Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Ophthalmology ; 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38336282

RESUMEN

PURPOSE: To identify factors for meeting prespecified criteria for switching from bevacizumab to aflibercept in eyes with center-involved diabetic macular edema (CI-DME) and moderate vision loss initially treated with bevacizumab in DRCR Retina Network protocol AC. DESIGN: Post hoc analysis of data from a randomized clinical trial. PARTICIPANTS: Two hundred seventy participants with one or both eyes harboring CI-DME with visual acuity (VA) letter score of 69 to 24 (Snellen equivalent, 20/50-20/320). METHODS: Eligible eyes were assigned to receive intravitreal aflibercept monotherapy (n = 158) or bevacizumab followed by aflibercept if prespecified criteria for switching were met between 12 weeks and 2 years (n = 154). MAIN OUTCOME MEASURES: Meeting switching criteria: (1) at any time, (2) at 12 weeks, and (3) after 12 weeks. Associations between meeting the criteria for switching and factors measured at baseline and 12 weeks were evaluated in univariable analyses. Stepwise procedures were used to select variables for multivariable models. RESULTS: In the group receiving bevacizumab first, older participants showed a higher risk of meeting the switching criteria at any time, with a hazard ratio (HR) for a 10-year increase in age of 1.32 (95% confidence interval [CI], 1.11-1.58). Male participants or eyes with worse baseline VA were more likely to switch at 12 weeks (for male vs. female: odds ratio [OR], 4.84 [95% CI, 1.32-17.81]; 5-letter lower baseline VA: OR, 1.30 [95% CI, 1.03-1.63]). Worse 12-week central subfield thickness (CST; 10-µm greater: HR, 1.06 [95% CI, 1.04-1.07]) was associated with increased risk of switching after 12 weeks. The mean ± standard deviation improvement in visual acuity after completing the switch to aflibercept was 3.7 ± 4.9 letters compared with the day of switching. CONCLUSIONS: The identified factors can be used to refine expectations regarding the likelihood that an eye will meet protocol criteria to switch to aflibercept when treatment is initiated with bevacizumab. Older patients are more likely to be switched. At 12 weeks, thicker CST was predictive of eyes most likely to be switched in the future. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.

2.
Retina ; 40(6): 1021-1028, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31567817

RESUMEN

PURPOSE: To investigate whether anti-vascular endothelial growth factor (anti-VEGF) for diabetic macular edema or proliferative diabetic retinopathy (PDR) increases the risk of traction retinal detachment (TRD) among eyes with PDR. METHODS: Pooled analysis of PDR eyes from Protocols I, J, N, S, or T with Early Treatment Diabetic Retinopathy Study level ≥61 (prompt vitrectomy was not planned) randomly assigned to the control group (laser photocoagulation, sham, or intravitreal saline; 396 eyes) or anti-VEGF (487 eyes). The primary outcome was investigator-identified TRD within 1 year of randomization. RESULTS: The 1-year cumulative probability of TRD was 6.8% (95% confidence interval: 4.6%-9.9%, 25 events) in control-group eyes and 4.8% (95% confidence interval: 3.2%-7.3%, 22 events) in anti-VEGF group eyes (hazard ratio = 0.95 [95% confidence interval: 0.54-1.66, P = 0.86]). The cumulative probability of vitrectomy for TRD was 4.4% (16 events) in control-group eyes and 2.2% (9 events) in anti-VEGF group eyes (P = 0.19). Percentage with TRD and vitrectomy for TRD were similar within strata of diabetic retinopathy severity. CONCLUSION: These findings do not support the hypothesis that anti-VEGF therapy for diabetic macular edema or PDR increases the risk of TRD among eyes with PDR similar to those enrolled in five DRCR Retina Network protocols for which prompt vitrectomy was not planned.


