OATP1B1 has reduced plasma membrane localization; no such data has been reported in physiologically relevant human liver tissue. Other potential changes, such as phosphorylation, of the V174A-OATP1B1 protein have not been explored. Current studies characterized the plasma membrane localization of V174A-OATP1B1 in genotyped human liver tissue and cell culture and compared the phosphorylation status of V174A- and wild-type (WT)-OATP1B1. METHODS Localization of V174A- and WT-OATP1B1 were determined in OATP1B1 c.521 T > C genotyped human liver tissue (n = 79) by immunohistochemistry and in transporter-overexpressing human embryonic kidney (HEK) 293 and HeLa cells by surface biotinylation and confocal microscopy. Phosphorylation and transport of OATP1B1 was determined using 32P-orthophosphate labeling and [3H]estradiol-17β-glucuronide accumulation, respectively. RESULTS All three methods demonstrated predominant plasma membrane localization of both V174A- and WT-OATP1B1 in human liver tissue and in cell culture. Compared to WT-OATP1B1, the V174A-OATP1B1 has significantly increased phosphorylation and reduced transport. CONCLUSIONS We report novel findings of increased phosphorylation, but not impaired membrane localization, in association with the reduced transport function of the V174A-OATP1B1.
Collapse
Affiliation(s)
- Alexandra Crowe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Wei Zheng
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jonathan Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Sonia Pahwa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Khondoker Alam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Erin Rubin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Feng Yin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kai Ding
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Wei Yue
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
34
|
Kolarić TO, Ninčević V, Smolić R, Smolić M, Wu GY. Mechanisms of Hepatic Cholestatic Drug Injury. J Clin Transl Hepatol 2019; 7:86-92. [PMID: 30944824 PMCID: PMC6441637 DOI: 10.14218/jcth.2018.00042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/18/2018] [Accepted: 02/08/2019] [Indexed: 12/12/2022] Open
Abstract
Drug-induced cholestasis represents a form of drug-induced liver disease that can lead to severe impairment of liver function. Numerous drugs have been shown to cause cholestasis and consequently bile duct toxicity. However, there is still lack of therapeutic tools that can prevent progression to advanced stages of liver injury. This review focuses on the various pathological mechanisms by which drugs express their hepatotoxic effects, as well as consequences of increased bile acid and toxin accumulation in the hepatocytes.
Collapse
Affiliation(s)
- Tea Omanović Kolarić
- Department of Pharmacology, Faculty of Medicine Osijek, Osijek, Croatia
- Department of Pharmacology, Faculty of Dental Medicine and Health, Osijek, Croatia
| | - Vjera Ninčević
- Department of Pharmacology, Faculty of Medicine Osijek, Osijek, Croatia
- Department of Pharmacology, Faculty of Dental Medicine and Health, Osijek, Croatia
| | - Robert Smolić
- Department of Pharmacology, Faculty of Medicine Osijek, Osijek, Croatia
| | - Martina Smolić
- Department of Pharmacology, Faculty of Medicine Osijek, Osijek, Croatia
- Department of Pharmacology, Faculty of Dental Medicine and Health, Osijek, Croatia
| | - George Y Wu
- Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
35
|
Characterization of non-radiolabeled Thyroxine (T 4) uptake in cryopreserved rat hepatocyte suspensions: Pharmacokinetic implications for PFOA and PFOS chemical exposure. Toxicol In Vitro 2019; 58:230-238. [PMID: 30930230 DOI: 10.1016/j.tiv.2019.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/28/2019] [Accepted: 03/18/2019] [Indexed: 11/22/2022]
Abstract
The alteration of thyroxine (T4) cellular uptake by an environmental chemical can serve as a contributing factor in thyroid hormone (TH) disruption. Herein, we describe a non-radiolabeled (LC-MS/MS) oil-filtration technique designed to characterize the mechanism(s) responsible for T4 cellular uptake in cryopreserved rat hepatocyte suspensions. The environmental chemicals perfluorooctanoate (PFOA) and perfluorooctane sulfonate (PFOS) were evaluated for their effect on T4 hepatic uptake. At 37 °C, hepatic assays demonstrated saturable kinetics with increasing T4 concentrations, while a linear uptake rate consistent with passive diffusion was detected at 4 °C. Carrier-mediated (37-4 °C) transport of T4 was the predominant hepatic uptake process versus passive diffusion. Cyclosporin A (CsA) chemically inhibited T4 hepatic uptake, whereas PFOA/PFOS displayed no inhibition of T4 translocation. Increasing PFOA/PFOS concentration levels with the T4 serum carrier-protein transthyretin (TTR) present resulted in a dose-response increase in T4 hepatic uptake rates, correlating with increased T4 free fraction values. Hepatic assays conducted in the presence of PFOA/PFOS and TTR displayed an enhanced first-order T4 hepatic uptake rate consistent with carrier-mediated transport. These in vitro findings characterizing increased T4 hepatic uptake provides mechanistic insight regarding decreased T4 serum levels (hypothyroxinemia) previously observed within in vivo rodent studies following perfluorinated chemical exposure.
