Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Exp Eye Res ; 235: 109638, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37657528

RESUMEN

Although mouse models are widely used in retinal drug development, pharmacokinetics in mouse eye is poorly understood. In this study, we applied non-invasive in vivo fluorophotometry to study pharmacokinetics of intravitreal fluorescein sodium (molecular weight 0.38 kDa) and fluorescein isothiocyanate-dextran (FD-150; molecular weight 150 kDa) in mice. Intravitreal half-lives of fluorescein and FD-150 in mouse eyes were 0.53 ± 0.06 h and 2.61 ± 0.86 h, respectively. These values are 8-230 times shorter than the elimination half-lives of similar compounds in the human vitreous. The apparent volumes of distribution in the mouse vitreous were close to the anatomical volume of the mouse vitreous (FD-150, 5.1 µl; fluorescein, 9.6 µl). Dose scaling factors were calculated from mouse to rat, rabbit, monkey and human translation. Based on pharmacokinetic modelling and compound concentrations in the vitreous and anterior chamber, fluorescein is mainly eliminated posteriorly across blood-retina barrier, but FD-150 is cleared via aqueous humour outflow. The results of this study improve the knowledge of intravitreal pharmacokinetics in mouse and facilitate inter-species scaling in ocular drug development.


Asunto(s)
Retina , Cuerpo Vítreo , Ratones , Ratas , Humanos , Animales , Conejos , Barrera Hematorretinal , Fluoresceína , Cámara Anterior , Inyecciones Intravítreas
2.
Exp Eye Res ; 224: 109237, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36096189

RESUMEN

Pathological angiogenesis related to neovascularization in the eye is mediated through vascular endothelial growth factors (VEGFs) and their receptors. Ocular neovascular-related diseases are mainly treated with anti-VEGF agents. In this study we evaluated the efficacy and safety of novel gene therapy using adeno associated virus 2 vector expressing a truncated form of soluble VEGF receptor-2 fused to the Fc-part of human IgG1 (AAV2-sVEGFR-2-Fc) to inhibit ocular neovascularization in laser induced choroidal neovascularization (CNV) in mice. The biological activity of sVEGFR-2-Fc was determined in vitro. It was shown that sVEGFR-2-Fc secreted from ARPE-19 cells was able to bind to VEGF-A165 and reduce VEGF-A165 induced cell growth and survival. A single intravitreal injection (IVT) of AAV2-sVEGFR-2-Fc (1 µl, 4.7 × 1012 vg/ml) one-month prior laser photocoagulation did not cause any changes in the retinal morphology and significantly suppressed fluorescein leakage at 7, 14, 21 and 28 days post-lasering compared to controls. Macrophage infiltration was observed after the injection of both AAV2-sVEGFR-2-Fc and PBS. Our findings indicate that AAV2 mediated gene delivery of the sVEGFR-2-Fc efficiently reduces formation of CNV and could be developed to a therapeutic tool for the treatment of retinal diseases associated with neovascularization.


Asunto(s)
Neovascularización Coroidal , Ratones , Humanos , Animales , Neovascularización Coroidal/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inyecciones Intravítreas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Dependovirus/genética , Vectores Genéticos , Ratones Endogámicos C57BL , Terapia Genética , Inhibidores de la Angiogénesis/uso terapéutico , Factores de Crecimiento Endotelial Vascular/genética , Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/uso terapéutico , Inmunoglobulina G/metabolismo , Fluoresceínas/metabolismo
3.
Mol Pharm ; 17(6): 1945-1953, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32320251

