Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Mater Sci Mater Med ; 34(1): 5, 2022 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-36586040

RESUMO

The administration of anti-vascular endothelial growth factor drugs in the posterior eye segment with sustained release through less invasive methods is a challenge in the treatment of age-related macular disease. We developed a flexible capsule device using porous poly(dimethylsiloxane) (PDMS) that was able to release ranibizumab. The porous PDMS sheet was fabricated by salt-leaching of a micro-sectioned PDMS sheet containing salt microparticles. Observation with scanning electron microscopy revealed that the pore densities could be adjusted by the concentration of salt. The in vitro release study showed that the release rate of fluorescein isothiocyanate-tagged albumin could be adjusted based on the pore density of the porous PDMS sheet. Ranibizumab could be released in a sustained-release manner for 16 weeks. The device was implanted on the sclera; its efficacy in terms of the suppression of laser-induced choroidal neovascularization (CNV) in rats was compared with that of monthly intravitreal injections of ranibizumab. At 8 and 18 weeks after implantation, the CNV area was significantly reduced in rats that received the ranibizumab-releasing device compared with those that received the placebo device. However, although monthly intravitreal injections of ranibizumab reduced CNV for 8 weeks, this reduction was not sustained for 18 weeks. In conclusion, we demonstrated a novel controlled-release device using a porous PDMS sheet that could suppress CNV via a less invasive transscleral route versus intravitreal injections. This device may also reduce the occurrence of side effects associated with frequent intravitreal injections.


Assuntos
Neovascularização de Coroide , Ranibizumab , Ratos , Animais , Ranibizumab/uso terapêutico , Porosidade , Neovascularização de Coroide/tratamento farmacológico , Lasers , Inibidores da Angiogênese/uso terapêutico
2.
Drug Dev Ind Pharm ; 42(1): 107-115, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25923417

RESUMO

The development of an effective sustained ocular drug delivery system remains a challenging task. The objective of the present study was to characterize a silicone pressure sensitive adhesive (PSA) episcleral implant system for transscleral drug delivery. Silicone PSA implants for dexamethasone, atenolol, and bovine serum albumin (BSA) were prepared at different polymer-to-drug mass ratios. Implant adhesion to human cadaver sclera was measured. Drug release experiments were conducted in well-stirred containers in vitro. The results were then analyzed using a pharmacokinetic model and in vitro-in vivo data comparison from previous studies. The silicone PSA episcleral implants in the present study had an average diameter of 3.5 mm and a thickness of 0.8 mm. Drug release from the silicone PSA implants was influenced by drug solubility, implant polymer content, and implant coating. Drug release from the implants was observed to follow the receding boundary release mechanism and was solubility dependent with the higher water solubility drug showing higher release rate than the low-solubility drug. Increasing polymer content in the implants led to a significant decrease in the drug release rate. Coated implants reduced the initial burst effect and provided lower release rates than the uncoated implants. These implants provided sustained drug release that could last up to several months in vitro and demonstrated the potential to offer drug delivery for chronic ocular diseases via the transscleral route.


Assuntos
Adesivos/química , Implantes de Medicamento/química , Implantes de Medicamento/farmacocinética , Liberação Controlada de Fármacos , Silicones/química , Administração Oftálmica , Atenolol/administração & dosagem , Atenolol/farmacocinética , Cadáver , Dexametasona/administração & dosagem , Dexametasona/farmacocinética , Oftalmopatias/tratamento farmacológico , Humanos , Técnicas In Vitro , Modelos Biológicos , Esclera , Solubilidade
3.
Surv Ophthalmol ; 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38986847

RESUMO

Ophthalmic treatment demands precision and consistency in delivering therapeutic agents over extended periods to address many conditions, from common eye disorders to complex diseases. This diversity necessitates a range of delivery strategies, each tailored to specific needs. We delve into various delivery cargos that are pivotal in ophthalmic care. These cargos encompass biodegradable implants that gradually release medication, nonbiodegradable implants for sustained drug delivery, refillable tools allowing flexibility in treatment, hydrogels capable of retaining substances while maintaining ocular comfort, and advanced nanotechnology devices that precisely target eye tissues. Within each cargo category, we explore cutting-edge research-level approaches and FDA-approved methods, providing a thorough overview of the current state of ophthalmic drug delivery. In particular, our focus on nanotechnology reveals the promising potential for gene delivery, cell therapy administration, and the implantation of active devices directly into the retina. These advancements hold the key to more effective, personalized, and minimally- invasive ophthalmic treatments, revolutionizing the field of eye care.

4.
Drug Deliv Transl Res ; 13(8): 2142-2158, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-35713782

RESUMO

The discovery of proteins that neutralise vascular endothelial growth factors, such as pegaptanib, ranibizumab and aflibercept, can inhibit the process of angiogenesis, thereby restoring eyesight in individuals with retinal vascular disorders. However, due to the posterior location and chronic nature of retinal diseases, a safe and effective intraocular protein delivery system is currently lacking. Thus, dissolving bilayer microneedles (MNs) with the potential to deliver proteins to the back of the eye in an efficient and minimally invasive manner were developed in this study. A model protein, ovalbumin (OVA), was incorporated into MNs fabricated from different polymers, including hyaluronic acid (HA), polyvinyl alcohol (PVA) and polyvinylpyrrolidone (PVP). Optimised PVA/PVP MNs were demonstrated to be robust enough to pierce porcine sclera with > 75% of the needle length penetrating the sclera and dissolving within 150 s. SDS-PAGE and OVA-specific ELISA revealed that the bioactivity of the model protein was maintained during the manufacture of MNs. In hen's egg-chorioallantoic membrane test, MNs fabricated from all chosen polymers were classified as non-irritants. Furthermore, ex vivo permeation studies showed that optimised MNs could permeate 86.99 ± 7.37% of OVA through the sclera, twice that of the needle-free patch (42.16 ± 3.95%), highlighting the capability of MNs to circumvent physical barriers and promote protein delivery to the posterior segment of the eye. In this work, a novel, efficient and safe intraocular protein delivery system was successfully established.


Assuntos
Galinhas , Ácido Hialurônico , Animais , Feminino , Suínos , Polímeros , Povidona , Sistemas de Liberação de Medicamentos , Administração Cutânea
5.
J Control Release ; 349: 744-755, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35901859

RESUMO

Cyclosporine is an immunomodulatory drug commonly used for the treatment of mild-to-severe dry eye syndrome as well as intermediate and posterior segment diseases as uveitis. The ocular administration is however hampered by its relatively high molecular weight and poor permeability across biological barriers. The aim of this work was to identify a micellar formulation with the ability to solubilize a considerable amount of cyclosporine and promote its transport across ocular barriers. Non-ionic amphiphilic polymers used for micelles preparation were tocopherol polyethylene glycol 1000 succinate (TPGS) and Solutol® HS15. Furthermore, the addition of alpha-linolenic acid was assessed. A second aim was to evaluate micelles fate in the ocular tissues (cornea and sclera) to shed light on penetration mechanisms. This was possible by extracting and quantifying both drug and polymer in the tissues, by studying TPGS hydrolysis in a bio-relevant environment and by following micelles penetration with two-photon microscopy. Furthermore, TPGS role as permeation enhancer on the cornea, with possible irreversible modifications of tissue permeability, was analyzed. Results showed that TPGS micelles (approx. 13 nm in size), loaded with 5 mg/ml of cyclosporine, promoted drug retention in both the cornea and the sclera. Data demonstrated that micelles behavior strictly depends on the tissue: micelles disruption occurs in contact with the cornea, while intact micelles diffuse in the interfibrillar pores of the sclera and form a reservoir that can sustain over time drug delivery to the deeper tissues. Finally, cornea quickly restore the barrier properties after TPGS removal from the tissue, demonstrating its potential good tolerability for ocular application.


Assuntos
Ciclosporina , Micelas , Córnea , Portadores de Fármacos , Polietilenoglicóis , Polímeros , Solubilidade , Succinatos , Tocoferóis , Vitamina E , Ácido alfa-Linolênico
6.
Biomater Adv ; 137: 212767, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35929230

RESUMO

Intravitreal injections (IVT) are regarded as the gold standard for effective delivery of hydrophobic drugs to the back of the eye. However, as a highly invasive procedure, the injection itself may lead to poor patient compliance and severe complications. In this research work, a hybrid system of nanosuspensions (NS) and dissolving microneedles (MNs) was developed as an alternative to conventional hypodermic needles used in IVT for minimally invasive transscleral delivery of hydrophobic drugs. NS of a hydrophobic drug, triamcinolone acetonide (TA), were fabricated using a wet milling technique. TA NS optimised by central composite factorial design had a proven diameter of 246.65 ± 8.55 nm. After optimisation, TA NS were incorporated into MN arrays to form a bilayer structure by high-speed centrifugation. TA NS-loaded MNs were robust enough to pierce excised porcine sclera with insertion depth higher than 80% of the needle height and showed rapid dissolution (<3 min). In contrast, the plain TA-loaded MNs exhibited poor mechanical and insertion performances and took more than 8 min to be fully dissolved in the scleral tissue. Importantly, transscleral deposition studies showed that 56.46 ± 7.76 µg/mm2 of TA was deposited into the sclera after 5 min of NS-loaded MN application, which was 4.5-fold higher than plain drug-loaded MNs (12.56 ± 2.59 µg/mm2). An ex vivo distribution study revealed that MN arrays could promote the transscleral penetration of hydrophobic molecules with higher drug concentrations observed in the deep layer of the sclera. Moreover, the developed TA NS-loaded MN array was biocompatible with ocular tissues, as demonstrated using the hens egg-chorioallantoic membrane assay and cytotoxicity test. The results presented here demonstrate that the hybrid system of NS and dissolving MNs can provide a novel and promising technology to alleviate retinal diseases in a therapeutically effective and minimally invasive manner.


Assuntos
Galinhas , Sistemas de Liberação de Medicamentos , Animais , Sistemas de Liberação de Medicamentos/métodos , Feminino , Agulhas , Preparações Farmacêuticas , Esclera , Suínos , Triancinolona Acetonida
7.
Mater Sci Eng C Mater Biol Appl ; 103: 109799, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31349439

RESUMO

Retinoblastoma (Rb) is the most common primary malignant intraocular tumor in children which develops from the retinal stem cells. Systemic chemotherapy is the typical therapeutic treatment and though most children survive Rb, they often lose their vision, or the eye needs to be enucleated. Regarding to the pure availability of the target tumor by systemic chemotherapy, the local anticancer drug administration would be advantageous to increase the local drug concentration and minimize adverse side effects of chemotherapy. The present paper describes a new hydrogel implant enabled to deliver therapeutically active doses of low molecular weight hydrophilic antitumor drugs topotecan and vincristine. The hydrogel implant is proposed as bi-layered with an inner hydrophilic layer from 2-hydroxyethyl methacrylate (HEMA) serving as a reservoir of the chemotherapeutic agent and an outer hydrophobic layer from 2-ethoxyethyl methacrylate (EOEMA) acting as a barrier to protect the surrounding vascularized tissue against cytotoxicity of the delivered chemotherapeutics. The experiments with enucleated pig eyes demonstrated the ability of tested drugs to diffuse through sclera and reach the vitreous humor. HEMA-based hydrogels were examined in terms of sorption, release and transport properties, showing the possibility of adjusting the loading capacity and diffusion of the drugs by the degree of crosslinking. The EOEMA-based gels proved to be an inert for drug sorption and diffusion. A chorioallantoic membrane assay demonstrated excellent biocompatibility of unloaded hydrogels, and in vitro experiments confirmed significant cytotoxicity of drug-loaded hydrogels against a Rb cell line; 2 days for those topotecan-loaded and a minimum of 6 days for vincristine-loaded hydrogels. The bi-layered hydrogel implant can be considered promising for local administration of active agents to eye-globe for the treatment of Rb and also other ocular disorders.


Assuntos
Portadores de Fármacos/química , Hidrogéis/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Estabilidade de Medicamentos , Olho/efeitos dos fármacos , Olho/metabolismo , Humanos , Cinética , Metacrilatos/química , Próteses e Implantes , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Suínos , Topotecan/química , Topotecan/metabolismo , Topotecan/farmacologia , Vincristina/química , Vincristina/metabolismo , Vincristina/farmacologia
8.
Eur J Pharm Biopharm ; 136: 184-191, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30690065

RESUMO

Continuous drug administration with better adherence to treatment and less invasive procedures is important in treating retinal diseases such as age-related macular disease. In this study, we report a drug-refillable device consisting of a silicone reservoir and an injectable gelatin/chitosan gel (iGel). The silicone reservoir was fabricated with polydimethylsiloxane (PDMS) using a computer-aided design and manufacturing to have micropores at a releasing side for uniaxial release to the sclera. A stainless steel wire and sheet were combined in the side and bottom of the reservoir to ensure flexibility and to fit on the curvature of the eyeball and prevent irritation to the sclera through the bottom of the reservoir. The drug was injected and formulated in the reservoir by in situ crosslinking of gelatin/chitosan gel with the crosslinker; 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride. The in vitro release study using fluorescein molecules showed that the release rate from encapsulated iGel in the reservoir was slower than that from the original iGel. After reinjecting the iGel into the reservoir, the same release profile as the first injection was observed. The reservoir containing iGel was placed on the sclera of a rabbit and the distribution of 150 kDa fluorescein isothiocyanate-dextran (FD150) in the retina and choroid/retinal pigment epithelium (choroid/RPE) was studied. The cryosections showed that FD150 was observed in the choroid/RPE. Homogenates of the retina and choroid/RPE showed fluorescence during 12 weeks implantation, indicating the drug could be delivered to the retina by using the device. The drug filling was successful into the reservoir implanted on the sclera through the conjunctiva by using a needle. In conclusion, the refillable drug delivery device is a promising tool to administer drugs long-term by reinjection with less invasiveness to intraocular tissues.


Assuntos
Quitosana/farmacocinética , Sistemas de Liberação de Medicamentos/instrumentação , Desenho de Equipamento , Gelatina/farmacocinética , Retina/metabolismo , Esclera/metabolismo , Animais , Quitosana/administração & dosagem , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/farmacocinética , Dimetilpolisiloxanos/química , Sistemas de Liberação de Medicamentos/métodos , Desenho de Equipamento/métodos , Gelatina/administração & dosagem , Masculino , Nylons/química , Coelhos , Retina/efeitos dos fármacos , Esclera/efeitos dos fármacos , Silicones/química
9.
Int J Pharm ; 567: 118458, 2019 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-31247277

RESUMO

Successful treatment of age-related macular diseases requires an effective controlled drug release system with less invasive route of administration in the eye to reduce the burden of frequent intravitreal injections for patients. In this study, we developed an episcleral implantable device for sustained release of ranibizumab, and evaluated its efficacy on suppression of laser-induced choroidal neovascularization (CNV) in rats. We tested both biodegradable and non-biodegradable sheet-type devices consisting of crosslinked gelatin/chitosan (Gel/CS) and photopolymerized poly(ethyleneglycol) dimethacrylate that incorporated collagen microparticles (PEGDM/COL). In vitro release studies of FITC-labeled albumin showed a constant release from PEGDM/COL sheets compared to Gel/CS sheets. The Gel/CS sheets gradually biodegraded in the sclera during the 24-week implantation; however, the PEGDM/COL sheets did not degrade. FITC-albumin was detected in the retina during 18 weeks implantation in the PEGDM/COL sheet-treated group, and was detected in the Gel/CS sheet-treated group during 6 weeks implantation. CNV was suppressed 18 weeks after application of ranibizumab-loaded PEGDM/COL sheets compared to a placebo PEGDM/COL sheet-treated group, and to the intravitreal ranibizumab-injected group. In conclusion, the PEGDM/COL sheet device suppressed CNV via a transscleral administration route for 18 weeks, indicating that prolonged sustained ranibizumab release could reduce the burden of repeated intravitreal injections.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Neovascularização de Coroide/tratamento farmacológico , Implantes de Medicamento/administração & dosagem , Ranibizumab/administração & dosagem , Inibidores da Angiogênese/química , Animais , Quitosana/administração & dosagem , Quitosana/química , Colágeno/administração & dosagem , Colágeno/química , Implantes de Medicamento/química , Liberação Controlada de Fármacos , Olho/efeitos dos fármacos , Olho/metabolismo , Olho/patologia , Fluoresceína-5-Isotiocianato/administração & dosagem , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/química , Gelatina/administração & dosagem , Gelatina/química , Lasers , Masculino , Metacrilatos/administração & dosagem , Metacrilatos/química , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Ranibizumab/química , Ratos Sprague-Dawley , Albumina Sérica/administração & dosagem , Albumina Sérica/química
10.
Acta Biomater ; 10(2): 680-7, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24239899

RESUMO

The design of drug delivery systems that can deliver multiple drugs to the posterior segment of the eye is a challenging task in retinal disease treatments. We report a polymeric device for multi-drug transscleral delivery at independently controlled release rates. The device comprises a microfabricated reservoir, controlled-release cover and three different fluorescent formulations, which were made of photopolymeized tri(ethyleneglycol)dimethacrylate (TEGDM) and poly(ethyleneglycol)dimethacrylate (PEGDM). The release rate of each fluorescent is controlled by varying the PEGDM/TEGDM ratio in its formulation and the cover. The release kinetics appeared to be related to the swelling ratio of the PEGDM/TEGDM polymers. When the devices were implanted onto rat sclerae, fluorescence was observable in the ocular tissues during 4 weeks' implantation and distributed locally around the implantation site. Our polymeric system, which can administer multiple compounds with distinct kinetics, provides prolonged action and less invasive transscleral administration, and is expected to provide new tools for the treatment of posterior eye diseases with new therapeutic modalities.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Metacrilatos/química , Polietilenoglicóis/química , Ácidos Polimetacrílicos/química , Segmento Posterior do Olho/metabolismo , Esclera/metabolismo , Animais , Difusão , Fluoresceína/metabolismo , Indóis/metabolismo , Permeabilidade , Implantação de Prótese , Ratos , Retina/metabolismo , Rodaminas/metabolismo
11.
Eur J Pharm Biopharm ; 88(1): 116-22, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24816128

RESUMO

The aim of this work was to study the penetration of cytochrome c, a positively charged model protein (MW 12.4 kDa, charge at pH 8.2: +9), across different ocular tissues, and to evaluate the potential of iontophoresis to enhance and control the transscleral transport. The passive transport of cytochrome c across the sclera and across the bilayer choroid-Bruch's membrane was evaluated using Franz diffusion cells and porcine tissues. The affinity of cytochrome c for melanin was measured by means of in vitro binding experiments. The iontophoretic (anodal) permeation was studied as a function of donor concentration (from 5 to 70 mg/ml) and current intensity (from 0.9 to 3.5 mA; density from 1.5 to 5.8 mA/cm(2)), and the contribution of electroosmosis on cytochrome c transport was evaluated by using a high molecular weight fluorescent dextran (FD-150, 149 kDa) as neutral marker. Finally, the possibility of tuning cytochrome c permeation rate was investigated on a 70 mg/ml cytochrome c solution, by alternating passive permeation and iontophoresis at different intensities. Cytochrome c permeated the sclera with a passive permeability coefficient of about 2.5 × 10(-6)cm/s, comparable to molecules of similar molecular radius. The choroid-Bruch's layer was an important barrier to penetration, since its presence reduced 5-7 times the amount permeated after 5h, also because of the presence of melanin that binds cytochrome. Iontophoresis (2.9 mA/cm(2)) enhanced cytochrome c penetration across the sclera at all the concentrations tested, increasing about ten times the amount permeated after 2h. The effect was proportional to current density: the enhancement factor (measured on a 10mg/ml solution), resulted 6.0 ± 4.3 (i=0.9 mA; density=1.5 mA/cm(2)), 10.6 ± 4.1 (i=1.75 mA; density=2.9 mA/cm(2)), 33.2 ± 8.3 (i=1.75 mA; density=5.8 mA/cm(2)). Iontophoretic (density=2.9 mA/cm(2)) experiments performed with FD-150, an electroosmotic flow (EO) marker, demonstrated that cytochrome c, at concentration higher that 1mg/ml, dramatically reduced the EO flow and that, despite the high MW, the main mechanism for cytochrome c iontophoretic permeation is electrorepulsion. Finally, by alternating in the same experiment passive permeation and iontophoresis at different current intensities, a precise modulation of cytochrome c release was obtained, thus indicating the possibility of tuning the release as a function of specific therapeutic needs.


Assuntos
Administração Tópica , Citocromos c/administração & dosagem , Iontoforese/métodos , Esclera/efeitos dos fármacos , Animais , Transporte Biológico , Bovinos , Corioide/efeitos dos fármacos , Difusão , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos , Olho/metabolismo , Técnicas In Vitro , Melaninas/administração & dosagem , Melaninas/química , Peso Molecular , Osmose , Permeabilidade , Suínos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa