1
|
Zeng W, Deng H, Luo Y, Zhong S, Huang M, Tomlinson B. Advances in statin adverse reactions and the potential mechanisms: A systematic review. J Adv Res 2024:S2090-1232(24)00598-8. [PMID: 39681285 DOI: 10.1016/j.jare.2024.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Elevated low-density lipoprotein (LDL) cholesterol is a major risk factor for cardiovascular disease. Statins are the cornerstone of preventing and treating cardiovascular disease and can reduce LDL cholesterol by more than 60%. Although statins have high tolerability and safety, as the number of users increases, their adverse reactions in the liver, kidneys, skeletal muscles, and their potential to induce diabetes have also received widespread attention. AIM OF REVIEW How to maximize the lipid-lowering effect of statins, reduce the incidence of adverse reactions, promote the rational application of statins in the clinic, and improve the risk-benefit level, in order to benefit more cardiovascular patients and provide reference for the related basic research of statins. Key scientific concepts of review: This article provides a comprehensive review of the clinical manifestations of statin-related adverse reactions (associated myopathy, hepatotoxicity, nephrotoxicity, glycemic effects, central nervous system, hemorrhagic stroke, etc.), risk factors for triggering adverse reactions, statin interactions with other drugs (food), potential etiopathological mechanisms and common interventions in the clinic. Genetic diversity is strongly associated with statin adverse effects, and thus, in the future genetic testing may also be key to mitigating statin adverse effects.
Collapse
Affiliation(s)
- Weiwei Zeng
- Department of Pharmacy, Shenzhen Longgang Second People's Hospital, Shenzhen 518112, Guangdong, China
| | - Huan Deng
- Department of Pharmacy, Shenzhen Longgang Second People's Hospital, Shenzhen 518112, Guangdong, China
| | - Yuning Luo
- Department of Pharmacy, Shenzhen Longgang Second People's Hospital, Shenzhen 518112, Guangdong, China
| | - Shilong Zhong
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangzhou, Guangdong, China
| | - Min Huang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Brian Tomlinson
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macau.
| |
Collapse
|
2
|
Kim JH, Hong JH, Jung JG, Jung WT, Nam KY, Roh JS, Choi YW, Bang J, Huh H, Lee HJ, Moon J, Kim J, Sunwoo J. Pharmacokinetic and Safety Comparison of Fixed-Dose Combination of Cilostazol/Rosuvastatin (200 + 20 mg) Versus Concurrent Administration of the Separate Components in Healthy Adults. Clin Pharmacol Drug Dev 2024; 13:842-851. [PMID: 38469999 DOI: 10.1002/cpdd.1390] [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: 12/11/2023] [Accepted: 01/23/2024] [Indexed: 03/13/2024]
Abstract
The combined cilostazol and rosuvastatin therapy is frequently used for coronary artery disease treatment. This open-label, 3 × 3 crossover clinical trial evaluated the pharmacokinetics and safety of a fixed-dose combination (FDC) of cilostazol/rosuvastatin (200 + 20 mg) versus a concurrent administration of the separate components (SCs) under both fasted and fed conditions. Among 48 enrolled healthy adults, 38 completed the study. Participants were administered a single oral dose of cilostazol/rosuvastatin (200 + 20 mg), either as an FDC or SCs in a fasted state, or FDC in a fed state, in each period of the trial. Blood samples were taken up to 48 hours after dosing, and plasma concentrations were analyzed using validated liquid chromatography-tandem mass spectrometry. The geometric mean ratios of FDC to SCs for area under the plasma concentration-time curve from time zero to the last quantifiable concentration (AUClast) and maximum plasma concentration (Cmax) were 0.94/1.05 and 1.06/1.15 for cilostazol and rosuvastatin, respectively (AUClast/Cmax). Compared with that during fasting, fed-state administration increased the AUClast and Cmax for cilostazol by approximately 72% and 160% and decreased these parameters for rosuvastatin by approximately 39% and 43%, respectively. To conclude, the FDC is bioequivalent to the SCs, with notable differences in pharmacokinetics when administered in a fed state. No significant safety differences were observed between the treatments.
Collapse
Affiliation(s)
- Jae Hoon Kim
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jang Hee Hong
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jin-Gyu Jung
- Department of Family Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Won Tae Jung
- Korea United Pharmaceutical Corporation, Seoul, Republic of Korea
| | - Kyu-Yeol Nam
- Korea United Pharmaceutical Corporation, Seoul, Republic of Korea
| | - Jae Seok Roh
- Korea United Pharmaceutical Corporation, Seoul, Republic of Korea
| | - Youn Woong Choi
- Korea United Pharmaceutical Corporation, Seoul, Republic of Korea
| | - Junbae Bang
- Korea United Pharmaceutical Corporation, Seoul, Republic of Korea
| | - Hyunwook Huh
- Korea United Pharmaceutical Corporation, Seoul, Republic of Korea
| | - Hye J Lee
- Caleb Multilab Corporation, Seoul, Republic of Korea
| | - JungHa Moon
- Caleb Multilab Corporation, Seoul, Republic of Korea
| | - Jaehee Kim
- Caleb Multilab Corporation, Seoul, Republic of Korea
| | - Jung Sunwoo
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Republic of Korea
| |
Collapse
|
3
|
Izat N, Kaplan O, Çelebier M, Sahin S. An Isolated Perfused Rat Liver Model: Simultaneous LC-MS Quantification of Pitavastatin, Coproporphyrin I, and Coproporphyrin III Levels in the Rat Liver and Bile. ACS OMEGA 2024; 9:19250-19260. [PMID: 38708282 PMCID: PMC11064166 DOI: 10.1021/acsomega.4c00109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 05/07/2024]
Abstract
The isolated perfused rat liver (IPRL) model provides a mechanistic understanding of the organic-anion-transporting polypeptide (OATP/Oatp)-mediated pharmacokinetics in the preclinical evaluation, which often requires the use of control substrates (i.e., pitavastatin) and monitoring endogenous biomarkers (coproporphyrin I and III). This study aimed to develop and validate an LC-MS method allowing the simultaneous quantification of pitavastatin, coproporphyrin I (CPI), and coproporphyrin III (CPIII) in rat liver perfusion matrices (perfusate, liver homogenate, bile). The analysis was performed on a C18 column at 60 °C with 20 μL of sample injection. The mobile phases consisted of water with 0.1% formic acid and acetonitrile with 0.1% formic acid with a gradient flow of 0.5 mL/min. The assay was validated according to the ICH M10 Bioanalytical Method Validation Guideline (2022) for selectivity, calibration curve and range, matrix effect, carryover, accuracy, precision, and reinjection reproducibility. The method allowing the simultaneous quantification of pitavastatin, CPI, and CPIII was selective without having carryover and matrix effects. The linear calibration curves were obtained within various calibration ranges for three analytes in different matrices. Accuracy and precision values fulfilled the required limits. After 60 min perfusion with pitavastatin (1 μM), the cumulative amounts of pitavastatin in the liver and bile were 5.770 ± 1.504 and 0.852 ± 0.430 nmol/g liver, respectively. CPIII was a more dominant marker than CPI in both liver (0.028 ± 0.017 vs 0.013 ± 0.008 nmol/g liver) and bile (0.016 ± 0.011 vs 0.009 ± 0.007 nmol/g liver). The novel and validated bioanalytical method can be applied in further IPRL preparations investigating Oatp-mediated pharmacokinetics and DDIs.
Collapse
Affiliation(s)
- Nihan Izat
- Department
of Pharmaceutical Technology, Hacettepe
University Faculty of Pharmacy, Ankara 06800, Turkey
| | - Ozan Kaplan
- Department
of Analytical Chemistry, Hacettepe University
Faculty of Pharmacy, Ankara 06100, Turkey
| | - Mustafa Çelebier
- Department
of Analytical Chemistry, Hacettepe University
Faculty of Pharmacy, Ankara 06100, Turkey
| | - Selma Sahin
- Department
of Pharmaceutical Technology, Hacettepe
University Faculty of Pharmacy, Ankara 06800, Turkey
| |
Collapse
|
4
|
Schelz Z, Muddather HF, Zupkó I. Repositioning of HMG-CoA Reductase Inhibitors as Adjuvants in the Modulation of Efflux Pump-Mediated Bacterial and Tumor Resistance. Antibiotics (Basel) 2023; 12:1468. [PMID: 37760764 PMCID: PMC10525194 DOI: 10.3390/antibiotics12091468] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Efflux pump (EP)-mediated multidrug resistance (MDR) seems ubiquitous in bacterial infections and neoplastic diseases. The diversity and lack of specificity of these efflux mechanisms raise a great obstacle in developing drugs that modulate efflux pumps. Since developing novel chemotherapeutic drugs requires large investments, drug repurposing offers a new approach that can provide alternatives as adjuvants in treating resistant microbial infections and progressive cancerous diseases. Hydroxy-methyl-glutaryl coenzyme-A (HMG-CoA) reductase inhibitors, also known as statins, are promising agents in this respect. Originally, statins were used in the therapy of dyslipidemia and for the prevention of cardiovascular diseases; however, extensive research has recently been performed to elucidate the functions of statins in bacterial infections and cancers. The mevalonate pathway is essential in the posttranslational modification of proteins related to vital eukaryotic cell functions. In this article, a comparative review is given about the possible role of HMG-CoA reductase inhibitors in managing diseases of bacterial and neoplastic origin. Molecular research and clinical studies have proven the justification of statins in this field. Further well-designed clinical trials are urged to clarify the significance of the contribution of statins to the lower risk of disease progression in bacterial infections and cancerous diseases.
Collapse
Affiliation(s)
| | | | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary; (Z.S.); (H.F.M.)
| |
Collapse
|
5
|
Dautović E, Rustemović-Čorbić M, Srabović N, Softić A, Smajlović A, Husejnović MŠ, Hatkić A, Halilčević D. Some pleiotropic effects of statins on hepatocellular carcinoma cells: Comparative study on atorvastatin, rosuvastatin and simvastatin. Adv Med Sci 2023; 68:258-264. [PMID: 37478516 DOI: 10.1016/j.advms.2023.07.002] [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: 02/19/2023] [Revised: 04/27/2023] [Accepted: 07/17/2023] [Indexed: 07/23/2023]
Abstract
PURPOSE For many years, statins have been the most commonly used drugs in cholesterol-lowering therapy. In addition to these therapeutic effects, statins exhibit other, pleiotropic effects that can be beneficial, but also harmful to cells and tissues. The aim of this research was to determine and compare the pleiotropic effects of structurally different statins: atorvastatin, simvastatin and rosuvastatin at different concentrations on hepatocellular carcinoma (HepG2) cells. MATERIALS AND METHODS The MTT assay was used to determine the cytotoxic effects of statins. The influence of statins on the production of reactive oxygen species (ROS) was determined by measuring fluorescent response of 2,7-dichlorofluorescein diacetate (DCFH-DA). The effect of statins on glucose production and excretion was determined with glucose production assay. RESULTS The obtained results confirmed that all tested statins exhibit cytotoxic effects, increase the production of ROS as well as the production and excretion of glucose from HepG2 cells. It was observed that all the mentioned effects are more pronounced with lipophilic statins, atorvastatin and simvastatin compared to hydrophilic rosuvastatin. CONCLUSION The less pronounced pleiotropic effects of rosuvastatin on HepG2 cells are probably due to differences in structure and solubility compared to atorvastatin and simvastatin. Transporter-dependent and a slower influx of rosuvastatin into cells compared to the tested lipophilic statins probably lead to a weaker accumulation of rosuvastatin in HepG2 cells, which results in less pronounced pleiotropic effects compared to lipophilic atorvastatin and simvastatin.
Collapse
Affiliation(s)
- Esmeralda Dautović
- Department of Biochemistry, Faculty of Pharmacy, University of Tuzla, Tuzla, Bosnia and Herzegovina.
| | | | - Nahida Srabović
- Department of Biochemistry, Faculty of Pharmacy, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Adaleta Softić
- Department of Biochemistry, Faculty of Pharmacy, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Aida Smajlović
- Department of Biochemistry, Faculty of Pharmacy, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Maida Šljivić Husejnović
- Department of Pharmaceutical Analytics, Faculty of Pharmacy, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Alen Hatkić
- Department of Biochemistry, Faculty of Pharmacy, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Dalila Halilčević
- Department of Biochemistry, Faculty of Pharmacy, University of Tuzla, Tuzla, Bosnia and Herzegovina
| |
Collapse
|
6
|
Elis A. Current and future options in cholesterol lowering treatments. Eur J Intern Med 2023; 112:1-5. [PMID: 36813611 DOI: 10.1016/j.ejim.2023.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/31/2023] [Accepted: 02/10/2023] [Indexed: 02/22/2023]
Abstract
The relative risk reduction of cardiovascular events is proportional to the absolute reduction in LDL-C levels, the primary target of therapy, no matter the way of reduction. During the last decades, the therapeutic regimens for reducing the LDL-C levels have been immerged and improved, with favorable effects on the atherosclerotic process and clinical benefits of various cardiovascular outcomes. From a practical view of point, this review is focusing only on the current available lipid lowering agents: statins, ezetimibe, anti PCSK9 monoclonal antibodies, the small interfering RNA (siRNA) agent, Inclisiran, and Bempedoic acid. The recent changes in lipid lowering regimens, including the early combination of lipid lowering agents and "Low LDL-C" levels <30 mg/dL for high/very high cardiovascular risk patients will also be discussed.
Collapse
Affiliation(s)
- Avishay Elis
- Department of Internal Medicine "C", Beilinson Hospital, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
7
|
Prado Y, Aravena D, Llancalahuen FM, Aravena C, Eltit F, Echeverría C, Gatica S, Riedel CA, Simon F. Statins and Hemostasis: Therapeutic Potential Based on Clinical Evidence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:25-47. [PMID: 37093420 DOI: 10.1007/978-3-031-26163-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Hemostasis preserves blood fluidity and prevents its loss after vessel injury. The maintenance of blood fluidity requires a delicate balance between pro-coagulant and fibrinolytic status. Endothelial cells (ECs) in the inner face of blood vessels maintain hemostasis through balancing anti-thrombotic and pro-fibrinolytic activities. Dyslipidemias are linked to hemostatic alterations. Thus, it is necessary a better understanding of the underlying mechanisms linking hemostasis with dyslipidemia. Statins are drugs that decrease cholesterol levels in the blood and are the gold standard for treating hyperlipidemias. Statins can be classified into natural and synthetic molecules, approved for the treatment of hypercholesterolemia. The classical mechanism of action of statins is by competitive inhibition of a key enzyme in the synthesis pathway of cholesterol, the HMG-CoA reductase. Statins are frequently administrated by oral ingestion and its interaction with other drugs and food supplements is associated with altered bioavailability. In this review we deeply discuss the actions of statins beyond the control of dyslipidemias, focusing on the actions in thrombotic modulation, vascular and cardiovascular-related diseases, metabolic diseases including metabolic syndrome, diabetes, hyperlipidemia, and hypertension, and chronic diseases such as cancer, chronic obstructive pulmonary disease, and chronic kidney disease. Furthermore, we were prompted to delved deeper in the molecular mechanisms by means statins regulate coagulation acting on liver, platelets, and endothelium. Clinical evidence show that statins are effective regulators of dyslipidemia with a high impact in hemostasis regulation and its deleterious consequences. However, studies are required to elucidate its underlying molecular mechanism and improving their therapeutical actions.
Collapse
Affiliation(s)
- Yolanda Prado
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Diego Aravena
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe M Llancalahuen
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Cristobal Aravena
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Eltit
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
- Vancouver Prostate Centre, Vancouver, Canada
| | - Cesar Echeverría
- Laboratory of Molecular Biology, Nanomedicine and Genomics, Faculty of Medicine, University of Atacama, Copiapo, Chile
| | - Sebastian Gatica
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Claudia A Riedel
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Simon
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Millennium Nucleus of Ion Channel-Associated Diseases, Santiago, Chile.
| |
Collapse
|
8
|
Tajbakhsh A, Gheibihayat SM, Askari H, Savardashtaki A, Pirro M, Johnston TP, Sahebkar A. Statin-regulated phagocytosis and efferocytosis in physiological and pathological conditions. Pharmacol Ther 2022; 238:108282. [DOI: 10.1016/j.pharmthera.2022.108282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
|
9
|
Zahedipour F, Butler AE, Eid AH, Sahebkar A. Pleiotropic properties of statins via angiogenesis modulation in cardiovascular disease. Drug Discov Today 2022; 27:103325. [PMID: 35872297 DOI: 10.1016/j.drudis.2022.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/30/2022] [Accepted: 07/19/2022] [Indexed: 12/15/2022]
Abstract
Inhibition of hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase by statins is affected by inhibiting the active site of the enzyme in a competitive manner. Statins reduce plasma cholesterol by inhibiting its de novo synthesis. In addition, statins impart 'pleiotropic' activities that do not directly relate to their ability to decrease cholesterol. The proangiogenic and antiangiogenic characteristics of statins are among these pleiotropic effects. These angiogenic-modifying properties could offer new therapeutic applications. Statins stimulate or suppress angiogenesis in a biphasic manner. Whereas low doses of statin stimulate angiogenesis, high doses reduce protein prenylation and limit cell development and angiogenesis. In this review, we discuss how statins impact angiogenesis, with a particular focus on angiogenesis in stroke and cardiovascular disease (CVD).
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, Adliya, Bahrain
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, WA, Australia; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Zeng W, Hu M, Lee HK, Wat E, Lau CBS, Ho CS, Wong CK, Tomlinson B. Effect of Green Tea Extract and Soy Isoflavones on the Pharmacokinetics of Rosuvastatin in Healthy Volunteers. Front Nutr 2022; 9:850318. [PMID: 35399656 PMCID: PMC8987933 DOI: 10.3389/fnut.2022.850318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Abstract
Background and Aim Green tea and soy products are extensively consumed in daily life. Research has shown that green tea catechins and soy isoflavones may influence the activity of drug metabolizing enzymes and drug transporters. We examined whether regular consumption of green tea extract or soy isoflavones affected the pharmacokinetics of a single dose of rosuvastatin in healthy subjects and whether any interactions were influenced by the polymorphism in the drug transporter ABCG2. Study Design This was an open-label, three-phase randomized crossover study with single doses of rosuvastatin. Methods Healthy Chinese male subjects were given a single dose of rosuvastatin 10 mg on 3 occasions: 1. without herbs; 2. with green tea extract; 3. with soy isoflavone extract. The green tea and soy isoflavone extract were given at a dose containing EGCG 800 mg once daily or soy isoflavones−80 mg once daily for 14 days before statin dosing and at the same time as the statin dosing with at least 4-weeks washout period between phases. Results Twenty healthy male subjects completed the study and the intake of green tea extract significantly reduced the systemic exposure to rosuvastatin by about 20% reducing AUC0−24h from [geometric mean (% coefficient of variation)] 108.7 (28.9) h·μg/L to 74.1 (35.3) h·μg/L and Cmax from 13.1 (32.2) μg/L to 7.9 (38.3) μg/L (P < 0.001 for both), without affecting the elimination half-life. The ABCG2 421C>A polymorphism had a significant effect on rosuvastatin exposure but no impact on the interaction with green tea. Soy isoflavones had no significant effect on rosuvastatin pharmacokinetics. Conclusion This study showed that repeated administration of green tea extract significantly reduced the systemic exposure of rosuvastatin in healthy volunteers. These effects might be predicted to either reduce or increase the lipid-lowering effect of rosuvastatin depending on the mechanism of the effect.
Collapse
Affiliation(s)
- Weiwei Zeng
- Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Miao Hu
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong SAR, China
| | - Hon Kit Lee
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Clinical Pathology, Tuen Mun Hospital, Hong Kong, Hong Kong SAR, China
| | - Elaine Wat
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Clara Bik San Lau
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Chung Shun Ho
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Chun Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Brian Tomlinson
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong SAR, China
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
- *Correspondence: Brian Tomlinson ; orcid.org/0000-0001-6717-5444
| |
Collapse
|
11
|
Loss of function polymorphisms in SLCO1B1 (c.521T>C, rs4149056) and ABCG2 (c.421C>A, rs2231142) genes are associated with adverse events of rosuvastatin: a case-control study. Eur J Clin Pharmacol 2021; 78:227-236. [PMID: 34668025 DOI: 10.1007/s00228-021-03233-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE The study aims to evaluate relationship between polymorphisms associated with a reduced function of two transporter proteins resulting in increased exposure to rosuvastatin - organic anion transporter 1B1 (OATP1B1) (SLCO1B1 c.521T>C) and ATP binding cassette subfamily G member 2 (ABCG2) (ABCG2 c.421C>A) and occurrence of rosuvastatin related myotoxicity/hepatotoxicity. METHODS In a case-control study, cases (rosuvastatin treated patients developing myotoxicity or hepatotoxicity) and controls (concurrent rosuvastatin treated patients free of adverse events) were prospectively recruited over a 2 year period in a single tertiary center specialized in treatment of metabolic disorders. Subjects were evaluated for clinical, comorbidity, and comedication characteristics and for genotype predicted metabolizing phenotypes regarding cytochrome P450 enzymes CYP2C9 and CYP2C19. Standard regression analysis and analysis in matched sets of cases and controls (optimal full matching) were undertaken by fitting frequentist and Bayesian models (covariates/matching variables: age, sex, diabetes, liver/renal disease, hypertension, CYP2C9 and C19 phenotype, use of CYP or transporter inhibitors, non evaluated transporter genotype). RESULTS A total of 88 cases (81 with myotoxicity, 6 with hepatotoxicity, 1 with both) and 129 controls were recruited. Odds of variant SLCO1B1 c.521T>C allele were 2.2-2.5 times higher in cases than in controls (OR = 2.45, 95% CI 1.34-4.48; Bayesian OR = 2.59, 95% CrI 1.42-4.90 in regression analysis; OR = 2.20, 1.10-4.42; Bayesian OR = 2.26, 1.28-4.41 in matched analysis). Odds of variant ABCG2 c.421C>A allele were 2.1-2.3 times higher in cases than in controls (OR = 2.24, 1.04-4.83; Bayesian OR = 2.35, 1.09-4.31 in regression analysis; OR = 2.10, 0.83-5.31; Bayesian OR = 2.17, 1.07-4.35 in matched analysis). CONCLUSION Loss of function polymorphisms in SLCO1B1 c.521T>C and ABCG2 c.421C>A genes are associated with the presence of rosuvastatin related myotoxicity and/or hepatotoxicity.
Collapse
|
12
|
Ahmadi M, Amiri S, Pecic S, Machaj F, Rosik J, Łos MJ, Alizadeh J, Mahdian R, da Silva Rosa SC, Schaafsma D, Shojaei S, Madrakian T, Zeki AA, Ghavami S. Pleiotropic effects of statins: A focus on cancer. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165968. [PMID: 32927022 DOI: 10.1016/j.bbadis.2020.165968] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/21/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023]
Abstract
The statin drugs ('statins') potently inhibit hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase by competitively blocking the active site of the enzyme. Statins decrease de novo cholesterol biosynthesis and thereby reduce plasma cholesterol levels. Statins exhibit "pleiotropic" properties that are independent of their lipid-lowering effects. For example, preclinical evidence suggests that statins inhibit tumor growth and induce apoptosis in specific cancer cell types. Furthermore, statins show chemo-sensitizing effects by impairing Ras family GTPase signaling. However, whether statins have clinically meaningful anti-cancer effects remains an area of active investigation. Both preclinical and clinical studies on the potential mechanisms of action of statins in several cancers have been reviewed in the literature. Considering the contradictory data on their efficacy, we present an up-to-date summary of the pleiotropic effects of statins in cancer therapy and review their impact on different malignancies. We also discuss the synergistic anti-cancer effects of statins when combined with other more conventional anti-cancer drugs to highlight areas of potential therapeutic development.
Collapse
Affiliation(s)
- Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran
| | - Shayan Amiri
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, R4046 - 351 Taché Ave, Winnipeg, Manitoba R2H 2A6, Canada; Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, CA, USA
| | - Filip Machaj
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada; Department of Pathology, Pomeranian Medical University in Szczecin, Poland
| | - Jakub Rosik
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada; Department of Pathology, Pomeranian Medical University in Szczecin, Poland
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Javad Alizadeh
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Reza Mahdian
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Simone C da Silva Rosa
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | | | - Shahla Shojaei
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran
| | - Amir A Zeki
- University of California, Davis School of Medicine. Division of Pulmonary, Critical Care, and Sleep Medicine. U.C. Davis Lung Center, Davis, California, USA; Veterans Affairs Medical Center, Mather, California, USA
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada; Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran; Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
13
|
Hahn HJ, Abagyan R, Podust LM, Roy S, Ali IKM, Debnath A. HMG-CoA Reductase Inhibitors as Drug Leads against Naegleria fowleri. ACS Chem Neurosci 2020; 11:3089-3096. [PMID: 32881478 DOI: 10.1021/acschemneuro.0c00428] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Primary amebic meningoencephalitis (PAM), caused by the free-living ameba Naegleria fowleri, has a fatality rate of over 97%. Treatment of PAM relies on amphotericin B in combination with other drugs, but few patients have survived with the existing drug treatment regimens. Therefore, development of effective drugs is a critical unmet need to avert deaths from PAM. Since ergosterol is one of the major sterols in the membrane of N. fowleri, disruption of isoprenoid and sterol biosynthesis by small-molecule inhibitors may be an effective intervention strategy against N. fowleri. The genome of N. fowleri contains a gene encoding HMG-CoA reductase (HMGR); the catalytic domains of human and N. fowleri HMGR share <60% sequence identity with only two amino acid substitutions in the active site of the enzyme. Considering the similarity of human and N. fowleri HMGR, we tested well-tolerated and widely used HMGR inhibitors, known as cholesterol-lowering statins, against N. fowleri. We identified blood-brain-barrier-permeable pitavastatin as a potent amebicidal agent against the U.S., Australian, and European strains of N. fowleri. Pitavastatin was equipotent to amphotericin B against the European strain of N. fowleri; it killed about 80% of trophozoites within 16 h of drug exposure. Pretreatment of trophozoites with mevalonate, the product of HMGR, rescued N. fowleri from inhibitory effects of statins, demonstrating that HMGR of N. fowleri is the target of statins. Because of the good safety profile and availability for both adult and pediatric uses, consideration should be given to repurposing the fast-acting pitavastatin for the treatment of PAM.
Collapse
Affiliation(s)
- Hye Jee Hahn
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0756, United States
| | - Ruben Abagyan
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0756, United States
| | - Larissa M. Podust
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0756, United States
| | - Shantanu Roy
- Free-Living and Intestinal Amebas (FLIA) Laboratory, Waterborne Disease Prevention Branch, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia 30329-4018, United States
| | - Ibne Karim M. Ali
- Free-Living and Intestinal Amebas (FLIA) Laboratory, Waterborne Disease Prevention Branch, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia 30329-4018, United States
| | - Anjan Debnath
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0756, United States
| |
Collapse
|
14
|
In Vitro Effect of Pitavastatin and Its Synergistic Activity with Isavuconazole against Acanthamoeba castellanii. Pathogens 2020; 9:pathogens9090681. [PMID: 32825652 PMCID: PMC7559540 DOI: 10.3390/pathogens9090681] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023] Open
Abstract
Acanthamoeba keratitis (AK) can occur in healthy individuals wearing contact lenses and it is a painful, blinding infection of the cornea caused by a free-living ameba Acanthamoeba. Current treatment for AK relies on a combination of chlorhexidine, propamidine isethionate, and polyhexamethylene biguanide. However, the current regimen includes an aggressive disinfectant and in 10% of cases recurrent infection ensues. Therefore, development of efficient and safe drugs is a critical unmet need to avert blindness. Acanthamoeba sterol biosynthesis includes two essential enzymes HMG-CoA reductase (HMGR) and sterol 14-demethylase (CYP51), and we earlier identified a CYP51 inhibitor isavuconazole that demonstrated nanomolar potency against A. castellanii trophozoites. In this study, we investigated the effect of well-tolerated HMGR inhibitors and identified pitavastatin that is active against trophozoites of three different clinical strains of A.castellanii. Pitavastatin demonstrated an EC50 of 0.5 to 1.9 µM, depending on strains. Combination of pitavastatin and isavuconazole is synergistic and led to 2- to 9-fold dose reduction for pitavastatin and 11- to 4000-fold dose reduction for isavuconazole to achieve 97% of growth inhibition. Pitavastatin, either alone or in combination with isavuconazole, may lead to repurposing for the treatment of Acanthamoeba keratitis.
Collapse
|
15
|
Hirota T, Fujita Y, Ieiri I. An updated review of pharmacokinetic drug interactions and pharmacogenetics of statins. Expert Opin Drug Metab Toxicol 2020; 16:809-822. [PMID: 32729746 DOI: 10.1080/17425255.2020.1801634] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Hydroxymethylglutaryl-coenzyme A reductase inhibitors (statins) lower cholesterol synthesis in patients with hypercholesterolemia. Increased statin exposure is an important risk factor for skeletal muscle toxicity. Potent inhibitors of cytochrome P450 (CYP) 3A4 significantly increase plasma concentrations of the active forms of simvastatin, lovastatin, and atorvastatin. Fluvastatin is metabolized by CYP2C9, whereas pravastatin, rosuvastatin, and pitavastatin are unaffected by inhibition by either CYP. Statins also have different affinities for membrane transporters involved in processes such as intestinal absorption, hepatic absorption, biliary excretion, and renal excretion. AREAS COVERED In this review, the pharmacokinetic aspects of drug-drug interactions with statins and genetic polymorphisms of CYPs and drug transporters involved in the pharmacokinetics of statins are discussed. EXPERT OPINION Understanding the mechanisms underlying statin interactions can help minimize drug interactions and reduce the adverse side effects caused by statins. Since recent studies have shown the involvement of drug transporters such as OATP and BCRP as well as CYPs in statin pharmacokinetics, further clinical studies focusing on the drug transporters are necessary. The establishment of biomarkers based on novel mechanisms, such as the leakage of microRNAs into the peripheral blood associated with the muscle toxicity, is important for the early detection of statin side effects.
Collapse
Affiliation(s)
- Takeshi Hirota
- Department of Clinical Pharmacokinetics, Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Kyushu University , Fukuoka, Japan
| | - Yuito Fujita
- Department of Clinical Pharmacokinetics, Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Kyushu University , Fukuoka, Japan
| | - Ichiro Ieiri
- Department of Clinical Pharmacokinetics, Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Kyushu University , Fukuoka, Japan
| |
Collapse
|
16
|
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
|
17
|
Tomlinson B, Chan P, Zhang Y, Liu Z, Lam CWK. Pharmacokinetics of current and emerging treatments for hypercholesterolemia. Expert Opin Drug Metab Toxicol 2020; 16:371-385. [PMID: 32223657 DOI: 10.1080/17425255.2020.1749261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Reduction of low-density-lipoprotein cholesterol (LDL-C) and other apolipoprotein B (apoB)-containing lipoproteins reduces cardiovascular (CV) events and greater reductions have greater benefits. Current lipid treatments cannot always achieve desirable LDL-C targets and additional or alternative treatments are often needed.Areas covered: In this article, we review the pharmacokinetics of the available and emerging treatments for hypercholesterolemia and focus on recently approved drugs and those at a late stage of development.Expert opinion: Statin pharmacokinetics are well known and appropriate drugs and doses can usually be chosen for individual patients to achieve LDL-C targets and avoid adverse effects and drug-drug interactions. Ezetimibe, icosapent ethyl and the monoclonal antibodies evolocumab and alirocumab have established efficacy and safety. Newer oral agents including pemafibrate and bempedoic acid have generally favorable pharmacokinetics supporting use in a wide range of patients. RNA-based therapies with antisense oligonucleotides are highly specific for their targets and those inhibiting apoB, apoCIII, angiopoietin-like protein 3 and lipoprotein(a) have shown promising results. The small-interfering RNA inclisiran has the notable advantage that a single subcutaneous administration may be effective for up to 6 months. The CV outcome trial results and long term safety data are eagerly awaited for these new agents.
Collapse
Affiliation(s)
- Brian Tomlinson
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Paul Chan
- Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan.,Research Center for Translational Medicine, Shanghai East Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yuzhen Zhang
- Research Center for Translational Medicine, Shanghai East Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Zhongmin Liu
- Research Center for Translational Medicine, Shanghai East Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | | |
Collapse
|
18
|
Dunkoksung W, Vardhanabhuti N, Siripong P, Jianmongkol S. Rhinacanthin-C Mediated Herb-Drug Interactions with Drug Transporters and Phase I Drug-Metabolizing Enzymes. Drug Metab Dispos 2019; 47:1040-1049. [PMID: 31399508 DOI: 10.1124/dmd.118.085647] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 07/15/2019] [Indexed: 11/22/2022] Open
Abstract
Rhinacanthin-C is a major active constituent in Rhinacanthus nasutus (L.) Kurz, a plant widely used in herbal remedies. Its potential for pharmacokinetic herb-drug interaction may exist with drug transporters and drug metabolizing enzymes. This study assessed the possibility for rhinacanthin-C-mediated drug interaction by determining its inhibitory effects against major human efflux and influx drug transporters as well as various human cytochrome P450(CYP) isoforms. Rhinacanthin-C demonstrated a moderate permeability through the Caco-2 monolayers [Papp (AP-to-BL) = 1.26 × 10-6 cm/s]. It significantly inhibited transport mediated by both P-glycoprotein (P-gp) (IC50 = 5.20 µM) and breast cancer resistance protein (BCRP) (IC50 = 0.83 µM) across Caco-2 and BCRP-overexpressing Madin-Darby canine kidney II cells (MDCKII) cells. This compound also strongly inhibited uptake mediated by organic anion-transporting polypeptide 1B1 (OATP1B1) (IC50 = 0.70 µM) and OATP1B3 (IC50 = 3.95 µM) in OATP1B-overexpressing HEK cells. In addition to its inhibitory effect on these drug transporters, rhinacanthin-C significantly inhibited multiple human CYP isoforms including CYP2C8 (IC50 = 4.56 µM), 2C9 (IC50 = 1.52 µM), 2C19 (IC50 = 28.40 µM), and 3A4/5 (IC50 = 53 µM for midazolam and IC50 = 81.20 µM for testosterone), but not CYP1A2, 2A6, 2B6, 2D6, and 2E1. These results strongly support a high propensity for rhinacanthin-C as a perpetrator of clinical herb-drug interaction via inhibiting various influx and efflux drug transporters (i.e., P-gp, BCRP, OATP1B1, and OATP1B3) and CYP isoforms (i.e., CYP2C8, CYP2C9, and CYP2C19). Thus, the potential for significant pharmacokinetic herb-drug interaction should be addressed when herbal products containing rhinacanthin-C are to be used in conjunction with other prescription drugs.
Collapse
Affiliation(s)
- Wilasinee Dunkoksung
- Departments of Pharmacology and Physiology (W.D., S.J.) and Pharmaceutics and Industrial Pharmacy (N.V.), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand; and National Cancer Institute, Bangkok, Thailand (P.S.)
| | - Nontima Vardhanabhuti
- Departments of Pharmacology and Physiology (W.D., S.J.) and Pharmaceutics and Industrial Pharmacy (N.V.), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand; and National Cancer Institute, Bangkok, Thailand (P.S.)
| | - Pongpun Siripong
- Departments of Pharmacology and Physiology (W.D., S.J.) and Pharmaceutics and Industrial Pharmacy (N.V.), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand; and National Cancer Institute, Bangkok, Thailand (P.S.)
| | - Suree Jianmongkol
- Departments of Pharmacology and Physiology (W.D., S.J.) and Pharmaceutics and Industrial Pharmacy (N.V.), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand; and National Cancer Institute, Bangkok, Thailand (P.S.)
| |
Collapse
|
19
|
Sirtori CR, Yamashita S, Greco MF, Corsini A, Watts GF, Ruscica M. Recent advances in synthetic pharmacotherapies for dyslipidaemias. Eur J Prev Cardiol 2019; 27:1576-1596. [DOI: 10.1177/2047487319845314] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite the demonstrated benefits of statins and injectable biologics, there is a need for new and safe oral agents for addressing classical lipid targets, low-density lipoprotein cholesterol (LDL-C), triglycerides and high-density lipoprotein cholesterol (HDL-C). LDL-C is unquestionably causal in the development of atherogenesis and atherosclerotic cardiovascular disease, but new options are required to address triglyceride-rich lipoproteins and lipoprotein(a). For hypercholesterolaemia, pitavastatin provides a very low dose and potent statin that does not adversely affect glucose metabolism; bempedoic acid acts at a biochemical step preceding hydroxymethylglutaryl-CoA reductase and is not associated with muscular side effects. For hypertriglyceridaemia, pemafibrate displays a unique and selective agonist activity on peroxisomal proliferator activated receptor-α that does not elevate homocysteine or creatinine. Although omega-3 fatty acids supplementation is not effective in secondary prevention, high dose eicosapentaenoic ethyl ester can lead to a remarkable fall in first and recurrent events in high risk patients with hypertriglyceridaemia/low HDL-C. Gemcabene, a dicarboxylic acid regulating apolipoprotein B-100, is effective in reducing both cholesterol and triglycerides. Among cholesteryl ester transfer protein antagonists that elevate HDL-C, only anacetrapib reduces cardiovascular events. Probucol stimulates reverse cholesteryl ester transport, lowers LDL-C stabilizing plaques and may lower incidence of cardiovascular events. These agents, which act through novel mechanisms, afford good and potentially safe treatment choices that may increase adherence and the attainment of therapeutic targets.
Collapse
Affiliation(s)
- Cesare R Sirtori
- Centro Dislipidemie, A.S.S.T. Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Shizuya Yamashita
- Rinku General Medical Centre, Izumisano, Japan
- Department of Community Medicine, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Maria Francesca Greco
- Department of Pharmacological and Bimolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Bimolecular Sciences, Università degli Studi di Milano, Milan, Italy
- Multimedica, IRCCS, Sesto San Giovanni, MI, Italy
| | - Gerald F Watts
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
- Lipid Disorders Clinic, Cardiometabolic Services, Department of Cardiology, Royal Perth Hospital, Australia
| | - Massimiliano Ruscica
- Department of Pharmacological and Bimolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
20
|
Yang J, Hasegawa J, Endo Y, Iitsuka K, Yamamoto M, Matsuda A. Pharmacokinetic Drug Interaction Between Rosuvastatin and Tanjin in Healthy Volunteers and Rats. Yonago Acta Med 2019. [PMID: 30962748 DOI: 10.33160/yam.2019.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Tanjin is an herbal medicine made from the root of salvia miltiorrhiza. It is predominantly given to arteriosclerotic patients as a supplement to ameliorate the clinical symptoms of cardiovascular diseases. In China, tanjin is used frequently in combination treatment for hypercholesterolemia. Thus, there is a high probability of combination of tanjin and statins in these arteriosclerotic patients. This study investigated the interaction between tanjin and rosuvastatin. Methods We performed a randomized single-blind, two-period crossover clinical trial on six healthy male volunteers. Volunteers were administered rosuvastatin with placebo or a tanjin-containing drug randomly. The blood samples were collected before drug administration, and at 0.5, 1, 1.5, 2, 4, 8, and 12 hours after administration. Lymphocytes were isolated from blood samples before and 12 hours after drug administration to measure mRNA. As an animal experiment, an in situ intestinal injection with portal vein sampling model was used to examine the interaction between tanjin and rosuvastatin during the absorption phase. Rosuvastatin or rosuvastatin combined with tanjin solution was injected into the intestine. After injection, blood from the portal vein was collected and the concentration of rosuvastatin was measured by LC/MS/MS analysis. A portion of the intestine and liver from the rats was collected and stored at -80°C for mRNA measurement. Results In the clinical trial, co-administration of tanjin decreased the maximum plasma concentration (Cmax) of rosuvastatin by 26.85% compared with rosuvastatin alone, and also decreased the area under the plasma concentration-time curve of rosuvastatin from 0 to 12 h (AUC0-12) by 19.43%. The relative expression of BCRP and OATP mRNA in human lymphocytes was increased by co-administration of tanjin. In the animal experiment, co-administration of tanjin extract reduced the concentration of rosuvastatin to 84.4, 64.4, and 50.0% at 15, 30, and 45 minutes, respectively. The tanjin-containing drug had a similar effect to tanjin extract. Furthermore, tanjin significantly reduced the absorption of rosuvastatin and the inhibitory effects lasted for at least 24 hours. Tanjin increased the relative expression of BCRP mRNA in the intestine, but it did not change the expression of OATP. Moreover, the concentration of rosuvastatin in the portal vein and systemic blood was reduced. In the liver, tanjin increased both BCRP and OATP mRNA expression, which was consistent with the results from human lymphocytes. Conclusion The clinical trial and animal experiment revealed that tanjin can significantly reduce the absorption of rosuvastatin. This interaction occurred, at least, at the absorption phase in the small intestine due to the enhanced efflux transport. Thus, as tanjin and rosuvastatin were found to interact, their combination needs to be paid attention to.