Asunto(s)
Retinopatía Diabética/complicaciones , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Retina/patología , Desprendimiento de Retina/tratamiento farmacológico , Agudeza Visual , Inhibidores de la Angiogénesis , Retinopatía Diabética/diagnóstico , Retinopatía Diabética/tratamiento farmacológico , Femenino , Angiografía con Fluoresceína/métodos , Fondo de Ojo , Humanos , Inyecciones Intravítreas , Masculino , Persona de Mediana Edad , Desprendimiento de Retina/diagnóstico , Desprendimiento de Retina/etiología , Tomografía de Coherencia Óptica/métodos , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
3.
Invest Ophthalmol Vis Sci ; 54(2): 1392-7, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23341018

RESUMEN

PURPOSE: Proliferative vitreoretinopathy (PVR) is a complication of retinal detachment that can lead to surgical failure and vision loss. Previous studies suggest that a variety of retinal cells, including RPE and Müller glia, may be responsible. Platelet-derived growth factor receptor alpha (PDGFRα) has been strongly implicated in the pathogenesis, and found to be intrinsic to the development of PVR in rabbit models. We examine whether SU9518, a tyrosine kinase inhibitor with PDGFRα specificity, can inhibit the development of PVR in fibroblast and Müller cell rabbit models of PVR. METHODS: SU9518 was injected in rabbit eyes along with fibroblasts, Müller cells (MIO-M1), or Müller cells transfected to increase their expression of PDGFRα (MIO-M1α). Indirect ophthalmoscopy and histopathology were used to assess efficacy and toxicity. RESULTS: SU9518 was an effective inhibitor of PVR in both fibroblast and Müller cell models of PVR. No toxic effects were identified by indirect ophthalmoscopy or histopathology. CONCLUSIONS: SU9518 is an effective and safe inhibitor of PVR in rabbit models, and could potentially be used in humans for the treatment of this and other proliferative diseases of the retina involving fibrosis and gliosis. Further animal studies need to be performed to examine retinal toxicity and sustained delivery mechanisms.


Asunto(s)
Indoles/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Epitelio Pigmentado de la Retina/patología , Vitreorretinopatía Proliferativa/tratamiento farmacológico , Cuerpo Vítreo/patología , Animales , División Celular , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Humanos , Conejos , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Vitreorretinopatía Proliferativa/metabolismo , Vitreorretinopatía Proliferativa/patología , Cuerpo Vítreo/efectos de los fármacos , Cuerpo Vítreo/metabolismo
4.
Stem Cells Int ; 2012: 106486, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22966235

RESUMEN

Purpose. Proliferative vitreoretinopathy (PVR) is a complication of retinal detachment characterized by redetachment of the retina as a result of membrane formation and contraction. A variety of retinal cells, including retinal pigment epithelial (RPE) and Müller glia, and growth factors may be responsible. Platelet-derived growth factor receptor alpha (PDGFRα) is found in large quantities in PVR membranes, and is intrinsic to the development of PVR in rabbit models. This study explores the expression of PDGFR in cocultures of RPE and Müller cells over time to examine how these two cell types may collaborate in the development of PVR. We also examine how changes in PDGFRα expression alter Müller cell pathogenicity. Methods. Human MIO-M1 Müller progenitor (MPC) and ARPE19 cells were studied in a transmembrane coculture system. Immunocytochemistry and Western blot were used to look at PDGFRα, PDGFRß, and GFAP expression. A transfected MPC line cell line expressing the PDGFRα (MIO-M1α) was generated, and tested in a rabbit model for its ability to induce PVR. Results. The expression of PDGFRα and PDGFRß was upregulated in MIO-M1 MPCs cocultured with ARPE19 cells; GFAP was slightly decreased. Increased expression of PDGFRα in the MIO-M1 cell line resulted in increased pathogenicity and enhanced ability to induce PVR in a rabbit model. Conclusions. Müller and RPE cell interaction can lead to upregulation of PDGFRα and increased Müller cell pathogenicity. Müller cells may play a more active role than previously thought in the development of PVR membranes, particularly when stimulated by an RPE-cell-rich environment. Additional studies of human samples and in animal models are warranted.

