Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters

Publication year range
1.
Environ Res ; 257: 119334, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38838750

ABSTRACT

In this study, bimetallic Cu-Fe nanoparticles were synthesized using the green approach with Piper betle leaves, and the removal efficiency of one of the pharmaceutical compounds, Atorvastatin, was investigated. UV, SEM, FTIR, EDAX, particle size, and zeta potential measurements were used to confirm nanoparticle fabrication. The removal efficiency of Atorvastatin (10 mg/L) by bimetallic Cu-Fe nanoparticles was 67% with a contact time of 30 min at pH 4, the adsorbent dosage of 0.2 g/L, and stirring at 100 rpm. Piper betle bimetallic Cu-Fe nanoparticles have demonstrated excellent stability, reusability, and durability, even after being reused five times. Furthermore, the synthesized bimetallic Cu-Fe nanoparticles demonstrated remarkable antimicrobial properties against gram-negative strains such as Escherichia coli and Klebsiella pneumoniae, gram-positive strains such as Staphylococcus aureus and Bacillus subtilis, and fungi such as Aspergillus niger. In addition, the antioxidant properties of the synthesized bimetallic Cu-Fe nanoparticles were assessed using the DPPH radical scavenging assay. The results indicated that the nanoparticles had good antioxidant activity. Thus, using Piper betle extract to make Cu-Fe nanoparticles made the procedure less expensive, chemical-free, and environmentally friendly, and the synthesized bimetallic Cu-Fe nanoparticles helped remove the pharmaceutical compound Atorvastatin from wastewater.


Subject(s)
Atorvastatin , Copper , Iron , Metal Nanoparticles , Piper betle , Plant Leaves , Water Pollutants, Chemical , Atorvastatin/chemistry , Plant Leaves/chemistry , Copper/chemistry , Iron/chemistry , Metal Nanoparticles/chemistry , Water Pollutants, Chemical/chemistry , Piper betle/chemistry , Pyrroles/chemistry
2.
Acta Pharmacol Sin ; 43(1): 240-250, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33686244

ABSTRACT

Cardiovascular safety assessment is vital for drug development, yet human cardiovascular cell models are lacking. In vitro mass-generated human pluripotent stem cell (hPSC)-derived cardiovascular cells are a suitable cell model for preclinical cardiovascular safety evaluations. In this study, we established a preclinical toxicology model using same-origin hPSC-differentiated cardiomyocytes (hPSC-CMs) and endothelial cells (hPSC-ECs). For validation of this cell model, alirocumab, a human antibody against proprotein convertase subtilisin kexin type 9 (PCSK9), was selected as an emerging safe lipid-lowering drug; atorvastatin, a common statin (the most effective type of lipid-lowering drug), was used as a drug with reported side effects at high concentrations, while doxorubicin was chosen as a positive cardiotoxic drug. The cytotoxicity of these drugs was assessed using CCK8, ATP, and lactate dehydrogenase release assays at 24, 48, and 72 h. The influences of these drugs on cardiomyocyte electrophysiology were detected using the patch-clamp technique, while their effects on endothelial function were determined by tube formation and Dil-acetylated low-density lipoprotein (Dil-Ac-LDL) uptake assays. We showed that alirocumab did not affect the cell viability or cardiomyocyte electrophysiology in agreement with the clinical results. Atorvastatin (5-50 µM) dose-dependently decreased cardiovascular cell viability over time, and at a high concentration (50 µM, ~100 times the normal peak serum concentration in clinic), it affected the action potentials of hPSC-CMs and damaged tube formation and Dil-Ac-LDL uptake of hPSC-ECs. The results demonstrate that the established same-origin hPSC-derived cardiovascular cell model can be used to evaluate lipid-lowering drug safety in cardiovascular cells and allow highly accurate preclinical assessment of potential drugs.


Subject(s)
Anticholesteremic Agents/pharmacology , Atorvastatin/pharmacology , Endothelial Cells/drug effects , Myocytes, Cardiac/drug effects , Anticholesteremic Agents/chemistry , Atorvastatin/chemistry , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
3.
Mol Med ; 27(1): 105, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34503440