RESUMEN

Because of poor ocular drug bioavailability, intravitreal injections have become the gold standard for drug delivery to the posterior eye. The prodrug approach can be used for optimizing the biopharmaceutical properties of intravitreal drugs. The preclinical screening of prodrugs' properties, such as hydrolysis and bioconversion, should be conducted in a resource-efficient way for an extensive set of synthesized compounds with validated methods. Our objective was to explore cassette dosing in in vitro prodrug hydrolysis and bioconversion studies in buffer, vitreous, and retinal pigment epithelium (RPE) homogenate for rapid medium-throughput screening. Moreover, our aim was to correlate the prodrug structure with hydrolytic behavior. We synthesized 18 novel ganciclovir prodrugs and first studied their hydrolysis in aqueous buffer and porcine vitreous in vitro with cassette dosing for 35 h. A method for vitreous homogenate pH equilibration to a physiological level by using buffer and incubation under 5% carbon dioxide was validated. The hydrolysis of the prodrugs was evaluated in porcine RPE homogenate in vitro with cassette dosing, and five prodrugs were assayed individually to examine their bioconversion into ganciclovir in RPE after 2 h. Lastly, the prodrugs' binding to melanin was studied in vitro. The prodrugs showed a wide spectrum of hydrolysis rates, ranging from a few percentages to 100% in the vitreous and RPE; in general, hydrolysis in RPE was faster than in vitreous. Prodrugs with long carbon chains and disubstitution showed lability in the tissue homogenates, whereas prodrugs with branched carbon chains and aromatic groups were stable. All five prodrugs chosen for the bioconversion study in RPE were hydrolyzed into ganciclovir, and their hydrolytic behavior matched results from the cassette mix experiment, supporting the cassette mix approach for hydrolysis and bioconversion studies. None of the prodrugs bound highly to melanin (<50% bound). In conclusion, cassette dosing proved useful for the rapid screening of prodrug hydrolysis and bioconversion properties. Analyzing several compounds simultaneously can complicate the analytics, and thus, choosing the compounds of the cassette mix should be done carefully to avoid mutual interference of the compounds with the results. The methodology and results of the work are applicable in ocular drug research and prodrug design.


Asunto(s)
Ganciclovir/química , Epitelio Pigmentado de la Retina/metabolismo , Animales , Antivirales/química , Sistemas de Liberación de Medicamentos/métodos , Profármacos/química , Porcinos , Espectrometría de Masas en Tándem
4.
Mol Pharm ; 16(9): 3968-3976, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31348666

RESUMEN

Lens is the avascular tissue in the eye between the aqueous humor and vitreous. Drug binding to the lens might affect ocular pharmacokinetics, and the binding may also have a pharmacological role in drug-induced cataract and cataract treatment. Drug distribution in the lens has been studied in vitro with many compounds; however, the experimental methods vary, no detailed information on distribution between the lens sublayers exist, and the partition coefficients are reported rarely. Therefore, our objectives were to clarify drug localization in the lens layers and establish partition coefficients for a wide range of molecules. Furthermore, we aimed to illustrate the effect of lenticular drug binding on overall ocular drug pharmacokinetics. We studied the distribution of 16 drugs and three fluorescent dyes in whole porcine lenses in vitro with imaging mass spectrometry and fluorescence microscopy techniques. Furthermore, we determined lens/buffer partition coefficients with the same experimental setup for 28 drugs with mass spectrometry. Finally, the effect of lenticular binding of drugs on aqueous humor drug exposure was explored with pharmacokinetic simulations. After 4 h, the drugs and the dyes distributed only to the outermost lens layers (capsule and cortex). The lens/buffer partition coefficients for the drugs were low, ranging from 0.05 to 0.8. On the basis of the pharmacokinetic simulations, a high lens-aqueous humor partition coefficient increases drug exposure in the lens but does not significantly alter the pharmacokinetics in the aqueous humor. To conclude, the lens seems to act mainly as a physical barrier for drug distribution in the eye, and drug binding to the lens affects mainly the drug pharmacokinetics in the lens.


Asunto(s)
Colorantes Fluorescentes/farmacocinética , Cristalino/efectos de los fármacos , Absorción Ocular/fisiología , Preparaciones Farmacéuticas/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Animales , Humor Acuoso/efectos de los fármacos , Humor Acuoso/metabolismo , Área Bajo la Curva , Tampones (Química) , Colorantes Fluorescentes/química , Cristalino/metabolismo , Microscopía Fluorescente , Peso Molecular , Absorción Ocular/efectos de los fármacos , Concentración Osmolar , Preparaciones Farmacéuticas/química , Porcinos , Distribución Tisular , Cuerpo Vítreo/efectos de los fármacos , Cuerpo Vítreo/metabolismo
5.
Bioorg Med Chem ; 27(21): 115086, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31515057

RESUMEN

Four inhibitors of human carbonic anhydrase II (hCA II) were designed based on the previously reported subnanomolar 1,3-oxazole-based sulfonamide inhibitors of the enzyme to incorporate primary and secondary amine functionality in the carboxamide side chain. The new hydrophilic compounds were found to inhibit the target isoform in sub-nanomolar to low nanomolar range with a good degree of selectivity to several other hCA isoforms. The hydrophilic character of these compounds is advantageous for intraocular residence time but not for corneal permeability which generally requires that a drug be sufficiently lipophilic. Two of the four compounds investigated, however, were found to exert comparable efficacy as 1% eye drops in PBS to that of the clinically used 2% dorzolamide (Trusopt®) eye drops. This indicated that the absorption of the compounds may occur via alternative route across conjunctiva and sclera.


Asunto(s)
Anhidrasa Carbónica II/antagonistas & inhibidores , Inhibidores de Anhidrasa Carbónica/farmacología , Presión Intraocular/efectos de los fármacos , Oxazoles/farmacología , Sulfonamidas/farmacología , Animales , Anhidrasa Carbónica II/metabolismo , Inhibidores de Anhidrasa Carbónica/síntesis química , Inhibidores de Anhidrasa Carbónica/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Simulación del Acoplamiento Molecular , Oxazoles/síntesis química , Oxazoles/metabolismo , Unión Proteica , Conejos , Sulfonamidas/síntesis química , Sulfonamidas/metabolismo , Porcinos
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 296-306, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29107807

RESUMEN

The RNA-binding protein, HuR, modulates mRNA processing and gene expression of several stress response proteins i.e. Hsp70 and p53 that have been postulated to be involved in the pathogenesis of glaucoma, a chronic optic neuropathy leading to irreversible blindness. We evaluated HuR protein expression in retinas and optic nerves of glaucomatous rats and human primary open angle glaucoma patients and its possible impact on stress response mechanisms. We found that the cytoplasmic content of HuR was reduced more extensively in glaucomatous retinas than in optic nerves and this was linked with a declined cytoplasmic Hsp70 level and p53 nuclear translocation. In the optic nerve, the p53 content was decreased as a feature of reactive gliosis. Based on our findings, we conclude that the alteration in the HuR content, observed both in rat glaucoma model and human glaucoma samples, affects post-transcriptionally the expression of genes crucial for maintaining cell homeostasis; therefore, we postulate that HuR may be involved in the pathogenesis of glaucoma.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , Hipertensión Ocular/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Proteína 1 Similar a ELAV/genética , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/metabolismo , Glaucoma de Ángulo Abierto/patología , Humanos , Presión Intraocular/genética , Masculino , Neuroprotección/genética , Hipertensión Ocular/genética , Hipertensión Ocular/patología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Ratas , Ratas Wistar , Células Ganglionares de la Retina/patología , Distribución Tisular
7.
Pharm Res ; 36(1): 17, 2018 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-30488131

RESUMEN

PURPOSE: The study aim was to evaluate the effect of Alzheimer's disease (AD) and inflammatory insult on the function of L-type amino acid transporter 1 (Lat1) at the mouse blood-brain barrier (BBB) as well as Lat1 function and expression in mouse primary astrocytes. METHODS: The Lat1 function and expression was determined in wildtype astrocytes with and without lipopolysaccharide (LPS)-induced inflammation and in LPS treated AD APP/PS1 transgenic astrocytes. The function of Lat1 at the BBB was evaluated in wildtype mice with and without LPS-induced neuroinflammation and APP/PS1 transgenic mice by in situ brain perfusion. RESULTS: There were 2.1 and 1.6 -fold decreases in Lat1 mRNA and protein expression in LPS-treated wildtype astrocytes compared to vehicle-treated astrocytes. In contrast, Lat1 mRNA and protein expression were increased by 1.7 and 1.2 -fold (not statistically significant) in the transgenic cells. A similar trend was observed in the cell uptake of [14C]-L-leucine. There were no statistically significant differences in [14C]-L-leucine BBB permeation between the groups. CONCLUSIONS: The results showed that neither LPS-induced inflammation or the presence of APP/PS1 mutations alters Lat1 function at the mouse BBB as well as Lat1 protein expression and function in mouse primary astrocytes.


Asunto(s)
Enfermedad de Alzheimer/patología , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Encefalitis/patología , Transportador de Aminoácidos Neutros Grandes 1/fisiología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Astrocitos/patología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Imidazoles/farmacología , Transportador de Aminoácidos Neutros Grandes 1/genética , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Presenilina-1/genética , Cultivo Primario de Células , Piridinas/farmacología , ARN Mensajero/metabolismo
8.
Pharm Res ; 34(12): 2652-2662, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28952054

RESUMEN

PURPOSE: Alzheimer's disease (AD) may disturb functions of the blood-brain barrier and change the disposition of drugs to the brain. This study assessed the disease-induced changes in drug transporters in the brain capillaries of transgenic AD mice. METHODS: Eighteen drug transporters and four tight junction-associated proteins were analyzed by RT-qPCR in cortex, hippocampus and cerebellum tissue samples of 12-16-month-old APdE9, Tg2576 and APP/PS1 transgenic mice and their healthy age-matched controls. In addition, microvessel fractions enriched from 1-3-month-old APdE9 mice were analyzed using RT-qPCR and Western blotting. Brain transport of methotrexate in APdE9 mice was assessed by in vivo microdialysis. RESULTS: The expression profiles of studied genes were similar in brain tissues of AD and control mice. Instead, in the microvessel fraction in APdE9 mice, >2-fold alterations were detected in the expressions of 11 genes but none at the protein level. In control mice strains, >5-fold changes between different brain regions were identified for Slc15a2, Slc22a3 and occludin. Methotrexate distribution into hippocampus of APdE9 mice was faster than in controls. CONCLUSIONS: The expression profile of mice carrying presenilin and amyloid precursor protein mutations is comparable to controls, but clear regional differences exist in the expression of drug transporters in brain.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Inhibidores Enzimáticos/farmacocinética , Proteínas de Transporte de Membrana/metabolismo , Metotrexato/farmacocinética , Proteínas de Uniones Estrechas/metabolismo , Enfermedad de Alzheimer/genética , Animales , Transporte Biológico , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Descubrimiento de Drogas , Inhibidores Enzimáticos/metabolismo , Humanos , Masculino , Proteínas de Transporte de Membrana/análisis , Proteínas de Transporte de Membrana/genética , Metotrexato/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Preparaciones Farmacéuticas/metabolismo , Proteínas de Uniones Estrechas/análisis , Proteínas de Uniones Estrechas/genética , Transcriptoma
9.
Mol Pharm ; 13(4): 1358-65, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-26950248

RESUMEN

Nanomedicines are widely studied for intracellular delivery of cancer drugs. However, the relationship between intracellular drug concentrations and drug responses are poorly understood. In this study, cellular and nuclear concentrations of doxorubicin were quantified with LC/MS after cell exposure with free and liposomal doxorubicin (pH-sensitive and pegylated liposomes). Cellular uptake of pegylated liposomes was low (∼3-fold extracellular concentrations) compared with doxorubicin in free form and pH-sensitive liposomes (up to 280-fold extracellular concentrations) in rat glioma (BT4C) and renal clear cell carcinoma (Caki-2) cells. However, after the cell exposure with pegylated liposomes, intracellular doxorubicin was distributed into the nuclear compartment in both cell types. Despite high drug concentrations in the nuclei, Caki-2 cells showed strong resistance toward doxorubicin. A model was successfully built to describe PK/PD relationship between drug concentrations in nucleus and cytotoxic responses in BT4C cells. This model is the first step to link target site concentration of doxorubicin into its effect and can be a useful part of more comprehensive future in vivo PK/PD models.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/farmacocinética , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacología , Doxorrubicina/farmacocinética , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Polietilenglicoles/farmacocinética , Polietilenglicoles/farmacología , Ratas
10.
Exp Cell Res ; 332(1): 89-101, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25597427

RESUMEN

Understanding mechanisms of endocytosis and trafficking of nanoparticles through endothelial and epithelial barriers leads potentially to improved efficacy of nanoparticulate drug delivery systems. Detailed characterizations of cell models with respect to endocytic pathway expression and activity (endocytic profiling) should facilitate data interpretation. We performed endocytic profiling of CaCo-2 and hCMEC/D3 cell lines, widely used as human intestinal and blood-brain barrier permeability models, respectively, during cell differentiation. Furthermore, we compared endocytic profiles of cell lines with those of primary cells. Expression of genes involved in specific endocytic pathways was analyzed at mRNA levels by quantitative real time PCR. Where possible, the respective protein levels were analyzed by Western blotting, and endocytic activities of cells were analyzed by flow cytometry. We showed that differentiated CaCo-2 cells formed tight, well polarized monolayers with reduced endocytic activity accompanied by reduced mRNA expression of most of the endocytosis-related genes. In contrast, hCMEC/D3 cells formed a leaky, less polarized barrier, and in vitro differentiation had little effect on either the expression of endocytosis-related genes or endocytic activity of these cells. Endocytic profiling of in vitro models and comparison with primary cells is an important measure to avoid misleading conclusions in nanoparticle permeation studies.


Asunto(s)
Endocitosis , Células Endoteliales/fisiología , Células Epiteliales/fisiología , Barrera Hematoencefálica/metabolismo , Células CACO-2 , Permeabilidad Capilar , Caveolina 1/genética , Caveolina 1/metabolismo , Diferenciación Celular , Polaridad Celular , Células Cultivadas , Impedancia Eléctrica , Expresión Génica , Humanos , Nanopartículas/metabolismo
11.
Xenobiotica ; 44(3): 283-94, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23924297

RESUMEN

1. Currently available in vitro blood-brain barrier models all have recognized restrictions. In addition to leakiness, inconsistent data about P-glycoprotein mediated efflux limit the attractiveness of the primary bovine brain microvessel endothelial cells (BBMECs). Therefore, we re-evaluated the role of P-glycoprotein mediated efflux with two culture conditions in BBMECs for prediction of drug permeability of potential P-glycoprotein substrates. 2. BBMECs were monocultured on filters on petri dishes and on filter inserts, and expression and localization of P-glycoprotein were compared by using western blot and confocal microscopy, respectively. The functionality of P-glycoprotein was assessed by using cellular uptake, calcein-AM and bidirectional transport assays. 3. P-glycoprotein expression was higher in BBMECs cultured on filter inserts decreasing the permeability of digoxin and paclitaxel, but not the permeability of vinblastine. However, the monocultured BBMECs were not able to demonstrate efflux in the bidirectional transport assays. Under certain culture conditions, occludin may not be correctly located, perhaps explaining in part the leakiness of BBMECs. 4. In conclusion, BBMECs, despite possessing a functional P-glycoprotein, under certain culture conditions may not be a suitable in vitro model for the bidirectional transport assays and for predicting the permeability of drugs and xenobiotics that are potential P-glycoprotein substrates.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/farmacocinética , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Células Endoteliales/metabolismo , Microvasos/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Western Blotting , Bovinos , Células Cultivadas , Procesamiento de Imagen Asistido por Computador , Técnicas In Vitro , Modelos Biológicos , Permeabilidad
12.
Eur J Pharm Biopharm ; 198: 114260, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38484852

RESUMEN

Even though subconjunctival injections are used in clinics, their quantitative pharmacokinetics has not been studied systematically. For this purpose, we evaluated the ocular and plasma pharmacokinetics of subconjunctival dexamethasone in rabbits. Intravenous injection was also given to enable a better understanding of the systemic pharmacokinetics. Dexamethasone concentrations in plasma (after subconjunctival and intravenous injections) and four ocular tissues (iris-ciliary body, aqueous humour, neural retina and vitreous) were analysed using LC-MS/MS. Population pharmacokinetic modelling for plasma data from both injection routes were used, and for first time the constant rate of absorption of dexamethasone from the subconjunctival space into plasma was estimated (ka,plasma = 0.043 min-1, i.e. absorption half-life of 17.3 min). Non-compartmental analysis was used for the ocular data analysis and resulting in ocular drug exposure of iris-ciliary body (AUC0-∞= 41984 min·ng/g) > neural retina (AUC0-∞= 25511 min·ng/g) > vitreous (AUC0-∞= 7319 min·ng/mL) > aqueous humour (AUC0-∞= 6146 min·ng/mL). The absolute bioavailability values after subconjunctival injection, reported for the first time, were 0.74 % in aqueous humour (comparable to topical dexamethasone suspensions), and 0.30 % in vitreous humour (estimated to be higher than in topical administration). These novel and comprehensive pharmacokinetic data provide valuable information on the potential for exploiting this route in ocular drug development for treating both, anterior and posterior segment ocular diseases. Moreover, the new generated dexamethasone-parameters are a step-forward in building predictive pharmacokinetic models to support the design of new subconjunctival dexamethasone formulations, which may sustain drug effect for longer period of time.


Asunto(s)
Espectrometría de Masas en Tándem , Cuerpo Vítreo , Animales , Conejos , Inyecciones Intravenosas , Cromatografía Liquida , Dexametasona
13.
Eur J Pharm Biopharm ; 184: 181-188, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36740104

RESUMEN

The retinal pigment epithelial (RPE) cell monolayer forms the outer blood-retinal barrier and has a crucial role in ocular pharmacokinetics. Although several RPE cell models are available, there have been no systematic comparisons of their barrier properties with respect to drug permeability. We compared the barrier properties of RPE secondary cell lines (ARPE19, and ARPE19mel) and both primary (hfRPE) and stem-cell derived RPE (hESC-RPE) cells by investigating the permeability of nine drugs (aztreonam, ciprofloxacin, dexamethasone, fluconazole, ganciclovir, ketorolac, methotrexate, voriconazole, and quinidine) across cell monolayers. ARPE19, ARPE19mel, and hfRPE cells displayed a narrow Papp value range, with relatively high permeation rates (5.2-26 × 10-6 cm/s). In contrast, hESC-RPE cells efficiently restricted the drug flux, and displayed even lower Papp values than those reported for bovine RPE-choroid, with the range of 0.4-32 cm-6/s. Therefore, ARPE19, ARPE19mel, and hfRPE cells failed to form a tight barrier, whereas hESC-RPE cells restricted the drug flux to a similar extent as bovine RPE-choroid. Therefore, hESC-RPE cells are valuable tools in ocular drug discovery.


Asunto(s)
Barrera Hematorretinal , Epitelio Pigmentado de la Retina , Humanos , Animales , Bovinos , Barrera Hematorretinal/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Células Madre , Coroides , Células Cultivadas
14.
Eur J Pharm Sci ; 181: 106366, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36565891

RESUMEN

Fosamprenavir is a phosphate ester prodrug that, upon dissolution, is cleaved to the poorly soluble yet readily absorbable parent drug amprenavir. In this study, a novel cell-free in vitro setup with quasi-continuous monitoring of the dynamic dissolution/bio-conversion/permeation of fosamprenavir was designed and tested. It consists of side-by-side diffusion cells, where the donor and acceptor compartments are separated by the biomimetic barrier PermeaPad®, and sampling from the donor compartment is accomplished via a microdialysis probe. Externally added bovine alkaline phosphatase induced bioconversion in the donor compartment. Microdialysis sampling allowed to follow the enzymatic conversion of fosamprenavir to amprenavir by the bovine alkaline phosphatase in an (almost) real-time manner eliminating the need to remove or inactivate the enzyme. Biomimetic conversion rates in the setup were established by adding appropriate amounts of the alkaline phosphatase. A substantial (6.5-fold) and persistent supersaturation of amprenavir was observed due to bioconversion at lower (500 µM) concentrations, resulting in a substantially increased flux across the biomimetic barrier, nicely reflecting the situation in vivo. At conditions with an almost 10-fold higher dose than the usual human dose, some replicates showed premature precipitation and collapse of supersaturation, while others did not. In conclusion, the proposed novel tool appears very promising in gaining an in-depth mechanistic understanding of the bioconversion/permeation interplay, including transient supersaturation of phosphate-ester prodrugs like fosamprenavir.


Asunto(s)
Profármacos , Animales , Bovinos , Humanos , Fosfatasa Alcalina , Biomimética , Ésteres , Microdiálisis , Organofosfatos , Fosfatos , Profármacos/metabolismo , Solubilidad
15.
J Control Release ; 360: 810-817, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37473807

RESUMEN

Choroidal neovascularization (CNV) is a common ocular pathology that may be associated in a variety of eye diseases. Although intravitreal injection treatment of anti-vascular endothelial growth factor (anti-VEGF) drugs shows significant clinical benefits in CNV treatment, the limitations of the current therapy need to be addressed. The aim of our study was to investigate the potential utility of three C-end Rule (CendR) peptides (RPARPAR, PL3, iRGD) for CNV targeting and to evaluate the efficacy of peptides for treating experimental CNV in mice. We observed that the CendR peptides localize to the CNV lesion sites after intravitreal injection and were mainly found in the outer nuclear cell layer (ONL) of the mouse retina. Interestingly, experimental therapy with tenascin-C (TNC-C) and neuropilin-1 (NRP-1)-targeting PL3 peptide, reduced angiogenesis and decreased vascular leakage. The results suggest that PL3 and potentially other CendR peptides could serve as affinity targeting ligands and therapeutics for ocular diseases that involve pathological CNV.


Asunto(s)
Neovascularización Coroidal , Ratones , Animales , Neovascularización Coroidal/tratamiento farmacológico , Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Péptidos/uso terapéutico , Inyecciones Intravítreas , Rayos Láser , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
16.
Pharmaceutics ; 15(5)2023 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-37242796

RESUMEN

Clozapine (CZP) is the only effective drug in schizophrenia resistant to typical antipsychotics. However, existing dosage forms (oral or orodispersible tablets, suspensions or intramuscular injection) show challenging limitations. After oral administration, CZP has low bioavailability due to a large first-pass effect, while the i.m. route is often painful, with low patient compliance and requiring specialised personnel. Moreover, CZP has a very low aqueous solubility. This study proposes the intranasal route as an alternative route of administration for CZP, through its encapsulation in polymeric nanoparticles (NPs) based on Eudragit® RS100 and RL100 copolymers. Slow-release polymeric NPs with dimensions around 400-500 nm were formulated to reside and release CZP in the nasal cavity, where it can be absorbed through the nasal mucosa and reach the systemic circulation. CZP-EUD-NPs showed a controlled release of CZP for up to 8 h. Furthermore, to reduce mucociliary clearance and increase the residence time of NPs in the nasal cavity to improve drug bioavailability, mucoadhesive NPs were formulated. This study shows that the NPs already exhibited strong electrostatic interactions with mucin at time zero due to the presence of the positive charge of the used copolymers. Furthermore, to improve the solubility, diffusion and adsorption of CZPs and the storage stability of the formulation, it was lyophilised using 5% (w/v) HP-ß-CD as a cryoprotectant. It ensured the preservation of the NPs' size, PDI and charge upon reconstitution. Moreover, physicochemical characterisation studies of solid-state NPs were performed. Finally, toxicity studies were performed in vitro on MDCKII cells and primary human olfactory mucosa cells and in vivo on the nasal mucosa of CD-1 mice. The latter showed non-toxicity of B-EUD-NPs and mild CZP-EUD-NP-induced tissue abnormalities.

17.
ACS Pharmacol Transl Sci ; 6(10): 1518-1530, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37854619

RESUMEN

Dry eye disease (DED), the most common ocular disorder, reduces the quality of life for hundreds of millions of people annually. In healthy eyes, the tear film lipid layer (TFLL) stabilizes the tear film and moderates the evaporation rate of tear fluid. In >80% of DED cases, these central features are compromised leading to tear film instability and excessive evaporation of tear fluid. Herein we assess the potential of liposomal formulations featuring phosphatidylcholines and tailored lipid species from the wax ester and O-acyl-ω-hydroxy fatty acid categories in targeting this defect. The developed lead formulation displays good evaporation-resistant properties and respreadability over compression-expansion cycles in our Langmuir model system and a promising safety and efficacy profile in vitro. Preclinical in vivo studies will in the future be required to further assess and validate the potential of this concept in the treatment of DED.

18.
Int J Pharm ; 624: 122028, 2022 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-35853567

RESUMEN

The number of patients affected by Dry Eye Disease (DED) had notably increased worldwide, addressing the need of novel therapeutic approaches. Polyphenols, quercetin (QUE) and resveratrol (RSV) show necessary antioxidant and anti-inflammatory properties to manage DED, but their application as topical eyedrops is restricted by low aqueous solubility and low chemical stability. Cyclodextrins (CD) are widely used to improve physicochemical characteristics of drugs. Consequently, the aim of this study was to make a comparison between binary complexes with quercetin, resveratrol and cyclodextrins and tertiary complexes adding hyaluronic acid (HA). Both complexes were able to enhance solubility and stability of QUE and RSV. AFM imaging and DLS measurements disclose the formation of spherical nanoaggregates within tertiary complexes of both QUE and RSV with mean diameters of 103 and 82 nm. Neither complex demonstrated cytotoxic effect in in vitro studies in corneal (HCE) and conjunctival (IM-ConjEpi) cell lines. In HCE cells, complexes containing QUE or RSV at their highest concentrations were able to scavenge more than 95 % of the ROS that were produced intracellularly (p < 0.005). Similar response was observed with IM-ConjEpi cells. The antioxidant effect was maintained in the complexes with HA. This confirmed their potential as viable topical treatment for DED.


Asunto(s)
Ciclodextrinas , Antioxidantes/química , Antioxidantes/farmacología , Conjuntiva , Ciclodextrinas/química , Humanos , Quercetina/química , Quercetina/farmacología , Resveratrol , Solubilidad
20.
Eur J Pharm Biopharm ; 172: 53-60, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35121080

RESUMEN

Rapid precorneal loss of topically applied eye drops limits ocular drug absorption. Controlling release and precorneal residence properties of topical formulations may improve ocular drug bioavailability and duration of action. In this study, we evaluated in vivo ocular pharmacokinetics of dexamethasone in rabbits after application of a drug solution (0.01%), suspension (Maxidex® 0.1%), and hydrogels of 2-hydroxyethyl methacrylate (HEMA) and acrylic acid (AAc) copolymers. The rabbits received a single eyedrop (solution or suspension) or dexamethasone-loaded hydrogel topically. Dexamethasone in tear fluid was sampled with glass capillaries and quantitated by LC-MS/MS. Higher dexamethasone exposure (AUC) in the tear fluid was observed with the suspension (≈3.6-fold) and hydrogel (12.8-fold) as compared to the solution. During initial 15 min post-application, the highest AUC of dissolved dexamethasone was seen after hydrogel application (368 min*µg/mL) followed by suspension (109.9 min*µg/mL) and solution (28.7 min*µg/mL. Based on kinetic simulations, dexamethasone release from hydrogels in vivo and in vitro is comparable. Our data indicate that prolonged exposure of absorbable dexamethasone in tear fluid is reached with hydrogels and suspensions. Pharmacokinetic understanding of formulation behavior in the lacrimal fluid helps in the design of dexamethasone delivery systems with improved ocular absorption and prolonged duration of action.


Asunto(s)
Hidrogeles , Espectrometría de Masas en Tándem , Animales , Cromatografía Liquida , Dexametasona , Liberación de Fármacos , Cinética , Conejos , Suspensiones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA