Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
J Fr Ophtalmol ; 46(7): 791-802, 2023 Sep.
Article in French | MEDLINE | ID: mdl-37277234

ABSTRACT

The central serous chorioretinopathy (CSCR) is characterized by serous retinal detachments SRD associated with one or several retinal pigment epithelium detachments/irregularities (PEDs). The choroid is thickened with dilated choroidal veins and choroidal hyperpermeability suggesting an underlying choroidopathy. CSCR belongs to the pachychoroid spectrum. CSCR affects mostly middle-aged men and the main risk factor is the corticosteroid intake. In most cases, the subretinal detachment resolves spontaneously with a good visual prognosis. However, recurrent or chronic form of the disease can lead to irreversible retinal damage and decreased visual acuity. Laser on an extra foveal leak point or half dose/half fluence photodynamic therapy are the first-line treatment options.


Subject(s)
Central Serous Chorioretinopathy , Retinal Detachment , Middle Aged , Male , Humans , Central Serous Chorioretinopathy/complications , Central Serous Chorioretinopathy/diagnosis , Central Serous Chorioretinopathy/epidemiology , Chronic Disease , Fluorescein Angiography , Retinal Detachment/diagnosis , Retinal Detachment/etiology , Retinal Detachment/therapy , Retina , Tomography, Optical Coherence , Retrospective Studies
2.
Sci Rep ; 10(1): 20175, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33214636

ABSTRACT

No systemic biomarker of Central Serous Chorioretinopathy (CSCR) has been identified. Lipocalin 2 (LCN2 or NGAL), alone or complexed with MMP-9 (NGAL/MMP-9), is increased in several retinal disorders. Serum levels of LCN2 and NGAL/MMP-9 were measured in CSCR patients (n = 147) with chronic (n = 76) or acute/recurrent disease (n = 71) and in age- and sex-matched healthy controls (n = 130). Samples with CRP > 5 mg/L, creatinine > 100 µmol/L, and/or urea > 7.5 mmol/L were excluded. Serum LCN2 was lower in CSCR patients than controls (81.4 ± 48.7 vs 107.3 ± 44.5 ng/ml, p < 0.0001), and lower in acute/recurrent CSCR than controls (p < 0.001) and chronic CSCR (p = 0.006). Serum NGAL/MMP-9 was lower in CSCR patients than controls (47.2 ± 40.7 vs 74.1 ± 42.6, p < 0.0001), and lower in acute/recurrent CSCR than controls (p < 0.001) and chronic CSCR (p = 0.002). A ROC curve showed that for LCN2 serum levels, the 80-ng/ml cutoff value allows to discriminate acute/recurrent CSCR from controls with 80.3% sensitivity and 75.8% specificity, and for NGAL/MMP-9 serum levels, a 38-ng/ml cutoff value allows to discriminate acute/recurrent CSCR from controls with 69.6% sensitivity and 80.3% specificity. In both acute and chronic CSCR, low serum LCN2 and NGAL/MMP-9, provide a biological link between the two CSCR forms, and potential susceptibility to oxidative stress and innate immune dysregulation in CSCR.


Subject(s)
Biomarkers/blood , Central Serous Chorioretinopathy/blood , Lipocalin-2/blood , Adult , Case-Control Studies , Female , Humans , Male , Matrix Metalloproteinase 9/blood , Middle Aged , ROC Curve , Retrospective Studies
3.
Nat Photonics ; 14(7): 439-445, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32607125

ABSTRACT

In-vivo observation of the human retina at the cellular level is crucial to detect the first signs of retinal diseases and properly treat them. Despite the phenomenal advances in adaptive optics (AO) systems, clinical imaging of many retinal cells is still elusive due to the low signal-to-noise ratio induced by transpupillary illumination. We present a transscleral optical phase imaging (TOPI) method, which relies on high-angle oblique illumination of the retina, combined with AO, to enhance cell contrast. Examination of eleven healthy volunteer eyes, without pupil dilation, shows the ability of this method to produce in-vivo images of retinal cells, from the retinal pigment epithelium to the nerve fibre layer. This method also allows the generation of high-resolution label-free ex-vivo phase images of flat-mounted retinas. The 4.4°x 4.4° field-of-view in-vivo images are recorded in less than 10 seconds, opening new avenues in the exploration of healthy and diseased retinas.

4.
J Expo Sci Environ Epidemiol ; 29(6): 753-764, 2019 10.
Article in English | MEDLINE | ID: mdl-30382242

ABSTRACT

The aim of this study was to assess solar ultraviolet radiation (UVR) doses received by the eyes in different exposure situations, and to predict the sun protection effectiveness provided by various styles of sunglasses at facial, periorbital, and ocular skin zones including the cornea and accounting for different head positions. A 3D numeric model was optimized to predict direct, diffuse and reflected erythemally weighted UVR doses received at various skin zones. Precisely defined facial, periorbital, and ocular skin zones, sunglasses (goggles, medium-, and large-sized sunglasses) and three head positions were modeled to simulate daily (08:00-17:00) and midday (12:00-14:00) UVR doses. The shading from sunglasses' frame and lenses' UVR transmission were used to calculate a predictive protection factor (PPF [%]). Highest ocular daily UVR doses were estimated at the uncovered cornea (1718.4 J/m2). Least sun protection was provided by middle-sized sunglasses with highest midday dose at the white lateral (290.8 J/m2) and lateral periorbital zones (390.9 J/m2). Goggles reached almost 100% protection at all skin zones. Large-sized sunglasses were highly effective in winter; however, their effectiveness depended on diffuse UVR doses received. In "looking-up" head positions highest midday UVR doses were received at the unprotected cornea (908.1 J/m2), totally protected when large-sized sunglasses are used. All tested sunglass lenses fully blocked UVR. Sunglasses' protection effectiveness is strongly influenced by geometry, wearing position, head positions, and exposure conditions. Sunglasses do not totally block UVR and should be combined with additional protection means. 3D modeling allows estimating UVR exposure of highly sensitive small skin zones, chronically exposed and rarely assessed.


Subject(s)
Environmental Exposure , Eye Protective Devices , Eye/radiation effects , Sunlight , Humans , Seasons
5.
Neuroscience ; 339: 296-307, 2016 Dec 17.
Article in English | MEDLINE | ID: mdl-27751961

ABSTRACT

To save energy, the European directives from the Eco-design of Energy Using Products (2005/32/CE) have recommended the replacement of incandescent lamps by more economic devices such as Light Emitting Diodes (LEDs). However, the emission spectrum of these devices is enriched in blue radiations, known to be potentially dangerous to the retina. Recent studies showed that light exposure contributes to the onset of early stages of age-related macular degeneration (AMD). Here, we investigate, in albinos and pigmented rats, the effects of different exposure protocols. Twenty-four hours exposure at high luminance was compared to a cyclic (dark/light) exposure at domestic levels for 1week and 1month, using different LEDs (Cold-white, blue and green), as well as fluorocompact bulbs and fluorescent tubes. The data suggest that the blue component of the white-LED may cause retinal toxicity at occupational domestic illuminance and not only in extreme experimental conditions, as previously reported. It is important to note that the current regulations and standards have been established on the basis of acute light exposure and do not take into account the effects of repeated exposure.


Subject(s)
Light/adverse effects , Lighting/adverse effects , Lighting/instrumentation , Retina/radiation effects , Retinal Degeneration/etiology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Dose-Response Relationship, Radiation , Drosophila Proteins , Electroretinography , Equipment Design , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Photic Stimulation/adverse effects , Photic Stimulation/instrumentation , Photoperiod , Rats, Long-Evans , Rats, Wistar , Retina/pathology , Retina/physiopathology , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Skin Pigmentation
6.
J Fr Ophtalmol ; 38(2): 159-67, 2015 Feb.
Article in French | MEDLINE | ID: mdl-25601517

ABSTRACT

Diabetic macular edema (DME) is a frequent complication of diabetic retinopathy and may cause severe visual loss. In this article, we examine the pathophysiology of DME and review various treatment options, such as laser photocoagulation, anti-vascular endothelial growth factor (VEGF) receptor antibodies, and steroids including ILUVIEN(®), which is a new sustained-release, non biodegradable, injectable, intravitreal micro-implant containing fluocinolone acetonide. The results of the FAME (Fluocinolone Acetonide in Diabetic Macular Edema) studies, conducted to evaluate the efficacy and safety of ILUVIEN(®) in DME, are discussed.


Subject(s)
Diabetic Retinopathy/drug therapy , Drug Implants , Fluocinolone Acetonide/administration & dosage , Macular Edema/drug therapy , Chronic Disease , Diabetic Retinopathy/complications , Humans , Intravitreal Injections , Macular Edema/etiology
7.
J Fr Ophtalmol ; 37(7): 566-79, 2014 Sep.
Article in French | MEDLINE | ID: mdl-25190312

ABSTRACT

Age related macular degeneration (AMD) is a pathological aging of the macula, brought about by the interaction of genetic and environmental factors. It induces geographic atrophy of the retina and/or choroidal neovascularization. In the latter, abnormal vessels develop from the choriocapillaris, with the involvement of VEGF (vascular endothelial growth factor). The VEGF family includes several factors, including VEGF-A, B, C, D, F and PlGF (placental growth factor). Their biological properties and their affinities to the VEGFR1, VEGFR2 and VEGFR3 receptors found on endothelial cells differ. Exudative AMD involves mainly VEGF-A and VEGF-R2. Anti-VEGF agents used in ophthalmology (ranibizumab, bevacizumab and aflibercept) are designed to primarily target this pathway. In vitro, all have sufficient affinity to their ligands. Their therapeutic efficacy must therefore be judged based on clinical criteria. In clinical practice, the minimum number of injections required for a satisfactory result appears to be comparable with all the three. The few available studies on therapeutic substitutions of anti-VEGF compounds suggest that some patients may benefit from substituting the anti-VEGF in cases of an unsatisfactory response to an initial molecule. Although local side effects, including increased risk of geographic atrophy, and systemic effects, including vascular accidents, have been suggested, these risks remain low, specially compared to the benefits of the treatment. Differences in safety between anti-VEGF are theoretically possible but unproven.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Macular Degeneration/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Aging/physiology , Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab , Choroidal Neovascularization/drug therapy , Drug Tolerance , Humans , Macular Degeneration/physiopathology , Randomized Controlled Trials as Topic , Ranibizumab , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Retina/physiology , Risk Factors
8.
Mol Vis ; 20: 908-20, 2014.
Article in English | MEDLINE | ID: mdl-24966662

ABSTRACT

PURPOSE: To evaluate whether anti-vascular endothelial growth factor (VEGF) neutralizing antibodies injected in the vitreous of rat eyes influence retinal microglia and macrophage activation. To dissociate the effect of anti-VEGF on microglia and macrophages subsequent to its antiangiogenic effect, we chose a model of acute intraocular inflammation. METHODS: Lewis rats were challenged with systemic lipopolysaccharide (LPS) injection and concomitantly received 5 µl of rat anti-VEGF-neutralizing antibody (1.5 mg/ml) in the vitreous. Rat immunoglobulin G (IgG) isotype was used as the control. The effect of anti-VEGF was evaluated at 24 and 48 h clinically (uveitis scores), biologically (cytokine multiplex analysis in ocular media), and histologically (inflammatory cell counts on eye sections). Microglia and macrophages were immunodetected with ionized calcium-binding adaptor molecule 1 (IBA1) staining and counted based on their differential shapes (round amoeboid or ramified dendritiform) on sections and flatmounted retinas using confocal imaging and automatic quantification. Activation of microglia was also evaluated with inducible nitric oxide synthase (iNOS) and IBA1 coimmunostaining. Coimmunolocalization of VEGF receptor 1 and 2 (VEGF-R1 and R2) with IBA1 was performed on eye sections with or without anti-VEGF treatment. RESULTS: Neutralizing rat anti-VEGF antibodies significantly decreased ocular VEGF levels but did not decrease the endotoxin-induced uveitis (EIU) clinical score or the number of infiltrating cells and cytokines in ocular media (interleukin [IL]-1ß, IL-6, tumor necrosis factor [TNF]-α, and monocyte chemoattractant protein [MCP]-1). Eyes treated with anti-VEGF showed a significantly decreased number of activated microglia and macrophages in the retina and the choroid and decreased iNOS-positive microglia. IBA1-positive cells expressed VEGF-R1 and R2 in the inflamed retina. CONCLUSIONS: Microglia and macrophages expressed VEGF receptors, and intravitreous anti-VEGF influenced the microglia and macrophage activation state. Taking into account that anti-VEGF drugs are repeatedly injected in the vitreous of patients with retinal diseases, part of their effects could result from unsuspected modulation of the microglia activation state. This should be further studied in other ocular pathogenic conditions and human pathology.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Macrophage Activation/drug effects , Microglia/pathology , Retina/pathology , Uveitis/drug therapy , Uveitis/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antibodies, Neutralizing/pharmacology , Calcium-Binding Proteins/metabolism , Cell Count , Disease Models, Animal , Humans , Lipopolysaccharides , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Neutralization Tests , Nitric Oxide Synthase Type II/metabolism , Rats , Receptors, Vascular Endothelial Growth Factor/metabolism , Retina/drug effects , Retina/enzymology , Vascular Endothelial Growth Factor A/metabolism
9.
Gene Ther ; 20(11): 1093-103, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23804076

ABSTRACT

Inhibition of vascular endothelial growth factor (VEGF) has become the standard of care for patients presenting with wet age-related macular degeneration. However, monthly intravitreal injections are required for optimal efficacy. We have previously shown that electroporation enabled ciliary muscle gene transfer results in sustained protein secretion into the vitreous for up to 9 months. Here, we evaluated the long-term efficacy of ciliary muscle gene transfer of three soluble VEGF receptor-1 (sFlt-1) variants in a rat model of laser-induced choroidal neovascularization (CNV). All three sFlt-1 variants significantly diminished vascular leakage and neovascularization as measured by fluorescein angiography (FA) and flatmount choroid at 3 weeks. FA and infracyanine angiography demonstrated that inhibition of CNV was maintained for up to 6 months after gene transfer of the two shortest sFlt-1 variants. Throughout, clinical efficacy was correlated with sustained VEGF neutralization in the ocular media. Interestingly, treatment with sFlt-1 induced a 50% downregulation of VEGF messenger RNA levels in the retinal pigment epithelium and the choroid. We demonstrate for the first time that non-viral gene transfer can achieve a long-term reduction of VEGF levels and efficacy in the treatment of CNV.


Subject(s)
Choroidal Neovascularization/genetics , Choroidal Neovascularization/therapy , Ciliary Body/metabolism , Genetic Therapy/methods , Transfection/methods , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Animals , Cell Line , Choroid/metabolism , Choroidal Neovascularization/metabolism , Disease Models, Animal , Electroporation , Female , Fluorescein Angiography , Gene Expression Regulation , Humans , Neovascularization, Pathologic/therapy , Plasmids , Rats , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
10.
Gene Ther ; 19(9): 886-98, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21993171

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) is one of the candidate molecules among neurotrophic factors proposed for a potential treatment of retinitis pigmentosa (RP). It must be administered repeatedly or through sustained releasing systems to exert prolonged neuroprotective effects. In the dystrophic Royal College of Surgeon's (RCS) rat model of RP, we found that endogenous GDNF levels dropped during retinal degeneration time course, opening a therapeutic window for GDNF supplementation. We showed that after a single electrotransfer of 30 µg of GDNF-encoding plasmid in the rat ciliary muscle, GDNF was produced for at least 7 months. Morphometric, electroretinographic and optokinetic analyses highlighted that this continuous release of GDNF delayed photoreceptors (PRs) as well as retinal functions loss until at least 70 days of age in RCS rats. Unexpectedly, increasing the GDNF secretion level accelerated PR degeneration and the loss of electrophysiological responses. This is the first report: (i) demonstrating the efficacy of GDNF delivery through non-viral gene therapy in RP; (ii) establishing the efficacy of intravitreal administration of GDNF in RP associated with a mutation in the retinal pigment epithelium; and (iii) warning against potential toxic effects of GDNF within the eye/retina.


Subject(s)
Electroporation , Genetic Therapy/methods , Glial Cell Line-Derived Neurotrophic Factor/genetics , Retinitis Pigmentosa/therapy , Animals , Ciliary Neurotrophic Factor/metabolism , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Photoreceptor Cells, Vertebrate/physiology , Plasmids , Rats , Retinal Degeneration/therapy
11.
Eur J Pharm Biopharm ; 80(3): 544-52, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22155591

ABSTRACT

Cyclosporine A (CsA) has been demonstrated to be effective for the treatment of a variety of ophthalmological conditions, including ocular surface disorders such as the dry eye disease (DED). Since CsA is characterised by its low water solubility, the development of a topical ophthalmic formulation represents an interesting pharmaceutical question. In the present study, two different strategies to address this challenge were studied and compared: (i) a water-soluble CsA prodrug formulated within an aqueous solution and (ii) a CsA oil-in-water emulsion (Restasis, Allergan Inc., Irvine, CA). First, the prodrug formulation was shown to have an excellent ocular tolerance as well as no influence on the basal tear production; maintaining the ocular surface properties remained unchanged. Then, in order to allow in vivo investigations, a specific analytical method based on ultra high pressure liquid chromatography coupled with triple quadrupole mass spectrometer (UHPLC-MS/MS) was developed and optimised to quantify CsA in ocular tissues and fluids. The CsA ocular kinetics in lachrymal fluid for both formulations were found to be similar between 15 min and 48 h. The CsA ocular distribution study evidenced the ability of the prodrug formulation to penetrate into the eye, achieving therapeutically active CsA levels in tissues of both the anterior and posterior segments. In addition, the detailed analysis of the in vivo data using a bicompartmental model pointed out a higher bioavailability and lower elimination rate for CsA when it is generated from the prodrug than after direct application as an emulsion. The interesting in vivo properties displayed by the prodrug solution make it a safe and suitable option for the treatment of DED.


Subject(s)
Cyclosporine/chemistry , Cyclosporine/pharmacology , Dry Eye Syndromes/drug therapy , Prodrugs/chemistry , Prodrugs/pharmacology , Animals , Biological Availability , Chemistry, Pharmaceutical/methods , Cyclosporine/pharmacokinetics , Dry Eye Syndromes/metabolism , Emulsions/chemistry , Emulsions/pharmacokinetics , Emulsions/pharmacology , Eye/drug effects , Eye/metabolism , Female , Kinetics , Ophthalmic Solutions/chemistry , Prodrugs/pharmacokinetics , Rabbits , Rats , Rats, Inbred Lew , Solubility , Tears/drug effects , Water/chemistry
12.
Arch Pediatr ; 18 Suppl 2: S79-85, 2011 Jul.
Article in French | MEDLINE | ID: mdl-21763979

ABSTRACT

Retinopathy of prematurity (ROP) is a major cause of visual impairment in premature infants. It is characterized by an arrest in normal retinal vascular development associated with microvascular degeneration, followed by an abnormal hypoxiainduced neovascularization. Recent studies point out that ROP is a multifactorial disease, implicating both oxygen-dependent and oxygen-independent mechanisms. Oxygen-dependent factors leading to microvascular degeneration include generation of reactive oxygen species and suppression of specific oxygen-regulated vascular survival factors, such as vascular endothelial growth factor (VEGF) and erythropoietin. The other major mechanism for the initial capillary loss is oxygen-independent and implicates a deficit in growth factor IGF-1/IGFBP3. The proliferative, second phase of ROP is triggered by increases in vascular growth factors concentrations, in an attempt to compensate for the hypoxic retina. Novel signaling pathways for vascular repair, implicating both metabolite signaling and inflammatory lipids signaling, represent new therapeutic avenues for ROP.


Subject(s)
Retinopathy of Prematurity/epidemiology , Retinopathy of Prematurity/physiopathology , Humans , Infant, Newborn , Insulin-Like Growth Factor I/metabolism , Oxidative Stress/physiology , Oxygen Inhalation Therapy/adverse effects , Reactive Oxygen Species/metabolism , Retinal Neovascularization , Vascular Endothelial Growth Factor A/metabolism
13.
Prog Retin Eye Res ; 30(4): 239-57, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21600300

ABSTRACT

Light-emitting diodes (LEDs) are taking an increasing place in the market of domestic lighting because they produce light with low energy consumption. In the EU, by 2016, no traditional incandescent light sources will be available and LEDs may become the major domestic light sources. Due to specific spectral and energetic characteristics of white LEDs as compared to other domestic light sources, some concerns have been raised regarding their safety for human health and particularly potential harmful risks for the eye. To conduct a health risk assessment on systems using LEDs, the French Agency for Food, Environmental and Occupational Health & Safety (ANSES), a public body reporting to the French Ministers for ecology, for health and for employment, has organized a task group. This group consisted physicists, lighting and metrology specialists, retinal biologist and ophthalmologist who have worked together for a year. Part of this work has comprised the evaluation of group risks of different white LEDs commercialized on the French market, according to the standards and found that some of these lights belonged to the group risk 1 or 2. This paper gives a comprehensive analysis of the potential risks of white LEDs, taking into account pre-clinical knowledge as well as epidemiologic studies and reports the French Agency's recommendations to avoid potential retinal hazards.


Subject(s)
Eye Diseases/etiology , Light , Lighting/methods , Semiconductors , Animals , Biomass , Circadian Rhythm/physiology , Environmental Exposure , Eye Diseases/pathology , Eye Diseases/physiopathology , Humans , Light/adverse effects , Reflex, Pupillary/physiology , Retina/pathology , Risk Assessment , Time Factors
14.
Ann Pharm Fr ; 69(2): 100-7, 2011 Mar.
Article in French | MEDLINE | ID: mdl-21440102

ABSTRACT

Intravitreal administration has been widely used since 20 years and has been shown to improve the treatment of diseases of the posterior segment of the eye with infectious origin or in edematous maculopathies. This route of administration allows to achieve high concentration of drug in the vitreous and avoids the problems resulting from systemic administration. However, two basic problems limit the use of intravitreal therapy. Many drugs are rapidly cleared from the vitreous humor; therefore, to reach and to maintain effective therapy repeated injections are necessary. Repeated intravitreal injections increase the risk of endophthalmitis, damage to lens, retinal detachment. Moreover, some drugs provoke a local toxicity at their effective dose inducing side-effects and possible retinal lesions. In this context, the development and the use of new drug delivery systems for intravitreal administration are necessary to treat chronic ocular diseases. Among them, particulate systems such as liposomes have been widely studied. Liposomes are easily injectable and permit to reduce the toxicity and to increase the residence time of several drugs in the eye. They are also able to protect in vivo poorly-stable molecules from degradation such as peptides and nucleic acids. Some promising results have been obtained for the treatment of retinitis induced by cytomegalovirus in human and more recently for the treatment of uveitis in animal. Finally, the fate of liposomes in ocular tissues and fluids after their injection into the vitreous and their elimination routes begin to be more known.


Subject(s)
Drug Carriers , Eye Diseases/drug therapy , Liposomes , Pharmaceutical Preparations/administration & dosage , Vitreous Body/physiology , Drug Carriers/pharmacokinetics , Drug Delivery Systems , Drug-Related Side Effects and Adverse Reactions , Eye/metabolism , Humans , Intravitreal Injections , Liposomes/pharmacokinetics
15.
Ann Pharm Fr ; 69(2): 116-23, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21440104

ABSTRACT

Due to the eye's specific anatomical and physiological conformation, the treatment of eye diseases is a real challenge for pharmaceutical therapy. The presence of efficient protective barriers (i.e., the conjunctival and corneal membranes) and protective mechanisms (i.e., blinking and nasolachrymal drainage) makes this organ particularly impervious to local drug therapy. To overcome these issues, numerous strategies have been envisioned using pharmaceutical technology. Many formulations currently on the market or still under development are emulsions or colloidal systems intended to enhance precorneal residence time and corneal penetration, causing a consequent increase in drug bioavailability after instillation. After a review of some recent developments in the field of cyclosporin A formulations for the eye, a novel micellar formulation of cyclosporine A based on a diblock methoxy-poly(ethylene glycol)-hexysubstituted poly(lactides) (MPEG-hexPLA) is described.


Subject(s)
Anterior Eye Segment , Cyclosporine/administration & dosage , Animals , Chemistry, Pharmaceutical , Colloids , Cyclosporine/therapeutic use , Drug Delivery Systems , Emulsions , Humans , Micelles , Nanoparticles , Ophthalmic Solutions , Polyesters , Polyethylene Glycols
16.
Ann Pharm Fr ; 69(2): 124-30, 2011 Mar.
Article in French | MEDLINE | ID: mdl-21440105

ABSTRACT

Retinal effects of systemically administered drugs are rare due to the hematoretinal barriers that protect the retina from circulating active principles. However, some compounds may have direct or indirect toxic effects on the retina through direct interaction with a specific receptor or due to their accumulation within pigment of uveal cells. In the latter case, toxicity is dose-dependent and may be observed years after cessation of medication, as observed with antimalarial drugs. Anti-infective and anti-inflammatory agents, particularly glucocorticoids, are currently injected peri- or intraocularly. The mechanisms and the exact toxicity of glucocorticoids on the retina remain poorly understood. More recently, anti-VEGF has been specifically developed for the treatment of retinal diseases. However, the long-term blockade of VEGF on normal retinal physiology should be determined taking into account VEGF and VEGF receptors expression in the normal and pathologic retina. Whilst enormous advances are made in the treatment of retinal diseases, basic research is still required to define more accurately the molecular targets of drugs to improve their benefits and reduce their potential side effects.


Subject(s)
Retina/drug effects , Retinal Diseases/chemically induced , Retinal Diseases/drug therapy , Adrenal Cortex Hormones/adverse effects , Animals , Anti-Inflammatory Agents/pharmacology , Blood-Retinal Barrier/physiology , Drug-Related Side Effects and Adverse Reactions , Humans , Ocular Physiological Phenomena/drug effects , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors
17.
Prog Retin Eye Res ; 29(6): 443-65, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20398784

ABSTRACT

Because the eye is protected by ocular barriers but is also easily accessible, direct intravitreous injections of therapeutic proteins allow for specific and targeted treatment of retinal diseases. Low doses of proteins are required in this confined environment and a long time of residency in the vitreous is expected, making the eye the ideal organ for local proteic therapies. Monthly intravitreous injection of Ranibizumab, an anti-VEGF Fab has become the standard of care for patients presenting wet AMD. It has brought the proof of concept that administering proteins into the physiologically low proteic concentration vitreous can be performed safely. Other antibodies, Fab, peptides and growth factors have been shown to exert beneficial effects on animal models when administered within the therapeutic and safe window. To extend the use of such biomolecules in the ophthalmology practice, optimization of treatment regimens and efficacy is required. Basic knowledge remains to be increased on how different proteins/peptides penetrate into the eye and the ocular tissues, distribute in the vitreous, penetrate into the retinal layers and/or cells, are eliminated from the eye or metabolized. This should serve as a basis for designing novel drug delivery systems. The later should be non-or minimally invasive and should allow for a controlled, scalable and sustained release of the therapeutic proteins in the ocular media. This paper reviews the actual knowledge regarding protein delivery for eye diseases and describes novel non-viral gene therapy technologies particularly adapted for this purpose.


Subject(s)
Drug Delivery Systems/methods , Proteins/administration & dosage , Retinal Diseases/therapy , Animals , Drug Administration Routes , Drug Compounding/methods , Drug Delivery Systems/trends , Eye/anatomy & histology , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Proteins/metabolism , Retinal Diseases/drug therapy , Retinal Diseases/metabolism , Vitreous Body/metabolism
18.
Exp Parasitol ; 126(2): 259-62, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20412796

ABSTRACT

Ocular toxoplasmosis is the principal cause of posterior uveitis and a leading cause of blindness. Animal models are required to improve our understanding of the pathogenesis of this disease. The method currently used for the detection of retinal cysts in animals involves the observation, under a microscope, of all the sections from infected eyes. However, this method is time-consuming and lacks sensitivity. We have developed a rapid, sensitive method for observing retinal cysts in mice infected with Toxoplasma gondii. This method involves combining the flat-mounting of retina - a compromise between macroscopic observation and global analysis of this tissue - and the use of an avirulent recombinant strain of T. gondii expressing the Escherichia coli beta-galactosidase gene, visually detectable at the submacroscopic level. Single cyst unilateral infection was found in six out of 17 mice killed within 28 days of infection, whereas a bilateral infection was found in only one mouse. There was no correlation between brain cysts number and ocular infection.


Subject(s)
Retina/parasitology , Toxoplasma/isolation & purification , Toxoplasmosis, Ocular/parasitology , Animals , Brain/parasitology , Disease Models, Animal , Female , Frozen Sections , Mice , Toxoplasmosis, Ocular/diagnosis
19.
Gene Ther ; 16(7): 862-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19440225

ABSTRACT

Intraocular inflammation has been recognized as a major factor leading to blindness. Because tumor necrosis factor-alpha (TNF-alpha) enhances intraocular cytotoxic events, systemic anti-TNF therapies have been introduced in the treatment of severe intraocular inflammation, but frequent re-injections are needed and are associated with severe side effects. We have devised a local intraocular nonviral gene therapy to deliver effective and sustained anti-TNF therapy in inflamed eyes. In this study, we show that transfection of the ciliary muscle by plasmids encoding for three different variants of the p55 TNF-alpha soluble receptor, using electrotransfer, resulted in sustained intraocular secretion of the encoded proteins, without any detection in the serum. In the eye, even the shorter monomeric variant resulted in efficient neutralization of TNF-alpha in a rat experimental model of endotoxin-induced uveitis, as long as 3 months after transfection. A subsequent downregulation of interleukin (IL)-6 and iNOS and upregulation of IL-10 expression was observed together with a decreased rolling of inflammatory cells in anterior segment vessels and reduced infiltration within the ocular tissues. Our results indicate that using a nonviral gene therapy strategy, the local self-production of monomeric TNF-alpha soluble receptors induces a local immunomodulation enabling the control of intraocular inflammation.


Subject(s)
Ciliary Body/metabolism , Genetic Therapy/methods , Muscle, Smooth/metabolism , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Uveitis/therapy , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Electroporation/methods , Endotoxins/adverse effects , Eye/metabolism , Female , Gene Transfer Techniques , Genes, Reporter , Humans , Immunomodulation , Interleukin-10/metabolism , Interleukin-6/metabolism , Lac Operon/genetics , Leukocyte Rolling , Microscopy, Confocal , Nitric Oxide Synthase Type II/metabolism , Plasmids , Rats , Rats, Inbred Lew , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transfection/methods , Tumor Necrosis Factor Decoy Receptors/metabolism , Tumor Necrosis Factor-alpha/adverse effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
20.
Mol Vis ; 15: 670-84, 2009.
Article in English | MEDLINE | ID: mdl-19347052

ABSTRACT

PURPOSE: To analyze the influence of age on retinochoroidal wound healing processes and on glial growth factor and cytokine mRNA expression profiles observed after argon laser photocoagulation. METHODS: A cellular and morphometric study was performed that used 44 C57Bl/6J mice: 4-week-old mice (group I, n=8), 6-week-old mice (group II, n=8), 10-12-week-old mice (group III, n=14), and 1-year-old mice (group IV, n=14). All mice in these groups underwent a standard argon laser photocoagulation (50 microm, 400 mW, 0.05 s). Two separated lesions were created in each retina using a slit lamp delivery system. At 1, 3, 7, 14, 60 days, and 4 months after photocoagulation, mice from each of the four groups were sacrificed by carbon dioxide inhalation. Groups III and IV were also studied at 6, 7, and 8 months after photocoagulation. At each time point the enucleated eyes were either mounted in Tissue Tek (OCT), snap frozen and processed for immunohistochemistry or either flat mounted (left eyes of groups III and IV). To determine, by RT-PCR, the time course of glial fibrillary acidic protein (GFAP), vascular endothelial growth factor (VEGF), and monocyte chemotactic protein-1 (MCP-1) gene expression, we delivered ten laser burns (50 microm, 400 mW, 0.05 s) to each retina in 10-12-week-old mice (group III', n=10) and 1-year-old mice (group IV', n=10). Animals from Groups III' and IV' had the same age than those from Groups III and IV, but they received ten laser impacts in each eye and served for the molecular analysis. Mice from Groups III and IV received only two laser impacts per eye and served for the cellular and morphologic study. Retinal and choroidal tissues from these treated mice were collected at 16 h, and 1, 2, 3, and 7 days after photocoagulation. Two mice of each group did not receive photocoagulation and were used as controls. RESULTS: In the cellular and morphologic study, the resultant retinal pigment epithelium interruption expanse was significantly different between the four groups. It was more concise and smaller in the oldest group IV (112.1 microm+/-11.4 versus 219.1 microm+/-12.2 in group III) p<0.0001 between groups III and IV. By contrast, while choroidal neovascularization (CNV) was mild and not readily identifiable in group I, at all time points studied, CNV was more prominent in the (1-year-old mice) Group IV than in the other groups. For instance, up to 14 days after photocoagulation, CNV reaction was statistically larger in group IV than in group III ((p=0.0049 between groups III and IV on slide sections and p<0.0001 between the same groups on flat mounts). Moreover, four months after photocoagulation, the CNV area (on slide sections) was 1,282 microm(2)+/-90 for group III and 2,999 microm(2)+/-115 for group IV (p<0.0001 between groups III and IV). Accordingly, GFAP, VEGF, and MCP-1 mRNA expression profiles, determined by RT-PCR at 16 h, 1, 2, 3, and 7 days postphotocoagulation, were modified with aging. In 1-year-old mice (group IV), GFAP mRNA expression was already significantly higher than in the younger (10-12 week) group III before photocoagulation. After laser burns, GFAP mRNA expression peaked at 16-24 h and on day 7, decreasing thereafter. VEGF mRNA expression was markedly increased after photocoagulation in old mice eyes, reaching 2.7 times its basal level at day 3, while it was only slightly increased in young mice (1.3 times its level in untreated young mice 3 days postphotocoagulation). At all time points after photocoagulation, MCP-1 mRNA expression was elevated in old mice, reaching high levels of expression at 16 h and day 3 respectively. CONCLUSIONS: Our results were based on the study of four different age groups and included not only data from morphological observations but also from a molecular analysis of the various alterations of cytokine signaling and expression. One-year-old mice demonstrated more extensive CNV formation and a slower pace of regression after laser photocoagulation than younger mice. These were accompanied by differences in growth factors and cytokine expression profiles indicate that aging is a factor that aggravates CNV. The above results may provide some insight into possible therapeutic strategies in the future.


Subject(s)
Aging/pathology , Argon , Choroid/pathology , Laser Coagulation , Retina/pathology , Retina/surgery , Wound Healing , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Choroid/blood supply , Choroidal Neovascularization/pathology , Choroidal Neovascularization/surgery , Gene Expression Regulation , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/surgery , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...