ABSTRACT

BACKGROUND: Vaccination programs have been launched worldwide to halt the spread of COVID-19. However, the identification of existing, safe compounds with combined treatment and prophylactic properties would be beneficial to individuals who are waiting to be vaccinated, particularly in less economically developed countries, where vaccine availability may be initially limited. METHODS: We used a data-driven approach, combining results from the screening of a large transcriptomic database (L1000) and molecular docking analyses, with in vitro tests using a lung organoid model of SARS-CoV-2 entry, to identify drugs with putative multimodal properties against COVID-19. RESULTS: Out of thousands of FDA-approved drugs considered, we observed that atorvastatin was the most promising candidate, as its effects negatively correlated with the transcriptional changes associated with infection. Atorvastatin was further predicted to bind to SARS-CoV-2's main protease and RNA-dependent RNA polymerase, and was shown to inhibit viral entry in our lung organoid model. CONCLUSIONS: Small clinical studies reported that general statin use, and specifically, atorvastatin use, are associated with protective effects against COVID-19. Our study corroborrates these findings and supports the investigation of atorvastatin in larger clinical studies. Ultimately, our framework demonstrates one promising way to fast-track the identification of compounds for COVID-19, which could similarly be applied when tackling future pandemics.


Subject(s)
Antiviral Agents/pharmacology , Atorvastatin/pharmacology , COVID-19 Drug Treatment , Lung/drug effects , Organoids/drug effects , SARS-CoV-2/drug effects , Antiviral Agents/chemistry , Atorvastatin/chemistry , COVID-19/prevention & control , Cell Line , Coronavirus 3C Proteases/chemistry , Coronavirus RNA-Dependent RNA Polymerase/chemistry , Doxycycline/pharmacology , Drug Approval , Drug Repositioning , Gene Expression Regulation/drug effects , Humans , Lung/virology , Models, Biological , Molecular Docking Simulation , Organoids/virology , Raloxifene Hydrochloride/chemistry , Raloxifene Hydrochloride/pharmacology , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/genetics , Trifluoperazine/chemistry , Trifluoperazine/pharmacology , United States , United States Food and Drug Administration , Vesiculovirus/genetics , Virus Internalization/drug effects
4.
Mol Pharm ; 18(9): 3378-3386, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34351158

ABSTRACT

Statins are 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors that are widely used to prevent cardiovascular diseases. However, a series of pleiotropic mechanisms have been associated with statins, particularly with atorvastatin. Therefore, the assessment of [18F]atorvastatin kinetics with positron emission tomography (PET) may elucidate the mechanism of action of statins and the impact of sexual dimorphism, which is one of the most debated interindividual variations influencing the therapeutic efficacy. [18F]Atorvastatin was synthesized via a previously optimized 18F-deoxyfluorination strategy, used for preclinical PET studies in female and male Wistar rats (n = 7 for both groups), and for subsequent ex vivo biodistribution assessment. PET data were fitted to several pharmacokinetic models, which allowed for estimating relevant kinetic parameters. Both PET imaging and biodistribution studies showed negligible uptake of [18F]atorvastatin in all tissues compared with the primary target organ (liver), excretory pathways (kidneys and small intestine), and stomach. Uptake of [18F]atorvastatin was 38 ± 3% higher in the female liver than in the male liver. The irreversible 2-tissue compartment model showed the best fit to describe [18F]atorvastatin kinetics in the liver. A strong correlation (R2 > 0.93) between quantitative Ki (the radiotracer's unidirectional net rate of influx between compartments) and semi-quantitative liver's SUV (standard uptake value), measured between 40 to 90 min, showed potential to use the latter parameter, which circumvents the need for blood sampling as a surrogate of Ki for monitoring [18F]atorvastatin uptake. Preclinical assays showed faster uptake and clearance for female rats compared to males, seemingly related to a higher efficiency for exchanges between the arterial input and the hepatic tissue. Due to the slow [18F]atorvastatin kinetics, equilibrium between the liver and plasma concentration was not reached during the time frame studied, making it difficult to obtain sufficient and accurate kinetic information to quantitatively characterize the radiotracer pharmacokinetics over time. Nevertheless, the reported results suggest that the SUV can potentially be used as a simplified measure, provided all scans are performed at the same time point. Preclinical PET-studies with [18F]atorvastatin showed faster uptake and clearance in female compared to male rats, apparently related to higher efficiency for exchange between arterial blood and hepatic tissue.


Subject(s)
Atorvastatin/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Positron-Emission Tomography/methods , Radiopharmaceuticals/analysis , Animals , Atorvastatin/administration & dosage , Atorvastatin/analysis , Atorvastatin/chemistry , Female , Fluorine Radioisotopes/administration & dosage , Fluorine Radioisotopes/analysis , Hepatobiliary Elimination , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/analysis , Male , Molecular Imaging/methods , Radiopharmaceuticals/administration & dosage , Rats , Rats, Wistar , Sex Factors , Tissue Distribution
5.
Int J Mol Sci ; 22(11)2021 May 24.
Article in English | MEDLINE | ID: mdl-34073952

ABSTRACT

Statins are the most effective cholesterol-lowering drugs. They also exert many pleiotropic effects, including anti-cancer and cardio- and neuro-protective. Numerous nano-sized drug delivery systems were developed to enhance the therapeutic potential of statins. Studies on possible interactions between statins and human proteins could provide a deeper insight into the pleiotropic and adverse effects of these drugs. Adenylate kinase (AK) was found to regulate HDL endocytosis, cellular metabolism, cardiovascular function and neurodegeneration. In this work, we investigated interactions between human adenylate kinase isoenzyme 1 (hAK1) and atorvastatin (AVS), fluvastatin (FVS), pravastatin (PVS), rosuvastatin (RVS) and simvastatin (SVS) with fluorescence spectroscopy. The tested statins quenched the intrinsic fluorescence of hAK1 by creating stable hAK1-statin complexes with the binding constants of the order of 104 M-1. The enzyme kinetic studies revealed that statins inhibited hAK1 with significantly different efficiencies, in a noncompetitive manner. Simvastatin inhibited hAK1 with the highest yield comparable to that reported for diadenosine pentaphosphate, the only known hAK1 inhibitor. The determined AK sensitivity to statins differed markedly between short and long type AKs, suggesting an essential role of the LID domain in the AK inhibition. Our studies might open new horizons for the development of new modulators of short type AKs.


Subject(s)
Adenylate Kinase/chemistry , Geobacillus stearothermophilus/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Adenylate Kinase/metabolism , Amino Acid Sequence , Atorvastatin/chemistry , Circular Dichroism , Fluvastatin/chemistry , Geobacillus stearothermophilus/chemistry , Geobacillus stearothermophilus/enzymology , Geobacillus stearothermophilus/genetics , Humans , Inhibitory Concentration 50 , Isoenzymes/chemistry , Kinetics , Ligands , Molecular Docking Simulation , Pravastatin/chemistry , Protein Binding , Recombinant Proteins , Rosuvastatin Calcium/chemistry , Sequence Alignment , Simvastatin/chemistry , Spectrometry, Fluorescence , Spectrophotometry , Static Electricity , Temperature
6.
Int J Mol Sci ; 22(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34681767

ABSTRACT

Atorvastatin ester (Ate) is a structural trim of atorvastatin that can regulate hyperlipidemia. The purpose of this study was to evaluate the lipid-lowering effect of Ate. Male Sprague Dawley (SD) rats were fed a high-fat diet for seven months and used as a hyperlipidemia model. The lipid level and liver function of the hyperlipidemia rats were studied by the levels of TG, TC, LDL, HDL, ALT, and AST in serum after intragastric administration with different doses of Ate. HE staining was used to observe the pathological changes of the rat liver and gastrocnemius muscle. The lipid deposits in the liver of rats were observed by staining with ORO. The genes in the rat liver were sequenced by RNA-sequencing. The results of the RNA-sequencing were further examined by qRT-PCR and western blotting. Biochemical test results indicated that Ate could obviously improve the metabolic disorder and reduce both the ALT and AST levels in serum of the hyperlipidemia rats. Pathological results showed that Ate could improve HFD-induced lipid deposition and had no muscle toxicity. The RNA-sequencing results suggested that Ate affected liver lipid metabolism and cholesterol, metabolism in the hyperlipidemia-model rats may vary via the PPAR-signaling pathway. The western blotting and qRT-PCR results demonstrated the Ate-regulated lipid metabolism in the hyperlipidemia model through the PPAR-signaling pathway and HMGCR expression. In brief, Ate can significantly regulate the blood lipid level of the model rats, which may be achieved by regulating the PPAR-signaling pathway and HMGCR gene expression.


Subject(s)
Atorvastatin/pharmacology , Hydroxymethylglutaryl CoA Reductases/metabolism , Hyperlipidemias/drug therapy , Lipid Metabolism/drug effects , PPAR gamma/metabolism , Animals , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/pharmacology , Atorvastatin/adverse effects , Atorvastatin/chemistry , Body Weight/drug effects , Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Rats, Sprague-Dawley , Signal Transduction/drug effects
7.
AAPS PharmSciTech ; 22(2): 59, 2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33517486

ABSTRACT

Development of fixed dose combinations is growing and many of these drug combinations are being legally marketed. However, the development of these requires careful investigation of possible physicochemical changes during co-processing. This requires investigation of the effect of co-processing of drug combination in absence of excipients to maximize the chance of interaction (if any). Accordingly, the aim was to investigate the effect of co-processing of ezetimibe and atorvastatin on drugs dissolution rate. The objective was extended to in vitro in vivo correlation. Drugs were subjected to wet co-processing in presence of ethanol after being mixed at different ratios. The prepared formulations were characterized using FTIR spectroscopy, X-ray powder diffraction, differential scanning calorimetry, scanning electron microscopy, and in vitro dissolution testing. These investigations proved the possibility of eutectic system formation after drugs co-processing. This was reflected on drugs dissolution rate which was significantly enhanced at dose ratio and 2:1 atorvastatin:ezetimibe molar ratio compared to the corresponding pure drugs. In vivo antihyperlipidemic effects of the co-processed drugs were monitored in albino mice which were subjected to hyperlipidemia induction using poloxamer 407. The results showed significant enhancement in pharmacological activity as revealed from pronounced reduction in cholesterol level in mice administering the co-processed form of both drugs. Besides, histopathological examinations of the liver showed marked decrease in hepatic vacuolation. In conclusion, co-processing of atorvastatin with ezetimibe resulted in beneficial eutexia which hastened the dissolution rate and pharmacological effects of both drugs.Graphical abstract.


Subject(s)
Anticholesteremic Agents/administration & dosage , Atorvastatin/administration & dosage , Ezetimibe/administration & dosage , Animals , Anticholesteremic Agents/pharmacology , Atorvastatin/chemistry , Atorvastatin/pharmacology , Drug Combinations , Drug Liberation , Ezetimibe/chemistry , Ezetimibe/pharmacology , Male , Mice
8.
AAPS PharmSciTech ; 22(5): 161, 2021 May 24.
Article in English | MEDLINE | ID: mdl-34031791

ABSTRACT

Atorvastatin (ATV) is a poorly water-soluble drug that exhibits poor oral bioavailability. Therefore, present research was designed to develop ATV solid dispersions (SDs) to enhance the solubility, drug release, and oral bioavailability. Various SDs of ATV were formulated by conventional and microwave-induced melting methods using Gelucire®48/16 as a carrier. The formulated SDs were characterized for different physicochemical characterizations, drug release, and oral bioavailability studies. The results obtained from the different physicochemical characterization indicate the molecular dispersion of ATV within various SDs. The drug polymer interaction results showed no interaction between ATV and used carrier. There was marked enhancement in the solubility (1.95-9.32 folds) was observed for ATV in prepared SDs as compare to pure ATV. The drug content was found to be in the range of 96.19% ± 2.14% to 98.34% ± 1.32%. The drug release results revealed significant enhancement in ATV release from prepared SDs compared to the pure drug and the marketed tablets. The formulation F8 showed high dissolution performance (% DE30 value of 80.65 ± 3.05) among the other formulations. Optimized Gelucire®48/16-based SDs formulation suggested improved oral absorption of atorvastatin as evidenced with improved pharmacokinetic parameters (Cmax 2864.33 ± 573.86 ng/ml; AUC0-t 5594.95 ± 623.3 ng/h ml) as compared to ATV suspension (Cmax 317.82 ± 63.56 ng/ml; AUC0-t 573.94 ± 398.9 ng/h ml) and marketed tablets (Cmax 852.72 ± 42.63 ng/ml; 4837.4 ± 174.7 ng/h ml). Conclusively, solid dispersion-based oral formulation of atorvastatin could be a promising approach for enhanced drug solubilization, dissolution, and subsequently improved absorption.


Subject(s)
Atorvastatin/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Administration, Oral , Animals , Atorvastatin/blood , Atorvastatin/chemistry , Biological Availability , Drug Carriers/chemistry , Drug Liberation , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , In Vitro Techniques , Rats , Solubility , Tablets
9.
World J Microbiol Biotechnol ; 37(11): 191, 2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34632522

ABSTRACT

The resistance of Candida albicans to azole drugs represents a great global challenge. This study investigates the potential fungicidal effects of atorvastatin (ATO) combinations with fluconazole (FLU), itraconazole (ITR), ketoconazole (KET) and voriconazole (VOR) against thirty-four multidrug-resistant (MDR) C. albicans using checkerboard and time-kill methods. Results showed that 94.12% of these isolates were MDR to ≥ two azole drugs, whereas 5.88% of them were susceptible to azole drugs. The tested isolates exhibited high resistance rates to FLU (58.82%), ITR (52.94%), VOR (47.06%) and KET (35.29%), whereas only three representative (8.82%) isolates were resistant to all tested azoles. Remarkably, the inhibition zones of these isolates were increased at least twofold with the presence of ATO, which interacted in a synergistic (FIC index ≤ 0.5) manner with tested azoles. In silico docking study of ATO and the four azole drugs were performed against the Lanosterol 14-alpha demethylase enzyme (ERG11) of C. albicans. Results showed that the mechanism of action of ATO against C. albicans is similar to that of azole compounds, with a docking score (-4.901) lower than azole drugs (≥5.0) due to the formation a single H-bond with Asp 225 and a pi-pi interaction with Thr 229. Importantly, ATO combinations with ITR, VOR and KET achieved fungicidal effects (≥ 3 Log10 cfu/ml reduction) against the representative isolates, whereas a fungistatic effect (≤ 3 Log10 cfu/ml reduction) was observed with FLU combination. Thus, the combination of ATO with azole drugs could be promising options for treating C. albicans infection.


Subject(s)
Atorvastatin/pharmacology , Azoles/pharmacology , Candida albicans/drug effects , Drug Resistance, Multiple, Fungal/drug effects , Fungicides, Industrial/pharmacology , Animals , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Atorvastatin/chemistry , Atorvastatin/therapeutic use , Azoles/chemistry , Azoles/therapeutic use , Candidiasis/drug therapy , Fluconazole/pharmacology , Fluconazole/therapeutic use , Fungicides, Industrial/chemistry , Fungicides, Industrial/therapeutic use , Humans , Itraconazole/pharmacology , Itraconazole/therapeutic use , Ketoconazole/pharmacology , Ketoconazole/therapeutic use , Kinetics , Microbial Sensitivity Tests , Molecular Docking Simulation , Voriconazole/pharmacology , Voriconazole/therapeutic use
10.
Mol Pharm ; 17(6): 1835-1847, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32315193

ABSTRACT

Inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase of the family of statins have been suggested as therapeutic options in various tumors. Atorvastatin is a statin with the potential to cross the blood-brain barrier; however, the concentrations necessary for a cytotoxic effect against cancer cells exceed the concentrations achievable via oral administration, which made the development of a novel atorvastatin formulation necessary. We characterized the drug loading and basic physicochemical characteristics of micellar atorvastatin formulations and tested their cytotoxicity against a panel of different glioblastoma cell lines. In addition, activity against tumor spheroids formed from mouse glioma and mouse cancer stem cells, respectively, was evaluated. Our results show good activity of atorvastatin against all tested cell lines. Interestingly, in the three-dimensional (3D) models, growth inhibition was more pronounced for the micellar formulation compared to free atorvastatin. Finally, atorvastatin penetration across a blood-brain barrier model obtained from human induced-pluripotent stem cells was evaluated. Our results suggest that the presented micelles may enable much higher serum concentrations than possible by oral administration; however, if transport across the blood-brain barrier is sufficient to reach the therapeutic atorvastatin concentration for the treatment of glioblastoma via intravenous administration remains unclear.


Subject(s)
Antineoplastic Agents/pharmacology , Atorvastatin/chemistry , Atorvastatin/pharmacology , Glioblastoma/drug therapy , Antineoplastic Agents/chemistry , Blood-Brain Barrier , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Drug Compounding , Dynamic Light Scattering , Glioblastoma/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Micelles , Nanomedicine/methods , Neoplastic Stem Cells/drug effects , Oxazoles/chemistry
11.
Magn Reson Chem ; 58(11): 1010-1017, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31469449

ABSTRACT

We report 43 Ca and 13 C solid-state nuclear magnetic resonance (NMR) spectroscopic studies of the ethylene glycol solvate of atorvastatin calcium. The 13 C and 43 Ca chemical shift and 43 Ca quadrupolar coupling tensor parameters are reported. The results are interpreted in terms of the reported X-ray diffraction crystal structure of the solvate and are compared with the NMR parameters of atorvastatin calcium trihydrate, the active pharmaceutical ingredient in Lipitor®. Hartree-Fock and density functional theory calculations of the NMR parameters based on a cluster model derived from the optimized X-ray diffraction crystal structure of the ethylene glycol solvate of atorvastatin calcium are in reasonable agreement with the experimental 43 Ca and 13 C NMR measurables.


Subject(s)
Atorvastatin/chemistry , Ethylene Glycol/chemistry , Calcium Isotopes , Carbon Isotopes , Crystallography, X-Ray , Magnetic Resonance Spectroscopy/standards , Models, Molecular , Molecular Structure , Reference Standards
12.
AAPS PharmSciTech ; 21(7): 263, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32978691

ABSTRACT

In the current research, the main focus was to overcome dermal delivery problems of atorvastatin. To this end, atorvastatin solid lipid nanoparticles (ATR-SLNs) were prepared by ultra-sonication technique. The prepared SLNs had a PDI value of ≤ 0.5, and the particle size of nanoparticles was in the range 71.07 ± 1.72 to 202.07 ± 8.40 nm. It was noticed that, when the concentration of lipid in ATR-SLNs increased, the size of nanoparticles and drug entrapment efficiency were also increased. Results showed that a reduction in the HLB of surfactants used in the preparation of SLN caused an increase in the particle size, zeta potential (better stability), and drug entrapment efficiency. Despite Tween and Span are non-ionic surfactants, SLNs containing these surfactants showed a negative zeta potential, and the absolute zeta potential increased when the concentration of Span 80 was at maximum. DSC thermograms, FTIR spectra, and x-ray diffraction (PXRD) pattern showed good incorporation of ATR in the nanoparticles without any chemical interaction. In vitro skin permeation results showed that SLN containing atorvastatin was capable of enhancing the dermal delivery of atorvastatin where a higher concentration of atorvastatin can be detected in skin layers. This is a hopeful promise which could be developed for clinical studies of the dermal delivery of atorvastatin nanoparticles as an anti-inflammatory agent.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Atorvastatin/administration & dosage , Drug Carriers/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Lipids/chemistry , Nanoparticles/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Atorvastatin/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Particle Size , Polysorbates , Skin/metabolism , Skin Absorption , Surface-Active Agents/metabolism , X-Ray Diffraction
13.
Phys Chem Chem Phys ; 21(12): 6319-6326, 2019 Mar 28.
Article in English | MEDLINE | ID: mdl-30720026

ABSTRACT

We combine experimental and computational determination of 43Ca solid-state NMR parameters (chemical shift tensors, quadrupolar coupling tensors, and Euler angles) to constrain the structure of the local calcium-ligand coordination environment. A new 43Ca NMR crystallographic approach which includes an extensive survey of the Cambridge Structural Database and a new symmetry benchmark is developed to enhance the selectivity of structural screening. The application of this method to quadrupolar NMR crystallographic investigations is demonstrated by unearthing the calcium local structure of the active pharmaceutical ingredient atorvastatin calcium trihydrate, the active ingredient in Lipitor®, in the absence of diffraction data. This method has been tested by applying it to calcium acetate monohydrate which has a known structure.


Subject(s)
Atorvastatin/chemistry , Calcium/chemistry , Magnetic Resonance Spectroscopy , Algorithms , Crystallography , Powder Diffraction
14.
Phys Chem Chem Phys ; 21(35): 19209-19215, 2019 Sep 21.
Article in English | MEDLINE | ID: mdl-31441478

ABSTRACT

Pure-shift NMR experiments provide highly resolved spectra, which could be perfect for precise monitoring of chemical shift variations under different conditions, such as temperature or concentration. However, their sensitivity is relatively low and signal sampling is time-consuming, which leads to long experimental times, making such serial acquisition problematic. In this paper we present a new method of NMR spectroscopy which improves the speed and sensitivity of serial pseudo-two-dimensional pure-shift experiments. The example of variable-temperature study of atorvastatin reveals the potential of the method in verifying the theoretical predictions of solvent-dependent spectral effects.


Subject(s)
Atorvastatin/chemistry , Chemistry Techniques, Analytical/methods , Magnetic Resonance Spectroscopy , Solvents/chemistry , Temperature , Chemistry Techniques, Analytical/standards
15.
Bioorg Chem ; 85: 413-419, 2019 04.
Article in English | MEDLINE | ID: mdl-30665035

ABSTRACT

Retinoid X receptor alpha (RXRα), a central member of the nuclear receptor superfamily and a key regulator of many signal transduction pathways, has been an attractive drug target. We previously discovered that an N-terminally truncated form of RXRα can be induced by specific ligands to form homotetramers, which, as a result of conformational selection, forms the basis for inhibiting the nongenomic activation of RXRα. Here, we report the identification and characterization of atorvastatin as a new RXRα tetramer stabilizer by using structure-based virtual screening and demonstrate that virtual library screening can be used to aid in identifying RXRα ligands that can induce its tetramerization. In this study, docking was applied to screen the FDA-approved small molecule drugs in the DrugBank 4.0 collection. Two compounds were selected and purchased for testing. We showed that the selected atorvastatin could bind to RXRα to promote RXRα-LBD tetramerization. We also showed that atorvastatin possessed RXRα-dependent apoptotic effects. In addition, we used a chemical approach to aid in the studies of the binding mode of atorvastatin.


Subject(s)
Atorvastatin/pharmacology , Protein Multimerization/drug effects , Retinoid X Receptor alpha/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Atorvastatin/chemistry , Atorvastatin/metabolism , Binding Sites , Drug Evaluation, Preclinical , Humans , Ligands , MCF-7 Cells , Protein Binding/drug effects , Protein Domains , Protein Stability/drug effects , Sulindac/analogs & derivatives , Sulindac/metabolism
16.
Molecules ; 24(23)2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31756986

ABSTRACT

There is an unmet need for late-stage 18F-fluorination strategies to label molecules with a wide range of relevant functionalities to medicinal chemistry, in particular (hetero)arenes, aiming to obtain unique in vivo information on the pharmacokinetics/pharmacodynamics (PK/PD) using positron emission tomography (PET). In the last few years, Cu-mediated oxidative radiofluorination of arylboronic esters/acids arose and has been successful in small molecules containing relatively simple (hetero)aromatic groups. However, this technique is sparsely used in the radiosynthesis of clinically significant molecules containing more complex backbones with several aromatic motifs. In this work, we add a new entry to this very limited database by presenting our recent results on the 18F-fluorination of an arylboronic ester derivative of atorvastatin. The moderate average conversion of [18F]F- (12%), in line with what has been reported for similarly complex molecules, stressed an overview through the literature to understand the radiolabeling variables and limitations preventing consistently higher yields. Nevertheless, the current disparity of procedures reported still hampers a consensual and conclusive output.


Subject(s)
Atorvastatin/chemistry , Boronic Acids/chemistry , Copper/chemistry , Fluorine Radioisotopes/chemistry , Halogenation , Radiopharmaceuticals , Catalysis , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry
17.
Biomed Microdevices ; 20(3): 53, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29946758

ABSTRACT

Atorvastatin is a lipid lowering agent with poor oral bioavailability (12%) because of poor solubility and extensive first pass hepatic metabolism. In order to overcome these issues, atorvastatin loaded solid lipid nanoparticles (ATOR-SLNs) were prepared by using glyceryl tripalmitate as lipid carrier, poloxamer 407 as surfactant and soya lecithin as emulsifier. The purpose of this work was to optimize the formulation with the application of response surface methodology to improve the physicochemical properties. The central composite rotatable design consisting of three factored factorial design with three levels was used for the optimization of the formulations. The optimized formulation was composed of drug/lipid ratio of 1:3.64, surfactant concentration of 1.5% with 5 min time for sonication. Fourier transforms infrared (FTIR) spectroscopy and differential scanning calorimetry (DSC) studies confirmed the compatibility of drug and lipid in the formulation. The optimized ATOR- SLNs showed almost spherical shape with a mean particle size of 338.5 nm, zeta potential of -24.7mV, DL of 17.7% and EE of 81.06% respectively. The in vitro drug release study showed a burst release at the initial stage followed by the prolongation of drug release from lipid matrix. Stability study revealed that ATOR-SLNs were more stable at 4±2˚C when compared with storage at 25±2˚C/60±5% RH during the six months storage period. These results indicated that the developed ATOR-SLNs is a promising approach for increment of bioavailability by improving the physicochemical properties.


Subject(s)
Atorvastatin/chemistry , Atorvastatin/pharmacology , Drug Carriers/chemistry , Nanoparticles/chemistry , Biological Availability , Calorimetry, Differential Scanning , Chemical Phenomena , Drug Liberation , Drug Stability , Lecithins/chemistry , Lipids/chemistry , Particle Size , Poloxamer/chemistry , Spectroscopy, Fourier Transform Infrared , Surface Properties , Surface-Active Agents/chemistry
18.
Mol Pharm ; 15(1): 247-255, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29232954

