Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
PLoS One ; 17(2): e0261925, 2022.
Article de Anglais | MEDLINE | ID: mdl-35143514

RÉSUMÉ

PURPOSE: Vitreous humor is a complex biofluid whose composition determines its structure and function. Vitreous viscosity will affect the delivery, distribution, and half-life of intraocular drugs, and key physiological molecules. The central pig vitreous is thought to closely match human vitreous viscosity. Diffusion is inversely related to viscosity, and diffusion is of fundamental importance for all biochemical reactions. Fluorescence Recovery After Photobleaching (FRAP) may provide a novel means of measuring intravitreal diffusion that could be applied to drugs and physiological macromolecules. It would also provide information about vitreous viscosity, which is relevant to drug elimination, and delivery. METHODS: Vitreous viscosity and intravitreal macromolecular diffusion of fluorescently labelled macromolecules were investigated in porcine eyes using fluorescence recovery after photobleaching (FRAP). Fluorescein isothiocyanate conjugated (FITC) dextrans and ficolls of varying molecular weights (MWs), and FITC-bovine serum albumin (BSA) were employed using FRAP bleach areas of different diameters. RESULTS: The mean (±standard deviation) viscosity of porcine vitreous using dextran, ficoll and BSA were 3.54 ± 1.40, 2.86 ± 1.13 and 4.54 ± 0.13 cP respectively, with an average of 3.65 ± 0.60 cP. CONCLUSIONS: FRAP is a feasible and practical optical method to quantify the diffusion of macromolecules through vitreous.


Sujet(s)
Redistribution de fluorescence après photoblanchiment/méthodes , Corps vitré/métabolisme , Animaux , Bévacizumab/composition chimique , Bévacizumab/métabolisme , Dextrane/composition chimique , Diffusion , Ficoll/composition chimique , Fluorescéine-5-isothiocyanate/analogues et dérivés , Fluorescéine-5-isothiocyanate/composition chimique , Ranibizumab/composition chimique , Ranibizumab/métabolisme , Récepteurs aux facteurs de croissance endothéliale vasculaire/composition chimique , Récepteurs aux facteurs de croissance endothéliale vasculaire/métabolisme , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/métabolisme , Sérumalbumine bovine/composition chimique , Suidae , Viscosité
2.
Adv Drug Deliv Rev ; 126: 113-126, 2018 02 15.
Article de Anglais | MEDLINE | ID: mdl-29288733

RÉSUMÉ

Aqueous gels formulated using hydrophilic polymers (hydrogels) along with those based on stimuli responsive polymers (in situ gelling or gel forming systems) continue to attract increasing interest for various eye health-related applications. They allow the incorporation of a variety of ophthalmic pharmaceuticals to achieve therapeutic levels of drugs and bioactives at target ocular sites. The integration of sophisticated drug delivery technologies such as nanotechnology-based ones with intelligent and environment responsive systems can extend current treatment duration to provide more clinically relevant time courses (weeks and months instead of hours and days) which will inevitably reduce dose frequency, increase patient compliance and improve clinical outcomes. Novel applications and design of contact lenses and intracanalicular delivery devices along with the move towards integrating gels into various drug delivery devices like intraocular pumps, injections and implants has the potential to reduce comorbidities caused by glaucoma, corneal keratopathy, cataract, diabetic retinopathies and age-related macular degeneration. This review describes ophthalmic gelling systems with emphasis on mechanism of gel formation and application in ophthalmology. It provides a critical appraisal of the techniques and methods used in the characterization of ophthalmic preformed gels and in situ gelling systems along with a thorough insight into the safety and biocompatibility of these systems. Newly developed ophthalmic gels, hydrogels, preformed gels and in situ gelling systems including the latest in the area of stimuli responsive gels, molecularly imprinted gels, nanogels, 3D printed hydrogels; 3D printed devices comprising ophthalmic gels are covered. Finally, new applications of gels in the production of artificial corneas, corneal wound healing and hydrogel contact lenses are described.


Sujet(s)
Solutions ophtalmiques/pharmacologie , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments , Gels/pharmacologie , Humains , Polymères/pharmacologie , Impression tridimensionnelle
3.
Mol Pharm ; 13(9): 2923-40, 2016 09 06.
Article de Anglais | MEDLINE | ID: mdl-27286558

RÉSUMÉ

Age-related macular degeneration (AMD) is the leading cause of certified vision loss worldwide. The standard treatment for neovascular AMD involves repeated intravitreal injections of therapeutic proteins directed against vascular endothelial growth factor, such as ranibizumab. Biodegradable polymers, such as poly(lactic-co-glycolic acid) (PLGA), form delivery vehicles which can be used to treat posterior segment eye diseases, but suffer from poor protein loading and release. This work describes a "system-within-system", PLGA microparticles incorporating chitosan-based nanoparticles, for improved loading and sustained intravitreal delivery of ranibizumab. Chitosan-N-acetyl-l-cysteine (CNAC) was synthesized and its synthesis confirmed using FT-IR and (1)H NMR. Chitosan-based nanoparticles composed of CNAC, CNAC/tripolyphosphate (CNAC/TPP), chitosan, chitosan/TPP (chit/TPP), or chit/TPP-hyaluronic acid (chit/TPP-HA) were incorporated in PLGA microparticles using a modified w/o/w double emulsion method. Nanoparticles and final nanoparticles-within-microparticles were characterized for their protein-nanoparticle interaction, size, zeta potential, morphology, protein loading, stability, in vitro release, in vivo antiangiogenic activity, and effects on cell viability. The prepared nanoparticles were 17-350 nm in size and had zeta potentials of -1.4 to +12 mV. Microscopic imaging revealed spherical nanoparticles on the surface of PLGA microparticles for preparations containing chit/TPP, CNAC, and CNAC/TPP. Ranibizumab entrapment efficiency in the preparations varied between 13 and 69% and was highest for the PLGA microparticles containing CNAC nanoparticles. This preparation also showed the slowest release with no initial burst release compared to all other preparations. Incorporation of TPP to this formulation increased the rate of protein release and reduced entrapment efficiency. PLGA microparticles containing chit/TPP-HA showed the fastest and near-complete release of ranibizumab. All of the prepared empty particles showed no effect on cell viability up to a concentration of 12.5 mg/mL. Ranibizumab released from all preparations maintained its structural integrity and in vitro activity. The chit/TPP-HA preparation enhanced antiangiogenic activity and may provide a potential biocompatible platform for enhanced antiangiogenic activity in combination with ranibizumab. In conclusion, the PLGA microparticles containing CNAC nanoparticles showed significantly improved ranibizumab loading and release profile. This novel drug delivery system may have potential for improved intravitreal delivery of therapeutic proteins, thereby reducing the frequency, risk, and cost of burdensome intravitreal injections.


Sujet(s)
Chitosane/composition chimique , Acide lactique/composition chimique , Nanoparticules/composition chimique , Acide polyglycolique/composition chimique , Ranibizumab/administration et posologie , Ranibizumab/composition chimique , Acétylcystéine/composition chimique , Lignée cellulaire , Mouvement cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Chitosane/analogues et dérivés , Systèmes de délivrance de médicaments/méthodes , Cellules endothéliales de la veine ombilicale humaine , Humains , Acide hyaluronique/composition chimique , Dégénérescence maculaire/traitement médicamenteux , Spectroscopie par résonance magnétique , Microscopie électronique à balayage , Microscopie électronique à transmission , Copolymère d'acide poly(lactique-co-glycolique) , Polyphosphates/composition chimique , Ranibizumab/usage thérapeutique , Spectroscopie infrarouge à transformée de Fourier
4.
Exp Eye Res ; 121: 121-9, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24530465

RÉSUMÉ

The aim of this study was to prepare and characterize cholesterol-poly(ethylene) glycol (chol-PEG) nanocarriers of two different molecular weights (1 and 5 kDa) and to determine their effect on the transscleral retention and permeation of a lipophilic multi-therapeutic agent, sirolimus (rapamycin), with potential application in angiogenic and immunogenic ocular diseases. Sirolimus-containing nanocarriers were prepared using the thin-film hydration method and characterized for their physicochemical properties including size, drug entrapment (EE) and loading (DL) efficiencies, stability, surface charge, morphology, critical micelle concentration (CMC) and thermal properties. Ussing chambers were used to determine the retention and permeability of sirolimus-containing nanocarriers in porcine sclera followed by ultrastructural tissue examination. Sirolimus-containing nanocarriers had an average size of 11.7 nm (chol-PEG 1 kDa) and 13.8 nm (chol-PEG 5 kDa) and zeta potentials of 0.41 and -1.05, respectively. Both nanocarriers had similar transscleral permeabilities (chol-PEG 1 kDa 6.44 × 10(-7) and 5 kDa 6.16 × 10(-7) cm2 s(-1)), and very high scleral retention compared with a free solution of sirolimus (chol-PEG 1 kDa 16.9 µg/g; chol-PEG 5 kDa 7.48 µg/g; free sirolimus 0.57 µg/g). The DL (EE) for chol-PEG 1 and 5 kDa were 2.93% (77.4%) and 3.10% (81.6%), respectively. The CMC values for the nanocarriers were similar to those previously reported in literature (3.85 × 10(-7) M for chol-PEG 1 kDa; 4.26 × 10(-7) M for chol-PEG 5 kDa). In conclusion, chol-PEG nanocarriers successfully loaded sirolimus and resulted in scleral permeation and high retention, which shows potential utility for the topical delivery of lipophilic ocular drugs.


Sujet(s)
Antibactériens/administration et posologie , Cholestérol/analogues et dérivés , Vecteurs de médicaments/composition chimique , Nanoparticules/composition chimique , Polyéthylène glycols/composition chimique , Sclère/métabolisme , Sirolimus/administration et posologie , Animaux , Antibactériens/pharmacocinétique , Calorimétrie différentielle à balayage , Chimie pharmaceutique , Cholestérol/composition chimique , Stabilité de médicament , Micelles , Microscopie électronique à balayage , Nanoparticules/ultrastructure , Taille de particule , Perméabilité , Préparations pharmaceutiques , Diffraction sur poudre , Sclère/ultrastructure , Sirolimus/pharmacocinétique , Suidae
5.
Int J Pharm ; 456(2): 400-7, 2013 Nov 18.
Article de Anglais | MEDLINE | ID: mdl-24012860

RÉSUMÉ

Iron (Fe) loaded solid lipid nanoparticles (SLN's) were formulated using stearic acid and iron absorption was evaluated in vitro using the cell line Caco-2 with intracellular ferritin formation as a marker of iron absorption. Iron loading was optimised at 1% Fe (w/w) lipid since an inverse relation was observed between initial iron concentration and SLN iron incorporation efficiency. Chitosan (Chi) was included to prepare chitosan coated SLN's. Particle size analysis revealed a sub-micron size range (300.3±31.75 nm to 495.1±80.42 nm), with chitosan containing particles having the largest dimensions. As expected, chitosan (0.1%, 0.2% and 0.4% w/v) conferred a net positive charge on the particle surface in a concentration dependent manner. For iron absorption experiments equal doses of Fe (20 µM) from selected formulations (SLN-FeA and SLN-Fe-ChiB) were added to Caco-2 cells and intracellular ferritin protein concentrations determined. Caco-2 iron absorption from SLN-FeA (583.98±40.83 ng/mg cell protein) and chitosan containing SLN-Fe-ChiB (642.77±29.37 ng/mg cell protein) were 13.42% and 24.9% greater than that from ferrous sulphate (FeSO4) reference (514.66±20.43 ng/mg cell protein) (p≤0.05). We demonstrate for the first time preparation, characterisation and superior iron absorption in vitro from SLN's, suggesting the potential of these formulations as a novel system for oral iron delivery.


Sujet(s)
Systèmes de délivrance de médicaments/méthodes , Composés du fer II/administration et posologie , Fer/administration et posologie , Lipides/administration et posologie , Nanoparticules/administration et posologie , Administration par voie orale , Cellules Caco-2 , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/physiologie , Composés du fer II/composition chimique , Humains , Fer/composition chimique , Lipides/composition chimique , Nanoparticules/composition chimique
6.
Invest Ophthalmol Vis Sci ; 53(13): 8105-11, 2012 Dec 13.
Article de Anglais | MEDLINE | ID: mdl-23049091

RÉSUMÉ

PURPOSE: We explored the potential of an amphiphilic chitosan derivative to facilitate the transscleral delivery of rapamycin, a potential multitherapeutic agent with poor water solubility. METHODS: The amphiphilic chitosan derivative, O-octanoyl-chitosan-polyethylene glycol (OChiPEG) graft copolymer, was analyzed using Fourier-transform infrared spectroscopy (FT-IR). OChiPEG micelles were prepared via the thin film method and characterized for their size using dynamic light scattering (DLS), zeta potential using laser Doppler velocimetry (LDV), morphology using transmission electron microscopy (TEM), drug entrapment efficiency (EE), and drug loading (DL) efficiency using reversed-phase high performance liquid chromatography (RP-HPLC), critical micelle concentration (CMC) using spectrofluorometry, and thermal properties using differential scanning calorimetry (DSC) and x-ray powder diffraction (XRPD). Scleral permeation and retention of rapamycin from the drug-loaded micelles were determined in porcine sclera clamped in Ussing chambers, using RP-HPLC. RESULTS: Conjugation of hydrophilic and hydrophobic groups to chitosan was confirmed using FT-IR. Rapamycin-loaded micelles of particle size 40.6 nm and zeta potential + 6.84 mV were prepared successfully. These carriers exhibited a high EE and DL of 85.6 and 16.3%, respectively, and a CMC of 16.6 µM. OChiPEG micelles showed a high rapamycin scleral retention (14.8 ± 0.81 µg/g) with successful transscleral permeation (5.57 ± 1.04 × 10(-8) cm(2) · s(-1)). CONCLUSIONS: Positively charged OChiPEG micelles loaded with rapamycin were prepared successfully. These showed a high scleral retention and successful permeation of rapamycin, and therefore may be useful for the topical delivery of other hydrophobic agents.


Sujet(s)
Antibactériens/administration et posologie , Matériaux biocompatibles/composition chimique , Chitosane/analogues et dérivés , Systèmes de délivrance de médicaments , Polyéthylène glycols/composition chimique , Sclère/effets des médicaments et des substances chimiques , Sirolimus/administration et posologie , Tensioactifs/composition chimique , Animaux , Calorimétrie différentielle à balayage , Chitosane/composition chimique , Chromatographie en phase liquide à haute performance , Fluxmétrie laser Doppler , Lumière , Micelles , Diffusion de rayonnements , Spectrométrie de fluorescence , Spectroscopie infrarouge à transformée de Fourier , Sus scrofa , Diffraction des rayons X
7.
Exp Eye Res ; 102: 85-92, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-22846670

RÉSUMÉ

The purpose of this study was to investigate the influence of molecular shape, conformability, net surface charge and tissue interaction on transscleral diffusion. Unfixed, porcine sclera was clamped in an Ussing chamber. Fluorophore-labelled neutral albumin, neutral dextran, or neutral ficoll were placed in one hemi-chamber and the rate of transscleral diffusion was measured over 24 h using a spectrophotometer. Experiments were repeated using dextrans and ficoll with positive or negative net surface charges. Fluorescence recovery after photobleaching (FRAP) was undertaken to compare transscleral diffusion with diffusion through a solution. All molecules were 70 kDa. With FRAP, the diffusion coefficient (D) of neutral molecules was highest for albumin, followed by ficoll, then dextran (p < 0.0001). Positive dextrans diffused fastest, followed by negative, then neutral dextrans (p = 0.0004). Neutral ficoll diffused the fastest, followed by positive then negative ficoll (p = 0.5865). For the neutral molecules, transscleral D was highest for albumin, followed by dextran, then ficoll (p < 0.0001). D was highest for negative ficoll, followed by neutral, then positive ficoll (p < 0.0001). By contrast, D was highest for positive dextran, followed by neutral, then negative dextran (p = 0.0021). In conclusion, diffusion in free solution does not predict transscleral diffusion and the molecular-tissue interaction is important. Molecular size, shape, and charge may all markedly influence transscleral diffusion, as may conformability to a lesser degree, but their effects may be diametrically opposed in different molecules, and their influence on diffusion is more complex than previously thought. Each variable cannot be considered in isolation, and the interplay of all these variables needs to be tested, when selecting or designing drugs for transscleral delivery.


Sujet(s)
Dextrane/pharmacocinétique , Ficoll/analogues et dérivés , Fluorescéine-5-isothiocyanate/analogues et dérivés , Complexes multiprotéiques/pharmacocinétique , Sclère/métabolisme , Sérumalbumine/pharmacocinétique , Animaux , Dextrane/composition chimique , Diffusion , Chambres de culture à diffusion , Ficoll/composition chimique , Ficoll/pharmacocinétique , Fluorescéine-5-isothiocyanate/composition chimique , Fluorescéine-5-isothiocyanate/pharmacocinétique , Fluxmétrie laser Doppler , Lumière , Masse moléculaire , Complexes multiprotéiques/composition chimique , Perméabilité , Conformation des protéines , Diffusion de rayonnements , Sérumalbumine/composition chimique , Spectrométrie de fluorescence , Suidae
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE