Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
J Mater Sci Mater Med ; 35(1): 29, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38884680

ABSTRACT

Mesoporous silica nanoparticles (MSNPs) coated by chitosan (CS) were shown to be a proper candidate as a carrier for drug delivery purposes. However, choosing the suitable drug-containing complexes to be applied on MSNPs-CS is of much greater importance to evaluate the possible candidate for an efficient combination of cell viability, drug release kinetics, and atherosclerosis prevention. In this regard, this study concentrates on the synthesis and assessment of coated MSNPs-CS designed for drug delivery purposes. The MSNPs are coated with polyelectrolyte complexes (PEC) composed of CS and dextran sulfate (MSNPs-CS-DX), serving as a versatile drug carrier with favorable biological characteristics. CS-DX is applied to MSNPs without requiring complex or multi-step synthesis procedures. Rosuvastatin, a cholesterol-lowering medication, is chosen for its therapeutic relevance. Additionally, CS-DX is found to relatively impede the uptake of low-density lipoproteins (LDLs) by macrophages, enhancing their potential therapeutic utility. FTIR pattern, FESEM, and TEM images prove MSNPs-CS-DX formation. DLS measurement demonstrates the average particle size of 110 nm for MSNPs, with the combined thickness of CS and DX layers ranging from 10 to 15 nm. BET test is carried out to evaluate the pore size and porosity of structure, showing outstanding results that cause an entrapment efficiency of 57% for MSNPs-CS-DX. Furthermore, the findings demonstrate the pH sensitivity of MSNPs-CS-DX on drug release kinetics. Notably, the CS-DX layer exhibits a significant enhancement in cell viability of human umbilical vein endothelial cells (HUVEC) by approximately 24% within a 24 h timeframe compared to MSNPs lacking CS-DX.


Subject(s)
Chitosan , Dextran Sulfate , Drug Delivery Systems , Silicon Dioxide , Chitosan/chemistry , Silicon Dioxide/chemistry , Hydrogen-Ion Concentration , Dextran Sulfate/chemistry , Porosity , Humans , Drug Carriers/chemistry , Nanoparticles/chemistry , Particle Size , Drug Liberation , Cell Survival/drug effects , Animals , Rosuvastatin Calcium/pharmacokinetics , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/administration & dosage , Rosuvastatin Calcium/pharmacology , Mice , Coated Materials, Biocompatible/chemistry , Lipoproteins, LDL , Macrophages/metabolism , Macrophages/drug effects , RAW 264.7 Cells
2.
ACS Appl Mater Interfaces ; 16(20): 26685-26712, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38722359

ABSTRACT

The ubiquitous presence of pharmaceutical pollutants in the environment significantly threatens human health and aquatic ecosystems. Conventional wastewater treatment processes often fall short of effectively removing these emerging contaminants. Therefore, the development of high-performance adsorbents is crucial for environmental remediation. This research utilizes molecular simulation to explore the potential of novel modified metal-organic frameworks (MOFs) in pharmaceutical pollutant removal, paving the way for the design of efficient wastewater treatment strategies. Utilizing UIO-66, a robust MOF, as the base material, we developed UIO-66 functionalized with chitosan (CHI) and oxidized chitosan (OCHI). These modified MOFs' physical and chemical properties were first investigated through various characterization techniques. Subsequently, molecular dynamics simulation (MDS) and Monte Carlo simulation (MCS) were employed to elucidate the adsorption mechanisms of rosuvastatin (ROSU) and simvastatin (SIMV), two prevalent pharmaceutical pollutants, onto these nanostructures. MCS calculations demonstrated a significant enhancement in the adsorption energy by incorporating CHI and OCHI into UIO-66. This increased ROSU from -14,522 to -16,459 kcal/mol and SIMV from -17,652 to -21,207 kcal/mol. Moreover, MDS reveals ROSU rejection rates in neat UIO-66 to be at 40%, rising to 60 and 70% with CHI and OCHI. Accumulation rates increase from 4 Å in UIO-66 to 6 and 9 Å in UIO-CHI and UIO-OCHI. Concentration analysis shows SIMV rejection surges from 50 to 90%, with accumulation rates increasing from 6 to 11 Å with CHI and OCHI in UIO-66. Functionalizing UIO-66 with CHI and OCHI significantly enhanced the adsorption capacity and selectivity for ROSU and SIMV. Abundant hydroxyl and amino groups facilitated strong interactions, improving performance over that of unmodified UIO-66. Surface functionalization plays a vital role in customizing the MOFs for pharmaceutical pollutant removal. These insights guide next-gen adsorbent development, offering high efficiency and selectivity for wastewater treatment.


Subject(s)
Chitosan , Metal-Organic Frameworks , Molecular Dynamics Simulation , Nanostructures , Rosuvastatin Calcium , Simvastatin , Water Pollutants, Chemical , Chitosan/chemistry , Metal-Organic Frameworks/chemistry , Simvastatin/chemistry , Rosuvastatin Calcium/chemistry , Adsorption , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/isolation & purification , Nanostructures/chemistry , Oxidation-Reduction , Phthalic Acids
3.
Int J Biol Macromol ; 266(Pt 1): 130995, 2024 May.
Article in English | MEDLINE | ID: mdl-38521323

ABSTRACT

Critical-size bone defects are one of the main challenges in bone tissue regeneration that determines the need to use angiogenic and osteogenic agents. Rosuvastatin (RSV) is a class of cholesterol-lowering drugs with osteogenic potential. Magnesium oxide (MgO) is an angiogenesis component affecting apatite formation. This study aims to evaluate 3D-printed Polycaprolactone/ß-tricalcium phosphate/nano-hydroxyapatite/ MgO (PCL/ß-TCP/nHA/MgO) scaffolds as a carrier for MgO and RSV in bone regeneration. For this purpose, PCL/ß-TCP/nHA/MgO scaffolds were fabricated with a 3D-printing method and coated with gelatin and RSV. The biocompatibility and osteogenicity of scaffolds were examined with MTT, ALP, and Alizarin red staining. Finally, the scaffolds were implanted in a bone defect of rat's calvaria, and tissue regeneration was investigated after 3 months. Our results showed that the simultaneous presence of RSV and MgO improved biocompatibility, wettability, degradation rate, and ALP activity but decreased mechanical strength. PCL/ß-TCP/nHA/MgO/gelatin-RSV scaffolds produced sustained release of MgO and RSV within 30 days. CT images showed that PCL/ß-TCP/nHA/MgO/gelatin-RSV scaffolds filled approximately 86.83 + 4.9 % of the defects within 3 months and improved angiogenesis, woven bone, and osteogenic genes expression. These results indicate the potential of PCL/ß-TCP/nHA/MgO/gelatin-RSV scaffolds as a promising tool for bone regeneration and clinical trials.


Subject(s)
Bone Regeneration , Gelatin , Magnesium Oxide , Osteogenesis , Printing, Three-Dimensional , Rosuvastatin Calcium , Tissue Scaffolds , Bone Regeneration/drug effects , Rosuvastatin Calcium/pharmacology , Rosuvastatin Calcium/chemistry , Tissue Scaffolds/chemistry , Gelatin/chemistry , Animals , Rats , Osteogenesis/drug effects , Magnesium Oxide/chemistry , Magnesium Oxide/pharmacology , Polyesters/chemistry , Drug Liberation , Durapatite/chemistry , Durapatite/pharmacology , Delayed-Action Preparations/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Skull/drug effects , Tissue Engineering/methods
4.
PLoS One ; 17(1): e0263026, 2022.
Article in English | MEDLINE | ID: mdl-35061861

ABSTRACT

The present study is aimed at enhancing the solubility of rosuvastatin (RST) by designing betacyclodextrin/polyvinypyrrolidone-co-poly (2-acrylamide-2-methylpropane sulphonic acid) crosslinked hydrophilic nanogels in the presence of crosslinker methylene bisacrylamide through free-radical polymerization method. Various formulations were fabricated by blending different amounts of betacyclodextrin, polyvinylpyrrolidone, 2-acrylamide-2-methylpropane sulphonic acid, and methylene bisacrylamide. The developed chemically crosslinked nanogels were characterized by FTIR, SEM, PXRD, TGA, DSC, sol-gel analysis, zeta size, micromeritics properties, drug loading percentage, swelling, solubility, and release studies. The FTIR spectrum depicts the leading peaks of resultant functional groups of blended constituents while a fluffy and porous structure was observed through SEM images. Remarkable reduction in crystallinity of RST in developed nanogels revealed by PXRD. TGA and DSC demonstrate the good thermal stability of nanogels. The size analysis depicts the particle size of the developed nanogels in the range of 178.5 ±3.14 nm. Drug loading percentage, swelling, solubility, and release studies revealed high drug loading, solubilization, swelling, and drug release patterns at 6.8 pH paralleled to 1.2 pH. In vivo experiments on developed nanogels in comparison to marketed brands were examined and better results regarding pharmacokinetic parameters were observed. The compatibility and non-toxicity of fabricated nanogels to biological systems was supported by a toxicity study that was conducted on rabbits. Efficient fabrication, excellent physicochemical properties, improved dissolution, high solubilization, and nontoxic nanogels might be a capable approach for the oral administration of poorly water-soluble drugs.


Subject(s)
Drug Carriers , Nanogels , Rosuvastatin Calcium , Animals , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Drug Evaluation, Preclinical , Nanogels/chemistry , Nanogels/therapeutic use , Rabbits , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacokinetics , Rosuvastatin Calcium/pharmacology , Solubility , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/pharmacokinetics , beta-Cyclodextrins/pharmacology
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.
Chem Phys Lipids ; 237: 105081, 2021 07.
Article in English | MEDLINE | ID: mdl-33811848

ABSTRACT

The aim of this research was to study the effect of marketed tablet (Crestor®) powder suspension (MTPS) and nanoparticle formulation of rosuvastatin calcium (RC) on the pharmacokinetic (PK) and pharmacodynamic (PD) parameters in hyperlipidemia rats. The hyperlipidemia is induced by intraperitoneal injection of Triton-X-100 in 0.9 %w/v saline solution. The marketed tablet was dispersed into suspension. The RC loaded nanoparticles (RC-NPs) are prepared by homogenization method. The prepared RC-NP formulation was characterized for size, drug excipient compatibility and crystallization by differential scanning calorimeter (DSC), morphology by SEM, stability at room temperature, in-vitro dissolution and in-situ absorption in rats. Further, the pharmacokinetic and pharmacodynamic studies were conducted in hyperlipidemia rats. The size of the RC-NP formulation was found to be 183.4 ± 4.5 nm and to be nearly spherical by SEM. DSC studies revealed that no interaction and RC converted to amorphous form in RC-NP formulation. RC-NP formulation was physically and chemically stable over two months at room temperature. The drug release was found to be 25.8 ± 2.5 and 89.96 ± 2.8 % in five mins, respectively from MTPS and RC-NP formulations. The Peff of MTPS and NP of RC was 1.8 ± 0.2 × 10-5 and 2.7 ± 0.3 × 10-5 cm/s, respectively. From the PK studies, the enhancement in the oral bioavailability was found to be 2.4-folds when compared to MTPS formulation and statistically significant (p < 0.05). PD study of RC-NP formulation in hyperlipidemic rats exhibited decrease in lipid profile for 24 h, while MTPS exhibited a decrease in lipid profile for 12 h. Therefore, the results conclusively demonstrate the nanoparticles of RC showed significant enhancement in the PK and PD parameters.


Subject(s)
Lipids/analysis , Nanoparticles/chemistry , Rosuvastatin Calcium/metabolism , Administration, Oral , Animals , Biological Availability , Drug Carriers/chemistry , Drug Compounding , Drug Liberation , Drug Stability , Half-Life , Hyperlipidemias/chemically induced , Hyperlipidemias/drug therapy , Hyperlipidemias/pathology , Male , Octoxynol/toxicity , Particle Size , Rats , Rats, Wistar , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacokinetics , Rosuvastatin Calcium/therapeutic use
7.
Biomed Mater ; 16(4)2021 06 04.
Article in English | MEDLINE | ID: mdl-33482656

ABSTRACT

Nowadays, heart disease, especially myocardial infarction, is one of the most astoundingly unfortunate causes of mortality in the world. That is why special attention has been paid toward tissue engineering techniques for curing and regeneration of heart tissue. In this study, poly(N-isopropyl acrylamide) (PNIPAAm), a temperature-sensitive injectable hydrogel, was selected as a minimally invasive scaffold to accommodate, carry, and release of niosomal rosuvastatin to the inflicted area for inducing angiogenesis and thus accelerating the healing process. The characteristics of PNIPAAm were studied by scanning electron microscopy, rheology tests, and Fourier transform infrared spectroscopy. The properties of the niosomal rosuvastatin release system, including particle size distribution, zeta potential, encapsulation efficiency (EE), and drug release, were also studied. The results showed that niosomes (358 nm) had a drug EE of 78% and a loading capacity of 53%. The drug was sustainably released from the system up to about 54% in 5 d. Cellular studies showed no toxicity to the endothelial cell lines, and the niosomal drug with a concentration of 7.5 nM enhanced cell proliferation, and cell migration increased from 72% to 90% compared to the control sample. Therefore, the controlled-release of niosomal rosuvastatin enhanced angiogenesis in a dose-dependent manner. Taken together, these advantages suggest that PNIPAAm-based niosomal hydrogel provides a promising candidate as an angiogentic injectable scaffold for potential cardiac tissue regeneration.


Subject(s)
Acrylic Resins/chemistry , Angiogenesis Inducing Agents , Drug Delivery Systems/methods , Hydrogels/chemistry , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/pharmacokinetics , Angiogenesis Inducing Agents/pharmacology , Cells, Cultured , Heart/physiology , Human Umbilical Vein Endothelial Cells , Humans , Injections , Neovascularization, Physiologic/drug effects , Regeneration , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacokinetics , Rosuvastatin Calcium/pharmacology , Tissue Engineering
8.
Drug Dev Ind Pharm ; 46(9): 1495-1506, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32804005

ABSTRACT

Despite being the most effective hypolipidemic agent, poor physicochemical properties of Rosuvastatin calcium (RCa) remain challenging obstacles in the development of pharmaceutical dosage forms. Inclusion complexes (ICs) of RCa with cyclodextrin (CD) derivatives; methyl-beta-cyclodextrin (M-ß-CD) and sulfobutylether-beta-cyclodextrin (SBE-ß-CD; Captisol®) were formulated by kneading and freeze-drying (lyophilization) methods. Pysicochemical properties of ICs were evaluated by SEM, DSC, XRD, FT-IR, 1H-NMR analyses. Entrapment efficiency (EE), water solubility, in vitro release analyses were also performed. Safety and efficacy of the ICs were analyzed by cytotoxicity and permeation studies on Caco-2 cell lines. Both CDs indicated AL type phase solubility diagrams showing that [1:1] molar ratio. Apparent stability constants (K1:1) were found to be 60.93 M-1 for M-ß-CD and 158.07 M-1 for Captisol®. High EE in the range of 93.50-105.40% was achieved. Molar solubility of RCa was increased 3.7- and 4.1-fold with M-ß-CD and Captisol® ICs, respectively. In vitro release analyses have indicated the equivalence of dissolution profiles for M-ß-CD and Captisol® based ICs to that of pure RCa (f2 > 50). Cytotoxicity studies on Caco-2 cell lines have revealed the safety of ICs for oral use. Permeability studies demonstrated that selected lyophilized F6 formulation has shown the best permeation rate with Papp value of 3.08 × 10-7 cm·s-1. Considering greater water solubility, lower toxicity, high efficiency of complexation as well as, RCa-like permeability and in vitro release behavior at pH 6.8; Captisol® based lyophilized F6 formulation was selected as the best IC to be used in oral dosage forms of RCa.


Subject(s)
Rosuvastatin Calcium/chemistry , beta-Cyclodextrins/chemistry , Caco-2 Cells , Humans , Solubility , Spectroscopy, Fourier Transform Infrared
9.
Int J Nanomedicine ; 15: 2391-2402, 2020.
Article in English | MEDLINE | ID: mdl-32308390

ABSTRACT

BACKGROUND: Oil-in-water drug nanoemulsion forms drug delivery systems with high oral bioavailability. The conventional fabrication methods of nanoemulsion are low energy emulsification methods and high energy emulsification methods. However, both two methods are not ideal for industrial production. The problem of low energy emulsification methods is the high dosage of surfactant and co-surfactant which has potential biosecurity issues. What is more, high energy emulsification methods have some disadvantages, like the destruction of drug components, the price of equipment and the difficulties of industrial production. Hence, there have been a few commercial drug nanoemulsions so far. METHODS: In this work, we reported a novel method for the fabrication of stable and transparent drug nanoemulsion which contains hydrophilic drug rosuvastatin (ROS) calcium or hydrophobic drug silybinin (SYN) by using high-gravity rotating packed bed (RPB). The drug nanoemulsion was systematically characterized by droplet size, size distribution, stability and in vitro drug release as well as Caco-2 cells permeability. RESULTS: Compared with the self-emulsification method (SE), high-gravity technology could reduce 75% amount of mixed surfactants. The as-prepared nanoemulsion exhibited a very narrow droplet size distribution with a size of 13.53 ± 0.53 nm and a polydispersity index of 0.073 ± 0.018. Meanwhile, the drug nanoemulsion was physicochemically stable at 25°C and 4°C for one-year storage. Furthermore, both ROS and SYN nanoemulsion displayed higher cell permeability and in vitro dissolution than that of commercial formulations. CONCLUSION: These results demonstrate that RPB can be a potential device to facilitate the industrial production of drug nanoemulsion.


Subject(s)
Drug Delivery Systems/methods , Nanostructures/chemistry , Administration, Oral , Biological Availability , Caco-2 Cells , Drug Liberation , Drug Stability , Emulsions/administration & dosage , Emulsions/chemistry , Emulsions/pharmacokinetics , Humans , Hydrophobic and Hydrophilic Interactions , Nanostructures/administration & dosage , Particle Size , Rosuvastatin Calcium/administration & dosage , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacokinetics , Silybin/administration & dosage , Silybin/chemistry , Silybin/pharmacokinetics , Surface-Active Agents/chemistry
10.
BMC Pharmacol Toxicol ; 21(1): 14, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32085802

ABSTRACT

BACKGROUND: Rosuvastatin (RSV) is a poorly water-soluble drug that has an absolute oral bioavailability of only 20%. The aim of this work was to prepare a positively charged chitosan coated flexible lipid-based vesicles (chitosomes) and compare their characteristics to the corresponding negatively charged flexible liposomal nanoparticles (NPs) in order to develop new RSV nanocarrier systems. METHODS: Three formulation factors affecting the development of chitosomes nano-formulation were optimized for their effects on the particles size, entrapment efficiency (EE) and zeta potential. The optimized flexible chitosomes and their corresponding liposomal NPs were characterized for morphology, in vitro release, flexibility and intestinal cell viability. The half maximum inhibitory concentrations (IC50) for both formulations were calculated. RESULTS: The drug to lipid molar ratio, edge activator percent and the chitosan concentration were significantly affecting the characteristics of NPs. The optimized chitosomes nano-formulation exhibited larger size, higher EE and greater zeta potential value when compared to the corresponding liposomal NPs. Both formulations showed a spherical shape nanostructure with a marked outer shell for the chitosomes nano-formulation. Chitosomes illustrated an extended drug release profile when compared with the corresponding liposomal NPs and the prepared drug suspension. Flexibility of both vesicles was confirmed with superiority of liposomal NPs over chitosomes. RSV loaded chitosomes nano-formulation exhibited lower IC50 values and higher therapeutic window while liposomal NPs were compatible with the intestinal cells. CONCLUSIONS: RSV loaded chitosomes nano-formulation could be considered as a promising nanocarrier system with a marked cytotoxic activity while, RSV loaded liposomal NPs are suitable nanocarrier to improve RSV activity in treatment of cardiovascular disorders.