Collapse
Affiliation(s)
- Jie Yang
- Division of Pharmacology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Junichi Hasegawa
- †National Hospital Organization Yonago Medical Center, Yonago 683-0006, Japan
| | - Yusuke Endo
- ‡Advanced Medicine, Innovation and Clinical Research Center, Tottori University Hospital, Yonago 683-8504, Japan
| | - Kazuhiko Iitsuka
- Division of Pharmacology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Miwa Yamamoto
- §Department of Adult and Elderly Nursing, School of Health of Science, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Akiko Matsuda
- Faculty of Nursing, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
21
|
Chan P, Shao L, Tomlinson B, Zhang Y, Liu ZM. An evaluation of pitavastatin for the treatment of hypercholesterolemia. Expert Opin Pharmacother 2018; 20:103-113. [PMID: 30482061 DOI: 10.1080/14656566.2018.1544243] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Paul Chan
- Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan
| | - Li Shao
- The VIP Department, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Brian Tomlinson
- Research Center for Translational Medicine, Shanghai East Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuzhen Zhang
- Research Center for Translational Medicine, Shanghai East Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Zhong-Min Liu
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
22
|
Werba JP, Misaka S, Giroli MG, Shimomura K, Amato M, Simonelli N, Vigo L, Tremoli E. Update of green tea interactions with cardiovascular drugs and putative mechanisms. J Food Drug Anal 2018; 26:S72-S77. [PMID: 29703388 PMCID: PMC9326886 DOI: 10.1016/j.jfda.2018.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/04/2018] [Accepted: 01/08/2018] [Indexed: 12/26/2022] Open
|
23
|
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
|
24
|
Telmisartan increases systemic exposure to rosuvastatin after single and multiple doses, and in vitro studies show telmisartan inhibits ABCG2-mediated transport of rosuvastatin. Eur J Clin Pharmacol 2016; 72:1471-1478. [DOI: 10.1007/s00228-016-2130-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/07/2016] [Indexed: 12/28/2022]
|
25
|
Abstract
Lipid-lowering medications, particularly statins, have been a popular target for pharmacogenetic studies. A handful of genes have shown promise for predicting response to therapy from the perspective of lipid lowering, as well as myopathy. A number of genes have been implicated and have biological plausibility based on their involvement with the pharmacokinetics or pharmacodynamics of statins or other lipid-lowering medications. The level of confidence and replication of these findings varies, although several associations are likely true. Novel classes of lipid-lowering therapy have opened up new possibilities in the treatment of severe inherited forms of dyslipidemia, making the identification of such mutations an important pharmacogenetic predictor of failure of standard therapy, with potential response to novel therapy. Advances in next-generation sequencing technology bring the application of pharmacogenetics even closer to routine clinical practice.
Collapse
|
26
|
Lin X, Xu R, Fang L, Chen W, Hu D, Zhao S, Wang Y, Li J, Yan X. Factors influencing low-density lipoprotein cholesterol target achievement in primary care - Results from DYSIS China. Int J Cardiol 2016; 222:51-56. [PMID: 27454615 DOI: 10.1016/j.ijcard.2016.07.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 07/04/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND Primary care facilities are the base for hypercholesterolemia treatment. However, data regarding the effectiveness of lipid management in primary care are lacking. METHODS AND RESULTS To evaluate lipid management in the primary care setting in China, we compared patients' characteristics and lipid management outcomes between 6100 outpatients treated at primary care versus 19,217 patients in non-primary settings using data from the DYSlipidemia International Study-China (DYSIS-CHINA). Compared to patients treated at non-primary hospitals, patients who received treatment at primary settings were younger, thinner, and were more likely to be female and to have a family history of premature CVD. Overall, 26.8% of very high-risk and 40.8% of high-risk patients achieved the LDL-C target with primary care treatment, whereas these target rates were 41.2% (p<0.001) and 58.6% (p<0.001), respectively, among patients treated at non-primary hospitals. High-dose statin therapy was underused in primary care patients compared to non-primary hospital patients (p<0.001). Logistic regression analysis showed that female gender, diabetes, and obesity were negative factors, whereas life-style modification and use of high-dose statin (40mg/d simvastatin equivalent) were favorable factors in predicting LDL-C target attainment in the primary care setting. CONCLUSION Sedentary life style, alcohol drinking, and use of suboptimal statin dosage are key factors that unfavorably affect the LDL-C target rate among patients treated at primary care facilities in China. Sufficient training for primary care physicians regarding proper statin use and support for the combined use of a statin with ezetimibe could promote LDL-C target attainment in primary care.
Collapse
Affiliation(s)
- Xue Lin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - RuiYi Xu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ligang Fang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Chen
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Dayi Hu
- Department of Cardiology, Peking University People's Hospital, No.11, Xi Zhi Men Nan Da Jie, Xicheng District, Beijing 100044, China
| | - Shuiping Zhao
- Department of Cardiology, Second Xiangya Hospital, Central South University, No.139, People Street (M.), Changsha 410011, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xi Li, Dongcheng District, Beijing 100050, China
| | - Jihu Li
- MSD China Holding Co., Ltd., No. 1601, Nanjing Rd.(W), JingAn District, Shanghai 20004, China
| | - Xiaowei Yan
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
27
|
Calza L, Colangeli V, Manfredi R, Bon I, Re MC, Viale P. Clinical management of dyslipidaemia associated with combination antiretroviral therapy in HIV-infected patients. J Antimicrob Chemother 2016; 71:1451-65. [PMID: 26846208 DOI: 10.1093/jac/dkv494] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The introduction of potent combination antiretroviral therapy (cART) has had a remarkable impact on the natural history of HIV infection, leading to a dramatic decline in the mortality rate and a considerable increase in the life expectancy of HIV-positive people. However, cART use is frequently associated with several metabolic complications, mostly represented by lipid metabolism alterations, which are reported very frequently among persons treated with antiretroviral agents. In particular, hyperlipidaemia occurs in up to 70%-80% of HIV-positive subjects receiving cART and is mainly associated with specific antiretroviral drugs belonging to three classes of antiretroviral agents: NRTIs, NNRTIs and PIs. The potential long-term consequences of cART-associated dyslipidaemia are not completely understood, but an increased risk of premature coronary heart disease has been reported in HIV-infected patients on cART, so prompt correction of lipid metabolism abnormalities is mandatory in this population. Dietary changes, regular aerobic exercise and switching to a different antiretroviral regimen associated with a more favourable metabolic profile are the first steps in clinical management, but lipid-lowering therapy with fibrates or statins is often required. In this case, the choice of hypolipidaemic drugs should take into account the potential pharmacokinetic interactions with many antiretroviral agents.
Collapse
Affiliation(s)
- Leonardo Calza
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, S. Orsola-Malpighi Hospital, University of Bologna, via G. Massarenti n.11, 40138 Bologna, Italy
| | - Vincenzo Colangeli
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, S. Orsola-Malpighi Hospital, University of Bologna, via G. Massarenti n.11, 40138 Bologna, Italy
| | - Roberto Manfredi
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, S. Orsola-Malpighi Hospital, University of Bologna, via G. Massarenti n.11, 40138 Bologna, Italy
| | - Isabella Bon
- Department of Specialized, Diagnostic and Experimental Medicine, Section of Microbiology, S. Orsola-Malpighi Hospital, University of Bologna, via G. Massarenti n.11, 40138 Bologna, Italy
| | - Maria Carla Re
- Department of Specialized, Diagnostic and Experimental Medicine, Section of Microbiology, S. Orsola-Malpighi Hospital, University of Bologna, via G. Massarenti n.11, 40138 Bologna, Italy
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, S. Orsola-Malpighi Hospital, University of Bologna, via G. Massarenti n.11, 40138 Bologna, Italy
| |
Collapse
|
28
|
Matusewicz L, Meissner J, Toporkiewicz M, Sikorski AF. The effect of statins on cancer cells--review. Tumour Biol 2015; 36:4889-904. [PMID: 26002574 DOI: 10.1007/s13277-015-3551-7] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/08/2015] [Indexed: 01/10/2023] Open
Abstract
Statins [3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoA reductase, abbreviated HMGCR) inhibitors], are well-known cholesterol-depleting agents. Since the early 1990 s, it has been known that statins could be successfully used in cancer therapy, but the exact mechanism(s) of statin activity remains unclear and is now an extensive focus of investigation. So far, it was proven that there are several mechanisms that are activated by statins in cancer cells; some of them are leading to cell death. Statins exert different effects depending on cell line, statin concentration, duration of exposure of cells to statins, and the type of statin being used. It was shown that statins may inhibit the cell cycle by influence on both expression and activity of proteins involved in cell-cycle progression such as cyclins, cyclin-dependent kinases (CDK), and/or inhibitors of CDK. Also, statins may induce apoptosis by both intrinsic and extrinsic pathways. Statin treatment may lead to changes in molecular pathways dependent on the EGF receptor, mainly via inhibition of isoprenoid synthesis. By inhibition of the synthesis of cholesterol, statins may destabilize the cell membrane. Moreover, statins may change the arrangement of transporter OATP1, the localization of HMGCR, and could induce conformational changes in GLUT proteins. In this review, we have tried to gather and compare most of the recent outcomes of the research in this field. We have also attempted to explain why hydrophilic statins are less effective than hydrophobic statins. Finally, we have gathered results from in vivo experiments, presenting the use of statins in combined therapies and discussed a number of molecular targets that could serve as biomarkers predisposing to statin therapy.
Collapse
Affiliation(s)
- Lucyna Matusewicz
- Laboratory of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland
| | | | | | | |
Collapse
|
29
|
Ding YL, Shih YH, Tsai FY, Leong MK. In silico prediction of inhibition of promiscuous breast cancer resistance protein (BCRP/ABCG2). PLoS One 2014; 9:e90689. [PMID: 24614353 PMCID: PMC3948701 DOI: 10.1371/journal.pone.0090689] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 02/03/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Breast cancer resistant protein has an essential role in active transport of endogenous substances and xenobiotics across extracellular and intracellular membranes along with P-glycoprotein. It also plays a major role in multiple drug resistance and permeation of blood-brain barrier. Therefore, it is of great importance to derive theoretical models to predict the inhibition of both transporters in the process of drug discovery and development. Hitherto, very limited BCRP inhibition predictive models have been proposed as compared with its P-gp counterpart. METHODOLOGY/PRINCIPAL FINDINGS An in silico BCRP inhibition model was developed in this study using the pharmacophore ensemble/support vector machine scheme to take into account the promiscuous nature of BCRP. The predictions by the PhE/SVM model were found to be in good agreement with the observed values for those molecules in the training set (n= 22, r2 =0.82, qCV2=0.73, RMSE= 0.40, s = 0.24), test set (n =97, q2=0.75-0.89, RMSE= 0.31, s= 0.21), and outlier set (n= 16, q2 =0.72-0.91, RMSE= 0.29, s=0.17). When subjected to a variety of statistical validations, the developed PhE/SVM model consistently met the most stringent criteria. A mock test by HIV protease inhibitors also asserted its predictivity. CONCLUSIONS/SIGNIFICANCE It was found that this accurate, fast, and robust PhE/SVM model can be employed to predict the BCRP inhibition of structurally diverse molecules that otherwise cannot be carried out by any other methods in a high-throughput fashion to design therapeutic agents with insignificant drug toxicity and unfavorable drug-drug interactions mediated by BCRP to enhance clinical efficacy and/or circumvent drug resistance.
Collapse
Affiliation(s)
- Yi-Lung Ding
- Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien, Taiwan
| | - Yu-Hsuan Shih
- Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien, Taiwan
| | - Fu-Yuan Tsai
- Center for General Education, Chang Gung University, Taoyuan, Taiwan
| | - Max K Leong
- Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien, Taiwan; Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien, Taiwan; Department of Medical Research and Teaching, Mennonite Christian Hospital, Hualien, Taiwan
| |
Collapse
|