Collapse
|
36
|
Takahashi T, Uno Y, Yamazaki H, Kume T. Functional characterization for polymorphic organic anion transporting polypeptides (OATP/SLCO
1B1, 1B3, 2B1) of monkeys recombinantly expressed with various OATP probes. Biopharm Drug Dispos 2019; 40:62-69. [DOI: 10.1002/bdd.2171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/15/2018] [Accepted: 01/02/2019] [Indexed: 11/08/2022]
Affiliation(s)
| | - Yasuhiro Uno
- Shin Nippon Biomedical Laboratories, Ltd., Kainan; Wakayama 642-0017 Japan
| | | | - Toshiyuki Kume
- Mitsubishi Tanabe Pharma Corporation, Toda; Saitama 335-8505 Japan
| |
Collapse
|
37
|
Smith DA, van Waterschoot RA, Parrott NJ, Olivares-Morales A, Lavé T, Rowland M. Importance of target-mediated drug disposition for small molecules. Drug Discov Today 2018; 23:2023-2030. [DOI: 10.1016/j.drudis.2018.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/04/2018] [Accepted: 06/12/2018] [Indexed: 11/30/2022]
|
38
|
Zhang B, Lauschke VM. Genetic variability and population diversity of the human SLCO (OATP) transporter family. Pharmacol Res 2018; 139:550-559. [PMID: 30359687 DOI: 10.1016/j.phrs.2018.10.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/14/2018] [Accepted: 10/17/2018] [Indexed: 01/12/2023]
Abstract
Organic anion transporting polypeptides (OATP) encoded by the SLCO gene family constitute clinically important transporters involved in the disposition of endogenous compounds and many commonly prescribed drugs, including statins, methotrexate and antihypertensive medications. Common genetic polymorphisms in SLCO genes are known to affect OATP function and modulate efficacy and safety of OATP substrates. However, current frequency data of these variants and haplotypes is generally based on few rather heterogenous populations of relatively small sample size. Furthermore, the genetic variability beyond these selected pharmacogenetic biomarkers has not been systematically analyzed. Here, we provide a global consolidated map of SLCO variability by leveraging fully compatible Next Generation Sequencing data from 138,632 unrelated individuals across seven major human populations. Overall, we find 9811 exonic single nucleotide variants and 155 copy number variations of which 99.3% were rare with frequencies <1%. Using orthogonal computational functionality predictors optimized for pharmacogenetic assessments, we find that four out of five individuals carry at least one deleterious variant in an SLCO transporter gene and rare variants contribute 23% to the genetically encoded functional variability. Moreover, 74.9% of all variants were found to be population-specific with important consequences for population-specific genotyping strategies and precision public health approaches. Combined, our analyses provide the most comprehensive data set of SLCO variability published to date and incentivize the integration of comprehensive NGS-based genotyping into personalized predictions of OATP substrate disposition.
Collapse
Affiliation(s)
- Boyao Zhang
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
39
|
Yorifuji K, Uemura Y, Horibata S, Tsuji G, Suzuki Y, Miyagawa K, Nakayama K, Hirata KI, Kumagai S, Emoto N. CHST3 and CHST13 polymorphisms as predictors of bosentan-induced liver toxicity in Japanese patients with pulmonary arterial hypertension. Pharmacol Res 2018; 135:259-264. [PMID: 30118797 DOI: 10.1016/j.phrs.2018.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 01/24/2023]
Abstract
Bosentan, an endothelin receptor antagonist, has been widely used as a first-line drug for the treatment of pulmonary arterial hypertension (PAH). In addition, bosentan is approved for patients with digital ulcers related to systemic sclerosis. Liver dysfunction is a major adverse effect of bosentan and may lead to discontinuation of therapy. The purpose of this study was to identify genomic biomarkers to predict bosentan-induced liver injury. A total of 69 PAH patients were recruited into the study. An exploratory analysis of 1936 single-nucleotide polymorphisms (SNPs) in 231 genes involved in absorption, distribution, metabolism, and elimination of multiple medications using Affimetrix DMET™ (Drug Metabolism Enzymes and Transporters) chips was performed. We extracted 16 SNPs (P < 0.05) using the Jonckheere-Terpstra trend test and multiplex logistic analysis; we identified two SNPs in two genes, CHST3 and CHST13, which are responsible for proteoglycan sulfation and were significantly associated with bosentan-induced liver injury. We constructed a predictive model for bosentan-induced liver injury (area under the curve [AUC]: 0.89, sensitivity: 82.61%, specificity: 86.05%) via receiver operating curve (ROC) analysis using 2 SNPs and 2 non-genetic factors. Two SNPs were identified as potential predictive markers for bosentan-induced liver injury in Japanese patients with pulmonary arterial hypertension. This is the first pharmacogenomics study linking proteoglycan sulfating genes to drug-induced liver dysfunction, a frequently observed clinical adverse effect of bosentan therapy. These results may provide a way to personalize PAH medicine as well as provide novel mechanistic insights to drug-induced liver dysfunction.
Collapse
Affiliation(s)
- Kennosuke Yorifuji
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama-kitamachi, Higashinada, Kobe 658-8558, Japan; The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan; Department of Pharmacy, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Yuko Uemura
- The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Shinji Horibata
- The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan; Department of Pharmacy, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Goh Tsuji
- The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan; Center for Rheumatic Diseases, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Yoko Suzuki
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama-kitamachi, Higashinada, Kobe 658-8558, Japan; Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 650-0017, Japan
| | - Kazuya Miyagawa
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama-kitamachi, Higashinada, Kobe 658-8558, Japan; Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 650-0017, Japan
| | - Kazuhiko Nakayama
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 650-0017, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 650-0017, Japan
| | - Shunichi Kumagai
- The Shinko Institute for Medical Research, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan; Center for Rheumatic Diseases, Shinko Hospital, 1-4-47, Wakinohama, Chuo, Kobe 651-0072, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama-kitamachi, Higashinada, Kobe 658-8558, Japan; Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 650-0017, Japan.
| |
Collapse
|
40
|
Yu Y, Li S, Liang W. Bona fide receptor for hepatitis B and D viral infections: Mechanism, research models and molecular drug targets. Emerg Microbes Infect 2018; 7:134. [PMID: 30050063 PMCID: PMC6062556 DOI: 10.1038/s41426-018-0137-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022]
Abstract
Hepatitis B infections have become a serious public health issue globally, and the current first-line antiviral treatment for this disease is not a true cure. Recently, sodium taurocholate cotransporting polypeptide (NTCP), a liver-specific bile acid transporter, was identified as a bona fide receptor for hepatitis B virus (HBV) and its satellite virus, hepatitis delta virus (HDV). Identification of the HBV receptor has led to the development of robust cell cultures and provides a potential target for new treatments. This review summarizes the process by which NTCP was discovered and describes its clinical significance as the receptor for HBV and HDV entry.
Collapse
Affiliation(s)
- Yueran Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.,Shengzhou People's Hospital, Shengzhou Branch of the First Affiliated Hospital of Zhejiang University, Shengzhou, 312400, China
| | - Shangda Li
- Department of Psychiatry, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Weifeng Liang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China. .,Shengzhou People's Hospital, Shengzhou Branch of the First Affiliated Hospital of Zhejiang University, Shengzhou, 312400, China.
| |
Collapse
|
41
|
Danielson ML, Sawada GA, Raub TJ, Desai PV. In Silico and in Vitro Assessment of OATP1B1 Inhibition in Drug Discovery. Mol Pharm 2018; 15:3060-3068. [DOI: 10.1021/acs.molpharmaceut.8b00168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
42
|
Sato M, Toshimoto K, Tomaru A, Yoshikado T, Tanaka Y, Hisaka A, Lee W, Sugiyama Y. Physiologically Based Pharmacokinetic Modeling of Bosentan Identifies the Saturable Hepatic Uptake As a Major Contributor to Its Nonlinear Pharmacokinetics. Drug Metab Dispos 2018; 46:740-748. [PMID: 29475833 DOI: 10.1124/dmd.117.078972] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/21/2018] [Indexed: 01/02/2023] Open
Abstract
Bosentan is a substrate of hepatic uptake transporter organic anion-transporting polypeptides (OATPs), and undergoes extensive hepatic metabolism by cytochrome P450 (P450), namely, CYP3A4 and CYP2C9. Several clinical investigations have reported a nonlinear relationship between bosentan doses and its systemic exposure, which likely involves the saturation of OATP-mediated uptake, P450-mediated metabolism, or both in the liver. Yet, the underlying causes for the nonlinear bosentan pharmacokinetics are not fully delineated. To address this, we performed physiologically based pharmacokinetic (PBPK) modeling analyses for bosentan after its intravenous administration at different doses. As a bottom-up approach, PBPK modeling analyses were performed using in vitro kinetic parameters, other relevant parameters, and scaling factors. As top-down approaches, three different types of PBPK models that incorporate the saturation of hepatic uptake, metabolism, or both were compared. The prediction from the bottom-up approach (models 1 and 2) yielded blood bosentan concentration-time profiles and their systemic clearance values that were not in good agreement with the clinically observed data. From top-down approaches (models 3, 4, 5-1, and 5-2), the prediction accuracy was best only with the incorporation of the saturable hepatic uptake for bosentan. Taken together, the PBPK models for bosentan were successfully established, and the comparison of different PBPK models identified the saturation of the hepatic uptake process as a major contributing factor for the nonlinear pharmacokinetics of bosentan.
Collapse
Affiliation(s)
- Masanobu Sato
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Kota Toshimoto
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Atsuko Tomaru
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Takashi Yoshikado
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Yuta Tanaka
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Akihiro Hisaka
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Wooin Lee
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Yuichi Sugiyama
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| |
Collapse
|
43
|
Chen Y, Chen L, Zhang H, Huang S, Xiong Y, Xia C. Interaction of Sulfonylureas with Liver Uptake Transporters OATP1B1 and OATP1B3. Basic Clin Pharmacol Toxicol 2018; 123:147-154. [PMID: 29498478 DOI: 10.1111/bcpt.12992] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/13/2018] [Indexed: 12/20/2022]
Abstract
Sulfonylureas (SUs) such as glibenclamide, gliclazide, glimepiride, glipizide and gliquidone are one of the first oral medicines available for the treatment of type 2 diabetes and are widely used for the treatment of hyperglycaemia. The hepatic transporters, organic anion transporting polypeptide 1B1 (OATP1B1) and organic anion transporting polypeptide 1B3 (OATP1B3), play an important role in the disposition of a variety of drugs by mediating their uptake from blood into hepatocytes. Drug-drug interactions mediated by OATP1B1/1B3 may result in the hepatic transporting change for drug substrates. The inhibitory effects of glibenclamide and glimepiride on sulfobromophthalein (BSP) uptake have been previously studied, and glibenclamide has been reported as the substrate of OATP1B3, but it remains unclear whether other SUs such as gliclazide, glipizide and gliquidone are substrates of OATP1B1 and OATP1B3. Here, we investigated the relationship between the five most commonly applied SUs (glibenclamide, gliclazide, glimepiride, glipizide, gliquidone) and OATP1B1 and OATP1B3. We performed uptake and inhibition assays in HEK293T cells stably expressing OATP1B1 or OATP1B3, respectively, and established a liquid chromatography-mass spectrometry (LC-MS) method for the simultaneous measurement of five SUs. We demonstrated that gliclazide and glimepiride are substrates of OATP1B1 and glibenclamide and glipizide are substrates of OATP1B3. We also confirmed the interaction between these SUs and rosuvastatin. No transporting was observed for gliquidone, suggesting that it is not a substrate of either transporter.
Collapse
Affiliation(s)
- Yu Chen
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China.,Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Lin Chen
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| | - Hong Zhang
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| | - Shibo Huang
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| | - Yuqing Xiong
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| | - Chunhua Xia
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| |
Collapse
|
44
|
Liu H, Stresser DM, Michmerhuizen MJ, Li X, Othman AA, Reed AD, Schrimpf MR, Sydor J, Lee AJ. Metabolism and Disposition of a Novel Selective α7 Neuronal Acetylcholine Receptor Agonist ABT-126 in Humans: Characterization of the Major Roles for Flavin-Containing Monooxygenases and UDP-Glucuronosyl Transferase 1A4 and 2B10 in Catalysis. Drug Metab Dispos 2018; 46:429-439. [PMID: 29348125 DOI: 10.1124/dmd.117.077511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 01/11/2018] [Indexed: 01/30/2023] Open
Abstract
Mass balance, metabolism, and excretion of ABT-126, an α7 neuronal acetylcholine receptor agonist, were characterized in healthy male subjects (n = 4) after a single 100-mg (100 μCi) oral dose. The total recovery of the administered radioactivity was 94.0% (±2.09%), with 81.5% (±10.2%) in urine and 12.4% (±9.3%) in feces. Metabolite profiling indicated that ABT-126 had been extensively metabolized, with 6.6% of the dose remaining as unchanged parent drug in urine. Parent drug accounted for 12.2% of the administered radioactivity in feces. The primary metabolic transformations of ABT-126 involved aza-adamantane N-oxidation (M1, 50.3% in urine) and aza-adamantane N-glucuronidation (M11, 19.9% in urine). M1 and M11 were also major circulating metabolites, accounting for 32.6% and 36.6% of the drug-related material in plasma, respectively. These results demonstrated that ABT-126 is eliminated primarily by hepatic metabolism, followed by urinary excretion. Enzymatic studies suggested that M1 formation is mediated primarily by human liver flavin-containing monooxygenase (FMO)3 and, to a lesser extent, by human kidney FMO1; M11 is generated mainly by human uridine 5'-diphospho-glucuronosyltransferase (UGT) 1A4, whereas UGT 2B10 also contributes to ABT-126 glucuronidation. Species-dependent formation of M11 was observed in hepatocytes; M11 was formed in human and monkey hepatocytes, but not in rat and dog hepatocytes, suggesting that monkeys constitute an appropriate model for predicting the fate of compounds undergoing significant N-glucuronidation. M1 and M11 are not expected to have clinically relevant on- or off-target pharmacologic activities. In summary, this study characterized ABT-126 metabolites in the circulation and excreta and the primary elimination pathways of ABT-126 in humans.
Collapse
Affiliation(s)
- Hong Liu
- Bioanalysis and Biotransformation (H.L., M.J.M., J.S., A.J.L.), DMPK and Translational Modeling (D.M.S., X.L.), Process Chemistry (A.D.R.), Discovery Chemistry and Technology (M.R.S.), and Clinical Pharmacology and Pharmacometrics (A.A.O.), Research and Development, AbbVie, North Chicago, Illinois
| | - David M Stresser
- Bioanalysis and Biotransformation (H.L., M.J.M., J.S., A.J.L.), DMPK and Translational Modeling (D.M.S., X.L.), Process Chemistry (A.D.R.), Discovery Chemistry and Technology (M.R.S.), and Clinical Pharmacology and Pharmacometrics (A.A.O.), Research and Development, AbbVie, North Chicago, Illinois
| | - Melissa J Michmerhuizen
- Bioanalysis and Biotransformation (H.L., M.J.M., J.S., A.J.L.), DMPK and Translational Modeling (D.M.S., X.L.), Process Chemistry (A.D.R.), Discovery Chemistry and Technology (M.R.S.), and Clinical Pharmacology and Pharmacometrics (A.A.O.), Research and Development, AbbVie, North Chicago, Illinois
| | - Xiaofeng Li
- Bioanalysis and Biotransformation (H.L., M.J.M., J.S., A.J.L.), DMPK and Translational Modeling (D.M.S., X.L.), Process Chemistry (A.D.R.), Discovery Chemistry and Technology (M.R.S.), and Clinical Pharmacology and Pharmacometrics (A.A.O.), Research and Development, AbbVie, North Chicago, Illinois
| | - Ahmed A Othman
- Bioanalysis and Biotransformation (H.L., M.J.M., J.S., A.J.L.), DMPK and Translational Modeling (D.M.S., X.L.), Process Chemistry (A.D.R.), Discovery Chemistry and Technology (M.R.S.), and Clinical Pharmacology and Pharmacometrics (A.A.O.), Research and Development, AbbVie, North Chicago, Illinois
| | - Aimee D Reed
- Bioanalysis and Biotransformation (H.L., M.J.M., J.S., A.J.L.), DMPK and Translational Modeling (D.M.S., X.L.), Process Chemistry (A.D.R.), Discovery Chemistry and Technology (M.R.S.), and Clinical Pharmacology and Pharmacometrics (A.A.O.), Research and Development, AbbVie, North Chicago, Illinois
| | - Michael R Schrimpf
- Bioanalysis and Biotransformation (H.L., M.J.M., J.S., A.J.L.), DMPK and Translational Modeling (D.M.S., X.L.), Process Chemistry (A.D.R.), Discovery Chemistry and Technology (M.R.S.), and Clinical Pharmacology and Pharmacometrics (A.A.O.), Research and Development, AbbVie, North Chicago, Illinois
| | - Jens Sydor
- Bioanalysis and Biotransformation (H.L., M.J.M., J.S., A.J.L.), DMPK and Translational Modeling (D.M.S., X.L.), Process Chemistry (A.D.R.), Discovery Chemistry and Technology (M.R.S.), and Clinical Pharmacology and Pharmacometrics (A.A.O.), Research and Development, AbbVie, North Chicago, Illinois
| | - Anthony J Lee
- Bioanalysis and Biotransformation (H.L., M.J.M., J.S., A.J.L.), DMPK and Translational Modeling (D.M.S., X.L.), Process Chemistry (A.D.R.), Discovery Chemistry and Technology (M.R.S.), and Clinical Pharmacology and Pharmacometrics (A.A.O.), Research and Development, AbbVie, North Chicago, Illinois
| |
Collapse
|
45
|
Li R, Niosi M, Johnson N, Tess DA, Kimoto E, Lin J, Yang X, Riccardi KA, Ryu S, El-Kattan AF, Maurer TS, Tremaine LM, Di L. A Study on Pharmacokinetics of Bosentan with Systems Modeling, Part 1: Translating Systemic Plasma Concentration to Liver Exposure in Healthy Subjects. Drug Metab Dispos 2018; 46:346-356. [PMID: 29330218 DOI: 10.1124/dmd.117.078790] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/08/2018] [Indexed: 02/13/2025] Open
Abstract
Understanding liver exposure of hepatic transporter substrates in clinical studies is often critical, as it typically governs pharmacodynamics, drug-drug interactions, and toxicity for certain drugs. However, this is a challenging task since there is currently no easy method to directly measure drug concentration in the human liver. Using bosentan as an example, we demonstrate a new approach to estimate liver exposure based on observed systemic pharmacokinetics from clinical studies using physiologically based pharmacokinetic modeling. The prediction was verified to be both accurate and precise using sensitivity analysis. For bosentan, the predicted pseudo steady-state unbound liver-to-unbound systemic plasma concentration ratio was 34.9 (95% confidence interval: 4.2, 50). Drug-drug interaction (i.e., CYP3A and CYP2B6 induction) and inhibition of hepatic transporters (i.e., bile salt export pump, multidrug resistance-associated proteins, and sodium-taurocholate cotransporting polypeptide) were predicted based on the estimated unbound liver tissue or plasma concentrations. With further validation and refinement, we conclude that this approach may serve to predict human liver exposure and complement other methods involving tissue biopsy and imaging.
Collapse
Affiliation(s)
- Rui Li
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Mark Niosi
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Nathaniel Johnson
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - David A Tess
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Emi Kimoto
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Jian Lin
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Xin Yang
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Keith A Riccardi
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Sangwoo Ryu
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Ayman F El-Kattan
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Tristan S Maurer
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Larry M Tremaine
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| | - Li Di
- Systems Modeling and Simulation, Medicine Design (R.L., D.A.T., T.S.M.) and Pharmacokinetics, Dynamics, and Metabolism (A.F.E.-K.), Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts; and Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (M.N., N.J., E.K., J.L., X.Y., K.A.R., S.R., L.M.T., L.D.)
| |
Collapse
|
46
|
Alam K, Crowe A, Wang X, Zhang P, Ding K, Li L, Yue W. Regulation of Organic Anion Transporting Polypeptides (OATP) 1B1- and OATP1B3-Mediated Transport: An Updated Review in the Context of OATP-Mediated Drug-Drug Interactions. Int J Mol Sci 2018. [PMID: 29538325 PMCID: PMC5877716 DOI: 10.3390/ijms19030855] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Organic anion transporting polypeptides (OATP) 1B1 and OATP1B3 are important hepatic transporters that mediate the uptake of many clinically important drugs, including statins from the blood into the liver. Reduced transport function of OATP1B1 and OATP1B3 can lead to clinically relevant drug-drug interactions (DDIs). Considering the importance of OATP1B1 and OATP1B3 in hepatic drug disposition, substantial efforts have been given on evaluating OATP1B1/1B3-mediated DDIs in order to avoid unwanted adverse effects of drugs that are OATP substrates due to their altered pharmacokinetics. Growing evidences suggest that the transport function of OATP1B1 and OATP1B3 can be regulated at various levels such as genetic variation, transcriptional and post-translational regulation. The present review summarizes the up to date information on the regulation of OATP1B1 and OATP1B3 transport function at different levels with a focus on potential impact on OATP-mediated DDIs.
Collapse
Affiliation(s)
- Khondoker Alam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| | - Alexandra Crowe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| | - Xueying Wang
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Pengyue Zhang
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kai Ding
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA.
| | - Lang Li
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA.
| | - Wei Yue
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| |
Collapse
|
47
|
Burbank MG, Sharanek A, Burban A, Mialanne H, Aerts H, Guguen-Guillouzo C, Weaver RJ, Guillouzo A. From the Cover: MechanisticInsights in Cytotoxic and Cholestatic Potential of the Endothelial Receptor Antagonists Using HepaRG Cells. Toxicol Sci 2018; 157:451-464. [PMID: 28369585 DOI: 10.1093/toxsci/kfx062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Several endothelin receptor antagonists (ERAs) have been developed for the treatment of pulmonary arterial hypertension (PAH). Some of them have been related to clinical cases of hepatocellular injury (sitaxentan [SIT]) and/or cholestasis (bosentan [BOS]). We aimed to determine if ambrisentan (AMB) and macitentan (MAC), in addition to BOS and SIT, could potentially cause liver damage in man by use of human HepaRG cells. Our results showed that like BOS, MAC-induced cytotoxicity and cholestatic disorders characterized by bile canaliculi dilatation and impairment of myosin light chain kinase signaling. Macitentan also strongly inhibited taurocholic acid and carboxy-2',7'-dichlorofluorescein efflux while it had a much lower inhibitory effect on influx activity compared to BOS and SIT. Moreover, these three drugs caused decreased intracellular accumulation and parallel increased levels of total bile acids (BAs) in serum-free culture media. In addition, all drugs except AMB variably deregulated gene expression of BA transporters. In contrast, SIT was hepatotoxic without causing cholestatic damage, likely via the formation of reactive metabolites and AMB was not hepatotoxic. Together, our results show that some ERAs can be hepatotoxic and that the recently marketed MAC, structurally similar to BOS, can also cause cholestatic alterations in HepaRG cells. The absence of currently known or suspected cases of cholestasis in patients suffering from PAH treated with MAC is rationalized by the lower therapeutic doses and Cmax, and longer receptor residence time compared to BOS.
Collapse
Affiliation(s)
- Matthew Gibson Burbank
- Inserm UMR 991, Foie, Métabolismes et Cancer, Rennes, France.,Université Rennes 1, Rennes, France.,Biologie Servier, Gidy, France
| | - Ahmad Sharanek
- Inserm UMR 991, Foie, Métabolismes et Cancer, Rennes, France.,Université Rennes 1, Rennes, France
| | - Audrey Burban
- Inserm UMR 991, Foie, Métabolismes et Cancer, Rennes, France.,Université Rennes 1, Rennes, France
| | | | | | | | | | - André Guillouzo
- Inserm UMR 991, Foie, Métabolismes et Cancer, Rennes, France.,Université Rennes 1, Rennes, France
| |
Collapse
|
48
|
Uchida M, Tajima Y, Kakuni M, Kageyama Y, Okada T, Sakurada E, Tateno C, Hayashi R. Organic Anion-Transporting Polypeptide (OATP)-Mediated Drug-Drug Interaction Study between Rosuvastatin and Cyclosporine A in Chimeric Mice with Humanized Liver. Drug Metab Dispos 2018; 46:11-19. [PMID: 29051147 DOI: 10.1124/dmd.117.075994] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 10/13/2017] [Indexed: 02/13/2025] Open
Abstract
The influence of transporters on the pharmacokinetics of drugs is being increasingly recognized, and DDIs via transporters may be a risk factor for adverse events. Cyclosporine A, a strong OATP inhibitor, has been reported to increase the systemic exposure of rosuvastatin, an OATP substrate, by 7.1-fold in clinical studies. PXB mice are chimeric mice with humanized livers that are highly repopulated with human hepatocytes and have been widely used for drug discovery in drug metabolism and pharmacokinetics studies. In the present study, we examined in vivo and in vitro DDIs between rosuvastatin and cyclosporine A in PXB mice and fresh human hepatocytes (PXB cells) obtained from PXB mice. We initially investigated the active transport of rosuvastatin into PXB cells, and found concentration-dependent uptake with a Michaelis-Menten constant value of 4.0 μmol/l and a Vmax value of 4.63 pmol/min per 106 cells. Cyclosporine A inhibited the uptake of rosuvastatin with an IC50 value of 0.21 μmol/l. We then examined in vivo DDIs, and the exposure of orally administered rosuvastatin increased by 3.3-fold and 11-fold in PXB mice pretreated with 10 and 50 mg/kg cyclosporine A, whereas it increased by 2.5-fold and 6.2-fold when rosuvastatin was administered intravenously, in studies that were conducted for considering gastrointestinal DDIs. The liver-to-blood concentration ratio of rosuvastatin was dose-dependently decreased by pretreatment with cyclosporine A in PXB mice and SCID mice. Observed DDIs in vivo were considered to be reasonable based on the estimated concentrations of cyclosporine A at the inlet to the liver and in the liver tissues of both mice. In conclusion, our results indicate that PXB mice might be a useful tool for predicting human OATP-mediated DDIs in drug discovery, and its limitation due to the differences of gastrointestinal condition from human should also be considered.
Collapse
Affiliation(s)
- Masashi Uchida
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Yoriko Tajima
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Masakazu Kakuni
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Yutaka Kageyama
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Taro Okada
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Eri Sakurada
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Chise Tateno
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| | - Ryoji Hayashi
- Pharmaceutical Research Laboratories, Toray Industries, Inc., Kamakura, Kanagawa, Japan (M.U., Y.T., E.S., R.H.); and PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima, Japan (M.K., Y.K., T.O., C.T.)
| |
Collapse
|
49
|
Sildenafil dosed concomitantly with bosentan for adult pulmonary arterial hypertension in a randomized controlled trial. BMC Cardiovasc Disord 2017; 17:239. [PMID: 28874133 PMCID: PMC5586020 DOI: 10.1186/s12872-017-0674-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 08/29/2017] [Indexed: 01/24/2023] Open
Abstract
Background Few controlled clinical trials exist to support oral combination therapy in pulmonary arterial hypertension (PAH). Methods Patients with PAH (idiopathic [IPAH] or associated with connective tissue disease [APAH-CTD]) taking bosentan (62.5 or 125 mg twice daily at a stable dose for ≥3 months) were randomized (1:1) to sildenafil (20 mg, 3 times daily; n = 50) or placebo (n = 53). The primary endpoint was change from baseline in 6-min walk distance (6MWD) at week 12, assessed using analysis of covariance. Patients could continue in a 52-week extension study. An analysis of covariance main-effects model was used, which included categorical terms for treatment, baseline 6MWD (<325 m; ≥325 m), and baseline aetiology; sensitivity analyses were subsequently performed. Results In sildenafil versus placebo arms, week-12 6MWD increases were similar (least squares mean difference [sildenafil–placebo], −2.4 m [90% CI: –21.8 to 17.1 m]; P = 0.6); mean ± SD changes from baseline were 26.4 ± 45.7 versus 11.8 ± 57.4 m, respectively, in IPAH (65% of population) and −18.3 ± 82.0 versus 17.5 ± 59.1 m in APAH-CTD (35% of population). One-year survival was 96%; patients maintained modest 6MWD improvements. Changes in WHO functional class and Borg dyspnoea score and incidence of clinical worsening did not differ. Headache, diarrhoea, and flushing were more common with sildenafil. Conclusions Sildenafil, in addition to stable (≥3 months) bosentan therapy, had no benefit over placebo for 12-week change from baseline in 6MWD. The influence of PAH aetiology warrants future study. Trial registration ClinicalTrials.gov NCT00323297 (registration date: May 5, 2006). Electronic supplementary material The online version of this article (10.1186/s12872-017-0674-3) contains supplementary material, which is available to authorized users.
Collapse
|
50
|
Wang C, Huo X, Wang C, Meng Q, Liu Z, Sun P, Cang J, Sun H, Liu K. Organic Anion–Transporting Polypeptide and Efflux Transporter–Mediated Hepatic Uptake and Biliary Excretion of Cilostazol and Its Metabolites in Rats and Humans. J Pharm Sci 2017; 106:2515-2523. [DOI: 10.1016/j.xphs.2017.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 01/24/2023]
|