ABSTRACT

Atorvastatin calcium (AC)-loaded nanoparticles (NPs) of mean particle diameter <100 nm and narrow distribution were prepared and characterized. Their in vivo PK as well as PD measures following oral administration in different dosage regimens in hyperlipidemic rats were evaluated. The results revealed that the oral bioavailability of two selected AC-NPs formulations was 235% and 169% relative to Lipitor. However, the treatment regimens were not superior in reducing serum total cholesterol (TC), low-density lipoproteins (LDL), and triglycerides (TG) levels compared to Lipitor. Moreover, the AC-NPs treatments were associated with significant adverse effects observed biochemically and histologically. These results were contradictory with those obtained from a previous study in which similarly formulated AC-NPs of mean particle diameter >200 nm were found to be more safe and effective in reducing TC, LDL, and TG levels when administered to hyperlipiemic rats at reduced dosing frequency compared to daily dose of Lipitor despite their lower oral bioavailability. The discrepant correlation between pharmacokinetics (PK) and pharmacodynamics (PD) results was suggested to pertain to the different biodistribution profiles of AC-NPs depending on their sizes. Hereby, we provide a simple approach of particle size modulation to enhance the efficacy and safety of atorvastatin.


Subject(s)
Atorvastatin/chemistry , Atorvastatin/pharmacokinetics , Nanoparticles/chemistry , Administration, Oral , Animals , Atorvastatin/administration & dosage , Cholesterol/blood , Lipoproteins, LDL/blood , Male , Rats , Tissue Distribution , Triglycerides/blood
19.
Biomed Chromatogr ; 32(7): e4224, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29498757

ABSTRACT

A robust, rapid and sensitive UPLC-MS/MS method has been developed, optimized and validated for the determination of amlodipine (AML) and atorvastatin (ATO) in human plasma using eplerenone as an internal standard (IS). Multiple-reaction monitoring in positive electrospray ionization mode was utilized in Xevo TQD LC-MS/MS. Double extraction was used in sample preparation using diethyl ether and ethyl acetate. The prepared samples were analyzed using an Acquity UPLC BEH C18 (50 × 2.1 mm, 1.7 µm) column. Ammonium formate and acetonitrile, pumped isocraticaly at a flow rate of 0.25 mL/min, were used as a mobile phase. Method validation was done as per the US Food and Drug Administration guidelines. Linearity was achieved in the range of 0.1-10 ng/mL for AML and 0.05-50 ng/mL for ATO. Intra-day and inter-day accuracy and precision were calculated and found to be within the acceptable range. A short run time, of <1.5 min, permits analysis of a large number of plasma samples per batch. The developed and validated method was applied to estimate AML and ATO in a bioequivalence study in healthy human volunteers.


Subject(s)
Amlodipine/blood , Atorvastatin/blood , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Adolescent , Adult , Amlodipine/chemistry , Amlodipine/pharmacokinetics , Atorvastatin/chemistry , Atorvastatin/pharmacokinetics , Humans , Linear Models , Middle Aged , Reproducibility of Results , Sensitivity and Specificity , Therapeutic Equivalency , Young Adult
20.
J Mater Sci Mater Med ; 29(11): 161, 2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30357534

ABSTRACT

Segmented polyurethanes were prepared with polycaprolactone diol as soft segment and various amounts of 4,4´-Methylenebis(cyclohexyl isocyanate) and atorvastatin, a statin used for lowering cholesterol, in order to obtain SPU with different content of rigid segments. Polyurethanes with 35% or 50% of rigid segment content were physicochemically characterized and their biocompatibility assessed with L929 fibroblasts. High concentrations of atorvastatin were incorporated by increasing the content of rigid segments as shown by FTIR, Raman, NMR, XPS and EDX. Thermal and mechanical characterization showed that polyurethanes containing atorvastatin and 35% of rigid segments were low modulus (13 MPa) semicrystalline polymers as they exhibited a glass transition temperature (Tg) at -38 °C, melting temperature (Tm) at 46 °C and crystallinity close to 35.9% as determined by DSC. In agreement with this, X-ray diffraction showed reflections at 21.3° and 23.6° for PCL without reflections for atorvastatin suggesting its presence in amorphous form with higher potential bioavailability. Low content of rigid segments led to highly degradable polymer in acidic, alkaline and oxidative media with an acceptable fibroblast cytotoxicity up to 7 days possibly due to low atorvastatin content.


Subject(s)
Atorvastatin/chemistry , Biocompatible Materials/chemistry , Cyanates/chemistry , Polyesters/chemistry , Polyurethanes/chemistry , Animals , Atorvastatin/toxicity , Biocompatible Materials/toxicity , Cell Line , Cell Survival/drug effects , Mice , Molecular Structure , Nonlinear Optical Microscopy , Polyesters/toxicity , Polyurethanes/toxicity , Spectrophotometry, Infrared , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL