Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 331
Filter
1.
Mol Pharm ; 18(5): 1992-2003, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33754729

ABSTRACT

Except for routine scaling and root planing, there are few effective nonsurgical therapeutic interventions for periodontitis and associated alveolar bone loss. Simvastatin (SIM), one of the 3-hydroxy-3-methylglutaryl-cosenzyme A reductase inhibitors, which is known for its capacity as a lipid-lowering medication, has been proven to be an effective anti-inflammatory and bone anabolic agent that has shown promising benefits in mitigating periodontal bone loss. The local delivery of SIM into the periodontal pocket, however, has been challenging due to SIM's poor water solubility and its lack of osteotropicity. To overcome these issues, we report a novel SIM formulation of a thermoresponsive, osteotropic, injectable hydrogel (PF127) based on pyrophosphorolated pluronic F127 (F127-PPi). After mixing F127-PPi with F127 at a 1:1 ratio, the resulting PF127 was used to dissolve free SIM to generate the SIM-loaded formulation. The thermoresponsive hydrogel's rheologic behavior, erosion and SIM release kinetics, osteotropic property, and biocompatibility were evaluated in vitro. The therapeutic efficacy of SIM-loaded PF127 hydrogel on periodontal bone preservation and inflammation resolution was validated in a ligature-induced periodontitis rat model. Given that SIM is already an approved medication for hyperlipidemia, the data presented here support the translational potential of the SIM-loaded PF127 hydrogel for better clinical management of periodontitis and associated pathologies.


Subject(s)
Alveolar Bone Loss/drug therapy , Drug Carriers/chemistry , Periodontitis/drug therapy , Simvastatin/administration & dosage , Alveolar Bone Loss/etiology , Alveolar Bone Loss/pathology , Alveolar Process/diagnostic imaging , Alveolar Process/drug effects , Animals , Drug Liberation , Female , Humans , Hydrogels/chemistry , Injections, Intralesional , Mice , Models, Animal , Periodontitis/complications , Periodontitis/pathology , Poloxamer/chemistry , RAW 264.7 Cells , Rats , Simvastatin/pharmacokinetics , Solubility , X-Ray Microtomography
2.
J Nanobiotechnology ; 19(1): 311, 2021 Oct 09.
Article in English | MEDLINE | ID: mdl-34627266

ABSTRACT

Triple-negative breast cancer (TNBC), a management of aggressive breast cancer, remains an unmet medical challenge. Although a wave of efforts had spurred to design novel therapeutic method of TNBC, unpredictable prognosis with lacking effective therapeutic targets along with the resistance to apoptosis seriously limited survival benefits. Ferroptosis is a non-apoptotic form of cell death that is induced by excessive lipid peroxidation, which provide an innovative way to combat cancer. Emerging evidence suggests that ferroptosis plays an important role in the treatment of TNBC cells. Herein, a novel ferroptosis nanomedicine was prepared by loading simvastatin (SIM), a ferroptosis drug, into zwitterionic polymer coated magnetic nanoparticles (Fe3O4@PCBMA) to improve the therapeutic effect of TNBC. The as-obtained Fe3O4@PCBMA-SIM nanoparticles demonstrated more cytotoxicity against MDA-MB-231 than MCF-7 due to the higher expression of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), which demonstrated that statins could effectively kill TNBC. Further experiments showed that SIM could inhibit the expression of HMGCR to downregulate the mevalonate (MVA) pathway and glutathione peroxidase 4 (GPX4), thereby inducing cancer cell ferroptosis. What's more, PCBMA endows Fe3O4@PCBMA longer blood circulation performance to enhance their accumulation at tumor sites. Given that Fe3O4 have proven for clinical applications by the U.S. Food and Drug Administration (FDA) and SIM could induce cancer cell ferroptosis, the developed Fe3O4@PCBMA-SIM nanosystem would have great potential in clinics for overcoming the drug resistance brought about by apoptotic drugs to cancer cells.


Subject(s)
Ferroptosis/drug effects , Simvastatin , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line, Tumor , Delayed-Action Preparations , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Female , Humans , MCF-7 Cells , Magnetite Nanoparticles/chemistry , Male , Mice, Nude , Signal Transduction/drug effects , Simvastatin/chemistry , Simvastatin/pharmacokinetics , Simvastatin/pharmacology
3.
AAPS PharmSciTech ; 22(5): 189, 2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34159457