Subject(s)
Antineoplastic Agents/administration & dosage , Chitosan/administration & dosage , Drug Carriers/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Nanoparticles/administration & dosage , Phospholipids/administration & dosage , Rosuvastatin Calcium/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Chitosan/chemistry , Drug Carriers/chemistry , Drug Liberation , HCT116 Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Intestines/cytology , Nanoparticles/chemistry , Phospholipids/chemistry , Rosuvastatin Calcium/chemistry
11.
Int J Pharm ; 578: 119106, 2020 Mar 30.
Article in English | MEDLINE | ID: mdl-32014599

ABSTRACT

Rosuvastatin calcium (RCa) is a very efficient antihyperlipidemic agent, however, being a BCS class II drug, results in poor oral bioavailability. The present study focused on the enhancement of oral bioavailability of RCa with solid lipid nanoparticles (SLNs). Physicochemical properties of the particles were evaluated by particle size (PS), polidispersity index (PDI), zeta potential (ZP), DSC, FT-IR, XRD, 1H NMR analyses. Entrapment efficiency (EE), drug loading capacity (DL), in vitro release and release kinetics were also analyzed. Safety and efficacy of the formulations were analyzed by cytotoxicity, permeability and pharmacokinetic studies. PS values were ranged between ∼134 and 351 nm with homogenous size distribution (PDI ∼ 0.130-0.33) and ZP data were valued within the range of ∼-17 mV to -41 mV. The SLN2 formulation showed the best cytotoxicity test results and had medium permeability (Papp 5.72 × 10-6 cm sec-1) while pure RCa resulted in low permeability (Papp 3.08 × 10-7 cm sec-1). According to the stability analyses (3 months) 5 ±â€¯3 °C and 25 ±â€¯2 °C were found suitable storage temperatures for SLNs. Pharmacokinetic studies confirmed significant improvement in Cmax (1.4 fold) and AUClast (8.5 fold) by SLNs in comparison with the pure drug indicating the enhanced biopharmaceutical performance of the RCa loaded SLNs.


Subject(s)
Lipids/chemistry , Nanoparticles/chemistry , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Biological Availability , Caco-2 Cells , Cell Line, Tumor , Drug Carriers/chemistry , Humans , Particle Size , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Spectroscopy, Fourier Transform Infrared/methods
12.
Bioprocess Biosyst Eng ; 43(1): 21-31, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31542820

ABSTRACT

tert-Butyl (3R,5S)-6-chloro-3,5-dihydroxyhexanoate [(3R,5S)-CDHH] is the key chiral intermediate to synthesize the side chain of the lipid-lowering drug rosuvastatin. Carbonyl reductases showed excellent activity for the biosynthesis of (3R,5S)-CDHH. The requirement of cofactor NADH/NADPH leads to high cost for the industrial application of carbonyl reductases. In this study, a self-sufficient biocatalyst based on carbonyl reductase and NADP+ co-immobilization strategy was developed on an amino resin carrier LX-1000HAA (SCR-NADP+@LX-1000HAA). The self-sufficient biocatalyst achieved in situ cofactor regeneration and showed the activity recovery of 77.93% and the specific activity of 70.45 U/g. Asymmetric synthesis of (3R,5S)-CDHH using SCR-NADP+@LX-1000HAA showed high enantioselectivity (> 99% e.e.) and yield (98.54%). Batch reactions were performed for ten cycles without extra addition of NADP+, and the total yield of (3R,5S)-CDHH achieved at 10.56 g/g biocatalyst. The present work demonstrated the potential of the self-sufficient biocatalyst for the asymmetric biosynthesis of rosuvastatin intermediate.


Subject(s)
Alcohol Oxidoreductases/chemistry , Biocatalysis , Candida/enzymology , Caproates/chemical synthesis , Enzymes, Immobilized/chemistry , Fungal Proteins/chemistry , NADP/chemistry , Alcohol Oxidoreductases/genetics , Candida/genetics , Caproates/chemistry , Enzymes, Immobilized/genetics , Fungal Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Rosuvastatin Calcium/chemical synthesis , Rosuvastatin Calcium/chemistry
13.
Drug Des Devel Ther ; 13: 4035-4051, 2019.
Article in English | MEDLINE | ID: mdl-31839704

ABSTRACT

BACKGROUND: Injectable in situ gel (ISG) systems suffer from high initial drug release that may result in toxic effects. OBJECTIVE: This work aimed to develop an injectable sustained release rosuvastatin (RSV) ISG formulation with minimum initial drug burst and improved hyperlipidemic efficacy. METHODS: Six formulation factors that affect RSV release after 0.5, 2 and 24 hrs have been screened and the significant ones were optimized utilizing an experimental design tool. The optimum ISG formulation components were physicochemically characterized. Kinetic treatment, dissolution efficiency and mean dissolution time were investigated for the developed ISG formulations. Pharmacodynamic effects of the optimized ISG formulation were studied and compared to ISG formulation loaded with free RSV and to a marketed oral drug product. RESULTS: The concentration polylactide-co-ε-caprolactone (25: 75), the surfactant hydrophilic lipophilic balance (HLB) and the ratio of surfactant to polyethylene glycol 400 were significantly affecting the release of RSV during the first 24 h. Physicochemical characterization demonstrated complete dispersion of RSV in the polymeric matrix with slight changes in the drug crystalline structure. The optimized formulation demonstrated acceptable syringeability, good flow rate and was able to extend the in vitro drug release for 34 days. The ISG formulations complied with Weibull model. Pharmacodynamic study revealed a sustained reduction in the lipid profile that lasted for 21 days with a marked decrease in the lipid level during the first 24 hrs from the ISG system loaded with free RSV. CONCLUSION: The optimized RSV ISG formulation could be considered a promising strategy due to a reduction in dosing frequency and enhancement in hypolipidemic efficacy.


Subject(s)
Hyperlipidemias/drug therapy , Polyesters/chemistry , Polyethylene Glycols/chemistry , Rosuvastatin Calcium/therapeutic use , Surface-Active Agents/chemistry , Animals , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemical synthesis , Delayed-Action Preparations/chemistry , Dose-Response Relationship, Drug , Drug Delivery Systems , Gels/administration & dosage , Gels/chemical synthesis , Gels/chemistry , Hyperlipidemias/chemically induced , Male , Molecular Structure , Poloxamer , Rats , Rats, Wistar , Rosuvastatin Calcium/chemistry , Structure-Activity Relationship
14.
J Pharm Pharm Sci ; 22(1): 567-575, 2019.
Article in English | MEDLINE | ID: mdl-31804919

ABSTRACT

PURPOSE: Probe substrates are used routinely to assess transporter function in vitro. Administration of multiple probe substrates together as a "cocktail" in sandwich-cultured human hepatocytes (SCHH) could increase the throughput of transporter function assessment in a physiologically-relevant in vitro system. This study was designed to compare transporter function between cocktail and single agent administration in SCHH. METHODS: Rosuvastatin, digoxin, and metformin were selected as probe substrates of hepatic transporters OATP1B1, OATP1B3, BCRP, P-gp, and OCT1. Total accumulation (Cells+Bile) and biliary excretion index (BEI) values derived from administration of the cocktail were compared to values obtained after administration of single agents in the absence and presence of a model inhibitor, erythromycin estolate. RESULTS: For rosuvastatin and metformin accumulation, the ratio of means [90% confidence interval (CI)] for cocktail to single agent administration was 100% [94%, 106%] and 90% [82%, 99%], respectively. Therefore, the cocktail and single-agent mode of administration were deemed equivalent per standard equivalence criterion of 80-120% for rosuvastatin and metformin accumulation, but not for digoxin accumulation (77% [62%, 92%]). The ratio of means [90% CI] for rosuvastatin BEI values between the two administration modes (105% [97%, 114%]) also was deemed equivalent. The ratio for digoxin BEI values between the two administration modes was 99% [78%, 120%]. In the presence of erythromycin estolate, the two administration modes were deemed equivalent for evaluation of rosuvastatin, digoxin, and metformin accumulation; the ratio of means [90% CI] was 104% [94%, 115%], 94% [82%, 105%], and 100% [88%, 111%], respectively. However, rosuvastatin and digoxin BEI values were low and quite variable in the presence of the inhibitor, so the BEI results were inconclusive. CONCLUSIONS: These data suggest that rosuvastatin and metformin can be administered as a cocktail to evaluate the function of OATP1B1, OATP1B3, BCRP, and OCT1 in SCHH, and that digoxin may not be an ideal component of such a cocktail.


Subject(s)
Cell Culture Techniques , Hepatocytes/metabolism , Membrane Transport Proteins/metabolism , Molecular Probes/chemistry , Biological Transport , Cells, Cultured , Digoxin/administration & dosage , Digoxin/chemistry , Digoxin/metabolism , Erythromycin Estolate/administration & dosage , Erythromycin Estolate/pharmacology , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Metformin/administration & dosage , Metformin/chemistry , Metformin/metabolism , Molecular Probes/administration & dosage , Molecular Probes/metabolism , Rosuvastatin Calcium/administration & dosage , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/metabolism
15.
Article in English | MEDLINE | ID: mdl-31374423