5.
Invest Ophthalmol Vis Sci ; 52(9): 5016-21, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21642621

RESUMEN

PURPOSE: Previous studies indicate that the expression of platelet-derived growth factor (PDGF) receptor α (PDGFRα) dramatically increases the ability of fibroblasts to induce experimental proliferative vitreoretinopathy (PVR). The purpose of this study was to determine whether PDGFRα contributed to the PVR potential of retinal pigment epithelial (RPE) cells, one of the most abundant cell types in PVR membranes. METHODS: PDGFRα expression in human ARPE19 cells was increased or decreased by stably expressing the PDGFRα cDNA or short hairpin (sh) RNA directed at PDGFRα, respectively. The level of PDGFRα expression in the resulting panel of cell lines was either barely detectable (KD), standard (similar to the level of primary RPE cells), or overexpressed approximately 80-fold. Western blot analysis was used to assess the level of p53 and the activation state of PDGFRα and Akt. The following cellular responses were monitored: proliferation, apoptosis, and contraction. The PVR potential of cells was tested in a rabbit model of PVR in which cells were coinjected with platelet-rich plasma into the vitreous. RESULTS: Comparison of KD and overexpressing cells indicated that high-level expression of PDGFRα dramatically augmented signaling events, cellular responses, and the PVR potential of ARPE19 cells. However, all these outcomes were also significantly increased, albeit not as robustly, by PDGFRα expression to the level typically present in RPE cells. CONCLUSIONS: Even though RPE cells express substantially less PDGFRα than fibroblasts, it significantly boosts PVR-related signaling events, cellular responses, and the PVR potential of ARPE19 cells. These studies suggest that inhibiting activation, signaling, or both by PDGFRα has the potential to prevent the development of PVR.


Asunto(s)
Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Epitelio Pigmentado de la Retina/fisiología , Vitreorretinopatía Proliferativa/genética , Vitreorretinopatía Proliferativa/fisiopatología , Animales , Apoptosis/fisiología , División Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica/fisiología , Humanos , Plasma Rico en Plaquetas , Conejos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Epitelio Pigmentado de la Retina/citología , Transducción de Señal/fisiología , Vitreorretinopatía Proliferativa/patología , Cuerpo Vítreo/metabolismo , Cuerpo Vítreo/patología
6.
Mol Cell Biol ; 31(9): 1788-99, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21357737

RESUMEN

In contrast to direct activation of platelet-derived growth factor (PDGF) receptor α (PDGFRα) via PDGF, indirect activation via growth factors outside the PDGF family failed to induce dimerization, internalization, and degradation of PDGFRα. Chronically activated, monomeric PDGFRα induced prolonged activation of Akt and suppressed the level of p53. These events were sufficient to promote both cellular responses (proliferation, survival, and contraction) that are intrinsic to proliferative vitreoretinopathy (PVR) and induce the disease itself. This signature signaling pathway appeared to extend beyond PVR since deregulating PDGFRα in ways that promote solid tumors also resulted in chronic activation of Akt and a decline in the level of p53.


Asunto(s)
Fibroblastos/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Vitreorretinopatía Proliferativa/metabolismo , Animales , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Conejos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/análisis , Proteínas Recombinantes/metabolismo , Transducción de Señal , Vitreorretinopatía Proliferativa/patología
7.
Am J Pathol ; 177(1): 132-40, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20489144

RESUMEN

Proliferative vitreoretinopathy (PVR) is a complication that develops in 5% to 10% of patients who undergo surgery to correct a detached retina. The only treatment option for PVR is surgical intervention, which has a limited success rate that diminishes in patients with recurring PVR. Our recent studies revealed that antioxidants prevented intracellular signaling events that were essential for experimental PVR. The purpose of this study was to test whether N-acetyl-cysteine (NAC), an antioxidant used in a variety of clinical settings, was capable of protecting rabbits from PVR. Vitreous-driven activation of PDGFRalpha and cellular responses intrinsic to PVR (contraction of collagen gels and cell proliferation) were blocked by concentrations of NAC that were well below the maximum tolerated dose. Furthermore, intravitreal injection of NAC effectively protected rabbits from developing retinal detachment, which is the sight-robbing phase of PVR. Finally, these observations with an animal model appear relevant to clinical PVR because NAC prevented human PVR vitreous-induced contraction of primary RPE cells derived from a human PVR membrane. Our observations demonstrate that antioxidants significantly inhibited experimental PVR, and suggest that antioxidants have the potential to function as a PVR prophylactic in patients undergoing retinal surgery to repair a detached retina.


Asunto(s)
Acetilcisteína/farmacología , Antioxidantes/uso terapéutico , Modelos Animales de Enfermedad , Depuradores de Radicales Libres/farmacología , Desprendimiento de Retina/prevención & control , Vitreorretinopatía Proliferativa/tratamiento farmacológico , Animales , Proliferación Celular , Células Cultivadas , Humanos , Conejos , Especies Reactivas de Oxígeno/metabolismo , Desprendimiento de Retina/complicaciones , Desprendimiento de Retina/cirugía , Epitelio Pigmentado de la Retina/citología , Vitreorretinopatía Proliferativa/etiología , Vitreorretinopatía Proliferativa/patología , Cuerpo Vítreo/metabolismo
8.
Invest Ophthalmol Vis Sci ; 50(7): 3394-403, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19324843

RESUMEN

PURPOSE: Proliferative vitreoretinopathy (PVR) is a recurring and problematic disease for which there is no pharmacologic treatment. Platelet-derived growth factor (PDGF) in the vitreous is associated with experimental and clinical PVR. Furthermore, PDGF receptors (PDGFRs) are present and activated in epiretinal membranes of patient donors, and they are essential for experimental PVR. These observations suggest that PVR arises at least in part from PDGF/PDGFR-driven events. The goal of this study was to determine whether PDGFs were a potential therapeutic target for PVR. METHODS: Experimental PVR was induced in rabbits by injecting fibroblasts. Vitreous specimens were collected from experimental rabbits or from patients undergoing vitrectomy to repair retinal detachment. A neutralizing PDGF antibody and a PDGF Trap were tested for their ability to prevent experimental PVR. Activation of PDGFR was monitored by antiphosphotyrosine Western blot analysis of immunoprecipitated PDGFRs. Contraction of collagen gels was monitored in vitro. RESULTS: Neutralizing vitreal PDGFs did not effectively attenuate PVR, even though the reagents used potently blocked PDGF-dependent activation of the PDGF alpha receptor (PDGFRalpha). Vitreal growth factors outside the PDGF family modestly activated PDGFRalpha and appeared to do so without engaging the ligand-binding domain of PDGFRalpha. This indirect route to activate PDGFRalpha had profound functional consequences. It promoted the contraction of collagen gels and appeared sufficient to drive experimental PVR. CONCLUSIONS: Although PDGF appears to be a poor therapeutic target, PDGFRalpha is particularly attractive because it can be activated by a much larger spectrum of vitreal growth factors than previously appreciated.


Asunto(s)
Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Vitreorretinopatía Proliferativa/metabolismo , Cuerpo Vítreo/metabolismo , Animales , Anticuerpos Monoclonales/uso terapéutico , Western Blotting , Técnicas de Cultivo de Célula , Colágeno Tipo I/metabolismo , Conjuntiva/citología , Modelos Animales de Enfermedad , Membrana Epirretinal/metabolismo , Membrana Epirretinal/prevención & control , Fibroblastos/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Conejos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , Tirosina/metabolismo , Vitrectomía , Vitreorretinopatía Proliferativa/prevención & control
9.
Invest Ophthalmol Vis Sci ; 49(1): 42-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18172073

RESUMEN

PURPOSE: Proliferative vitreoretinopathy (PVR) is the primary cause of failure of retinal reattachment surgery. Growth factors such as platelet-derived growth factor (PDGF) are strongly associated with PVR. Of the five PDGF family members, PDGF-C predominates in the vitreous of experimental and clinical PVR. PDGF-C is secreted as a latent protein that requires proteolytic processing for activation. Although tissue plasminogen activator (tPA) is primarily responsible for processing PDGF-C in cultured cells, it constitutes a minority of the processing activity in the vitreous of experimental animals and in patients with PVR. Identifying the major PDGF-C processing protease was the purpose of this study. METHODS: The presence of serum proteins in the vitreous was detected by Coomassie blue staining and Western blotting. PDGF-C processing activity was detected in an in vitro processing assay using either native or recombinant PDGF-C as the substrate. Plasmin activity was blocked using alpha(2)-plasmin inhibitor. Phosphorylation of the PDGF receptor (PDGFR) was monitored by antiphosphotyrosine Western blotting. Vitreous specimens were collected from experimental rabbits or from patients undergoing vitrectomy to repair retinal detachment or for other reasons. RESULTS: A number of prominent serum proteins (albumin and IgG) were detected in the vitreous of all patients undergoing retinal surgery. The level of these proteins markedly increased in the vitreous of rabbits as they developed PVR. These observations suggested that serum-borne proteases are also likely to be present in the vitreous. Indeed, plasmin (a protease capable of processing PDGF-C) was present in the vitreous from PVR rabbits and retinal surgery patients. Plasmin was dramatically more effective than tPA in processing PDGF-C in an in vitro assay. Blocking plasmin activity eliminated most of the processing activity in the vitreous of patients and rabbits with PVR. CONCLUSIONS: Plasmin was the major PDGF-C processing protease in the vitreous of PVR rabbits and patients undergoing retinal surgery. Blocking plasmin prevented the generation of active PDGF-C, which is the major PDGF isoform relevant for PVR. These observations are the first report of an in vivo protease responsible for processing PDGF-C. In addition, plasmin was identified as a novel therapeutic target for patients with PVR.


Asunto(s)
Fibrinolisina/metabolismo , Fibrinolíticos/metabolismo , Linfocinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Vitreorretinopatía Proliferativa/metabolismo , Cuerpo Vítreo/metabolismo , Animales , Antifibrinolíticos/farmacología , Proteínas Sanguíneas/metabolismo , Western Blotting , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Fibrinolisina/antagonistas & inhibidores , Humanos , Immunoblotting , Inmunoglobulina G/metabolismo , Inmunoprecipitación , Fosforilación , Conejos , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Activador de Tejido Plasminógeno/metabolismo , alfa 2-Antiplasmina/farmacología
10.
Ocul Immunol Inflamm ; 15(2): 63-70, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17558830

RESUMEN

PURPOSE: To investigate the safety and efficacy of daclizumab (Zenapax, humanized anti-Tac, HAT) in controlling the ocular manifestations of Behçet's disease. DESIGN: Randomized, placebo-controlled, double-masked clinical trial. PARTICIPANTS: Seventeen participants with Behçet's disease experiencing at least two prior ocular attacks and requiring treatment with immunosuppressive agents for the ocular complications of Behçet's disease. METHODS: Participants received either intravenous placebo or daclizumab (1 mg/kg) infusions every two weeks for six weeks, then every four weeks while continuing their standard immunosuppressive regimens. If clinically indicated, tapering of the standard immunosuppressive medications was allowed after six months of study enrollment. Complete ocular and physical examinations and an adverse event assessment were performed at baseline and prior to each study infusion. MAIN OUTCOME MEASURES: Primary safety endpoints were the development of a life-threatening complication or a severe opportunistic infection. Primary efficacy outcomes were the number of ocular attacks and an assessment of systemic immunosuppressive medications required during the study, including the ability to taper concomitant immunosuppressive therapy. RESULTS: Nine participants randomized to daclizumab and eight to placebo were followed monthly. Follow-up ranged from one to 34 months, with a median follow-up of 15 months. Two participants randomized to daclizumab discontinued study therapy prior to the end of the study for personal reasons. No participant experienced a safety endpoint, and visual acuity remained stable in all participants during the course of the study. Ten participants (six daclizumab, four placebo) experienced ocular attacks requiring therapy. The median ocular attack rate during the study was greater in the daclizumab arm than the placebo arm (median 1.27 vs. 0.17 attacks/year, respectively). Participants in the placebo arm also experienced a greater reduction in the immunosuppressive medication score compared to participants receiving daclizumab (median -4.0 vs. -1.0, respectively). CONCLUSIONS: The observed results in the placebo group demonstrate that careful follow-up and treatment with standard combination immunosuppressive therapy can be effective for the management of the ocular complications of Behçet's disease. In our small study, there was no suggestion that daclizumab was beneficial in comparison with placebo. However, the low observed attack rate limited our ability to make a definitive treatment group comparison.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Síndrome de Behçet/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Inmunosupresores/uso terapéutico , Uveítis/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Síndrome de Behçet/diagnóstico , Niño , Daclizumab , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Angiografía con Fluoresceína , Estudios de Seguimiento , Fondo de Ojo , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/efectos adversos , Inmunosupresores/administración & dosificación , Inmunosupresores/efectos adversos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Factores de Tiempo , Resultado del Tratamiento , Uveítis/diagnóstico
11.
Invest Ophthalmol Vis Sci ; 48(5): 2335-42, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17460299

RESUMEN

PURPOSE: Proliferative vitreoretinopathy (PVR) is a disorder characterized by the formation of cellular membranes on both surfaces of the retina and within the vitreous cavity. It occurs in 5% to 10% of patients who undergo retinal reattachment surgery. In the rabbit model of the disease, the platelet-derived growth factor alpha receptor (PDGFRalpha) is dramatically more capable of promoting PVR than is closely related PDGFRbeta. To test the ligand hypothesis (i.e., that this phenomenon can be explained by a predominance of PDGFRalpha-specific ligands) this study was conducted to determine the profile of PDGF ligands expressed by cells that induce PVR and in the vitreous of rabbits that have PVR. In addition, we examined which PDGF isoforms were present in the vitreous of patients with PVR, to assess the relevance of the rabbit model to the clinical setting. METHODS: PDGF isoforms were detected and quantified by Western blot analysis and ELISA. An assay was performed of conditioned medium from mouse embryo fibroblasts expressing the PDGFRalpha (Falpha) and rabbit conjunctival fibroblasts (RCFs), both of which cause PVR in the experimental model, and from human retinal pigment epithelial cells (ARPE-19). Because PDGF-C is secreted in a latent form and must be proteolytically processed to become biologically active, a PDGF-C processing assay was established, and conditioned medium was tested from these cells lines, for processing activity. Vitreous specimens, from control and PVR rabbits and from patients undergoing vitrectomy surgery, either to repair retinal detachment or for other reasons, were also tested for PDGF isoforms and for PDGF-C processing activity. RESULTS: PDGF isoforms that activate PDGFRbeta (PDGF-B and -D) were either undetectable or were present at very low levels in all the samples tested. Relatively low levels of PDGF-A and -AB were detected, whereas PDGF-C was the predominant isoform. Falpha, RCFs, and ARPE-19 cells accumulated PDGF-C in the conditioned medium at an average rate of 2.0 +/- 0.2, 2.9 +/- 0.3, and 71.3 +/- 6.0 ng/mL per day, respectively. Although there was no detectable PDGF-C in the vitreous of control rabbits (n = 8), there was an average of 1784 +/- 1150 ng/mL latent PDGF-C in the vitreous from rabbits with PVR (n= 14). Of the patients with PVR, eight of nine contained PDGF-C (range, 50-1000 ng/mL). In contrast, PDGF-C was detected in only 1 of 16 of the patients without PVR. In both conditioned medium and vitreous samples, the latent (instead of the active) form of PDGF-C was detected, even though processing activity was present in all the samples tested. CONCLUSIONS: The predominance of PDGF isoforms that activate PDGFRalpha support the ligand hypothesis as an explanation of why PDGFRalpha is more capable of inducing PVR than is PDGFRbeta. Furthermore, the profile of PDGF isoforms observed in the rabbit model accurately reflected the clinical specimens from patients with PVR. Finally, these findings implicate one of the new PDGF family members as an important contributor to experimental and clinical PVR.


Asunto(s)
Linfocinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Vitreorretinopatía Proliferativa/metabolismo , Animales , Western Blotting , Técnicas de Cultivo de Célula , Conjuntiva/citología , Modelos Animales de Enfermedad , Embrión de Mamíferos/citología , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/metabolismo , Humanos , Immunoblotting , Ligandos , Ratones , Ratones Noqueados , Epitelio Pigmentado Ocular/metabolismo , Isoformas de Proteínas/metabolismo , Conejos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Cuerpo Vítreo/metabolismo
12.
J Autoimmun ; 21(3): 283-93, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14599854

RESUMEN

Therapy for severe uveitis is frequently long-term immunosuppression using systemic corticosteroids and cytotoxic agents, but side effects make long-term therapy difficult. A long-term (>4 year) Phase I/II single armed interventional study using intravenous anti-IL-2 receptor alpha treatments (daclizumab) and a short-term Phase II study evaluating the use of a subcutaneous daclizumab formulation were conducted. Patients were tapered off their systemic immunosuppressive therapy and received daclizumab infusions or subcutaneous injections at intervals varying from 2 to 6 weeks. In the long-term study, seven of ten enrolled patients were tapered from their original immunosuppressive medications and maintained exclusively on repeated daclizumab infusions for control of their uveitis for over 4 years. No patient was permanently removed from therapy for an adverse event ascribed to the medication. The use of 6-week infusion intervals led to recurrence of uveitis, while 2- to 4-week intervals did not. Only one patient developed measurable anti-daclizumab antibodies but this disappeared when subcutaneous therapy was begun. In the short-term study, four of the five patients receiving the subcutaneous formulation met the study endpoints for success within the first 12 weeks. All five were successful by 26 weeks. These studies provide preliminary evidence that regularly administered long-term daclizumab therapy can be given in lieu of standard immunosuppression for years to treat severe uveitis and that subcutaneously administered daclizumab appeared to be a clinically viable treatment strategy. These studies suggest that anti-IL-2 receptor blockade could be useful in the treatment of Th1-mediated autoimmune conditions.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoglobulina G/uso terapéutico , Inmunosupresores/uso terapéutico , Uveítis/tratamiento farmacológico , Adolescente , Adulto , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales Humanizados , Antígenos CD/análisis , Apoptosis/efectos de los fármacos , Enfermedades Autoinmunes/tratamiento farmacológico , Daclizumab , Femenino , Citometría de Flujo , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/sangre , Inmunohistoquímica , Inmunosupresores/administración & dosificación , Inmunosupresores/sangre , Infusiones Intravenosas , Inyecciones Subcutáneas , Subunidad alfa del Receptor de Interleucina-2 , Ganglios Linfáticos/química , Masculino , Persona de Mediana Edad , Selección de Paciente , Receptores de Interleucina/inmunología , Receptores de Interleucina-2/análisis , Receptores de Interleucina-2/inmunología , Linfocitos T/química , Linfocitos T/inmunología , Resultado del Tratamiento , Agudeza Visual/efectos de los fármacos
13.
Ophthalmic Surg Lasers ; 33(4): 329-33, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12134997

RESUMEN

A 70-year-old patient with recurrent bilateral primary intraocular lymphoma (PIOL) was treated with local injections of methotrexate and periocular dexamethasone phosphate to both eyes over the course of 5 months. Local therapy consisted of a cycle to each eye of 3 intravitreal injections of methotrexate (200 microg in a total volume of 0.1 cc) administered on days 1, 5, and 8, followed by a subtenon injection of dexamethasone phosphate (7.5 mg in a total volume of 0.3 cc) on day 9. This treatment cycle was administered 4 times for the right eye and 3 times for the left eye, at 4 to 6 week intervals. Electroretinography was used to assess retinal function prior to and during each treatment regimen. Complete regression of the lymphomatous infiltrates and resolution of the vitritis was observed with preservation of visual acuity and no changes on electroretinography. The effect was sustained for 24 months after termination of treatment in the absence of systemic chemotherapy, radiation treatments, or maintenance intravitreal injections. Local combined chemotherapy can be used to treat recurrent PIOL, and can serve as successful palliative therapy in patients for whom further treatment with systemic chemotherapy or radiation is contraindicated.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Ojo/tratamiento farmacológico , Linfoma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Anciano , Antimetabolitos Antineoplásicos/administración & dosificación , Antineoplásicos Hormonales/administración & dosificación , Dexametasona/administración & dosificación , Neoplasias del Ojo/patología , Femenino , Fondo de Ojo , Humanos , Linfoma/patología , Metotrexato/administración & dosificación , Recurrencia Local de Neoplasia/patología , Resultado del Tratamiento
14.
Retina ; 22(1): 37-43, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11884876

RESUMEN

PURPOSE: Primary intraocular lymphoma (PIOL), also known as primary central nervous system lymphoma, is a rare yet blinding and fatal disease. Often presenting with ocular involvement, it can masquerade as posterior or intermediate uveitis, thus delaying diagnosis. A noninvasive ancillary test such as fluorescein angiography could be helpful in raising the level of suspicion in the diagnosis of this disease. METHODS: Results of fluorescein angiography (FA) and clinical characteristics of 17 patients (31 eyes) who presented to the National Eye Institute with the diagnosis of PIOL (confirmed by histopathologic analysis) were reviewed. RESULTS: The most common angiographic characteristics included disturbances at the level of the retinal pigment epithelium (RPE), such as granularity (19 eyes [61%]), blockage (17 eyes [55%]), and late staining (14 eyes [45%]). These changes are well correlated to histopathologic findings of lymphoma cells located between the RPE and Bruchs membrane. Perivascular staining or leakage and cystoid macular edema were rare. Other less common findings included pigment epithelial detachments and punctate hyperfluorescent lesions. Clinical characteristics found in eyes for which results of FA were available included vitreitis (29 eyes [94%]), subretinal infiltrates (19 eyes [61%]), and anterior chamber cells (10 eyes [32%]). In some cases, clinical examination did not correlate with FA findings. CONCLUSIONS: Although PIOL may present with a normal angiographic phenotype, extensive RPE changes demonstrated by FA, combined with the absence of perivascular staining or leakage and macular edema, may be associated with and distinctive of PIOL.


Asunto(s)
Angiografía con Fluoresceína , Linfoma de Células B/diagnóstico , Neoplasias de la Retina/diagnóstico , Adulto , Anciano , Humanos , Edema Macular/diagnóstico , Persona de Mediana Edad , Epitelio Pigmentado Ocular/patología , Cuerpo Vítreo/patología
15.
Semin Ophthalmol ; 17(3-4): 187-95, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12759849

RESUMEN

Both cataract and age-related macular degeneration (ARMD) are common findings in our aging population. At issue is the progression of ARMD following cataract extraction. The purpose of this paper is to review the data relevant to this subject and to offer suggestions as to the management of cataract associated with macular degeneration. Published data are reviewed and an attempt is made to interpret the results. Topics discussed include the epidemiology and association between the incidence of cataract and ARMD, the role of photo-oxidative damage and inflammation in the pathogenesis of ARMD, and the possible contribution of cataract extraction to the exacerbation and acceleration of ARMD through increased photo-oxidative damage and intraocular inflammation. Recommendations for the cataract surgeon and specialist are formulated on the basis of existing literature.


Asunto(s)
Extracción de Catarata/efectos adversos , Degeneración Macular , Catarata/complicaciones , Catarata/fisiopatología , Progresión de la Enfermedad , Humanos , Degeneración Macular/diagnóstico , Degeneración Macular/etiología , Degeneración Macular/fisiopatología , Degeneración Macular/terapia , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...