ABSTRACT

Phospholipid complexation, despite being a successful, versatile, and burgeoning strategy, stickiness of phospholipids leads to suboptimal dissolution rate of drugs. This work was undertaken to fabricate simvastatin-phospholipid complex (SIM-PLC)-loaded matrix dispersion (SIM-PLC-MD) using Soluplus® as carrier material, to augment dispersibility and dissolution of SIM-PLC without altering complexation between simvastatin (SIM) and phospholipid. SIM-PLC and SIM-PLC-MD were prepared using solvent evaporation and discontinuous solvent evaporation techniques, respectively. The successful complexation was substantiated by FTIR method. Besides, PXRD and SEM studies disclosed the absence of crystallinity of SIM in both SIM-PLC and SIM-PLC-MD. The TEM analysis monitored the self-assembly of SIM-PLC and SIM-PLC-MD into colloidal structures, which could be correlated with redispersion in GIT fluids upon oral administration. The considerable increase in hydrophilicity of SIM-PLC-MD and SIM-PLC as evident from partition coefficient experiment can further be correlated with their remarkably improved solubility profiles in the following pattern: SIM-PLC-MD˃SIM-PLC˃SIM. Correspondingly, improved dispersibility of SIM-PLC-MD in comparison to SIM-PLC can be accountable for accelerated dissolution rate by 2.53-fold and 1.5-fold in pH 1.2 and 6.8 conditions, respectively. The oral pharmacokinetic evaluation in Sprague Dawley (SD) rats revealed 3.19-fold enhancement in oral bioavailability of SIM through SIM-PLC-MD when compared with plain SIM, whereas 1.83-fold increment was observed in the case of SIM-PLC. Finally, the efficacy experimentation in SD rats revealed that SIM-PLC-MD significantly reduced triglycerides and cholesterol levels in comparison to SIM and SIM-PLC. These outcomes suggest that a matrix dispersion strategy improves oral bioavailability and hypolipidemic activity of SIM.


Subject(s)
Phospholipids/chemistry , Phospholipids/pharmacokinetics , Simvastatin/chemistry , Simvastatin/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Female , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyvinyls/administration & dosage , Polyvinyls/chemistry , Polyvinyls/pharmacokinetics , Rats , Rats, Sprague-Dawley , Simvastatin/administration & dosage , Solubility , Solvents/administration & dosage , Solvents/chemistry , Solvents/pharmacokinetics
4.
Pharm Biol ; 59(1): 880-883, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34214011

ABSTRACT

CONTEXT: Baicalein and simvastatin possess similar pharmacological activities and indications. The risk of their co-administration was unclear. OBJECTIVE: The interaction between baicalein and simvastatin was investigated to provide reference and guidance for the clinical application of the combination of these two drugs. MATERIALS AND METHODS: The pharmacokinetics of simvastatin was investigated in Sprague-Dawley rats (n = 6). The rats were pre-treated with 20 mg/kg baicalein for 10 days and then administrated with 40 mg/kg simvastatin. The single administration of simvastatin was set as the control group. The rat liver microsomes were employed to assess the metabolic stability and the effect of baicalein on the activity of CYP3A4. RESULTS: Baicalein significantly increased the AUC(0-t) (2018.58 ± 483.11 vs. 653.05 ± 160.10 µg/L × h) and Cmax (173.69 ± 35.49 vs. 85.63 ± 13.28 µg/L) of simvastatin. The t1/2 of simvastatin was prolonged by baicalein in vivo and in vitro. The metabolic stability of simvastatin was also improved by the co-administration of baicalein. Baicalein showed an inhibitory effect on the activity of CYP3A4 with the IC50 value of 12.03 µM, which is responsible for the metabolism of simvastatin. DISCUSSION AND CONCLUSION: The co-administration of baicalein and simvastatin may induce drug-drug interaction through inhibiting CYP3A4. The dose of baicalein and simvastatin should be adjusted when they are co-administrated.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A/drug effects , Flavanones/pharmacology , Simvastatin/pharmacokinetics , Animals , Area Under Curve , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Drug Interactions , Flavanones/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Inhibitory Concentration 50 , Male , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley
5.
J Cell Mol Med ; 24(18): 11024-11029, 2020 09.
Article in English | MEDLINE | ID: mdl-32767644

ABSTRACT

The chronopharmacology refers to the utilization of physiological circadian rhythms to optimize the administration time of drugs, thus increasing their efficacy and safety, or reducing adverse effects. Simvastatin is one of the most widely prescribed drugs for the treatment of hypercholesterolaemia, hyperlipidemia and coronary artery disease. There are conflicting statements regarding the timing of simvastatin administration, and convincing experimental evidence remains unavailable. Thus, we aimed to examine whether different administration times would influence the efficacy of simvastatin. High-fat diet-fed mice were treated with simvastatin at zeitgeber time 1 (ZT1) or ZT13, respectively, for nine weeks. Simvastatin showed robust anti-hypercholesterolaemia and anti-hyperlipidemia effects on these obese mice, regardless of administration time. However, simvastatin administrated at ZT13, compared to ZT1, was more functional for decreasing serum levels of total cholesterol, triglycerides, non-esterified free fatty acids and LDL cholesterol, as well as improving liver pathological characteristics. In terms of possible mechanisms, we found that simvastatin did not alter the expression of hepatic circadian clock gene in vivo, although it failed to change the period, phase and amplitude of oscillation patterns in Per2::Luc U2OS and Bmal1::Luc U2OS cells in vitro. In contrast, simvastatin regulated the expression of Hmgcr, Mdr1 and Slco2b1 in a circadian manner, which potentially contributed to the chronopharmacological function of the drug. Taken together, we provide solid evidence to suggest that different administration times affect the lipid-lowering effects of simvastatin.


Subject(s)
Circadian Rhythm Signaling Peptides and Proteins/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hyperlipidemias/drug therapy , Simvastatin/pharmacokinetics , Animals , Chronopharmacokinetics , Circadian Clocks/drug effects , Circadian Rhythm Signaling Peptides and Proteins/biosynthesis , Circadian Rhythm Signaling Peptides and Proteins/genetics , Diet, High-Fat/adverse effects , Drug Chronotherapy , Gene Expression Regulation/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Obese , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Simvastatin/administration & dosage , Simvastatin/therapeutic use
6.
Ann Pharmacother ; 54(2): 171-177, 2020 02.
Article in English | MEDLINE | ID: mdl-31441337

ABSTRACT

Objective: To review the available literature that provides evidence for the absence of statin interactions with tacrolimus compared with cyclosporine. Data Sources: A literature search of PubMed was performed (1990 to June 2019) using the following search terms: calcineurin inhibitors, tacrolimus, cyclosporine, statins, atorvastatin, simvastatin, and drug interactions. Clinical practice guidelines, article bibliographies, drug interaction database references, and product monographs were also reviewed. Study Selection and Data Extraction: Relevant English-language studies describing the mechanism of interaction, the magnitude of pharmacokinetic alterations, and safety were evaluated. In vitro data and studies conducted in adult humans were considered. Data Synthesis: Studies demonstrate pharmacokinetic differences between cyclosporine and tacrolimus, particularly with regard to inhibition of 2 hepatic transporters: P-glycoprotein and organic anion transporting polypeptide (OATP). Compared with cyclosporine, tacrolimus does not affect these transporters, does not enhance statin exposure, and does not increase statin-associated safety events. Relevance to Patient Care and Clinical Practice: Clinical practice guidelines allude to the need to reduce statin doses in the setting of tacrolimus. Some providers have adopted this practice, and doing so may prevent transplant recipients from attaining cardiovascular benefit, especially when increased or high-intensity doses are required. The pharmacokinetic differences between tacrolimus and cyclosporine highlight different interaction potential with statins. Conclusions: Clinicians need to be aware that tacrolimus and cyclosporine are not the same with regard to causing drug interactions with statins. Tacrolimus can be used with statins without the need for dose adjustments because of lack of an interaction.


Subject(s)
Cyclosporine/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Immunosuppressive Agents/therapeutic use , Organ Transplantation , Tacrolimus/therapeutic use , Adult , Atorvastatin/administration & dosage , Atorvastatin/adverse effects , Atorvastatin/pharmacokinetics , Atorvastatin/therapeutic use , Cyclosporine/administration & dosage , Cyclosporine/adverse effects , Cyclosporine/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacokinetics , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Simvastatin/administration & dosage , Simvastatin/adverse effects , Simvastatin/pharmacokinetics , Simvastatin/therapeutic use , Tacrolimus/administration & dosage , Tacrolimus/adverse effects , Tacrolimus/pharmacokinetics
7.
J Nanobiotechnology ; 18(1): 179, 2020 Dec 07.
Article in English | MEDLINE | ID: mdl-33287831

ABSTRACT

Inflammation and oxidative stress are two major factors that are involved in the pathogenesis of atherosclerosis. A smart drug delivery system that responds to the oxidative microenvironment of atherosclerotic plaques was constructed in the present study. Simvastatin (SIM)-loaded biodegradable polymeric micelles were constructed from hyaluronic acid (HA)-coated poly(ethylene glycol)-poly(tyrosine-ethyl oxalyl) (PEG-Ptyr-EO) for the purpose of simultaneously inhibiting macrophages and decreasing the level of reactive oxygen species (ROS) to treat atherosclerosis. HA coating endows the micelle system the ability of targeting CD44-positive inflammatory macrophages. Owing to the ROS-responsive nature of PEG-Ptyr-EO, the micelles can not only be degraded by enzymes, but also consumes ROS by itself at the pathologic sites, upon which the accumulation of pro-inflammatory macrophages is effectively suppressed and oxidative stress is alleviated. Consequently, the cellular uptake experiment demonstrated that SIM-loaded HA-coated micelles can be effectively internalized by LPS-induced RAW264.7 cells and showed high cytotoxicity against the cells, but low cytotoxicity against LO2 cells. In mouse models of atherosclerosis, intravenously SIM-loaded HA-coated micelles can effectively reduce plaque content of cholesterol, resulting in remarkable therapeutic effects. In conclusion, the SIM-loaded micelle system provides a promising and innovative option against atherosclerosis.


Subject(s)
Antioxidants , Atherosclerosis/metabolism , Hyaluronic Acid/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Oxidative Stress/drug effects , Animals , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Disease Models, Animal , Hydrogen Peroxide/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Micelles , Polyethylene Glycols/chemistry , RAW 264.7 Cells , Simvastatin/chemistry , Simvastatin/pharmacokinetics , Simvastatin/pharmacology
8.
Xenobiotica ; 50(9): 1115-1120, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32150479

ABSTRACT

Apatinib, a small molecule anti-angiogenic tyrosine kinase inhibitor is used extensively to treat advanced gastric cancer and simvastatin (SV) is often co-prescribed to treat cardiovascular disease in cancer patients. As both apatinib and SV are metabolized primarily by cytochrome P450 variant CYP3A4, they are likely to interact. Therefore, the potential effect of SV co-administration on pharmacokinetics of apatinib in Sprague-Dawley male rats is demonstrated for the first time.Sixteen rats were randomly divided into two groups (n = 8), 2 mg/kg SV orally co-administrated for seven days (group B) and the corresponding control group (group A). Apatinib concentrations of rat plasma samples were detected by ultra-performance liquid chromatography tandem mass spectrometry. Pharmacokinetic parameters were calculated using non compartmental methods.Co-administration of SV for seven days significantly increased area under curve (AUC(0-t)), AUC(0-∞) and maximum plasma concentration of apatinib by 2.4-, 2.4-, and 2.7-fold, respectively while decreasing apparent volume of distribution and clearance by 81.7 and 73.9%, respectively.These findings suggest that concomitant administration of SV with 7 days may have inhibited the metabolism of apatinib in rats.


Subject(s)
Pyridines/pharmacokinetics , Simvastatin/pharmacokinetics , Animals , Area Under Curve , Chromatography, Liquid , Cytochrome P-450 CYP3A , Protein Kinase Inhibitors , Pyridines/administration & dosage , Rats , Rats, Sprague-Dawley , Simvastatin/administration & dosage
9.
AAPS PharmSciTech ; 21(6): 223, 2020 Aug 04.
Article in English | MEDLINE | ID: mdl-32749629

ABSTRACT

This study aimed to enhance the dissolution of simvastatin (SMV) through its formulation in liquisolid tablets (LSTs) to improve its bioavailability and hypolipidemic activity after oral administration. SMV-LSTs were optimized using Box-Behnken design to maximize the rate and extent of SMV dissolution. The optimized SMV-LST was evaluated for pharmacokinetic parameters and potential hypolipidemic activity on induced hyperlipidemic rats. The dissolution parameters revealed a shortening of mean dissolution time from 10.99 to 6.82 min, increasing of dissolution rate during the first 10 min from 1253.15 to 1667.31 µg/min, and enhancing of dissolution efficiency after 60 min from 71.92 to 86.93% for SMV-LSTs versus the commercial SMV tablets. The obtained data reflected an improvement in the relative bioavailability of SMV with 148.232% which was confirmed by the significant reduction of the levels of circulating total cholesterol, triglycerides that reached the normal level after 12 h. In particular, the optimized SMV-LSTs reduced serum low-density lipoproteins (LDL) by 44.6% which was significantly different from the commercial SMV tablets. In contrast, the level of serum high-density lipoprotein (HDL) was significantly augmented after 4 h in rats treated with the optimized SMV-LSTs by 47.6%. Finally, the optimized SMV-LSTs showed a significant lower atherosclerotic index value which could maximize its potential in decreasing the risk of coronary disease and atherosclerosis. Overall enhancement in pharmacokinetics and pharmacodynamics in comparison with the commercial tablets confers the potential of the liquisolid approach as a promising alternative for improved oral bioavailability, hypolipidemic, and cardioprotective effects of SMV. Graphical abstract.


Subject(s)
Hypolipidemic Agents/pharmacology , Simvastatin/pharmacology , Animals , Biological Availability , Male , Poloxamer/toxicity , Rats , Rats, Wistar , Simvastatin/chemistry , Simvastatin/pharmacokinetics , Solubility , Tablets
10.
Pak J Pharm Sci ; 33(6): 2489-2495, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33867321

ABSTRACT

Because of the low solubility, the oral bioavailability of simvastatin (SV) was poor, which restricted the application in clinic. In order to increase the dissolution and the oral absorption of simvastatin, we prepared a novel solid nanomatrix of SV with pharmaceutical acceptable nano-sized silica and Eudragit®. The nanomatrix was prepared using solvent evaporate method and the formulation was optimized. The X-ray diffraction (XRD) and differential scanning calorimetry (DSC) were used to analyze the physicochemical characterization of the SV nanomatrix. The results indicated that the SV existed in the nanomatrix was in a state of molecule or amorphous form. The optimal formulation, consisted of SV, Eudragit® L100-55 and Sylysia 350 (1:5:5, w/w/w), significantly enhanced the dissolution of SV compared with Zocor. And the relative bioavailability was 272% to Zocor. The oral absorption of simvastatin was enhanced markedly. The SV nanomatrix after storage for 1 year displayed similar performance in vitro and in vivo with the freshly prepared nanomatrix. The stability of SV nanomatrix achieved the desired objectives. In conclusion, the nanomatrix system described here had superior performance in vitro and in vivo and was expected to have a promising future as an alternative oral drug delivery system for SV.