ABSTRACT

Thanks to highly active antiretroviral treatments, HIV infection is now considered as a chronic condition. Consequently, people living with HIV (PLWH) live longer and encounter more age-related chronic co-morbidities, notably cardiovascular diseases, leading to polypharmacy. As the management of drug-drug interactions (DDIs) constitutes a key aspect of the care of PLWH, the magnitude of pharmacokinetic DDIs between cardiovascular and anti-HIV drugs needs to be more thoroughly characterized. To that endeavour, an UHPLC-MS/MS bioanalytical method has been developed for the simultaneous determination in human plasma of amlodipine, metoprolol, pravastatin, rosuvastatin, atorvastatin and its active metabolites. Plasma samples were subjected to protein precipitation with methanol, followed by evaporation at room temperature under nitrogen of the supernatant, allowing to attain measurable plasma concentrations down to sub-nanogram per milliliter levels. Stable isotope-labelled analytes were used as internal standards. The five drugs and two metabolites were analyzed using a 6-min liquid chromatographic run coupled to electrospray triple quadrupole mass spectrometry detection. The method was validated over the clinically relevant concentrations ranging from 0.3 to 480 ng/mL for amlodipine, atorvastatin and p-OH-atorvastatin, and 0.4 to 480 ng/mL for pravastatin, 0.5 to 480 ng/mL for rosuvastatin and o-OH-atorvastatin, and 3 to 4800 ng/mL for metoprolol. Validation performances such as trueness (95.4-110.8%), repeatability (1.5-13.4%) and intermediate precision (3.6-14.5%) were in agreement with current international recommendations. Accuracy profiles (total error approach) were lying within the limits of ±30% accepted in bioanalysis. This rapid and robust UHPLC-MS/MS assay allows the simultaneous quantification in plasma of the major currently used cardiovascular drugs and offers an efficient analytical tool for clinical pharmacokinetics as well as DDIs studies.


Subject(s)
Amlodipine/blood , Atorvastatin/blood , HIV Infections , Metoprolol/blood , Pravastatin/blood , Rosuvastatin Calcium/blood , Amlodipine/chemistry , Amlodipine/metabolism , Amlodipine/pharmacokinetics , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Atorvastatin/chemistry , Atorvastatin/metabolism , Atorvastatin/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Drug Interactions , HIV Infections/drug therapy , HIV Infections/metabolism , Humans , Linear Models , Metoprolol/chemistry , Metoprolol/metabolism , Metoprolol/pharmacokinetics , Pravastatin/chemistry , Pravastatin/metabolism , Pravastatin/pharmacokinetics , Reproducibility of Results , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/metabolism , Rosuvastatin Calcium/pharmacokinetics , Sensitivity and Specificity , Tandem Mass Spectrometry/methods
16.
Int J Nanomedicine ; 14: 4625-4636, 2019.
Article in English | MEDLINE | ID: mdl-31303752

ABSTRACT

PURPOSE: Rosuvastatin calcium (ROSCa) nanoparticles were fabricated by planetary ball mill to enhance ROSCa dissolution rate and bioavailability. METHODS: Milling time factors (milling cycle time and number as well as pause time) were explored. The effect of different milling ball size, speed, and solid-to-solvent ratio were also studied using Box-Behnken factorial design. The fabricated nanoparticles were evaluated in term of physicochemical properties and long-term stability. RESULTS: The obtained data revealed that the integrated formulation and process factors should be monitored to obtain desirable nanoparticle attributes in terms of particle size, zeta potential, dissolution rate, and bioavailability. The optimized ROSCa nanoparticles prepared by milling technique showed a significant enhancement in the dissolution rate by 1.3-fold and the plasma concentration increased by 2-fold (P<0.05). Moreover, stability study showed that the optimized formula of ROSCa nanoparticles exhibits higher stability in long-term stability conditions at 30°C with humidity of 60%. CONCLUSION: Formulation of ROSCa as nanoparticles using milling technique showed a significant enhancement in both dissolution rate and plasma concentration as well as stability compared with untreated drug.


Subject(s)
Nanoparticles/chemistry , Nanotechnology/methods , Rosuvastatin Calcium/chemistry , Animals , Drug Stability , Nanoparticles/ultrastructure , Particle Size , Rabbits , Rosuvastatin Calcium/blood , Rosuvastatin Calcium/pharmacokinetics , Static Electricity , Time Factors
17.
Drug Dev Ind Pharm ; 45(10): 1635-1645, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31342792

ABSTRACT

Objective: The aim of the study was to formulate, cyclodextrin (CD)-polyanhydride (PA) nanoparticles (CPNs) with rosuvastatin calcium (RCa) in order to enhance the poor oral bioavailability. Methods: CPNs containing RCa/CD complexes were prepared by a modified solvent displacement method and morphological analyses, particle size (PS), polydispersity index (PDI), zeta potential (ZP), encapsulation efficiency (EE), DSC, FT-IR, XRD, 1H-NMR analyses were performed. In vitro release properties, release kinetics, cytotoxicity, in vitro permeability and pharmacokinetic studies were also studied. The stability of the formulations were evaluated during the storage period of 3 months. Results: The physicochemical studies showed that the RCa/CD complexes were well incorporated into CPNs resulted in nanosized particles (215.22 and 189.13 nm) with homogenous size distribution (PDI: 0.203 and 0.182) with relatively high incorporation capacity (76.11 and 68.18%) for the CPN1 and CPN2 formulations respectively. Sustained release of RCa from CPNs were achieved. The cytotoxicity values showed that the safety of the formulations. According to permeability studies, pure RCa had lowest permeability data (3.08 × 10-7 cm⋅s-1 Papp value) while CPNs gained higher permeability data (1.36 × 10-5 and 1.12 × 10-5 cm⋅s-1 Papp values) for the CPN1 and CPN2 formulations respectively. CPN2 formulation was selected for pharmacokinetic studies and analyses results demonstrated that approximately 8-fold relative oral bioavailability enhancement compared to the pure RCa was achieved. Conclusion: Considering the analyses results of the study, CPNs can be regarded as suitable, safe, functional oral delivery systems for RCa with enhanced oral bioavailability.


Subject(s)
Cyclodextrins/chemistry , Cyclodextrins/pharmacokinetics , Nanoparticles/chemistry , Polyanhydrides/chemistry , Polyanhydrides/pharmacokinetics , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacokinetics , Animals , Biological Availability , Caco-2 Cells , Cell Line, Tumor , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Drug Carriers/chemistry , Drug Delivery Systems/methods , Humans , Male , Particle Size , Permeability/drug effects , Rats , Rats, Sprague-Dawley
19.
Biomed Chromatogr ; 33(10): e4607, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31141832

ABSTRACT

A simple, precise and accurate HPLC method was developed, optimized and validated for simultaneous determination of rosuvastatin and candesartan in rat plasma using atorvastatin as an internal standard. Solid-phase extraction was used for sample cleanup and its subsequent optimization was carried out to achieve higher extraction efficiency and to eliminate matrix effect. A quality by design approach was used, wherein three-level factorial design was applied for optimization of mobile phase composition and for assessing the effect of pH of the mobile phase using Design Expert Software. Adequate separation for both analytes was achieved with a Waters C18 column (250 × 4.6 mm, 5 µm) using acetonitrile-5 mm sodium acetate buffer (70:30, v/v; pH adjusted to 3.5 with acetic acid) as a mobile phase at a flow rate of 1.0 mL/min and wavelength of 254 nm. The calibration curves were linear over the concentration ranges 5-150 and 10-300 ng/mL for rosuvastatin (ROS) and candesartan (CAN), respectively. The validated method was successfully applied to a pharmacokinetic study in Wistar rats and the data did not reveal any evidence for a potential drug-drug interaction between ROS and CAN. This information provides evidence for clinical rational use of ROS and CAN.


Subject(s)
Benzimidazoles/blood , Chromatography, High Pressure Liquid/methods , Rosuvastatin Calcium/blood , Tetrazoles/blood , Animals , Benzimidazoles/chemistry , Benzimidazoles/pharmacokinetics , Biphenyl Compounds , Linear Models , Male , Rats , Rats, Wistar , Reproducibility of Results , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacokinetics , Sensitivity and Specificity , Solid Phase Extraction , Tetrazoles/chemistry , Tetrazoles/pharmacokinetics
20.
Colloids Surf B Biointerfaces ; 177: 541-549, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30825846

ABSTRACT

The present work entails development of novel phospholipid-based self-nanoemulsifying systems (SNES) of rosuvastatin calcium for improving the oral biopharmaceutical performance via intestinal lymphatic pathways. The phospholipid complex-loaded SNES exhibited emulsification time of 142 s, particle size of 182.5 nm, polydispersity index of 0.35, zeta potential of -22.5 mV and complete in vitro drug release within 3 h. Cell line study on Caco-2 indicated absence of cytotoxicity and excellent cellular uptake of PL-SNES vis-à-vis plain SNES. Permeability study revealed >85% enhancement in the permeation, while intestinal perfusion study showed 2.9 and 3.5-fold increase in the permeation and absorption of the drug from the optimized PL-SNES over the pure drug suspension. Nearly 2.2 and 7.2-folds improvement in AUC0-t and Cmax, and 0.33-fold reduction in the Tmax of drug was observed for PL-SNES vis-à-vis the pure drug suspension during pharmacokinetic study. Moreover, PL-SNES also showed superior antihyperlipidemic activity over the pure drug suspension during pharmacodynamic study. Overall, the developed nanoformulation yielded significant improvement in the oral deliverability of the explored drug candidate.


Subject(s)
Chylomicrons/chemistry , Hyperlipidemias/drug therapy , Nanoparticles/chemistry , Rosuvastatin Calcium/administration & dosage , Rosuvastatin Calcium/therapeutic use , Animals , Caco-2 Cells , Cell Survival/drug effects , Colloids/chemistry , Drug Carriers/chemistry , Humans , Hyperlipidemias/pathology , Male , Rats , Rats, Wistar , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...