1
|
Fashe MM, Tiley JB, Lee CR. Mechanisms of altered hepatic drug disposition during pregnancy: small molecules. Expert Opin Drug Metab Toxicol 2025; 21:445-462. [PMID: 39992297 PMCID: PMC11961323 DOI: 10.1080/17425255.2025.2470792] [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: 12/06/2024] [Revised: 02/01/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Pregnancy alters the systemic exposure and clearance of many hepatically cleared drugs that are commonly used by obstetric patients. Understanding the molecular mechanisms underlying the changes in factors that affect hepatic drug clearance (blood flow, protein binding, and intrinsic clearance) is essential to more precisely predict systemic drug exposure and dose requirements in obstetric patients. AREAS COVERED This review (1) summarizes the anatomic, physiologic, and biochemical changes in maternal hepatic, cardiovascular, endocrine, and renal systems relevant to hepatic drug clearance and (2) reviews the molecular mechanisms underlying the altered hepatic metabolism and intrinsic clearance of drugs during pregnancy via a comprehensive PubMed search. It also identifies knowledge gaps in the molecular mechanisms and factors that modulate hepatic drug clearance during pregnancy. EXPERT OPINION Pharmacokinetic studies have shown that pregnancy alters systemic exposure, protein binding, and clearance of many drugs during gestation in part due to pregnancy-associated decreases in plasma albumin, increases in organ blood flow, and changes in the activity of drug-metabolizing enzymes (DMEs) and transporters. The changes in the activity of certain DMEs and transporters during pregnancy are likely driven by hormonal-changes that inhibit their activity or alter the expression of these proteins through activation of transcription factors.
Collapse
Affiliation(s)
- Muluneh M. Fashe
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Jacqueline B. Tiley
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| |
Collapse
|
2
|
Krohmer E, Rohr BS, Stoll F, Gümüs KS, Bergamino M, Mikus G, Sauter M, Burhenne J, Weiss J, Meid AD, Czock D, Blank A, Haefeli WE. Influence of a Short Course of Ritonavir Used as Booster in Antiviral Therapies Against SARS-CoV-2 on the Exposure of Atorvastatin and Rosuvastatin. Cardiovasc Drugs Ther 2025; 39:325-335. [PMID: 38112932 PMCID: PMC11954719 DOI: 10.1007/s10557-023-07538-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 12/21/2023]
Abstract
PURPOSE Early antiviral treatment with nirmatrelvir/ritonavir is recommended for SARS-CoV-2-infected patients at high risk for severe courses. Such patients are usually chronically ill and susceptible to adverse drug interactions caused by ritonavir. We investigated the interactions of short-term low-dose ritonavir therapy with atorvastatin and rosuvastatin, two statins commonly used in this population. METHOD We assessed exposure changes (area under the concentration-time curve (AUC∞) and maximum concentration (Cmax)) of a single dose of 10 mg atorvastatin and 10 mg rosuvastatin before and on the fifth day of ritonavir treatment (2 × 100 mg/day) in healthy volunteers and developed a semi-mechanistic pharmacokinetic model to estimate dose adjustment of atorvastatin during ritonavir treatment. RESULTS By the fifth day of ritonavir treatment, the AUC∞ of atorvastatin increased 4.76-fold and Cmax 3.78-fold, and concurrently, the concentration of atorvastatin metabolites decreased to values below the lower limit of quantification. Pharmacokinetic modelling indicated that a stepwise reduction in atorvastatin dose during ritonavir treatment with a stepwise increase up to 4 days after ritonavir discontinuation can keep atorvastatin exposure within safe and effective margins. Rosuvastatin pharmacokinetics were only mildly modified; ritonavir significantly increased the Cmax 1.94-fold, while AUC∞ was unchanged. CONCLUSION Atorvastatin doses should likely be adjusted during nirmatrelvir/ritonavir treatment. For patients on a 20-mg dose, we recommend half of the original dose. In patients taking 40 mg or more, a quarter of the dose should be taken until 2 days after discontinuation of nirmatrelvir/ritonavir. Patients receiving rosuvastatin do not need to change their treatment regimen. TRIAL REGISTRATION EudraCT number: 2021-006634-39. DRKS00027838.
Collapse
Affiliation(s)
- Evelyn Krohmer
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Brit Silja Rohr
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Felicitas Stoll
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Katja S Gümüs
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Mariano Bergamino
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Gerd Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Max Sauter
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Andreas D Meid
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - David Czock
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Antje Blank
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Walter E Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Medical Faculty of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| |
Collapse
|
3
|
Sun Y, Tan H, Wang F, Hu J, Duan X, Bai W, Wu J, Bai J, Hu J. Inhibitory Effects of Alkaloids on OATP1B1 In Vitro and In Vivo: Prediction for Food/Herb-Drug Interactions and Hepatoprotective Effects Based on Structure-Activity Relationships. Chem Res Toxicol 2025; 38:281-295. [PMID: 39899883 DOI: 10.1021/acs.chemrestox.4c00418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Alkaloids, a class of low-molecular-weight nitrogenous compounds, attract a great deal of interest because of their biological activities and therapeutic potential. Yet, surprisingly little is known about their interactions with drug transporters, especially Organic Anion Transporting Polypeptide 1B1 (OATP1B1), a liver-specific uptake transporter, which is closely associated with drug-induced liver injury (DILI). This study aims to investigate the inhibitory effects of 160 alkaloids on OATP1B1, assess the hepatoprotective effects against bosentan-induced liver injury, and elucidate the structure-activity relationships of alkaloids with OATP1B1. Four alkaloids, including dihydroberberine, deacetyltaxol, dihydrocapsaicin, and tetrahydropalmatine, significantly inhibited OATP1B1 transport activity in OATP1B1-HEK293 cells (>50%), which reduced the OATP1B1-mediated uptake of methotrexate and microcystin-LR, and consequently decreased their cell toxicity. In bosentan-induced liver injury models, 4 alkaloids reduced serum total bile acid (TBA) levels and liver concentration of bosentan to different degrees, especially deacetyltaxol, which exhibited the most potent hepatoprotective effect against bosentan. The pharmacophore model suggested that the critical pharmacophores of alkaloid inhibitors are hydrogen bond acceptors and hydrophobic groups. Our findings pave the way for predicting the potential risks of alkaloids-containing food/herb-drug interactions in humans and optimizing the alkaloid structure for alleviating OATP1B1-related DILI.
Collapse
Affiliation(s)
- Yanhong Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Huixin Tan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Fenghe Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jiahuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiaoyan Duan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Wanting Bai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jinjin Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jie Bai
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Jinping Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
4
|
Korkak FA, Alkaç ZK, Arkalı G, Güngör İH, Yüksel BF, İncili CA, Tanyıldızı S, Dağoğlu G. Co-administration of rifampicin and Boswellia serrata mitigates testicular toxicity caused by Aflatoxin B1. Toxicon 2025; 255:108238. [PMID: 39824464 DOI: 10.1016/j.toxicon.2025.108238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
The current study was aimed to investigate the effect of rifampicin (Rif), a stimulator of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), on limiting the passage of AFB1 (Aflatoxin B1) into testicular tissue. The second objective was to examine the potential protective effects of Boswellia serrata extract (BSE), which exhibits a strong antioxidant capacity, alone or incombination with Rif against testicular damage induced by AFB1. A total of 49 male Sprague-Dawley rats were randomly divided into seven experimental groups as follows: control (placebo), Rif (10 mg/kg), BSE (500 mg/kg), AFB1 (0.75 mg/kg), AFB1+Rif, AFB1+BSE, and AFB1+Rif + BSE. The rats were administered AFB1, Rif, and BSE for seven days. The result of this study indicated that Rif decreased the amount of AFB1 permeating the testicular tissue by stimulating the expression of P-gp and BCRP. The administration of the combination of BSE and Rif resulted in a reduction of oxidative stress, apoptosis, improvement in sperm function parameters, and an increase in serum testosterone levels. These effects contributed to the improvement of impaired testicular structure. The result of this study revealed that the Rif can potentially serve as an efficacious therapeutic agent and the administration of BSE exhibited a reduction in testicular damage induced by AFB1. However, the combination of BSE and Rif provided more effective protection than using alone.
Collapse
Affiliation(s)
- Fatih Ahmet Korkak
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey.
| | - Zeliha Keskin Alkaç
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Firat University, Elazig, Turkey
| | - Gözde Arkalı
- Department of Physiology, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| | - İbrahim Halil Güngör
- Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Firat University, Turkey
| | - Burak Fatih Yüksel
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Firat University, Turkey
| | - Canan Akdeniz İncili
- Department of Pathology, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| | - Sadettin Tanyıldızı
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| | - Gürdal Dağoğlu
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
5
|
Wen J, Zhao M, Xiao Y, Li S, Hu W. OATP1A2 mediates Aβ 1-42 transport and may be a novel target for the treatment of Alzheimer's disease. Front Pharmacol 2024; 15:1443789. [PMID: 39600371 PMCID: PMC11588442 DOI: 10.3389/fphar.2024.1443789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with an unknown cause. Many studies have suggested that the imbalance between the clearance and accumulation of β-amyloid protein (Aβ) in the brain of AD patients is the main cause of AD development of AD. Meanwhile, drug transporters play a key role in the transport of drugs and endogenous substances in vivo as well as in the development of many diseases. Could they be related to the imbalance between Aβ clearance and accumulation? OATP1A2 is the most abundant subfamily of organic anion transporting polypeptides (OATPs) that transport amphipathic substrates. Its high bilateral expression in brain endothelial cells suggests it plays a crucial role in delivering drugs and neuroactive peptides to brain tissue. Could it also be involved in mediating the production and accumulation of Aβ in the central system? This could lead to an imbalance between Aβ clearance and accumulation, ultimately resulting in AD development. This hypothesis would be bold and novel in the field of science. In this study, we successfully established the OATP1A2-HEK293T transgenic cell model, and found that the uptake of Aβ1-42 by OATP1A2-HEK293T cells was significantly higher than that of NC-HEK293T control cells and human astrocytes by adding different concentrations of Aβ1-42 to the cells of each group, suggesting that OATP1A2 expressed in the human brain is involved in Aβ amyloid protein transport.
Collapse
Affiliation(s)
- Jinhua Wen
- Department of GCP/Psychosomatic Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Menghua Zhao
- Department of GCP/Psychosomatic Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuwei Xiao
- School of Pharmacy, Nanchang University, Nanchang, China
| | - Sihong Li
- School of Pharmacy, Nanchang University, Nanchang, China
| | - Weiqiang Hu
- School of Pharmacy, Nanchang University, Nanchang, China
| |
Collapse
|
6
|
Ryu H, Kim HC, Jeon I, Jang IJ, Cho JY, Kim KT, Oh J. Pharmacokinetic Interactions Between the Fixed-Dose Combination of Ezetimibe/Rosuvastatin 10/20 Mg and the Fixed-Dose Combination of Telmisartan/Amlodipine 80/5 Mg in Healthy Subjects. Drug Des Devel Ther 2024; 18:2641-2652. [PMID: 38974125 PMCID: PMC11225994 DOI: 10.2147/dddt.s465652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
Background Management of hypertension and hyperlipidemia, which are common comorbid risk factors for cardiovascular diseases, require multiple medications. The development of a fixed-dose combination (FDC) containing ezetimibe, rosuvastatin, telmisartan, and amlodipine aims to enhance patient adherence and persistence, but the potential interactions among the four medications have not been studied. This study aimed to evaluate the pharmacokinetic (PK) interactions between the FDC of ezetimibe/rosuvastatin 10/20 mg (ER) and the FDC of telmisartan/amlodipine 80/5 mg (TA). Methods An open-label, single-sequence, three-period, three-treatment crossover study was conducted in healthy male subjects. All subjects received ER for 7 days, TA for 9 days and ER combined with TA for 7 days during each treatment period. For PK analysis of total/free ezetimibe, rosuvastatin, telmisartan, and amlodipine, serial blood samples were collected for 24 hours at steady state. Safety profiles were assessed throughout the study. Results Thirty-eight subjects were enrolled, and 34 subjects completed the study. The systemic exposure to each active ingredient after coadministration of the two FDCs was similar to that after each FDC alone. The geometric mean ratios and 90% confidence intervals for the maximum plasma concentration (µg/L) and the area under the plasma concentration-time curve (h·µg/L) of the combination therapy to monotherapy, assessed at steady state, were as follows: total ezetimibe, 1.0264 (0.8765-1.2017) and 0.9359 (0.7847-1.1163); free ezetimibe, 1.5713 (1.2821-1.9257) and 0.9941 (0.8384-1.1788); rosuvastatin, 2.1673 (1.7807-2.6379) and 1.1714 (0.9992-1.3733); telmisartan, 1.0745 (0.8139-1.4186) and 1.1057 (0.8379-1.4591); and amlodipine, 0.9421 (0.8764-1.0126) and 0.9603 (0.8862-1.0405). Both combination therapy and monotherapy were well tolerated by the subjects. Conclusion The coadministration of ezetimibe/rosuvastatin 10/20 mg and ezetimibe/rosuvastatin 10/20 mg was well tolerated in healthy subjects, and the PK interaction between those two FDCs was not clinically significant.
Collapse
Affiliation(s)
- Hyunwook Ryu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Hyun Chul Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Inseung Jeon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung Tae Kim
- Addpharma, Inc., Yongin-si, Gyeonggi-do, Republic of Korea
| | - Jaeseong Oh
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Department of Pharmacology, Jeju National University College of Medicine, Jeju, Republic of Korea
- Clinical Research Institute, Jeju National University Hospital, Jeju, Republic of Korea
| |
Collapse
|
7
|
Naritaka H, Aoki Y, Obata Y, Mimuro S, Nakajima Y. Rhabdomyolysis in a Long-Term Statin User Without Traditional Risk Factors: A Case Report. Cureus 2023; 15:e46069. [PMID: 37900543 PMCID: PMC10607640 DOI: 10.7759/cureus.46069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
We report a rare case of rhabdomyolysis in a 64-year-old man who had been receiving long-term statin therapy for hyperlipidemia. The patient initially presented with symptoms of acute appendicitis, which later progressed to acute renal failure and rhabdomyolysis. No commonly identified risk factors for rhabdomyolysis, including drug interactions and statin doses, were observed. The patient was urgently admitted to the intensive care unit where the relevant medications were discontinued in a timely manner and infusion resuscitation was performed. Renal function and serum creatine kinase levels gradually stabilized without the need for hemodialysis. After four days, the patient was transferred to a general ward and was fully discharged from the hospital 13 days after admission. This case highlights the importance of considering rhabdomyolysis as a possible complication among patients receiving statin therapy, even in the absence of traditional risk factors.
Collapse
Affiliation(s)
- Hiroyuki Naritaka
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, JPN
| | - Yoshitaka Aoki
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, JPN
| | - Yukako Obata
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, JPN
| | - Soichiro Mimuro
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, JPN
| | - Yoshiki Nakajima
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, JPN
| |
Collapse
|
8
|
Said S, Dardik A, Chaar CIO. What are the benefits and drawbacks of statins in carotid artery disease? A perspective review. Expert Rev Cardiovasc Ther 2023; 21:763-777. [PMID: 37994875 PMCID: PMC11493559 DOI: 10.1080/14779072.2023.2286011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023]
Abstract
INTRODUCTION The prevalence of carotid artery stenosis in the general population is approximately 3%, but approximately 20% among people with acute ischemic stroke. Statins are recommended by multiple international guidelines as the drug of choice for lipid control in people with asymptomatic or symptomatic carotid artery stenosis due to their lipid-lowering and other pleiotropic effects. AREAS COVERED This review discusses the guidelines for statin usage as a cornerstone in the prevention and management of atherosclerotic carotid artery disease and the impact of statins on stroke incidence and mortality. Statin side effects, alternative therapy, and genetic polymorphisms are reviewed. EXPERT OPINION Statin therapy is associated with a decreased incidence of stroke and mortality as well as improved outcomes for patients treated with carotid revascularization. Statins are a safe and effective class of medications, but the initiation of therapy warrants close monitoring to avoid rare and potentially serious side effects. Lack of clinical efficacy or the presence of side effects suggests a need for treatment with an alternative therapy such as PCSK9 inhibitors. Understanding the interplay between the mechanisms of statins and PCSK9 inhibition therapies will allow optimal benefits while minimizing risks. Future research into genetic polymorphisms may improve patient selection for personalized therapy.
Collapse
Affiliation(s)
- Shreef Said
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, CT
| | - Alan Dardik
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT
| | - Cassius Iyad Ochoa Chaar
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, CT
| |
Collapse
|
9
|
Chen M, Yang Y, Ying Y, Huang J, Sun M, Hong M, Wang H, Xie S, Chen D. ABC Transporters and CYP3A4 Mediate Drug Interactions between Enrofloxacin and Salinomycin Leading to Increased Risk of Drug Residues and Resistance. Antibiotics (Basel) 2023; 12:antibiotics12020403. [PMID: 36830313 PMCID: PMC9952136 DOI: 10.3390/antibiotics12020403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/31/2022] [Accepted: 01/05/2023] [Indexed: 02/22/2023] Open
Abstract
Enrofloxacin (ENR) is one of the most common drugs used in poultry production to treat bacterial diseases, and there is a high risk of drug interactions (DDIs) between polyether anticoccidial drugs added to poultry feed over time. This may affect the efficacy of antibiotics or lead to toxicity, posing a potential risk to the environment and food safety. This study aimed to investigate the DDI of ENR and salinomycin (SAL) in broilers and the mechanism of their DDI. We found that SAL increased the area under the curve and elimination half-life of ENR and ciprofloxacin (CIP) by 1.3 and 2.4 times, 1.2 and 2.5 times, respectively. Cytochrome 3A4 (CYP3A4), p-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) were important factors for the DDI between ENR and SAL in broilers. ENR and SAL are substrates of CYP3A4, P-gp and BCRP in broilers; ENR and SAL inhibited the expression of CYP3A4 activity in a time- and concentration-dependent. Meanwhile, ENR downregulated the expression of P-gp and BCRP in a time- and concentration-dependent manner. A single oral administration of SAL inhibited CYP3A4, P-gp, and BCRP, but long-term mixed feeding upregulated the expression of CYP3A4, P-gp, and BCRP. Molecular docking revealed that ENR and SAL compete with each other for CYP3A4 to affect hepatic metabolism, and compete with ATP for P-gp and BCRP binding sites to inhibit efflux. ENR and SAL in broilers can lead to severe DDI. Drug residues and resistance following co-administration of ENR and SAL and other SAL-based drug-feed interactions warrant further study.
Collapse
Affiliation(s)
- Min Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
| | - Yujuan Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
| | - Yupeng Ying
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
| | - Jiamin Huang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
| | - Mengyuan Sun
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
| | - Mian Hong
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
| | - Haizhen Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: (S.X.); (D.C.); Tel.: +86-027-8728-7323 (D.C.)
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: (S.X.); (D.C.); Tel.: +86-027-8728-7323 (D.C.)
| |
Collapse
|
10
|
Richardson LS, K Kammala A, Costantine MM, Fortunato SJ, Radnaa E, Kim S, Taylor RN, Han A, Menon R. Testing of drugs using human feto-maternal interface organ-on-chips provide insights into pharmacokinetics and efficacy. LAB ON A CHIP 2022; 22:4574-4592. [PMID: 36322152 PMCID: PMC9682442 DOI: 10.1039/d2lc00691j] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/07/2022] [Indexed: 06/01/2023]
Abstract
Objectives: To improve preclinical drug testing during pregnancy, we developed multiple microfluidic organ-on-chip (OOC) devices that represent the structure, functions, and responses of the two feto-maternal interfaces (FMis) in humans (fetal membrane [FMi-OOC] and placenta [PLA-OOC]). This study utilized feto-maternal interface OOCs to test the kinetics and efficacy of drugs during pregnancy. Study design: The FMi-OOC contained amnion epithelial, mesenchymal, chorion trophoblast, and decidual cells. The PLA-OOC contained cytotrophoblasts (BeWo), syncytiotrophoblasts (BeWo + forskolin), and human umbilical vein endothelial cell lines. Therapeutic concentrations of either pravastatin or rosuvastatin (200 ng mL-1), a model drug for these experiments, were applied to either decidua (in FMi-OOC) and syncytiotrophoblasts (in PLA-OOC) chambers under normal and oxidative stress conditions (induced by cigarette smoke extract [CSE 1 : 25]) to evaluate maternal drug exposure during normal pregnancy or oxidative stress (OS) associated pathologies, respectively. We determined statin pharmacokinetics and metabolism (LC-MS/MS), drug-induced cytotoxicity (LDH assay), and efficacy to reduce OS-induced inflammation (multiplex cytokine assay). Results: Both OOCs mimicked two distinct human feto-maternal interfaces. The drugs tested permeated the maternal-fetal cell layers of the FMi-OOC and PLA-OOC within 4 hours and generated cell and time-specific statin metabolites from various cell types without causing any cytotoxicity. OS-induced pro-inflammatory cytokines were effectively reduced by statins by increasing anti-inflammatory cytokine response across the FMi-OOC and PLA-OOC. Conclusion: Two distinct feto-maternal interface OOCs were developed, tested, and validated for their utility to conduct preclinical trials during pregnancy. We demonstrated that the placenta and fetal membranes-decidual interface both are able to transport and metabolize drugs and that the safety and efficacy of a drug can be determined using the anatomical structures recreated on OOCs.
Collapse
Affiliation(s)
- Lauren S Richardson
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, Division of Basic Science and Translational Medicine, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, Texas, USA.
| | - Ananth K Kammala
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, Division of Basic Science and Translational Medicine, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, Texas, USA.
| | - Maged M Costantine
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Stephen J Fortunato
- Obstetrics and Gynecology, Maternal-Fetal Medicine, Ochsner Medical Center, New Orleans, LA, USA
| | - Enkhtuya Radnaa
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, Division of Basic Science and Translational Medicine, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, Texas, USA.
| | - Sungjin Kim
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA.
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA.
| | - Ramkumar Menon
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, Division of Basic Science and Translational Medicine, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, Texas, USA.
| |
Collapse
|
11
|
Sun Q, Li L, Zhou Q. Effects of Ethanolic Extract of Schisandra sphenanthera on the Pharmacokinetics of Rosuvastatin in Rats. Drug Des Devel Ther 2022; 16:1473-1481. [PMID: 35607596 PMCID: PMC9123906 DOI: 10.2147/dddt.s364234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Wuzhi capsule (WZ) is a proprietary Chinese medicine prepared from the ethanolic extract of Schisandra sphenanthera that is commonly used to treat liver injury. Statins are widely used in patients with hyperlipidemia, coronary heart disease, metabolic syndrome, type 2 diabetes mellitus, and nonalcoholic fatty liver disease. Co-administration of statins with WZ is possible in clinical practice. WZ has obvious inhibitory effects on the bioavailability of atorvastatin and simvastatin; however, the drug–herb interactions between WZ and rosuvastatin have not been addressed. We explored the effects of WZ on the pharmacokinetics of rosuvastatin in Sprague-Dawley rats to promote a rational use of statins. Methods Eighteen male rats were randomly and evenly divided into three groups: control group (gavage feeding of rosuvastatin 10 mg·kg−1), single dose group (gavage feeding of a single dose of WZ 150 mg·kg−1 followed by rosuvastatin 10 mg·kg−1) and multiple doses group (gavage feeding of WZ 150 mg·kg−1 for 7 days followed by rosuvastatin 10 mg·kg−1 on the seventh day). Plasma samples were collected at different times before and after rosuvastatin administration. The other 18 female rats were tested the same way as the male rats. All samples were analyzed by a validated LC-MS/MS method, and the pharmacokinetic parameters were calculated using a non-compartmental model. Results In both male and female rats, there were no statistically significant differences in rosuvastatin pharmacokinetic parameters between the control group, the single dose group, and the multi-dose group. Conclusion Acute or long-term intake of WZ had no obvious effect on the pharmacokinetics of rosuvastatin, and therefore rosuvastatin could be used as an alternative to atorvastatin and simvastatin when WZ is clinically required in conjunction with statins. An appropriate pharmacodynamic study is needed to encourage the safe use of this combination.
Collapse
Affiliation(s)
- Qing Sun
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, People’s Republic of China
| | - Li Li
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, People’s Republic of China
- Li Li, Department of Pharmacy, Zhejiang Hospital, No. 12 Lingyin Road, Hangzhou, 310013, Zhejiang Province, People’s Republic of China, Email
| | - Quan Zhou
- Department of Pharmacy, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- Correspondence: Quan Zhou, Department of Pharmacy, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road No. 88, Shangcheng District, Hangzhou, 310009, Zhejiang Province, People’s Republic of China, Tel +86 571 89713473, Email
| |
Collapse
|
12
|
Siew KSW, Abdul Hafidz MI, Mohd Zaidan FZB, Hadi MFB. Approaches to management of rhabdomyolysis as the adverse effect of drug interaction between atorvastatin and sacubitril/valsartan: a case report. Eur Heart J Case Rep 2022; 6:ytac091. [PMID: 35261962 PMCID: PMC8895310 DOI: 10.1093/ehjcr/ytac091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/06/2021] [Accepted: 02/18/2022] [Indexed: 12/11/2022]
Abstract
Background Atorvastatin and sacubitril/valsartan (Entresto™) have been cornerstones in managing patients with coronary artery disease and heart failure (HF). We report a case of life-threatening rhabdomyolysis associated with the co-administration of atorvastatin and sacubitril/valsartan. Case summary A 58-year-old male with coronary heart disease and chronic HF treated with the optimal dose of atorvastatin and other cardiovascular medications was frequently admitted for acute decompensation of HF. We decided to optimize his condition by adding sacubitril/valsartan to his treatment regime. He presented to our outpatient clinic with worsening myalgia and oliguria 6 days later. He was readmitted with markedly elevated serum creatinine kinase (CK) (94 850 U/L; normal range 32–294 U/L), deranged liver function tests, and acute kidney injury. We withheld atorvastatin and sacubitril/valsartan and treated him with renal replacement therapy. Discussion Sacubitril inhibits the excretion of statins, thereby elevating serum statin concentration and increasing the likelihood of developing muscle-related toxicity. Co-administration of atorvastatin and sacubitril/valsartan should be monitored closely with laboratory investigations of CK and liver and renal function. The physician may consider starting low-dose atorvastatin at 20 mg daily in combination with sacubitril/valsartan 24 mg/26 mg twice daily and titrating accordingly to optimal doses. Rosuvastatin could be an alternative to atorvastatin, as it has less drug–drug interaction with sacubitril, thereby reducing the adverse effect.
Collapse
Affiliation(s)
- Kelvin Shenq Woei Siew
- Division of Cardiology, Department of Medicine, University Malaya Medical Centre, Jln Profesor Diraja Ungku Aziz, 59100 Kuala Lumpur, Malaysia
| | - Muhammad Imran Abdul Hafidz
- Division of Cardiology, Department of Medicine, University Malaya Medical Centre, Jln Profesor Diraja Ungku Aziz, 59100 Kuala Lumpur, Malaysia
| | - Fatimah Zahrah Binti Mohd Zaidan
- Department of Primary Care Medicine, University Malaya Medical Centre, Jln Profesor Diraja Ungku Aziz, 59100 Kuala Lumpur, Malaysia
| | - Mohd Firdaus bin Hadi
- Division of Cardiology, Department of Medicine, University Malaya Medical Centre, Jln Profesor Diraja Ungku Aziz, 59100 Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Groenendaal-van de Meent D, den Adel M, Kerbusch V, van Dijk J, Shibata T, Kato K, Schaddelee M. Effect of Roxadustat on the Pharmacokinetics of Simvastatin, Rosuvastatin, and Atorvastatin in Healthy Subjects: Results From 3 Phase 1, Open-Label, 1-Sequence, Crossover Studies. Clin Pharmacol Drug Dev 2022; 11:486-501. [PMID: 35182045 PMCID: PMC9306950 DOI: 10.1002/cpdd.1076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/27/2021] [Indexed: 12/17/2022]
Abstract
Roxadustat inhibits breast cancer resistance protein and organic anion transporting polypeptide 1B1, which can affect coadministered statin concentrations. Three open‐label, 1‐sequence crossover phase 1 studies in healthy subjects were conducted to assess effects from steady‐state 200‐mg roxadustat on pharmacokinetics and tolerability of 40‐mg simvastatin (CL‐0537 and CL‐0541), 40‐mg atorvastatin (CL‐0538), or 10‐mg rosuvastatin (CL‐0537). Statins were dosed concomitantly with roxadustat in 28 (CL‐0537) and 24 (CL‐0538) healthy subjects, resulting in increases of maximum plasma concentration (Cmax) and area under the plasma concentration–time curve from the time of dosing extrapolated to infinity (AUCinf) 1.87‐ and 1.75‐fold for simvastatin, 2.76‐ and 1.85‐fold for simvastatin acid, 4.47‐ and 2.93‐fold for rosuvastatin, and 1.34‐ and 1.96‐fold for atorvastatin, respectively. Additionally, simvastatin dosed 2 hours before, and 4 and 10 hours after roxadustat in 28 (CL‐0541) healthy subjects, resulted in increases of Cmax and AUCinf 2.32‐ to 3.10‐fold and 1.56‐ to 1.74‐fold for simvastatin and 2.34‐ to 5.98‐fold and 1.89‐ to 3.42‐fold for simvastatin acid, respectively. These increases were not attenuated by time‐separated statin dosing. No clinically relevant differences were observed for terminal elimination half‐life. Concomitant 200‐mg roxadustat and a statin was generally well tolerated during the study period. Roxadustat effects on statin Cmax and AUCinf were statin and administration time dependent. When coadministered with roxadustat, statin‐associated adverse reactions and the need for statin dose reduction should be evaluated.
Collapse
Affiliation(s)
| | | | | | - Jan van Dijk
- Astellas Pharma Europe B.V., Leiden, The Netherlands
| | | | | | | |
Collapse
|
14
|
Silveira AMR, Duarte GHB, Fernandes AMADP, Garcia PHD, Vieira NR, Antonio MA, Carvalho PDO. Serum Predose Metabolic Profiling for Prediction of Rosuvastatin Pharmacokinetic Parameters in Healthy Volunteers. Front Pharmacol 2021; 12:752960. [PMID: 34867363 PMCID: PMC8633954 DOI: 10.3389/fphar.2021.752960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/13/2021] [Indexed: 11/23/2022] Open
Abstract
Rosuvastatin is a well-known lipid-lowering agent generally used for hypercholesterolemia treatment and coronary artery disease prevention. There is a substantial inter-individual variability in the absorption of statins usually caused by genetic polymorphisms leading to a variation in the corresponding pharmacokinetic parameters, which may affect drug therapy safety and efficacy. Therefore, the investigation of metabolic markers associated with rosuvastatin inter-individual variability is exceedingly relevant for drug therapy optimization and minimizing side effects. This work describes the application of pharmacometabolomic strategies using liquid chromatography coupled to mass spectrometry to investigate endogenous plasma metabolites capable of predicting pharmacokinetic parameters in predose samples. First, a targeted method for the determination of plasma concentration levels of rosuvastatin was validated and applied to obtain the pharmacokinetic parameters from 40 enrolled individuals; then, predose samples were analyzed using a metabolomic approach to search for associations between endogenous metabolites and the corresponding pharmacokinetic parameters. Data processing using machine learning revealed some candidates including sterols and bile acids, carboxylated metabolites, and lipids, suggesting the approach herein described as promising for personalized drug therapy.
Collapse
Affiliation(s)
| | | | | | | | - Nelson Rogerio Vieira
- Integrated Unit of Pharmacology and Gastroenterology (UNIFAG), São Francisco University-USF, Bragança Paulista, Brazil
| | - Marcia Aparecida Antonio
- Integrated Unit of Pharmacology and Gastroenterology (UNIFAG), São Francisco University-USF, Bragança Paulista, Brazil
| | | |
Collapse
|
15
|
Mohamed MEF, Coppola S, Feng T, Camp HS, Kim E, Othman AA. Effect of Upadacitinib on the Pharmacokinetics of Rosuvastatin or Atorvastatin in Healthy Subjects. Clin Pharmacol Drug Dev 2021; 10:1335-1344. [PMID: 34109764 PMCID: PMC8596765 DOI: 10.1002/cpdd.957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/28/2021] [Indexed: 11/08/2022]
Abstract
This phase 1, 2‐part, 2‐period, open‐label, drug‐drug interaction study evaluated the potential for pharmacokinetic interactions between upadacitinib and rosuvastatin, an organic anion transporting polypeptide (OATP) 1B1 and breast cancer resistance protein substrate, or atorvastatin, a cytochrome P450 3A, OATP1B1, and OATP1B3 substrate, in 36 healthy volunteers. During period 1, a single dose of rosuvastatin (5 mg; part 1) or atorvastatin (10 mg; part 2) was administered on day 1, followed by a washout period of 5 days. During period 2, once‐daily doses of upadacitinib extended‐release (30 mg) were administered on days 1 to 10, and a single dose of rosuvastatin (5 mg; part 1) or atorvastatin (10 mg; part 2) was administered 1 hour after the upadacitinib dose on day 7. Serial blood samples were collected for assays of drug concentrations. In Part 1, rosuvastatin maximum observed plasma concentration (Cmax) and area under the plasma concentration–time curve from time 0 to infinity (AUCinf) were 23% and 33% lower, respectively, when administered with upadacitinib relative to when administered alone. In part 2, atorvastatin Cmax and AUCinf was 11% and 23% lower, respectively, when administered with upadacitinib relative to when administered alone. The Cmax and AUCinf of the active metabolite ortho‐hydroxyatorvastatin remained unchanged. Administration of a single 5‐mg dose of rosuvastatin or a single 10‐mg dose of atorvastatin had no relevant effect on upadacitinib Cmax or area under the plasma concentration–time curve. These results demonstrated that upadacitinib has no clinically relevant effect on the pharmacokinetics of rosuvastatin and atorvastatin or on substrates transported by OATP1B or breast cancer resistance protein.
Collapse
Affiliation(s)
| | | | - Tian Feng
- AbbVie Inc., North Chicago, Illinois, USA
| | | | - Elaine Kim
- AbbVie Inc., North Chicago, Illinois, USA
| | | |
Collapse
|
16
|
Gebremichael LG, Suppiah V, Wiese MD, Mackenzie L, Phillips C, Williams DB, Roberts MS. Efficacy and safety of statins in ethnic differences: a lesson for application in Indigenous Australian patient care. Pharmacogenomics 2021; 22:553-571. [PMID: 34120458 DOI: 10.2217/pgs-2020-0152] [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: 10/06/2020] [Accepted: 03/29/2021] [Indexed: 11/21/2022] Open
Abstract
Although statins are effective in treating high cholesterol, adverse effects do occur with their use. Efficacy and tolerability vary among statins in different ethnic groups. Indigenous Australians have a high risk for cardiovascular and kidney diseases. Prescribing statins to Indigenous Australians with multi-morbidity requires different strategies to increase efficacy and reduce their toxicity. Previous studies have reported that Indigenous Australians are more susceptible to severe statin-induced myopathies. However, there is a lack of evidence in the underlying genetic factors in this population. This review aims to identify: inter-ethnic differences in the efficacy and safety of statins; major contributing factors accounting for any identified differences; and provide an overview of statin-induced adverse effects in Indigenous Australians.
Collapse
Affiliation(s)
- Lemlem G Gebremichael
- UniSA Clinical & Health Science, University of South Australia, Adelaide, SA 5000, Australia
| | - Vijayaprakash Suppiah
- UniSA Clinical & Health Science, University of South Australia, Adelaide, SA 5000, Australia
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA 5000, Australia
| | - Michael D Wiese
- UniSA Clinical & Health Science, University of South Australia, Adelaide, SA 5000, Australia
| | - Lorraine Mackenzie
- UniSA Clinical & Health Science, University of South Australia, Adelaide, SA 5000, Australia
| | - Craig Phillips
- UniSA Clinical & Health Science, University of South Australia, Adelaide, SA 5000, Australia
| | - Desmond B Williams
- UniSA Clinical & Health Science, University of South Australia, Adelaide, SA 5000, Australia
| | - Michael S Roberts
- UniSA Clinical & Health Science, University of South Australia, Adelaide, SA 5000, Australia
- Therapeutics Research Centre, Diamantina Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
- Basil Hetzel Institute for Translational Medical Research, The Queen Elizabeth Hospital, 28 Woodville Rd, Woodville, SA 5011, Australia
| |
Collapse
|
17
|
Brailovski E, Kim RB, Juurlink D. Rosuvastatin Myotoxicity After Starting Canagliflozin Treatment. Ann Intern Med 2021; 174:432. [PMID: 33721527 DOI: 10.7326/l20-1456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | - Richard B Kim
- University of Western Ontario, London, Ontario, Canada
| | | |
Collapse
|
18
|
Sane R, Cheung KWK, Kovács P, Farasyn T, Li R, Bui A, Musib L, Kis E, Plise E, Gáborik Z. Calibrating the In Vitro-In Vivo Correlation for OATP-Mediated Drug-Drug Interactions with Rosuvastatin Using Static and PBPK Models. Drug Metab Dispos 2020; 48:1264-1270. [PMID: 33037044 DOI: 10.1124/dmd.120.000149] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/28/2020] [Indexed: 02/13/2025] Open
Abstract
Organic anion-transporting polypeptide (OATP) 1B1/3-mediated drug-drug interaction (DDI) potential is evaluated in vivo with rosuvastatin (RST) as a probe substrate in clinical studies. We calibrated our assay with RST and estradiol 17-β-D-glucuronide (E217βG)/cholecystokinin-8 (CCK8) as in vitro probes for qualitative and quantitative prediction of OATP1B-mediated DDI potential for RST. In vitro OATP1B1/1B3 inhibition using E217βG and CCK8 yielded higher area under the curve (AUC) ratio (AUCR) values numerically with the static model, but all probes performed similarly from a qualitative cutoff-based prediction, as described in regulatory guidances. However, the magnitudes of DDI were not captured satisfactorily. Considering that clearance of RST is also mediated by gut breast cancer resistance protein (BCRP), inhibition of BCRP was also incorporated in the DDI prediction if the gut inhibitor concentrations were 10 × IC50 for BCRP inhibition. This combined static model closely predicted the magnitude of RST DDI with root-mean-square error values of 0.767-0.812 and 1.24-1.31 with and without BCRP inhibition, respectively, for in vitro-in vivo correlation of DDI. Physiologically based pharmacokinetic (PBPK) modeling was also used to simulate DDI between RST and rifampicin, asunaprevir, and velpatasvir. Predicted AUCR for rifampicin and asunaprevir was within 1.5-fold of that observed, whereas that for velpatasvir showed a 2-fold underprediction. Overall, the combined static model incorporating both OATP1B and BCRP inhibition provides a quick and simple mathematical approach to quantitatively predict the magnitude of transporter-mediated DDI for RST for routine application. PBPK complements the static model and provides a framework for studying molecules when a dynamic model is needed. SIGNIFICANCE STATEMENT: Using 22 drugs, we show that a static model for organic anion-transporting polypeptide (OATP) 1B1/1B3 inhibition can qualitatively predict potential for drug-drug interaction (DDI) using a cutoff-based approach, as in regulatory guidances. However, consideration of both OATP1B1/3 and gut breast cancer resistance protein inhibition provided a better prediction of the magnitude of the transporter-mediated DDI of these inhibitors with rosuvastatin. Based on these results, we have proposed an empirical mechanistic-static approach for a more reliable prediction of transporter-mediated DDI liability with rosuvastatin that drug development teams can leverage.
Collapse
Affiliation(s)
- Rucha Sane
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Kit Wun Kathy Cheung
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Péter Kovács
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Taleah Farasyn
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Ruina Li
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Annamaria Bui
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Luna Musib
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Emese Kis
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Emile Plise
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Zsuzsanna Gáborik
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| |
Collapse
|
19
|
Sidharta PN, Dingemanse J. Effects of Multiple‐Dose Administration of Aprocitentan on the Pharmacokinetics of Rosuvastatin. Clin Pharmacol Drug Dev 2020; 9:995-1002. [DOI: 10.1002/cpdd.815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/20/2020] [Indexed: 01/07/2023]
Affiliation(s)
- Patricia N. Sidharta
- Department of Clinical Pharmacology Idorsia Pharmaceuticals Ltd Allschwil Switzerland
| | - Jasper Dingemanse
- Department of Clinical Pharmacology Idorsia Pharmaceuticals Ltd Allschwil Switzerland
| |
Collapse
|
20
|
Huang S, Xu Q, Liu L, Bian Y, Zhang S, Huang C, Miao L. Effect of Green Tea and (-)-Epigallocatechin Gallate on the Pharmacokinetics of Rosuvastatin. Curr Drug Metab 2020; 21:471-478. [PMID: 32407265 DOI: 10.2174/1389200221666200514133355] [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] [Received: 12/23/2019] [Revised: 03/24/2020] [Accepted: 04/09/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Green tea can inhibit OATPs, so it may interact with the substrate of OATPs, such as rosuvastatin. OBJECTIVE This study aimed to investigate the effects of green tea on the pharmacokinetics of rosuvastatin and its mechanism. METHODS Male Sprague-Dawley rats received different doses of green tea extract (GTE) and (-)- epigallocatechin-3- gallate (EGCG). Caco-2 cells and OATP1B1-HEK293T cells were used in drug uptake and transport assay. The matrix concentrations of rosuvastatin and catechins were determined by ultra-performance liquid chromatographytandem mass spectrometry (UPLC-MS/MS). RESULTS GTE and EGCG were both found to increase the area under the plasma concentration-time curve (AUC0-∞) of rosuvastatin ((p<0.050). In the Caco-2 cell model, the uptake and transport of rosuvastatin in the GTE groups were 1.94-fold (p<0.001) and 2.11-fold (p<0.050) higher, respectively, than those of the control group. However, in the EGCG group, the uptake and transport of rosuvastatin were decreased by 22.62% and 44.19%, respectively (p<0.050). In the OATP1B1- HEK293T cell model, the OATP1B1-mediated rosuvastatin uptake was decreased by GTE to 35.02% of that in the control (p<0.050) and was decreased by EGCG to 45.61% of that in the control (p<0.050). CONCLUSION GTE increased the systemic rosuvastatin exposure in rats. The mechanism may include an increase in rosuvastatin absorption and a decrease in liver distribution by inhibiting OATP1B1. EGCG may be the main ingredient of green tea that affects the pharmacokinetic parameters of rosuvastatin. Our results showed the importance of conducting green tea-rosuvastatin study.
Collapse
Affiliation(s)
- Shenjia Huang
- Department of Pharmaceutics, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Qingqing Xu
- Department of Pharmaceutics, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Linsheng Liu
- Department of Clinical Pharmacology, The Frist Affiliated Hospital of Soochow University, Suzhou, China
| | - Yicong Bian
- Department of Clinical Pharmacology, The Frist Affiliated Hospital of Soochow University, Suzhou, China
| | - Shichao Zhang
- Department of Pharmaceutics, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Chenrong Huang
- Department of Clinical Pharmacology, The Frist Affiliated Hospital of Soochow University, Suzhou, China
| | - Liyan Miao
- Department of Clinical Pharmacology, The Frist Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
21
|
Peng Y, Chen L, Ye S, Kang Y, Liu J, Zeng S, Yu L. Research and development of drug delivery systems based on drug transporter and nano-formulation. Asian J Pharm Sci 2020; 15:220-236. [PMID: 32373201 PMCID: PMC7193453 DOI: 10.1016/j.ajps.2020.02.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/16/2020] [Accepted: 02/29/2020] [Indexed: 12/11/2022] Open
Abstract
In recent years, the continuous occurrence of multi-drug resistance in the clinic has made people pay more attention to the transporter. Changes in the expression and activity of transporters can cause corresponding changes in drug pharmacokinetics and pharmacodynamics. The drug-drug interactions (DDI) caused by transporters can seriously affect drug effectiveness and toxicity. In the development of pharmaceutical preparations, people have increasingly concerned about the effects and regulation of transporters in drug effects. To improve the targeting and physicochemical properties of drugs, the development of targeted agents is very rapid. Among them, novel nano-formulations are the best. With the continuous innovation and development of nano-formulation, its application has become more and more extensive. Nano-formulation has exerted certain advantages in the drug development based on transporters, and is also involved in the combination of targeted transporters. This review focuses on the application of novel nano-agents targeting transporters and the introduction of drug-transporter-based nano-formulations.
Collapse
Affiliation(s)
- Yi Peng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lu Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sheng Ye
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yu Kang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junqing Liu
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lushan Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
22
|
Identification of Functional Transcriptional Binding Sites within Chicken Abcg2 Gene Promoter and Screening Its Regulators. Genes (Basel) 2020; 11:genes11020186. [PMID: 32050731 PMCID: PMC7073639 DOI: 10.3390/genes11020186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/02/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer resistance protein (BCRP), an ATP-binding cassette (ABC) half transporter encoded by the Abcg2 gene, is reported to influence the pharmacokinetics of substrate drugs during clinical therapy. The aim of this study was to clarify the mechanisms that regulate the transcription of the chicken Abcg2 gene through cloning and characterization of its promoter region. Results showed that the Abcg2 gene is transcribed by a TATA-less promoter with several putative Sp1 sites upstream from two putative CpG islands. A luciferase reporter assay conducted both in chicken leghorn male hepatoma (LMH) cells and chicken primary hepatocytes mapped a basal promoter to nucleotides -110 to +30, which is responsible for the constitutive expression of Abcg2. The 5'-region upstream of the basal promoter was characterized by both positive and negative regulatory domains. Further, using the cell-based reporter gene assay combined with RT-PCR and drug accumulation analysis, we found that four xenobiotics, daidzein, clotrimazole, ivermectin, and lipopolysaccharide (LPS), influence the expression and function of BCRP through significant regulation of the Abcg2 gene promoter. Interaction sites with the Abcg2 gene promoter of these four selected regulators were clarified by progressive deletions and mutation assays. This study shed some light on the regulatory mechanisms involved in chicken Abcg2 gene expression and the results may have far-reaching significance regarding the usage and development of veterinary drugs.
Collapse
|
23
|
Influence of OATP1B1 and BCRP polymorphisms on the pharmacokinetics and pharmacodynamics of rosuvastatin in elderly and young Korean subjects. Sci Rep 2019; 9:19410. [PMID: 31857620 PMCID: PMC6923423 DOI: 10.1038/s41598-019-55562-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/26/2019] [Indexed: 02/04/2023] Open
Abstract
A lack of information regarding whether genetic polymorphisms of SLCO1B1 and ABCG2 affect the pharmacokinetics (PKs)/pharmacodynamics (PDs) of rosuvastatin in elderly subjects prevents optimal individualized pharmacotherapy of rosuvastatin in clinical settings. This study aimed to investigate the effect of age and genetic polymorphisms and possible differences in genetic effects on the PKs/PDs of rosuvastatin between elderly and young subjects. Two separate clinical studies designed as open-label, one-sequence studies with multiple-dose administration for elderly (n = 20) and young (n = 32) subjects were conducted. All subjects received 20 mg of rosuvastatin once daily for 21 days. The exposure to rosuvastatin, characterized by the area under the time curve (AUC), increased by 23% in the elderly subjects compared with that of young subjects, which was not significant. When compared to the subjects with breast cancer resistance protein (BCRP) normal function, the exposure to rosuvastatin increased by 44% in young subjects (p = 0.0021) with BCRP intermediate function (IF) and by 35% and 59% (p > 0.05 for both) in elderly subjects with BCRP IF and low function, respectively. SLCO1B1 521T > C was also partially associated with a higher AUC of rosuvastatin in young subjects and a less pronounced increasing trend in elderly subjects (p > 0.05 for both). The lipid-lowering effect of rosuvastatin was less pronounced in the elderly subjects than in the young subjects, and genetic polymorphisms of neither SLCO1B1 nor ABCG2 significantly affected the PDs of rosuvastatin. The ABCG2 421C > A polymorphism was associated with the PKs of rosuvastatin and was identified as a more important determinant than the SLCO1B1 521T > C polymorphism in both elderly and young subjects.
Collapse
|
24
|
Ericsson H, Nelander K, Heijer M, Kjaer M, Lindstedt EL, Albayaty M, Forte P, Lagerström-Fermér M, Skrtic S. Phase 1 Pharmacokinetic Study of AZD5718 in Healthy Volunteers: Effects of Coadministration With Rosuvastatin, Formulation and Food on Oral Bioavailability. Clin Pharmacol Drug Dev 2019; 9:411-421. [PMID: 31793171 PMCID: PMC7187334 DOI: 10.1002/cpdd.756] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022]
Abstract
AZD5718 is a first‐in‐class small‐molecule anti‐inflammatory drug with the potential to reduce the residual risk of cardiovascular events after myocardial infarction in patients receiving lipid‐lowering statin therapy. Leukotrienes are potent proinflammatory and vasoactive mediators synthesized in leukocytes via 5‐lipoxygenase and 5‐lipoxygenase‐activating protein (FLAP). AZD5718 is a FLAP inhibitor that dose‐dependently reduced leukotriene biosynthesis in a first‐in‐human study. We enrolled 12 healthy men in a randomized, open‐label, crossover, single‐dose phase 1 pharmacokinetic study of AZD5718 to investigate a potential drug‐drug interaction with rosuvastatin, and the effects of formulation and food intake (ClinicalTrials.gov identifier: NCT02963116). Rosuvastatin (10 mg) were absorbed more rapidly when coadministered with AZD5718 (200 mg), probably owing to weak inhibition of hepatic statin uptake, but relative bioavailability was unaffected (geometric least‐squares mean ratio [GMR], 100%; 90% confidence interval [CI], 86%‐116%). AZD5718 pharmacokinetics were unaffected by coadministration of rosuvastatin. AZD5718 (200 mg) was absorbed less rapidly when formulated as tablets than oral suspension, with reduced relative bioavailability (GMR, 72%; 90%CI, 64%‐80%). AZD5718 absorption was slower when 200‐mg tablets were taken after a high‐fat breakfast than after fasting, but relative bioavailability was unaffected (GMR, 96%; 90%CI, 87%‐106%). In post hoc pharmacodynamic simulations, plasma leukotriene B4 levels were inhibited by >90% throughout the day following once‐daily AZD5718, regardless of formulation or administration with food. AZD5718 was well tolerated, with no severe or serious adverse events. These data supported the design of a phase 2a efficacy study of AZD5718 in patients with coronary artery disease.
Collapse
Affiliation(s)
- Hans Ericsson
- Clinical Pharmacology, ADME and AI, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Karin Nelander
- Clinical Pharmacology, ADME and AI, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Heijer
- Clinical Pharmacology Biologics and Bioanalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Magnus Kjaer
- Early Biometrics and Statistical Innovation, Data Science & AI, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eva-Lotte Lindstedt
- Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Pablo Forte
- Parexel, Early Phase Clinical Unit, Harrow, UK
| | - Maria Lagerström-Fermér
- Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stanko Skrtic
- Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
25
|
Kim Y, Hatley O, Rhee SJ, Yi S, Lee HA, Yoon S, Chung JY, Yu KS, Lee H. Development of a Korean-specific virtual population for physiologically based pharmacokinetic modelling and simulation. Biopharm Drug Dispos 2019; 40:135-150. [PMID: 30921829 DOI: 10.1002/bdd.2178] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/07/2019] [Accepted: 03/21/2019] [Indexed: 01/19/2023]
Abstract
Physiologically based pharmacokinetic (PBPK) modelling and simulation is a useful tool in predicting the PK profiles of a drug, assessing the effects of covariates such as demographics, ethnicity, genetic polymorphisms and disease status on the PK, and evaluating the potential of drug-drug interactions. We developed a Korean-specific virtual population for the SimCYP® Simulator (version 15 used) and evaluated the population's predictive performance using six substrate drugs (midazolam, S-warfarin, metoprolol, omeprazole, lorazepam and rosuvastatin) of five major drug metabolizing enzymes (DMEs) and two transporters. Forty-three parameters including the proportion of phenotypes in DMEs and transporters were incorporated into the Korean-specific virtual population. The simulated concentration-time profiles in Koreans were overlapped with most of the observed concentrations for the selected substrate drugs with a < 2-fold difference in clearance. Furthermore, we found some drug models within the SimCYP® library can be improved, e.g., the minor allele frequency of ABCG2 and the fraction metabolized by UGT2B15 should be incorporated for rosuvastatin and lorazepam, respectively. The Korean-specific population can be used to evaluate the impact of ethnicity on the PKs of a drug, particularly in various stages of drug development.
Collapse
Affiliation(s)
- Yun Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | | | - Su-Jin Rhee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | - Sojeong Yi
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | - Hyun A Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Sumin Yoon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | - Jae-Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Bundang Hospital, Seongnam, South Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | - Howard Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| |
Collapse
|
26
|
Maekawa Y, Furuie H, Kato M, Myobatake Y, Kamiyama E, Watanabe A, Shiosakai K, Taguchi T, Bass R, Zhou J, Dishy V, Warren V, Vashi V, Ishizuka H. Effect of DS-8500a, a Novel G Protein-Coupled Receptor 119 Agonist, on the Pharmacokinetics of Rosuvastatin and Atorvastatin in Healthy Subjects. Clin Drug Investig 2019; 39:967-978. [DOI: 10.1007/s40261-019-00825-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
27
|
Najjar N, McColl ER, Weckman A, Kain KC, Piquette-Miller M. Dysregulation of solute carrier transporters in malaria-infected pregnant mice. Parasite Immunol 2019; 41:e12614. [PMID: 30703256 DOI: 10.1111/pim.12614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 12/18/2022]
Abstract
AIMS Malaria in pregnancy (MiP) alters the expression of ATP-binding cassette efflux transporters in maternal and foetal tissues, as well as the placenta. Malaria induces oxidative stress, and pregnancy is associated with arginine deficiency. We hypothesized that reducing oxidative stress during MiP by supplementation with L-arginine, a NO precursor, would attenuate transcriptional changes in a second superfamily of transporters, solute carrier (SLC) transporters, and improve pregnancy outcomes. METHODS AND RESULTS Pregnant BALB/c mice receiving L-arginine (1.2%) in water, or water alone, were infected with Plasmodium berghei ANKA on gestational day 13 and sacrificed on gestational day 19. Compared to controls, the mRNA of numerous SLC transporters was downregulated in maternal and foetal tissues, as well as in the placentas of infected mice. While supplementation with L-arginine did improve foetal viability, it did not improve the mRNA expression of oxidative stress markers, transporters nor other indices of foetal and maternal health. Moreover, amino acid uptake transporters were downregulated upon infection, which could potentially contribute to decreased foetal birthweight. CONCLUSIONS Malaria in pregnancy significantly alters the expression of SLC transporters in maternal and foetal tissues as well as the placenta, regardless of L-arginine supplementation. Further studies to investigate methods of reducing oxidative stress in MiP are warranted.
Collapse
Affiliation(s)
- Najwa Najjar
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Eliza R McColl
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Weckman
- Sandra Rotman Centre for Global Health, University Health Network: Toronto General Hospital, Toronto, Ontario, Canada
| | - Kevin C Kain
- Sandra Rotman Centre for Global Health, University Health Network: Toronto General Hospital, Toronto, Ontario, Canada
| | - Micheline Piquette-Miller
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Han W, Wu Q, Zhang X, Duan Z. Innovation for hepatotoxicity in vitro research models: A review. J Appl Toxicol 2018; 39:146-162. [PMID: 30182494 DOI: 10.1002/jat.3711] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022]
Abstract
Many categories of drugs can induce hepatotoxicity, so improving the prediction of toxic drugs is important. In vitro models using human hepatocytes are more accurate than in vivo animal models. Good in vitro models require an abundance of metabolic enzyme activities and normal cellular polarity. However, none of the in vitro models can completely simulate hepatocytes in the human body. There are two ways to overcome this limitation: enhancing the metabolic function of hepatocytes and changing the cultural environment. In this review, we summarize the current state of research, including the main characteristics of in vitro models and their limitations, as well as improved technology and developmental prospects. We hope that this review provides some new ideas for hepatotoxicity research.
Collapse
Affiliation(s)
- Weijia Han
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Qiao Wu
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Xiaohui Zhang
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Zhongping Duan
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| |
Collapse
|
29
|
Zeng X, Su W, Liu H, Zheng Y, Chen T, Zhang W, Yan Z, Bai Y, Yao H. Simultaneous determination of rosuvastatin, naringin and naringenin in rat plasma by RRLC-MS/MS and its application to a pharmacokinetic drug interaction study. J Chromatogr Sci 2018; 56:611-618. [PMID: 29701749 DOI: 10.1093/chromsci/bmy034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 04/04/2018] [Indexed: 01/12/2023]
Abstract
A rapid resolution liquid chromatography tandem mass spectrometry method was developed and validated for simultaneous determination of rosuvastatin, naringin and naringenin in rat plasma. Chromatographic separation of analytes and internal standard (fluvastatin for rosuvastatin, while isoquercitrin for naringin and naringenin) was performed on Agilent Poroshell 120 EC-C18 column (3.0 × 50 mm, 2.7 μm) using gradient elution with a mobile phase of methanol and water, both with 0.1% formic acid (v/v). The detection was operated in multiple reaction monitoring mode to monitor the precursor-to-product ion transitions of m/z 579.1→270.8 for naringin, m/z 270.9→150.7 for naringenin, m/z 463.1→299.8 for isoquercitrin in negative ionization mode, and m/z 482.2→258.1 for rosuvastatin, m/z 412.1→224.1 for fluvastatin in positive ionization mode. Polarity switch (negative-positive-negative ionization mode) was performed in a total runtime of 5.0 min. The method was validated over a concentration range of 10-2,000 ng/mL for the above three analytes. The intra-day and inter-day precisions and accuracies of the quality control samples at low, medium and high concentration levels exhibited relative standard deviations <10% and the accuracy values ranged from -7.2% to 8.4%. The proposed method was successfully applied to the pharmacokinetic drug interaction study of rosuvastatin combined with naringin in rats.
Collapse
Affiliation(s)
- Xuan Zeng
- Guangdong Key Laboratory of Plant Resources, Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, People's Republic of China
| | - Weiwei Su
- Guangdong Key Laboratory of Plant Resources, Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, People's Republic of China
| | - Hong Liu
- Guangdong Key Laboratory of Plant Resources, Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, People's Republic of China
| | - Yuying Zheng
- Guangdong Key Laboratory of Plant Resources, Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, People's Republic of China
| | - Taobin Chen
- Guangdong Key Laboratory of Plant Resources, Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, People's Republic of China
| | - Weijian Zhang
- Guangdong Key Laboratory of Plant Resources, Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, People's Republic of China
| | - Zenghao Yan
- Guangdong Key Laboratory of Plant Resources, Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, People's Republic of China
| | - Yang Bai
- Guangdong Key Laboratory of Plant Resources, Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, People's Republic of China
| | - Hongliang Yao
- Guangdong Key Laboratory of Plant Resources, Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, People's Republic of China
| |
Collapse
|
30
|
Lee J, Rhee SJ, Lee S, Yu KS. Evaluation of drug interactions between fimasartan and rosuvastatin after single and multiple doses in healthy Caucasians. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:787-794. [PMID: 29670335 PMCID: PMC5896677 DOI: 10.2147/dddt.s145339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Objectives As hypercholesterolemia is often accompanied by hypertension, statins are usually prescribed with angiotensin receptor blockers in clinical practice. This study was performed to evaluate the pharmacokinetics and safety of fimasartan and rosuvastatin when coadministered or administered alone as a single dose or as multiple doses to healthy Caucasians. Methods Thirty-six subjects were enrolled into an open-labeled, randomized, 6-sequence, 3-period, 3-way crossover study, and randomly received fimasartan (120 mg), rosuvastatin (20 mg) or both. Blood samples for pharmacokinetics were collected up to 48 hours for fimasartan and 72 hours for rosuvastatin after the last dosing and plasma concentrations of study drugs were determined by liquid chromatography-tandem mass spectrometry. Maximum plasma concentration (Cmax), area under the concentration-time curve (AUC) from 0 to the last measurable time (AUClast), maximum plasma concentration at steady state (Cmax,ss) and AUC to the end of the dosing period at steady state (AUCτ,ss) were estimated using a non-compartmental method. Safety and tolerability were evaluated throughout the study. Results Thirty subjects completed the study. After single dose administration, the geometric mean ratio (GMR) and 90% confidence intervals (CIs) of fimasartan with or without rosuvastatin were 0.95 (0.80–1.14) and 0.98 (0.91–1.07) for Cmax and AUClast, respectively. The corresponding values for rosuvastatin with or without fimasartan were 1.32 (1.16–1.50) and 0.97 (0.89–1.05), respectively. After administration of multiple doses, the GMRs (90% CIs) for Cmax,ss and AUCτ,ss of fimasartan with or without rosuvastatin were 0.94 (0.74–1.20) and 1.07 (0.90–1.16), respectively. The corresponding values for rosuvastatin with or without fimasartan were 1.16 (1.02–1.32) and 0.86 (0.79–0.94), respectively. A total of 74 adverse events (AEs) were reported and incidences of AEs did not increase significantly with co-administration. Conclusion Co-administration of fimasartan and rosuvastatin did not result in clinically relevant changes in the systemic exposure of fimasartan or rosuvastatin after single and multiple administrations, and they were well tolerated.
Collapse
Affiliation(s)
- Jieon Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Su-Jin Rhee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
31
|
Zha W. Transporter-mediated natural product-drug interactions for the treatment of cardiovascular diseases. J Food Drug Anal 2017; 26:S32-S44. [PMID: 29703385 PMCID: PMC9326887 DOI: 10.1016/j.jfda.2017.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/18/2017] [Accepted: 11/21/2017] [Indexed: 12/17/2022] Open
Abstract
The growing use of natural products in cardiovascular (CV) patients has been greatly raising the concerns about potential natural product–CV drug interactions. Some of these may lead to unexpected cardiovascular adverse effects and it is, therefore, essential to identify or predict potential natural product–CV drug interactions, and to understand the underlying mechanisms. Drug transporters are important determinants for the pharmacokinetics of drugs and alterations of drug transport has been recognized as one of the major causes of natural product–drug interactions. In last two decades, many CV drugs (e.g., angiotensin II receptor blockers, beta-blockers and statins) have been identified to be substrates and inhibitors of the solute carrier (SLC) transporters and the ATP-binding cassette (ABC) transporters, which are two major transporter superfamilies. Meanwhile, in vitro and in vivo studies indicate that a growing number of natural products showed cardioprotective effects (e.g., gingko biloba, danshen and their active ingredients) are also substrates and inhibitors of drug transporters. Thus, to understand transporter-mediated natural product–CV drug interactions is important and some transporter-mediated interactions have already shown to have clinical relevance. In this review, we review the current knowledge on the role of ABC and SLC transporters in CV therapy, as well as transporter modulation by natural products used in CV diseases and their induced natural product–CV drug interactions through alterations of drug transport. We hope our review will aid in a comprehensive summary of transporter-mediated natural product–CV drug interactions and help public and physicians understand these type of interactions.
Collapse
Affiliation(s)
- Weibin Zha
- MyoKardia, South San Francisco, CA, USA.
| |
Collapse
|
32
|
Kim TE, Shin D, Gu N, Jung BH, Kim J, Cho YM, Yu KS, Cho JY. The Effect of Genetic Polymorphisms in SLCO2B1
on the Lipid-Lowering Efficacy of Rosuvastatin in Healthy Adults with Elevated Low-Density Lipoprotein. Basic Clin Pharmacol Toxicol 2017. [DOI: 10.1111/bcpt.12826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Tae-Eun Kim
- Department of Clinical Pharmacology; Konkuk University Medical Center; Seoul Korea
- Department of Clinical Pharmacology and Therapeutics; Seoul National University College of Medicine and Hospital; Seoul Korea
| | - Dongseong Shin
- Department of Clinical Pharmacology and Therapeutics; Seoul National University College of Medicine and Hospital; Seoul Korea
- Clinical Trials Center; Gachon University Gil Medical Center; Incheon Korea
| | - Namyi Gu
- Department of Clinical Pharmacology and Therapeutics; Seoul National University College of Medicine and Hospital; Seoul Korea
- Department of Clinical Pharmacology and Therapeutics; Dongguk University College of Medicine and Ilsan Hospital; Goyang Gyeonggi-do Korea
| | - Byung Hwa Jung
- Molecular Recognition Research Center; Korea Institute of Science and Technology; Seoul Korea
| | - Jayoun Kim
- Research Coordinating Center; Konkuk University Medical Center; Seoul Korea
| | - Young Min Cho
- Department of Internal Medicine; Seoul National University College of Medicine and Hospital; Seoul Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics; Seoul National University College of Medicine and Hospital; Seoul Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics; Seoul National University College of Medicine and Hospital; Seoul Korea
| |
Collapse
|
33
|
Arrigoni E, Del Re M, Fidilio L, Fogli S, Danesi R, Di Paolo A. Pharmacogenetic Foundations of Therapeutic Efficacy and Adverse Events of Statins. Int J Mol Sci 2017; 18:ijms18010104. [PMID: 28067828 PMCID: PMC5297738 DOI: 10.3390/ijms18010104] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 12/11/2022] Open
Abstract
Background: In the era of precision medicine, more attention is paid to the search for predictive markers of treatment efficacy and tolerability. Statins are one of the classes of drugs that could benefit from this approach because of their wide use and their incidence of adverse events. Methods: Literature from PubMed databases and bibliography from retrieved publications have been analyzed according to terms such as statins, pharmacogenetics, epigenetics, toxicity and drug–drug interaction, among others. The search was performed until 1 October 2016 for articles published in English language. Results: Several technical and methodological approaches have been adopted, including candidate gene and next generation sequencing (NGS) analyses, the latter being more robust and reliable. Among genes identified as possible predictive factors associated with statins toxicity, cytochrome P450 isoforms, transmembrane transporters and mitochondrial enzymes are the best characterized. Finally, the solute carrier organic anion transporter family member 1B1 (SLCO1B1) transporter seems to be the best target for future studies. Moreover, drug–drug interactions need to be considered for the best approach to personalized treatment. Conclusions: Pharmacogenetics of statins includes several possible genes and their polymorphisms, but muscular toxicities seem better related to SLCO1B1 variant alleles. Their analysis in the general population of patients taking statins could improve treatment adherence and efficacy; however, the cost–efficacy ratio should be carefully evaluated.
Collapse
Affiliation(s)
- Elena Arrigoni
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Leonardo Fidilio
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Stefano Fogli
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy.
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| | - Antonello Di Paolo
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| |
Collapse
|
34
|
Roth AD, Lee MY. Idiosyncratic Drug-Induced Liver Injury (IDILI): Potential Mechanisms and Predictive Assays. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9176937. [PMID: 28133614 PMCID: PMC5241492 DOI: 10.1155/2017/9176937] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/29/2016] [Indexed: 12/16/2022]
Abstract
Idiosyncratic drug-induced liver injury (IDILI) is a significant source of drug recall and acute liver failure (ALF) in the United States. While current drug development processes emphasize general toxicity and drug metabolizing enzyme- (DME-) mediated toxicity, it has been challenging to develop comprehensive models for assessing complete idiosyncratic potential. In this review, we describe the enzymes and proteins that contain polymorphisms believed to contribute to IDILI, including ones that affect phase I and phase II metabolism, antioxidant enzymes, drug transporters, inflammation, and human leukocyte antigen (HLA). We then describe the various assays that have been developed to detect individual reactions focusing on each of the mechanisms described in the background. Finally, we examine current trends in developing comprehensive models for examining these mechanisms. There is an urgent need to develop a panel of multiparametric assays for diagnosing individual toxicity potential.
Collapse
Affiliation(s)
- Alexander D. Roth
- Department of Chemical & Biomedical Engineering, Cleveland State University, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| | - Moo-Yeal Lee
- Department of Chemical & Biomedical Engineering, Cleveland State University, 1960 East 24th Street, Cleveland, OH 44115-2214, USA
| |
Collapse
|
35
|
Shin E, Shin N, Oh JH, Lee YJ. High-Dose Metformin May Increase the Concentration of Atorvastatin in the Liver by Inhibition of Multidrug Resistance-Associated Protein 2. J Pharm Sci 2016; 106:961-967. [PMID: 27964938 DOI: 10.1016/j.xphs.2016.11.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/04/2016] [Accepted: 11/29/2016] [Indexed: 12/22/2022]
Abstract
In this study, we evaluated the effect of coadministered metformin on the biliary excretion and liver concentration of atorvastatin. To investigate the inhibitory effect of metformin on biliary efflux transporters, the transport of atorvastatin in MDCKII-MDR1, BCRP, and MRP2 was evaluated. The effects of metformin on the steady state liver concentration and biliary excretion of atorvastatin and 2-hydroxyatorvastatin were evaluated in SDR and Mrp2-deficient EHBR. Metformin did not inhibit the transport of atorvastatin via BCRP and MDR1. However, metformin significantly inhibited the transport of atorvastatin and 2-hydroxyatorvastatin via MRP2 (apparent IC50 = 12 and 2 μM). Coadministered metformin significantly increased the Kp,liver and Cliver (1.7- and 1.6-fold) and decreased the biliary clearance of atorvastatin (2.7-fold) in SDR, but it did not affect the plasma concentration and total clearance of atorvastatin. Similar effects by metformin were observed for 2-hydroxyatorvastatin. In addition, coadministered metformin did not have any effect in EHBR. Therefore, coadministered metformin increases the liver concentration of atorvastatin via inhibition of the Mrp2 in rats, without affecting the plasma concentration. This "silent interaction" by metformin in atorvastatin and metformin combination therapy may be related to the unnoticeable pharmacological synergism or unpredicted side effects of atorvastatin in the liver.
Collapse
Affiliation(s)
- Eunjung Shin
- Division of Biopharmaceutics, College of Pharmacy, Kyung Hee University, Seoul, South Korea; Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, South Korea
| | - Naree Shin
- Division of Biopharmaceutics, College of Pharmacy, Kyung Hee University, Seoul, South Korea; Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, South Korea
| | - Ju-Hee Oh
- Division of Biopharmaceutics, College of Pharmacy, Kyung Hee University, Seoul, South Korea
| | - Young-Joo Lee
- Division of Biopharmaceutics, College of Pharmacy, Kyung Hee University, Seoul, South Korea; Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
36
|
Wiggins BS, Saseen JJ, Page RL, Reed BN, Sneed K, Kostis JB, Lanfear D, Virani S, Morris PB. Recommendations for Management of Clinically Significant Drug-Drug Interactions With Statins and Select Agents Used in Patients With Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation 2016; 134:e468-e495. [DOI: 10.1161/cir.0000000000000456] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
Son M, Guk J, Kim Y, Woo Chae D, Heo YA, Soh D, Park K. Pharmacokinetic Interaction Between Rosuvastatin, Telmisartan, and Amlodipine in Healthy Male Korean Subjects: A Randomized, Open-label, Multiple-dose, 2-period Crossover Study. Clin Ther 2016; 38:1845-57. [PMID: 27422590 DOI: 10.1016/j.clinthera.2016.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/03/2016] [Accepted: 06/10/2016] [Indexed: 11/29/2022]
Abstract
PURPOSE Rosuvastatin, a hydroxy methylglutaryl coenzyme A reductase inhibitor; telmisartan, an angiotensin receptor blocker; and amlodipine, a calcium channel inhibitor, are commonly prescribed together for the treatment of hypertension nonresponsive to monotherapy and accompanied by dyslipidemia. However, the pharmacokinetic interactions among these 3 substances are not well understood. The aim of this study was to investigate the pharmacokinetic drug-drug interactions among rosuvastatin, telmisartan, and amlodipine in a healthy Korean male population. METHODS In both parts of this randomized, open-label, multiple-dose, 2-part, 2-period crossover study, subjects aged 19 to 55 years were enrolled. In part 1, each subject received rosuvastatin 20 mg with and without 2 fixed-dose combination (FDC) tablets of telmisartan/amlodipine 40/5 mg, once daily for 9 consecutive days. In part 2, each subject received 2 FDC tablets of telmisartan/amlodipine 40/5 mg with and without rosuvastatin 20 mg, once daily for 9 consecutive days. In both parts, there was a 13-day washout period between treatments. Pharmacokinetic samples were collected up to 72 hours after the last dose in subjects who received rosuvastatin only, and up to 144 hours after the last dose in subjects who received telmisartan/amlodipine with or without rosuvastatin. Adverse events (AEs) were assessed via interviews and physical examinations. FINDINGS Forty-eight subjects were enrolled, of whom 19 in part 1 and 22 in part 2 completed the study. In Part 1, the 90% CIs of the geometric mean ratios (GMRs) (coadministration of rosuvastatin and telmisartan/amlodipine to monotherapy with rosuvastatin) of the primary pharmacokinetic parameters (AUCτ and Cmax,ss) were: rosuvastatin, 1.1436 to 1.3059 and 1.8970 to 2.3514, respectively; and N-desmethyl rosuvastatin, 0.8441 to 1.0200 and 1.1971 to 1.5457. In part 2, the 90% CIs of the GMRs (coadministration to monotherapy with telmisartan/amlodipine) were: telmisartan, 1.1204 to 1.4228 and 0.9940 to 1.5940; amlodipine, 0.9705 to 1.0636 and 0.9813 to 1.0779. There were no significant differences in the prevalences of AEs between the treatments, and all reported AEs were mild or moderate. IMPLICATIONS These results demonstrate that when rosuvastatin, telmisartan, and amlodipine are coadministered to healthy male subjects, pharmacokinetic exposure increases with respect to rosuvastatin and telmisartan, whereas no change occurs with respect to amlodipine. However, based on previous analyses, the degree of increase in the exposure observed was not regarded as clinically significant. All treatments were well-tolerated.
Collapse
Affiliation(s)
- Mijeong Son
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Jinju Guk
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Yukyung Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Dong Woo Chae
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Young-A Heo
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Dongjun Soh
- Jeil Pharmaceutical Co, Ltd, Seoul, Republic of Korea
| | - Kyungsoo Park
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Dallas S, Salphati L, Gomez-Zepeda D, Wanek T, Chen L, Chu X, Kunta J, Mezler M, Menet MC, Chasseigneaux S, Declèves X, Langer O, Pierre E, DiLoreto K, Hoft C, Laplanche L, Pang J, Pereira T, Andonian C, Simic D, Rode A, Yabut J, Zhang X, Scheer N. Generation and Characterization of a Breast Cancer Resistance Protein Humanized Mouse Model. Mol Pharmacol 2016; 89:492-504. [PMID: 26893303 DOI: 10.1124/mol.115.102079] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/17/2016] [Indexed: 12/17/2022] Open
Abstract
Breast cancer resistance protein (BCRP) is expressed in various tissues, such as the gut, liver, kidney and blood brain barrier (BBB), where it mediates the unidirectional transport of substrates to the apical/luminal side of polarized cells. Thereby BCRP acts as an efflux pump, mediating the elimination or restricting the entry of endogenous compounds or xenobiotics into tissues and it plays important roles in drug disposition, efficacy and safety. Bcrp knockout mice (Bcrp(-/-)) have been used widely to study the role of this transporter in limiting intestinal absorption and brain penetration of substrate compounds. Here we describe the first generation and characterization of a mouse line humanized for BCRP (hBCRP), in which the mouse coding sequence from the start to stop codon was replaced with the corresponding human genomic region, such that the human transporter is expressed under control of the murineBcrppromoter. We demonstrate robust human and loss of mouse BCRP/Bcrp mRNA and protein expression in the hBCRP mice and the absence of major compensatory changes in the expression of other genes involved in drug metabolism and disposition. Pharmacokinetic and brain distribution studies with several BCRP probe substrates confirmed the functional activity of the human transporter in these mice. Furthermore, we provide practical examples for the use of hBCRP mice to study drug-drug interactions (DDIs). The hBCRP mouse is a promising model to study the in vivo role of human BCRP in limiting absorption and BBB penetration of substrate compounds and to investigate clinically relevant DDIs involving BCRP.
Collapse
Affiliation(s)
- Shannon Dallas
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Laurent Salphati
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - David Gomez-Zepeda
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Thomas Wanek
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Liangfu Chen
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Xiaoyan Chu
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Jeevan Kunta
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Mario Mezler
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Marie-Claude Menet
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Stephanie Chasseigneaux
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Xavier Declèves
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Oliver Langer
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Esaie Pierre
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Karen DiLoreto
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Carolin Hoft
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Loic Laplanche
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Jodie Pang
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Tony Pereira
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Clara Andonian
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Damir Simic
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Anja Rode
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Jocelyn Yabut
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Xiaolin Zhang
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| | - Nico Scheer
- DMPK and Bioanalytical Research, Abbvie Deutschland GmbH & Co. KG, Ludwigshafen, Germany (M.M., C.H., L.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA (L.S., J.P., X.Z.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania (L.C., C.A., E.P.); Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (T.W., O.L.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria (O.L.), Preclinical Development and Safety, Janssen Research and Development, LLC, Spring House, PA (S.D., J.K., K.D., D.S.). Merck Sharp and Dohme Corporation, Whitehouse Station, New Jersey (X.C., T.P., J.Y.); Université Paris Descartes, UMR-S 1144, Paris, France (D.G.-Z., M.-C.M., S.C., X.D.); Taconic Biosciences GmbH, Koeln, Germany (A.R., N.S.)
| |
Collapse
|
39
|
Guo M, Dai X, Hu D, Zhang Y, Sun Y, Ren W, Wang L. Potential pharmacokinetic effect of rifampicin on enrofloxacin in broilers: Roles of P-glycoprotein and BCRP induction by rifampicin. Poult Sci 2016; 95:2129-35. [PMID: 27118859 DOI: 10.3382/ps/pew148] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2016] [Indexed: 12/11/2022] Open
Abstract
P-glycoprotein ( P-GP: , encoding gene Abcb1) and Breast Cancer Resistance Protein ( BCRP: , encoding gene Abcg2) are transport proteins that play a major role in modulating the bioavailability of oral drugs in humans and rodents. It has been shown that rifampicin is the typical inducer of P-gp in rodents by activating the nuclear receptor. However, its effect on Abcb1, Abcg2, CYP3A, and chicken xenobiotic-sensing orphan nuclear receptor ( CXR: ) mRNA expression in broilers is poorly understood. This study explored the effect of rifampicin on mRNA expression of Abcb1, Abcg2, CYP3A37, CXR as well as its effect on the pharmacokinetics of enrofloxacin in broilers. The mRNA levels of Abcb1, Abcg2, CYP3A37, and CXR were significantly increased in the liver (except Abcg2), kidney, jejunum, and ileum (P < 0.05) but not significantly changed in the duodenum (P > 0.05) after treated with rifampicin. Further analysis revealed that the variation tendencies of Abcb1, Abcg2, and CYP3A37 expression levels were significantly correlated with CXR mRNA expression levels in liver, kidney, jejunum, and ileum. Coadministration of rifampicin significantly changed the pharmacokinetic behavior of enrofloxacin orally administered by showing clearly lower AUC0-∞, AUC0-t, and Cmax as well as longer Tmax. The bioavailability of orally administered enrofloxacin was decreased from 72.5% to 24.8% by rifampicin. However, rifampicin did not significantly change the pharmacokinetics of enrofloxacin following intravenous administration. Our study shows that rifampicin up-regulated the small intestinal level of P-gp and BCRP and suggests that P-gp and BCRP are key factors that affected pharmacokinetic behavior of orally administered enrofloxacin by limiting its absorption from the intestine in broilers.
Collapse
Affiliation(s)
- Mengjie Guo
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, PR China Center for Safety Evaluation of Drugs, Science and Technology Division, Nanjing University of Traditional Chinese Medicine, Nanjing 210029, China
| | - Xiaohua Dai
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, PR China College of Food Science and Pharmacy, Xinjiang Agricultural University, Urumqi 830052, China
| | - Dongmin Hu
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, PR China
| | - Yu Zhang
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, PR China
| | - Yong Sun
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, PR China
| | - Weilong Ren
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, PR China
| | - Liping Wang
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, PR China
| |
Collapse
|
40
|
Pharmacokinetic drug interactions of the selective androgen receptor modulator GTx-024(Enobosarm) with itraconazole, rifampin, probenecid, celecoxib and rosuvastatin. Invest New Drugs 2016; 34:458-67. [PMID: 27105861 DOI: 10.1007/s10637-016-0353-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/14/2016] [Indexed: 02/06/2023]
Abstract
GTx-024 (also known as enobosarm) is a first in class selective androgen receptor modulator being developed for diverse indications in oncology. Preclinical studies of GTx-024 supported the evaluation of several potential drug-drug interactions in a clinical setting. A series of open-label Phase I GTx-024 drug-drug interaction studies were designed to interrogate potential interactions with CYP3A4 inhibitor (itraconazole), a CYP3A4 inducer (rifampin), a pan-UGT inhibitor (probenecid), a CYP2C9 substrate (celecoxib) and a BCRP substrate (rosuvastatin). The plasma pharmacokinetics of GTx-024, its major metabolite (GTx-024 glucuronide), and each substrate were characterized in detail. Itraconazole administration had no effect on GTx-024 pharmacokinetics. Likewise, GTx-024 administration did not significantly change the pharmacokinetics of celecoxib or rosuvastatin. Rifampin administration had the largest impact on GTx-024 pharmacokinetics of any co-administered agent and reduced the maximal plasma concentration (Cmax) by 23 % and the area under the curve (AUC∞) by 43 %. Probenecid had a complex interaction with GTx-024 whereby both GTx-024 plasma levels and GTx-024 glucuronide plasma levels (AUC∞) were increased by co-administration of the UGT inhibitor (50 and 112 %, respectively). Overall, GTx-024 was well tolerated and poses very little risk of generating clinically relevant drug-drug interactions.
Collapse
|
41
|
Li Q, Yang H, Guo D, Zhang T, Polli JE, Zhou H, Shu Y. Effect of Ondansetron on Metformin Pharmacokinetics and Response in Healthy Subjects. Drug Metab Dispos 2016; 44:489-94. [PMID: 26825640 PMCID: PMC4810771 DOI: 10.1124/dmd.115.067223] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/19/2016] [Indexed: 01/11/2023] Open
Abstract
The 5-hydroxytryptamine-3 (5-HT3) receptor antagonists such as ondansetron have been used to prevent and treat nausea and vomiting for over 2 decades. This study was to determine whether ondansetron could serve as a perpetrator drug causing transporter-mediated drug-drug interactions in humans. Twelve unrelated male healthy Chinese volunteers were enrolled into a prospective, randomized, double-blind, crossover study to investigate the effects of ondansetron or placebo on the pharmacokinetics of and the response to metformin, a well-characterized substrate of organic cation transporters and multidrug and toxin extrusions (MATEs). Ondansetron treatment caused a statistically significantly higher Cmax of metformin compared with placebo (18.3 ± 5.05 versus 15.2 ± 3.23; P = 0.006) and apparently decreased the renal clearance of metformin by 37% as compared with placebo (P = 0.001). Interestingly, ondansetron treatment also statistically significantly improved glucose tolerance in subjects, as indicated by the smaller glucose area under the curve in the oral glucose tolerance test (10.4 ± 1.43) as compared with placebo (11.5 ± 2.29 mmol∙mg/l) (P = 0.020). It remains possible that ondansetron itself may affect glucose homeostasis in human subjects, but our clinical study, coupled with our previous findings in cells and in animal models, indicates that ondansetron can cause a drug-drug interaction via its potent inhibition of MATE transporters in humans.
Collapse
Affiliation(s)
- Qing Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (Q.L., H.Y., D.G., J.E.P., Y.S.); Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Hunan, People's Republic of China (Q.L., T.Z., H.Z.)
| | - Hong Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (Q.L., H.Y., D.G., J.E.P., Y.S.); Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Hunan, People's Republic of China (Q.L., T.Z., H.Z.)
| | - Dong Guo
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (Q.L., H.Y., D.G., J.E.P., Y.S.); Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Hunan, People's Republic of China (Q.L., T.Z., H.Z.)
| | - Taolan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (Q.L., H.Y., D.G., J.E.P., Y.S.); Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Hunan, People's Republic of China (Q.L., T.Z., H.Z.)
| | - James E Polli
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (Q.L., H.Y., D.G., J.E.P., Y.S.); Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Hunan, People's Republic of China (Q.L., T.Z., H.Z.)
| | - Honghao Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (Q.L., H.Y., D.G., J.E.P., Y.S.); Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Hunan, People's Republic of China (Q.L., T.Z., H.Z.)
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (Q.L., H.Y., D.G., J.E.P., Y.S.); Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Hunan, People's Republic of China (Q.L., T.Z., H.Z.)
| |
Collapse
|
42
|
The effects of rosuvastatin on lipid-lowering, inflammatory, antioxidant and fibrinolytics blood biomarkers are influenced by Val16Ala superoxide dismutase manganese-dependent gene polymorphism. THE PHARMACOGENOMICS JOURNAL 2016; 16:501-506. [PMID: 26882122 DOI: 10.1038/tpj.2015.91] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 10/29/2015] [Accepted: 11/13/2015] [Indexed: 12/28/2022]
Abstract
Rosuvastatin is a cholesterol-lowering drug that also attenuates the inflammatory process and oxidative stress via the reduction of superoxide anion production. Superoxide anions are metabolized by manganese-dependent superoxide dismutase (MnSOD or SOD2) in the mitochondria. In humans, there is a gene polymorphism where a change of alanine (Ala) to valine (Val) occurs at the 16th amino acid (Ala16Val-SOD2). The VV genotype has been associated with the risk of developing several metabolic diseases, such as hypercholesterolemia. Thus, to further explore this phenomenon, this study investigated the influence of the Val16Ala-SOD2 polymorphism on the lipid profile and inflammatory and fibrinolytic biomarkers of 122 hypercholesterolemic patients undergoing the first pharmacological cholesterol-lowering therapy who were treated with 20 mg rosuvastatin for 120 days. The findings indicate that the VV patients who present a low-efficiency SOD2 enzyme exhibit an attenuated response to rosuvastatin compared with the A-allele patients. The effect of rosuvastatin on inflammatory and fibrinolytic biomarkers was also less intense in the VV patients. These results suggest some pharmacogenetic effects of Val16Ala-SOD2 in hypercholesterolemia treatment.
Collapse
|
43
|
Lu XF, Zhou Y, Bi KS, Chen XH. Mixed effects of OATP1B1, BCRP and NTCP polymorphisms on the population pharmacokinetics of pravastatin in healthy volunteers. Xenobiotica 2016; 46:841-9. [PMID: 26744986 DOI: 10.3109/00498254.2015.1130881] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
1. Pravastatin is a 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor used for the treatment of hyperlipidaemia. This study aims to investigate the effects of genetic polymorphisms in OATP1B1, BCRP and NTCP on pravastatin population pharmacokinetics in healthy Chinese volunteers using a non-linear mixed-effect modelling (NONMEM) approach. A two-compartment model with a first-order absorption and elimination described plasma pravastatin concentrations well. 2. Genetic polymorphisms of rs4149056 (OATP1B1) and rs2306283 (OATP1B1) were found to be associated with a significant (p < 0.01) decrease in the apparent clearance from the central compartment (CL/F), while rs2296651 (NTCP) increased CL/F to a significant degree (p < 0.01). The combination of these three polymorphisms reduced the inter-individual variability of CL/F by 78.8%. 3. There was minimal effect of rs2231137 (BCRP) and rs2231142 (BCRP) on pravastatin pharmacokinetics (0.01 < p < 0.05), whereas rs11045819 (OATP1B1), rs1061018 (BCRP) and rs61745930 (NTCP) genotypes do not appear to be associated with pravastatin pharmacokinetics based on the population model (p > 0.05). 4. The current data suggest that the combination of rs4149056, rs2306283 and rs2296651 polymorphisms is an important determinant of pravastatin pharmacokinetics.
Collapse
Affiliation(s)
- Xue-Feng Lu
- a Department of Pharmaceutical Analysis , School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , China and
| | - Yang Zhou
- b Department of Measurement and Control , School of Physics, Liaoning University , Shenyang , China
| | - Kai-Shun Bi
- a Department of Pharmaceutical Analysis , School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , China and
| | - Xiao-Hui Chen
- a Department of Pharmaceutical Analysis , School of Pharmacy, Shenyang Pharmaceutical University , Shenyang , China and
| |
Collapse
|
44
|
Park WS, Jang D, Han S, Yim DS. Mixed–effects analysis of increased rosuvastatin absorption by coadministered telmisartan. Transl Clin Pharmacol 2016. [DOI: 10.12793/tcp.2016.24.1.55] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Wan-Su Park
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, PIPET (Pharmacometrics Institute for Practical Education and Training), College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Dooyeon Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, PIPET (Pharmacometrics Institute for Practical Education and Training), College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seunghoon Han
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, PIPET (Pharmacometrics Institute for Practical Education and Training), College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Dong-Seok Yim
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, PIPET (Pharmacometrics Institute for Practical Education and Training), College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
45
|
Wen JH, Wei XH, Sheng XY, Zhou DQ, Peng HW, Lu YN, Zhou J. Effect of Ursolic Acid on Breast Cancer Resistance Protein-mediated Transport of Rosuvastatin In Vivo and Vitro. CHINESE MEDICAL SCIENCES JOURNAL = CHUNG-KUO I HSUEH K'O HSUEH TSA CHIH 2015; 30:218-25. [PMID: 26960302 DOI: 10.1016/s1001-9294(16)30004-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To evaluate whether ursolic acid can inhibit breast cancer resistance protein (BCRP)-mediated transport of rosuvastatin in vivo and in vitro. METHODS Firstly, we explored the pharmacokinetics of 5-fluorouracil (5-FU, a substrate of BCRP) in rats in the presence or absence of ursolic acid. Secondly, we studied the pharmacokinetics of rosuvastatin in rats in the presence or absence of ursolic acid or Ko143 (inhibitor of BCRP). Finially, the concentration-dependent transport of rosuvastatin and the inhibitory effects of ursolic acid and Ko143 were examined in Madin-Darby Canine Kidney (MDCK) 2-BCRP421CC (wild type) cells and MDCK2-BCRP421AA (mutant type) cells. RESULTS As a result, significant changes in pharmacokinetics parameters of 5-FU were observed in rats following pretreatment with ursolic acid. Both ursolic acid and Ko143 could significantly affect the pharmacokinetics of rosuvastatin. The rosuvastatin transport in the BCRP overexpressing system was increased in a concentration-dependent manner. However, there was no statistical difference in BCRP-mediated transport of rosuvastatin betweent the wild type cells and mutant cells. The same as Ko143, ursolic acid inhibited BCRP-mediated transport of rosuvastatin in vitro. CONCLUSION Ursolic acid appears to be a potent modulator of BCRP that affects the pharmacokinetic of rosuvastatin in vivo and inhibits the transport of rosuvastatin in vitro.
Collapse
Affiliation(s)
- Jin-hua Wen
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiao-hua Wei
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiang-yuan Sheng
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - De-qing Zhou
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Hong-wei Peng
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yan-ni Lu
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jian Zhou
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
46
|
You Q, Ma ZC, Wang YG, Hu DH, Liang QD, Xiao CR, Tan HL, Tang XL, Gao Y. Panax ginseng inhibits intestinal absorption of toxic Aconitum carmichaeli alkaloids in Vitro. Chin J Integr Med 2015. [PMID: 26272549 DOI: 10.1007/s11655-015-2282-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To evaluate the rationality and compatibility of Shenfu Formula (, SFF), a typical Chinese medicine (CM) comprised of Panax ginseng and Aconitum carmichaeli. METHODS Caco-2 cells were used to study the permeability of Aconitum carmichaeli marker compounds when the CM preparation was combined with Panax ginseng. P-glycoprotein (P-gp) activity and protein as well as multidrug resistance 1 (MDR1) mRNA were analyzed with rhodamine123 efflflux, western blot and real time quantitative polymerase chain reaction. RESULTS Aconitine (AC), mesaconitine (MA), hypaconitine (HA) and fifive other active alkaloids in Aconitum carmichaeli were selected as marker compounds. Panax ginseng inhibited intestinal absorption of highly toxic AC, MA and HA from Aconitum carmichaeli in Caco-2 cells. P-gp and breast cancer resistance protein (BCRP) were observed to be involved in AC, MA and HA efflflux. Panax ginseng induced P-gp activity in Caco-2 cells via increased MDR1/P-gp expression. Thus, Panax ginseng facilitated P-gp-mediated efflflux of toxic Aconitum carmichaeli alkaloids and restricted their intestinal absorption without inflfluencing other active components. CONCLUSION Future studies to elucidate mechanism of reduced toxicity of Aconitum carmichaeli when combined with Panax ginseng will guide future formula optimization.
Collapse
Affiliation(s)
- Qing You
- Beijing Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing, 100850, China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Jung JA, Lee SY, Kim JR, Ko JW, Jang SB, Nam SY, Huh W. A pharmacokinetic and pharmacodynamic drug interaction between rosuvastatin and valsartan in healthy subjects. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:745-52. [PMID: 25767372 PMCID: PMC4354399 DOI: 10.2147/dddt.s76942] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
PURPOSE Valsartan, an angiotensin-receptor blocker, and rosuvastatin, a competitive inhibitor of the 3-hydroxy-3-methylglutaryl coenzyme A reductase, are frequently coadministered to treat patients with hypertension and dyslipidemia. The study reported here sought to evaluate the pharmacokinetic and pharmacodynamic interactions between rosuvastatin and valsartan in healthy Korean subjects. SUBJECTS AND METHODS Thirty healthy male Korean subjects were administered with rosuvastatin (20 mg/day), valsartan (160 mg/day), and both drugs concomitantly for 4 days in a randomized, open-label, multiple-dose, three-treatment, three-period crossover study. Plasma concentrations of rosuvastatin, N-desmethyl rosuvastatin, and valsartan were determined using validated high-performance liquid chromatography with tandem mass spectrometry. Lipid profiles and vital signs (systolic and diastolic blood pressure and pulse rate) were measured for the pharmacodynamic assessment. RESULTS For rosuvastatin, the geometric mean ratios (90% confidence intervals [CIs]) of coadministration to mono-administration were 0.8809 (0.7873-0.9857) for maximum plasma concentration at steady state and 0.9151 (0.8632-0.9701) for area under the concentration-time curve (AUC) over a dosing interval at steady state. For valsartan, the geometric mean ratios (90% CIs) of those were 0.9300 (0.7946-1.0884) and 1.0072 (0.8893-1.1406), respectively. There were no significant differences in the metabolic ratio of N-desmethyl rosuvastatin AUC to rosuvastatin AUC between coadministration and rosuvastatin alone. No interaction was found in terms of systolic or diastolic blood pressure or lipid profiles. Combined treatment with valsartan and rosuvastatin was generally well tolerated without serious adverse events. CONCLUSION The pharmacokinetic profiles of rosuvastatin and valsartan in combination were comparable with those of rosuvastatin and valsartan administered individually, suggesting that their individual pharmacokinetics were not affected by their coadministration. No dose adjustment was required and the results are supportive of a study in a larger patient population.
Collapse
Affiliation(s)
- Jin Ah Jung
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Sungkyunkwan University, Seoul, Kore
| | - Soo-Yun Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea
| | - Jung-Ryul Kim
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Sungkyunkwan University, Seoul, Kore
| | - Jae-Wook Ko
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Sungkyunkwan University, Seoul, Kore ; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea
| | - Seong Bok Jang
- Yuhan Research Institute, Yuhan Corporation, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Su Youn Nam
- Yuhan Research Institute, Yuhan Corporation, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Wooseong Huh
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Sungkyunkwan University, Seoul, Kore ; Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
48
|
Sheng J, Tian X, Xu G, Wu Z, Chen C, Wang L, Pan L, Huang C, Pan G. The hepatobiliary disposition of timosaponin b2 is highly dependent on influx/efflux transporters but not metabolism. Drug Metab Dispos 2015; 43:63-72. [PMID: 25336752 DOI: 10.1124/dmd.114.059923] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The purpose of this study was to characterize the hepatobiliary disposition of timosaponin B2 (TB-2), a natural saponin. Although TB-2 has multiple pharmacologic activities, the mechanism of its hepatobiliary disposition has not been explored. Because the metabolism of TB-2 is limited and the accumulation of TB-2 in primary hepatocytes is highly temperature dependent (93% of its accumulation is due to active uptake), the contribution of hepatic transporters was investigated. Organic anion-transporting polypeptide (OATP) 1B1- and OATP1B3-transfected human embryonic kidney 293 cells were employed. TB-2 serves as a substrate for OATP1B1 and OATP1B3, with the former playing a predominant role in the hepatic uptake of TB-2. An inhibition study in sandwich-cultured rat hepatocytes suggested that TB-2 is a substrate for both breast cancer resistance protein (Bcrp) and multidrug resistance-associated protein 2 (Mrp2), consistent with its high biliary excretion index (43.1-44.9%). This hypothesis was further verified in BCRP and MRP2 membrane vesicles. The cooperation of uptake and efflux transporters in TB-2 hepatic disposition could partially explain the double-peak phenomenon observed in rat plasma and liver and biliary clearance, which accounted for 70% of the total TB-2 clearance. Moreover, TB-2 significantly increased the rosuvastatin concentration in rat plasma in a concentration-dependent manner and decreased its biliary excretion, which corresponded to reductions in rosuvastatin accumulation in hepatocytes and the biliary excretion index in sandwich-cultured rat hepatocytes, representing a perfect example of a potential saponin-statin drug-drug interaction. These studies demonstrate that transporters (Oatp, Bcrp/Mrp2), but not metabolism, contribute significantly to rat TB-2 hepatobiliary disposition.
Collapse
Affiliation(s)
- Jingjing Sheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.T., Z.W., L.W., C.H., G.P.); Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (J.S., G.X., L.P.); and Institute of Life Sciences, Jiangsu University, Zhenjiang, People's Republic of China (C.C.)
| | - Xiaoting Tian
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.T., Z.W., L.W., C.H., G.P.); Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (J.S., G.X., L.P.); and Institute of Life Sciences, Jiangsu University, Zhenjiang, People's Republic of China (C.C.)
| | - Guanglin Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.T., Z.W., L.W., C.H., G.P.); Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (J.S., G.X., L.P.); and Institute of Life Sciences, Jiangsu University, Zhenjiang, People's Republic of China (C.C.)
| | - Zhitao Wu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.T., Z.W., L.W., C.H., G.P.); Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (J.S., G.X., L.P.); and Institute of Life Sciences, Jiangsu University, Zhenjiang, People's Republic of China (C.C.)
| | - Chen Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.T., Z.W., L.W., C.H., G.P.); Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (J.S., G.X., L.P.); and Institute of Life Sciences, Jiangsu University, Zhenjiang, People's Republic of China (C.C.)
| | - Le Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.T., Z.W., L.W., C.H., G.P.); Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (J.S., G.X., L.P.); and Institute of Life Sciences, Jiangsu University, Zhenjiang, People's Republic of China (C.C.)
| | - Lili Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.T., Z.W., L.W., C.H., G.P.); Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (J.S., G.X., L.P.); and Institute of Life Sciences, Jiangsu University, Zhenjiang, People's Republic of China (C.C.)
| | - Chenggang Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.T., Z.W., L.W., C.H., G.P.); Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (J.S., G.X., L.P.); and Institute of Life Sciences, Jiangsu University, Zhenjiang, People's Republic of China (C.C.)
| | - Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (X.T., Z.W., L.W., C.H., G.P.); Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (J.S., G.X., L.P.); and Institute of Life Sciences, Jiangsu University, Zhenjiang, People's Republic of China (C.C.)
| |
Collapse
|
49
|
Mei Y, Wang SY, Li Y, Yi SQ, Wang CY, Yang M, Duan KM. Role of SLCO1B1, ABCB1, and CHRNA1 gene polymorphisms on the efficacy of rocuronium in Chinese patients. J Clin Pharmacol 2014; 55:261-8. [PMID: 25279974 DOI: 10.1002/jcph.405] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022]
Abstract
This study explored the role of SLCO1B1, ABCB1, and CHRNA1 gene polymorphisms on the efficacy and duration of action of rocuronium in Chinese patients. Two hundred seven unrelated Chinese patients scheduled for elective surgery were recruited, and 200 completed the study. Their ABCB1, SLCO1B1, and CHRNA1 genotypes were determined. Demographic and clinical non-genetic data also were collected. The SLCO1B1, ABCB1, and CHRNA1 variants did not affect the onset time of rocuronium. Clinical duration and recovery time of rocuronium were prolonged in patients with the ABCB1 rs1128503TT and SLCO1B1 rs2306283 AG and GG genotypes. We demonstrate that the SLCO1B1 and ABCB1 gene variants could affect the pharmacodynamics of rocuronium. The ABCB1 rs1128503 C>T genotype was the most important factor on the efficacy of rocuronium.
Collapse
Affiliation(s)
- Yang Mei
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Sai-Ying Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.,Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, People's Republic of China
| | - Yang Li
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Shuang-Qiang Yi
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Chun-Yan Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Mi Yang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Kai-Ming Duan
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
50
|
Hua WJ, Hua WX, Nan FY, Jiang WA, Yan C. The influence of herbal medicine ursolic acid on the uptake of rosuvastatin mediated by OATP1B1*1a and *5. Eur J Drug Metab Pharmacokinet 2014; 39:221-30. [PMID: 24736980 PMCID: PMC4142139 DOI: 10.1007/s13318-014-0187-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 03/07/2014] [Indexed: 02/08/2023]
Abstract
Chinese herbal medicines such as hawthorn, salvia, etc., are frequently combined with statins so as to treat cardiovascular diseases more effectively. Chinese herbal medicines contain many kinds of active components, which may have drug-drug interactions with statins. This study aims to explore the effect and mechanism by which ursolic acid affects OATP1B1-mediated transport of rosuvastatin. This study will explore the effect of ursolic acid on OAPT1B1-mediated transport of rosuvastatin in the different cell systems. Given the genetic polymorphisms of OATP1B1, simultaneously, this study will further explore the effect of ursolic acid on OATP1B1 (521T>C)-mediated transport of rosuvastatin. When the concentration of ursolic acid was 1.8 and 18 µM, it showed that ursolic acid significantly inhibits the uptake of rosuvastatin in both OATP1B1*1a-HEK 293T cells and OATP1B1*5-HEK 293T cells. The reduction of OATP1B1*1a transport of rosuvastatin were 34.60 ± 2.99 and 66.08 ± 1.83 %, and for OATP1B1*5 were 34.27 ± 7.08 % and 66.95 ± 1.14 %. Inhibitory parameters of IC50 were 6.25 ± 0.42 and 6.07 ± 0.57 µM, respectively. This study suggests that ursolic acid can affect the uptake of rosuvastatin in hepatocytes by inhibiting the transport of OATP1B1, and gene mutation of OATP1B1 may cause different effects on its transport of rosuvastatin.
Collapse
Affiliation(s)
- Wen Jin Hua
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China,
| | | | | | | | | |
Collapse
|