Subject(s)
Drug Delivery Systems/methods , Nanostructures/chemistry , Simvastatin/administration & dosage , Simvastatin/pharmacokinetics , Acrylic Resins/chemistry , Administration, Oral , Animals , Biological Availability , Calorimetry, Differential Scanning , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Storage , Male , Nanostructures/administration & dosage , Rats, Sprague-Dawley , Silicon Dioxide/chemistry , Simvastatin/chemistry , X-Ray Diffraction
11.
Pulm Pharmacol Ther ; 57: 101809, 2019 08.
Article in English | MEDLINE | ID: mdl-31195091

ABSTRACT

Fevipiprant, a prostaglandin D2 receptor 2 antagonist, is in clinical development as a treatment for asthma. The goal of this study was to assess the potential of fevipiprant to cause drug-drug interactions (DDI) as a perpetrator, that is, by altering the pharmacokinetics (PK) of co-medications. In vitro drug interaction studies of clinically relevant drug metabolizing enzymes and transporters were conducted for fevipiprant and its acyl glucuronide (AG) metabolite. Comparison of Ki values with unbound systemic or portal vein steady-state plasma exposure of fevipiprant and its AG metabolite revealed the potential for inhibition of organic anion transporting polypeptide 1B1 (OATP1B1) transporters (R-value of 5.99), while other targets including cytochrome P450 enzymes were not, or only marginally, inhibited. Consequently, an open-label, two-part, two-period, single-sequence clinical study assessed the effect of fevipiprant 450 mg QD on the pharmacokinetics of simvastatin 20 mg and rosuvastatin 20 mg, two statins with different dependency in OATP1B1-mediated hepatic uptake, in healthy adult volunteers. The study also assessed the pharmacogenetics of the SLCO1B1 gene, which encodes OATP1B1. Clinically, fevipiprant 450 mg QD showed a low potential for interaction and increased the peak concentrations of simvastatin acid and rosuvastatin by 2.23- and 1.87-fold, respectively, with little or no impact on total exposure. Genotype analysis confirmed that SLCO1B1 genotype influences statin pharmacokinetics to a similar extent either with or without fevipiprant co-administration. In summary, fevipiprant at 450 mg QD has only minor liabilities as a perpetrator for DDI.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Indoleacetic Acids/pharmacology , Liver-Specific Organic Anion Transporter 1/genetics , Pyridines/pharmacology , Rosuvastatin Calcium/pharmacokinetics , Simvastatin/pharmacokinetics , Adult , Drug Interactions , Female , Genotype , Healthy Volunteers , Humans , Male , Middle Aged , Organic Anion Transporters , Pharmacogenetics , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors
12.
Eur J Clin Pharmacol ; 75(9): 1227-1235, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31172248

ABSTRACT

PURPOSE: Poor adherence to dietary/behaviour modifications as interventions for hypercholesterolemia in paediatric patients often necessitates the initiation of statin therapy. The aim of this study was to develop a joint population pharmacokinetic model for simvastatin and four metabolites in children and adolescents to investigate sources of variability in simvastatin acid exposure in this patient population, in addition to SLCO1B1 genotype status. METHODS: Plasma concentrations of simvastatin and its four metabolites, demographic and polymorphism data for OATP1B1 and CYP3A5 were analysed utilising a population pharmacokinetic modelling approach from an existing single oral dose (10 mg < 17 years and 20 mg ≥ 18 years) pharmacokinetic dataset of 32 children and adolescents. RESULTS: The population PK model included a one compartment disposition model for simvastatin with irregular oral absorption described by two parallel absorption processes each consisting of sequential zero and first-order processes. The data for each metabolite were described by a one-compartment disposition model with the formation and elimination apparent parameters estimated. The model confirmed the statistically significant effect of c.521T>C (rs4149056) on the pharmacokinetics of the active metabolite simvastatin acid in children/adolescents, consistent with adult data. In addition, age was identified as a covariate affecting elimination clearances of 6-hydroxymethyl simvastatin acid and 3, 5 dihydrodiol simvastatin metabolites. CONCLUSION: The model developed describes the pharmacokinetics of simvastatin and its metabolites in children/adolescents capturing the effects of both c.521T>C and age on variability in exposure in this patient population. This joint simvastatin metabolite model is envisaged to facilitate optimisation of simvastatin dosing in children/adolescents.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Models, Biological , Simvastatin/pharmacokinetics , Adolescent , Adult , Child , Cytochrome P-450 CYP3A/genetics , Female , Genotype , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Liver-Specific Organic Anion Transporter 1/genetics , Male , Simvastatin/blood , Young Adult
13.
J Mater Sci Mater Med ; 30(4): 47, 2019 Apr 12.
Article in English | MEDLINE | ID: mdl-30980130

ABSTRACT

The objective of this study was to fabricate and characterize chitosan combined with different amounts of simvastatin-loaded nanoparticles and to investigate their potential for guided bone regeneration in vitro and in vivo. Different SIM-CSN formulations were combined into a chitosan scaffold (SIM-CSNs-S), and the morphology, simvastatin release profile, and effect on cell proliferation and differentiation were investigated. For in vivo experiments, ectopic osteogenesis and the critical-size cranial defect model in SD rats were chosen to evaluate bone regeneration potential. All three SIM-CSNs-S formulations had a porous structure and exhibited sustained simvastatin release. CSNs-S showed excellent degradation and biocompatibility characteristics. The 4 mg SIM-CSNs-S formulation stimulated higher BMSC ALP activity levels, demonstrated significantly earlier collagen enhancement, and led to faster bone regeneration than the other formulations. SIM-CSNs-S should have a significant effect on bone regeneration.


Subject(s)
Bone Regeneration/drug effects , Chitosan/chemistry , Guided Tissue Regeneration/methods , Nanoparticles/chemistry , Nanoparticles/metabolism , Simvastatin/pharmacokinetics , Tissue Scaffolds/chemistry , Animals , Bone and Bones/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/metabolism , Delayed-Action Preparations , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Compounding , Male , Materials Testing , Microspheres , Osteogenesis/drug effects , Osteogenesis/physiology , Rats , Rats, Sprague-Dawley , Simvastatin/administration & dosage , Surface Properties , Tissue Engineering/methods
14.
Drug Dev Ind Pharm ; 45(11): 1725-1739, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31432703

ABSTRACT

Objective and methods: This study predicted the nature of chitosan interactions and effects of this interaction on drug release mechanism in simvastatin-loaded chitosan nanoformulation using molecular docking, spectroscopic and thermal analysis. Significance: This work explains in depth the molecular mechanism of simvastatin and chitosan bond formation in nanoformulation. Results: The effective encapsulation and sustain release properties of chitosan were indicated by increase in melting endotherm of simvastatin. Intermolecular hydrogen bond between third hydroxyl group pyranone ring of simvastatin and amino group of chitosan represented the stability of active lactone moiety that was not cleaved during formulation which is prerequisite for biological activity. UV-vis spectroscopic characterization, shift in infrared vibration wavenumber of simvastatin and chitosan, ligand titration, 1HNMR and 13C-NMR analyses confirmed this interaction pattern. The pharmacokinetic evaluation in mouse model revealed the sustain release property of nanoformulation. Conclusion: Thus formation of intermolecular hydrogen bond in nanoformulation contributed to modified physicochemical properties and improved in vivo performance of simvastatin.


Subject(s)
Chitosan/chemistry , Drug Carriers/chemistry , Drug Liberation , Hyperlipidemias/drug therapy , Simvastatin/pharmacokinetics , Administration, Oral , Animals , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/adverse effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Disease Models, Animal , Drug Administration Schedule , Drug Compounding , Female , Humans , Hyperlipidemias/blood , Hyperlipidemias/etiology , Mice , Molecular Docking Simulation , Nanoparticles/chemistry , Particle Size , Proton Magnetic Resonance Spectroscopy , Simvastatin/administration & dosage , Spectroscopy, Fourier Transform Infrared
15.
Drug Dev Ind Pharm ; 45(5): 745-753, 2019 May.
Article in English | MEDLINE | ID: mdl-30632800

ABSTRACT

Despite the ongoing extensive research, cancer therapeutics still remains an area with unmet needs which is hampered by shortfall in the development of newer medicines. The present study discusses a nano-based combinational approach for treating solid tumor. Dual-loaded nanoparticles encapsulating gemcitabine HCl (GM) and simvastatin (SV) were fabricated by double emulsion solvent evaporation method and optimized. Optimized nanoparticles showed a particle size of 258 ± 2.4 nm, polydispersity index of 0.32 ± 0.052, and zeta potential of -12.5 mV. The size and the morphology of the particles wee further confirmed by transmission electron microscopy (TEM) and scanning electron microscopy, respectively of the particles. The entrapment efficiency of GM and SV in the nanoparticles was 38.5 ± 4.5% and 72.2 ± 5.6%, respectively. The in vitro release profile was studied for 60 h and showed Higuchi release pattern. The cell toxicity was done using MTT assay and lower IC50 was obtained with the nanoparticles as compared to the pure drug. The bioavailability of GM and SV in PLGA nanoparticles was enhanced by 1.4-fold and 1.3-fold respectively, compared to drug solution. The results revealed that co-delivery of GM and SV could be used for its oral delivery for the effective treatment of pancreatic cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Carriers/chemistry , Pancreatic Neoplasms/drug therapy , Simvastatin/administration & dosage , Administration, Oral , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Biological Availability , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Drug Compounding/methods , Drug Liberation , Drug Screening Assays, Antitumor , Emulsions , Humans , Inhibitory Concentration 50 , Nanoparticles/chemistry , Pancreatic Neoplasms/pathology , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rats , Rats, Wistar , Simvastatin/pharmacokinetics , Gemcitabine
16.
Molecules ; 24(9)2019 Apr 28.
Article in English | MEDLINE | ID: mdl-31035343

ABSTRACT

Herein, the effect of silymarin pretreatment on the pharmacokinetics of simvastatin in rats was evaluated. To ensure the accuracy of the results, a rapid and sensitive UPLC-MS/MS method was established for simultaneous quantification of simvastatin (SV) and its active metabolite simvastatin acid (SVA). This method was applied for studying the pharmacokinetic interactions in rats after oral co-administration of silymarin (45 mg/kg) and different concentrations of SV. The major pharmacokinetic parameters, including Cmax, tmax, t1/2, mean residence time (MRT), elimination rate constant (λz) and area under the concentration-time curve (AUC0-12h), were calculated using the non-compartmental model. The results showed that the co-administration of silymarin and SV significantly increased the Cmax and AUC0-12h of SVA compared with SV alone, while there was no significant difference with regards to Tmax and t1/2. However, SV pharmacokinetic parameters were not significantly affected by silymarin pretreatment. Therefore, these changes indicated that drug-drug interactions may occur after co-administration of silymarin and SV.


Subject(s)
Drug Interactions , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Metabolomics , Silymarin/pharmacology , Simvastatin/pharmacokinetics , Animals , Metabolomics/methods , Molecular Structure , Rats
17.
Pharm Dev Technol ; 24(6): 663-670, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30516412

ABSTRACT

PURPOSE: In this study, methoxy poly (ethylene glycol)-poly (ε-caprolactone) (mPEG-PCL) di-block copolymers were synthesized. The purpose of this work is to investigate the in vivo anti-inflammatory effects of simvastatin-loaded micelles. METHODS: The structure of synthesized copolymers was characterized by using HNMR, FTIR, and GPC techniques. Simvastatin was encapsulated in micelles through a single-step nano-precipitation method, leading to the formation of simvastatin-loaded mPEG-PCL (simvastatin-mPEG-PCL) micelles. In this study, the anti-inflammatory effects of simvastatin/mPEG-PCL micelles versus indomethacin were investigated in acute inflammation-induced rats. The paw edema thickness was measured 1, 2, 3, and 4 h after injection of formulation. The inhibition of edema in various groups were calculated and reported by percentages. RESULTS: The results showed that the zeta potential of micelles was about -14.9 ± 0.47 mV and the average size was in range of 66.10 ± 0.34 nm. Simvastatin was encapsulated in mPEG-PCL micelles with a loading capacity of 9.63 ± 0.87% and an encapsulation efficiency of 64.20 ± 0.79%. Simvastatin and simvastatin-mPEG-PCL micelles showed significant anti-inflammatory activity in the present study. CONCLUSIONS: This study revealed that simvastatin and simvastatin/mPEG-PCL micelles both have anti-inflammatory effects and suggested that statins have potential anti-inflammatory activity along with their lipid lowering properties.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Anticholesteremic Agents/administration & dosage , Drug Carriers/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Simvastatin/administration & dosage , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Anticholesteremic Agents/pharmacokinetics , Anticholesteremic Agents/therapeutic use , Drug Delivery Systems , Edema/drug therapy , Male , Micelles , Rats, Wistar , Simvastatin/pharmacokinetics , Simvastatin/therapeutic use
18.
Br J Clin Pharmacol ; 84(12): 2877-2888, 2018 12.
Article in English | MEDLINE | ID: mdl-30171779

ABSTRACT

AIM: We report on two Phase 1, open-label, single-arm studies assessing the effect of osimertinib on simvastatin (CYP3A substrate) and rosuvastatin (breast cancer resistance protein substrate [BCRP] substrate) exposure in patients with advanced epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer who have progressed after treatment with an EGFR tyrosine kinase inhibitor, to determine, upon coadministration, whether osimertinib could affect the exposure of these agents. METHODS: Fifty-two patients in the CYP3A study (pharmacokinetic [PK] analysis, n = 49), and 44 patients in the BCRP study were dosed (PK analysis, n = 44). In the CYP3A study, patients received single doses of simvastatin 40 mg on Days 1 and 31, and osimertinib 80 mg once daily on Days 3-32. In the BCRP study, single doses of rosuvastatin 20 mg were given on Days 1 and 32, and osimertinib 80 mg once daily on Days 4-34. RESULTS: Geometric least squares mean (GLSM) ratios (90% confidence intervals) of simvastatin plus osimertinib for area under the plasma concentration-time curves from zero to infinity (AUC) were 91% (77-108): entirely contained within the predefined no relevant effect limits, and Cmax of 77% (63, 94) which was not contained within the limits. GLSM ratios of rosuvastatin plus osimertinib for AUC were 135% (115-157) and Cmax were 172 (146, 203): outside the no relevant effect limits. CONCLUSIONS: Osimertinib is unlikely to have any clinically relevant interaction with CYP3A substrates and has a weak inhibitory effect on BCRP. No new safety concerns were identified in either study.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Rosuvastatin Calcium/pharmacokinetics , Simvastatin/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/physiology , Acrylamides/administration & dosage , Acrylamides/adverse effects , Adult , Aged , Aged, 80 and over , Aniline Compounds/administration & dosage , Aniline Compounds/adverse effects , Area Under Curve , Cytochrome P-450 CYP3A/physiology , Female , Humans , Hydroxycholesterols/blood , Male , Middle Aged , Neoplasm Proteins/physiology
19.
Pharm Res ; 35(11): 199, 2018 Aug 30.
Article in English | MEDLINE | ID: mdl-30167890

ABSTRACT

PURPOSE: In this study, a new modified nanoprecipitation approach that more efficient and simpler than conventional approach was developed to synthesize D-alpha-Tocopheryl polyethylene glycol 1000 succinate stabilized liposome-PLGA hybrid nanoparticle, loaded with simvastatin (ST-TLPN). METHODS: The optimum formulation was screened via investigation of the impact of TPGS mass within polymeric core and lipid shell on the physicochemical properties of nanoparticles respectively. FTIR, and drug release of ST-TLPN were also systematically determined. Finally, the cellular internalization was evaluated using the murine macrophage cell line, in vivo pharmacokinetic behavior and antiatherogenic efficacies were elaborately examined in atherosclerotic rabbit models. RESULTS: With the weight ratio of TPGS-to-PLGA in organic phase of 30% and TPGS-to-lipid in aqueous phase of 35%, ST-TLPN exhibited core-shell structure, sub-100 nm size, EE% of over 90% and a slow release profile. The excellent cellular uptake was displayed in RAW264.7 cell line. Improved pharmacokinetic behavior, and enhanced antiatherogenic efficacy of ST-TLPN in the model animals were also revealed compared with ST-loaded PLGA nanoparticles. CONCLUSION: These findings suggest the modified nanoprecipitation method holds great potential for fabricating LPN, aided by the multiple functions of TPGS. And the prepared TLPN is a promising delivery system for use in the pharmaceutical field.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liposomes/chemistry , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Simvastatin/pharmacology , Vitamin E/chemistry , Animals , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Drug Carriers , Drug Liberation , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Mice , Oxidative Stress/drug effects , Particle Size , RAW 264.7 Cells , Rabbits , Simvastatin/chemistry , Simvastatin/pharmacokinetics , Surface Properties
20.
Drug Dev Ind Pharm ; 44(2): 276-288, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29043860

ABSTRACT

The aim of this study was to develop hydroxypropyl methyl cellulose (HPMC)/chitosan gel containing polymeric micelles loaded with simvastatin (Sim) and evaluates its wound healing properties in rats. An irregular full factorial design was employed to evaluate the effects of various formulation variables including polymer/drug ratio, hydration temperature, hydration time, and organic solvent type on the physicochemical characteristics of pluronic F127-cholesterol nanomicelles prepared using the film hydration method. Among single studied factors, solvent type had the most impact on the amount of drug loading and zeta potential. Particle size and release efficiency was more affected by hydration temperature. The optimized formulation suggested by desirability of 93.5% was prepared using 1 mg of Sim, 10 mg of copolymer, dichloromethane as the organic solvent, hydration time of 45 min and hydration temperature of 25 °C. The release of the drug from nanomicelles was found to be biphasic and showed a rapid release in the first stage followed by a sustained release for 96 h. The gel-contained nanomicelles exhibited pseudo-plastic flow and more sustained drug release profile compared to nanomicelles. In excision wound model on normal rats, the wound closure of the group treated by Sim loaded micelles-gel was superior to other groups. Taken together, Sim loaded micelles-gel may represent a novel topical formulation for wound healing.


Subject(s)
Chitosan/chemistry , Hypromellose Derivatives/chemistry , Micelles , Simvastatin/administration & dosage , Simvastatin/pharmacology , Wound Healing/drug effects , Animals , Chemistry, Pharmaceutical , Cholesterol/chemistry , Drug Carriers/chemistry , Drug Liberation , Gels , Male , Nanoparticles/chemistry , Particle Size , Poloxamer/chemistry , Polyethylene Glycols/chemistry , Rats , Rats, Wistar , Rheology , Simvastatin/pharmacokinetics , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL