Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
PLoS One ; 19(1): e0294526, 2024.
Article in English | MEDLINE | ID: mdl-38241211

ABSTRACT

PURPOSE: VitreoDx is an experimental device enabling push-button collection of a neat vitreous liquid biopsy incidental to an intravitreal injection. We explored the ability of the device to collect a sample usable for proteomic biomarker discovery and testing. DESIGN: Pilot study using ex vivo human eyes. METHODS: Non-vitrectomized, human eyes from nine donors 75-91 years of age were refrigerated in BSS and used within 5 days of death. Four VitreoDx devices fitted with 25G needles, and four staked needle insulin syringes with 30G needles, were inserted at equal intervals through the pars plana of each eye and held in place by a fixture. The sampling mode of each VitreoDx device was triggered to attempt to acquire a liquid biopsy up to 70 µL. The plunger of each insulin syringe was retracted to attempt to obtain a liquid biopsy with a maximum volume of 50 µL. Samples acquired with the VitreoDx were extracted to polypropylene cryovials, refrigerated to -80 ºC, and sent for offsite proteomic analysis by proximity extension assay with a focus on panels containing approved and pipelined drug targets for neovascular disease and inflammatory factors. RESULTS: Of the attempted liquid biopsies with the novel 25G VitreoDx, 92% (66 of 72) resulted in successful acquisition (>25 µL) while 89% (64 of 72) attempted by a traditional 30G needle resulted in a successful acquisition. Sample volume sufficient for proteomics array analysis was acquired by the VitreoDx for every eye. Detectable protein was found for 151 of 166 unique proteins assayed in at least 25% of eyes sampled by VitreoDx. CONCLUSIONS: The high acquisition rate achieved by the prototype was similar to that achieved in previous clinical studies where a standard syringe was used with a 25G needle to biopsy vitreous fluid directly prior to standard intravitreal injection. Successful aspiration rates were likewise high for 30G needles. Together, these suggest that it is possible to routinely acquire liquid vitreous biopsies from patients who typically receive intravitreal injections with an injection device using a standard size needle without a vitreous cutter. Protein analysis shows that proteins of interest survive the sampling mechanism and may have potential to direct care in the future.


Subject(s)
Insulins , Proteomics , Humans , Infant, Newborn , Intravitreal Injections , Feasibility Studies , Pilot Projects , Vitreous Body/metabolism , Biopsy , Needles , Liquid Biopsy , Insulins/metabolism
2.
Curr Opin Ophthalmol ; 33(3): 131-136, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35266895

ABSTRACT

PURPOSE OF REVIEW: This review provides background on the remaining unmet needs with antivascular endothelial growth factor (VEGF) therapies for the treatment of neovascular age-related macular degeneration (nAMD). We also discuss the developmental story of the Port Delivery System with ranibizumab (PDS; SUSVIMO, Genentech, Inc., South San Francisco, CA, USA). RECENT FINDINGS: Real-world studies have shown that undertreatment is a major reason for continued vision loss in the anti-VEGF era. As a result, there is a need for long-acting anti-VEGF treatment options for patients with nAMD, diabetic macular edema, and other retinal diseases. The PDS is a solid state, refillable, intraocular long-acting drug delivery system that continuously delivers a customized formulation of ranibizumab into the vitreous for 6 months. In a phase 3 trial, the PDS showed equivalent visual acuity improvements with monthly ranibizumab injections in patients with nAMD and adverse events associated with the PDS were well understood and manageable. SUMMARY: The PDS is the first US Food and Drug Administration-approved treatment for nAMD that provides continuous delivery of an anti-VEGF molecule. The PDS offers a unique drug delivery system that has the potential to serve as a platform to be used with other molecules in the future.


Subject(s)
Diabetic Retinopathy , Macular Degeneration , Macular Edema , Wet Macular Degeneration , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Diabetic Retinopathy/drug therapy , Humans , Intravitreal Injections , Macular Degeneration/drug therapy , Macular Edema/drug therapy , Ranibizumab/therapeutic use , Treatment Outcome , Wet Macular Degeneration/drug therapy
3.
Lancet ; 399(10326): 741-755, 2022 02 19.
Article in English | MEDLINE | ID: mdl-35085503

ABSTRACT

BACKGROUND: To reduce treatment burden and optimise patient outcomes in diabetic macular oedema, we present 1-year results from two phase 3 trials of faricimab, a novel angiopoietin-2 and vascular endothelial growth factor-A bispecific antibody. METHODS: YOSEMITE and RHINE were randomised, double-masked, non-inferiority trials across 353 sites worldwide. Adults with vision loss due to centre-involving diabetic macular oedema were randomly assigned (1:1:1) to intravitreal faricimab 6·0 mg every 8 weeks, faricimab 6·0 mg per personalised treatment interval (PTI), or aflibercept 2·0 mg every 8 weeks up to week 100. PTI dosing intervals were extended, maintained, or reduced (every 4 weeks up to every 16 weeks) based on disease activity at active dosing visits. The primary endpoint was mean change in best-corrected visual acuity at 1 year, averaged over weeks 48, 52, and 56. Efficacy analyses included the intention-to-treat population (non-inferiority margin 4 Early Treatment Diabetic Retinopathy Study [ETDRS] letters); safety analyses included patients with at least one dose of study treatment. These trials are registered with ClinicalTrials.gov (YOSEMITE NCT03622580 and RHINE NCT03622593). FINDINGS: 3247 patients were screened for eligibility in YOSEMITE (n=1532) and RHINE (n=1715). After exclusions, 940 patients were enrolled into YOSEMITE between Sept 5, 2018, and Sept 19, 2019, and 951 patients were enrolled into RHINE between Oct 9, 2018, and Sept 20, 2019. These 1891 patients were randomly assigned to faricimab every 8 weeks (YOSEMITE n=315, RHINE n=317), faricimab PTI (n=313, n=319), or aflibercept every 8 weeks (n=312, n=315). Non-inferiority for the primary endpoint was achieved with faricimab every 8 weeks (adjusted mean vs aflibercept every 8 weeks in YOSEMITE 10·7 ETDRS letters [97·52% CI 9·4 to 12·0] vs 10·9 ETDRS letters [9·6 to 12·2], difference -0·2 ETDRS letters [-2·0 to 1·6]; RHINE 11·8 ETDRS letters [10·6 to 13·0] vs 10·3 ETDRS letters [9·1 to 11·4] letters, difference 1·5 ETDRS letters [-0·1 to 3·2]) and faricimab PTI (YOSEMITE 11·6 ETDRS letters [10·3 to 12·9], difference 0·7 ETDRS letters [-1·1 to 2·5]; RHINE 10·8 ETDRS letters [9·6 to 11·9], difference 0·5 ETDRS letters [-1·1 to 2·1]). Incidence of ocular adverse events was comparable between faricimab every 8 weeks (YOSEMITE n=98 [31%], RHINE n=137 [43%]), faricimab PTI (n=106 [34%], n=119 [37%]), and aflibercept every 8 weeks (n=102 [33%], n=113 [36%]). INTERPRETATION: Robust vision gains and anatomical improvements with faricimab were achieved with adjustable dosing up to every 16 weeks, demonstrating the potential for faricimab to extend the durability of treatment for patients with diabetic macular oedema. FUNDING: F Hoffmann-La Roche.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Bispecific/administration & dosage , Diabetic Retinopathy/drug therapy , Edema/drug therapy , Aged , Angiogenesis Inhibitors/adverse effects , Angiopoietin-2/antagonists & inhibitors , Antibodies, Bispecific/adverse effects , Diabetic Retinopathy/diagnosis , Double-Blind Method , Drug Administration Schedule , Edema/etiology , Female , Humans , Intravitreal Injections , Macula Lutea/diagnostic imaging , Macula Lutea/drug effects , Male , Middle Aged , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Visual Acuity/drug effects
4.
Ophthalmology ; 129(3): 295-307, 2022 03.
Article in English | MEDLINE | ID: mdl-34597713

ABSTRACT

PURPOSE: To evaluate the safety and efficacy of the Port Delivery System with ranibizumab (PDS) for the treatment of neovascular age-related macular degeneration (nAMD). DESIGN: Phase 3, open-label, randomized, visual acuity assessor-masked noninferiority and equivalence trial. PARTICIPANTS: Patients with nAMD diagnosed within 9 months of screening previously treated with and responsive to anti-vascular endothelial growth factor therapy. METHODS: Patients were randomized 3:2 to treatment with the PDS with ranibizumab 100 mg/ml with fixed 24-week (Q24W) refill-exchanges (PDS Q24W) or intravitreal ranibizumab 0.5-mg injections every 4 weeks (monthly ranibizumab). MAIN OUTCOME MEASURES: Primary end point was change in best-corrected visual acuity (BCVA) Early Treatment Diabetic Retinopathy Study letter (letters) score from baseline averaged over weeks 36 and 40 (noninferiority margin,-4.5 letters; equivalence margin, ±4.5 letters). RESULTS: Archway enrolled 418 patients; 251 were randomized to and 248 received treatment with the PDS Q24W, and 167 were randomized to and received treatment with monthly ranibizumab. Baseline BCVA was 74.4 letters (PDS Q24W arm) and 75.5 letters (monthly ranibizumab arm; Snellen equivalent, 20/32). Adjusted mean change in BCVA score from baseline averaged over weeks 36 and 40 was +0.2 letters (standard error [SE], 0.5 letters) in the PDS Q24W arm and +0.5 letters (SE, 0.6 letters) in the monthly ranibizumab arm (difference, -0.3 letters; 95% confidence interval, -1.7 to 1.1 letters). PDS Q24W was both noninferior and equivalent to monthly ranibizumab. Of 246 PDS-treated patients assessed for supplemental ranibizumab treatment, 242 (98.4%) did not receive supplemental ranibizumab treatment before the first refill-exchange procedure, including 4 patients who discontinued treatment before the first refill-exchange procedure. Prespecified ocular adverse events of special interest were reported in 47 patients (19.0%) in the PDS Q24W arm and 10 patients (6.0%) in the monthly ranibizumab arm, which included, in the former arm, 4 (1.6%) endophthalmitis cases, 2 (0.8%) retinal detachments, 13 (5.2%) vitreous hemorrhages, 6 (2.4%) conjunctival erosions, and 5 (2.0%) conjunctival retractions. Most ocular adverse events in the PDS Q24W arm occurred within 1 month of implantation. CONCLUSIONS: Archway met its primary objective and PDS Q24W demonstrated noninferior and equivalent efficacy to monthly ranibizumab, with 98.4% of PDS-treated patients not receiving supplemental treatment in the first 24-week interval.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Choroidal Neovascularization/drug therapy , Drug Delivery Systems , Ranibizumab/administration & dosage , Vitreous Body/drug effects , Wet Macular Degeneration/drug therapy , Aged , Aged, 80 and over , Choroidal Neovascularization/diagnosis , Choroidal Neovascularization/physiopathology , Female , Humans , Male , Middle Aged , Treatment Outcome , Vascular Access Devices , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Visual Acuity/physiology , Wet Macular Degeneration/diagnosis , Wet Macular Degeneration/physiopathology
5.
Eye (Lond) ; 34(11): 1966-1972, 2020 11.
Article in English | MEDLINE | ID: mdl-32541890

ABSTRACT

This article aims to identify key opportunities for improvement in the diagnosis and treatment of retinal disease, and describe recent innovations that will potentially facilitate improved outcomes with existing intravitreal vascular endothelial growth factor (VEGF) therapies and lay the groundwork for new treatment approaches. The review begins with a summary of the key discoveries that led to the development of anti-VEGF therapies and briefly reviews their impact on clinical practice. Opportunities for improvements in diagnosis, real-world outcomes with existing therapies, long-acting therapeutics and personalised health care are discussed, as well as the need to identify new targets for therapeutic intervention. Low-cost, remote patient screening and monitoring using artificial intelligence (AI)-based technologies can help improve diagnosis rates and enable remote disease monitoring with minimal patient burden. AI-based tools can be applied to generate patient-level prognostic data and predict individual treatment needs, reducing the time needed to optimise a patient's treatment regimen. Long-acting therapeutics can help improve visual outcomes by reducing the treatment burden. When paired with AI-generated prognoses, long-acting therapeutics enable the possibility of vision loss prevention. Dual-acting drugs may help improve efficacy and/or durability beyond what is possible with anti-VEGF agents alone. Recent developments and ongoing innovations will help build upon the success of anti-VEGF therapies to further reduce vision loss owing to retinal disease while lowering the overall burden of care.


Subject(s)
Ranibizumab , Vascular Endothelial Growth Factor A , Angiogenesis Inhibitors/therapeutic use , Artificial Intelligence , Bevacizumab , Humans
6.
Ophthalmology ; 126(8): 1141-1154, 2019 08.
Article in English | MEDLINE | ID: mdl-30946888

ABSTRACT

PURPOSE: To evaluate the safety and efficacy of the Port Delivery System with ranibizumab (PDS) for neovascular age-related macular degeneration (nAMD) treatment. DESIGN: Phase 2, multicenter, randomized, active treatment-controlled clinical trial. PARTICIPANTS: Patients diagnosed with nAMD within 9 months who had received 2 or more prior anti-vascular endothelial growth factor intravitreal injections and were responsive to treatment. METHODS: Patients were randomized 3:3:3:2 to receive the PDS filled with ranibizumab 10 mg/ml, 40 mg/ml, 100 mg/ml, or monthly intravitreal ranibizumab 0.5-mg injections. MAIN OUTCOME MEASURES: Time to first implant refill assessed when the last enrolled patient completed the month 9 visit (primary efficacy end point), improvement in best-corrected visual acuity (BCVA) and central foveal thickness (CFT), and safety. RESULTS: The primary analysis population was 220 patients, with 58, 62, 59, and 41 patients in the PDS 10-mg/ml, PDS 40-mg/ml, PDS 100-mg/ml, and monthly intravitreal ranibizumab 0.5-mg arms, respectively. Median time to first implant refill was 8.7, 13.0, and 15.0 months in the PDS 10-mg/ml, PDS 40-mg/ml, and PDS 100-mg/ml arms, respectively. At month 9, the adjusted mean BCVA change from baseline was ‒3.2 Early Treatment Diabetic Retinopathy Study (ETDRS) letters, ‒0.5 ETDRS letters, +5.0 ETDRS letters, and +3.9 ETDRS letters in the PDS 10-mg/ml, PDS 40-mg/ml, PDS 100-mg/ml, and monthly intravitreal ranibizumab 0.5-mg arms, respectively. At month 9, the adjusted mean CFT change from baseline was similar in the PDS 100-mg/ml and monthly intravitreal ranibizumab 0.5-mg arms. The optimized PDS implant insertion and refill procedures were generally well tolerated. After surgical procedure optimization, postoperative vitreous hemorrhage rate was 4.5% (7/157; 1 event classified as serious). There was no evidence of implant clogging. CONCLUSIONS: In the phase 2 Ladder trial, the PDS was generally well tolerated and demonstrated a dose response across multiple end points in patients with nAMD. The PDS 100-mg/ml arm showed visual and anatomic outcomes comparable with monthly intravitreal ranibizumab 0.5-mg injections but with a reduced total number of ranibizumab treatments. The PDS has the potential to reduce treatment burden in nAMD while maintaining vision.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Delayed-Action Preparations/administration & dosage , Drug Implants , Macular Degeneration/drug therapy , Ranibizumab/administration & dosage , Aged , Aged, 80 and over , Female , Humans , Intravitreal Injections , Male , Middle Aged
7.
Invest Ophthalmol Vis Sci ; 60(4): 852-857, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30821810

ABSTRACT

Purpose: To develop deep learning (DL) models for the automatic detection of optical coherence tomography (OCT) measures of diabetic macular thickening (MT) from color fundus photographs (CFPs). Methods: Retrospective analysis on 17,997 CFPs and their associated OCT measurements from the phase 3 RIDE/RISE diabetic macular edema (DME) studies. DL with transfer-learning cascade was applied on CFPs to predict time-domain OCT (TD-OCT)-equivalent measures of MT, including central subfield thickness (CST) and central foveal thickness (CFT). MT was defined by using two OCT cutoff points: 250 µm and 400 µm. A DL regression model was developed to directly quantify the actual CFT and CST from CFPs. Results: The best DL model was able to predict CST ≥ 250 µm and CFT ≥ 250 µm with an area under the curve (AUC) of 0.97 (95% confidence interval [CI], 0.89-1.00) and 0.91 (95% CI, 0.76-0.99), respectively. To predict CST ≥ 400 µm and CFT ≥ 400 µm, the best DL model had an AUC of 0.94 (95% CI, 0.82-1.00) and 0.96 (95% CI, 0.88-1.00), respectively. The best deep convolutional neural network regression model to quantify CST and CFT had an R2 of 0.74 (95% CI, 0.49-0.91) and 0.54 (95% CI, 0.20-0.87), respectively. The performance of the DL models declined when the CFPs were of poor quality or contained laser scars. Conclusions: DL is capable of predicting key quantitative TD-OCT measurements related to MT from CFPs. The DL models presented here could enhance the efficiency of DME diagnosis in tele-ophthalmology programs, promoting better visual outcomes. Future research is needed to validate DL algorithms for MT in the real-world.


Subject(s)
Deep Learning , Diabetic Retinopathy/diagnostic imaging , Image Interpretation, Computer-Assisted/methods , Macula Lutea/pathology , Macular Edema/diagnostic imaging , Photography/methods , Tomography, Optical Coherence/methods , Angiogenesis Inhibitors/therapeutic use , Diabetic Retinopathy/drug therapy , Diagnostic Techniques, Ophthalmological , False Positive Reactions , Female , Fundus Oculi , Humans , Intravitreal Injections , Macular Edema/drug therapy , Male , Middle Aged , Neural Networks, Computer , Predictive Value of Tests , Randomized Controlled Trials as Topic , Ranibizumab/therapeutic use , Retrospective Studies , Sensitivity and Specificity , Vascular Endothelial Growth Factor A/antagonists & inhibitors
8.
Nat Rev Drug Discov ; 15(6): 385-403, 2016 06.
Article in English | MEDLINE | ID: mdl-26775688

ABSTRACT

The targeting of vascular endothelial growth factor A (VEGFA), a crucial regulator of both normal and pathological angiogenesis, has revealed innovative therapeutic approaches in oncology and ophthalmology. The first VEGFA inhibitor, bevacizumab, was approved by the US Food and Drug Administration in 2004 for the first-line treatment of metastatic colorectal cancer, and the first VEGFA inhibitors in ophthalmology, pegaptanib and ranibizumab, were approved in 2004 and 2006, respectively. To mark this tenth anniversary of anti-VEGFA therapy, we discuss the discovery of VEGFA, the successes and challenges in the development of VEGFA inhibitors and the impact of these agents on the treatment of cancers and ophthalmic diseases.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Humans , Neoplasms/blood supply , Time Factors
9.
Dev Ophthalmol ; 55: 28-37, 2016.
Article in English | MEDLINE | ID: mdl-26502333

ABSTRACT

Systematic study of the mechanisms underlying pathological ocular neovascularization has yielded a wealth of knowledge about pro- and anti-angiogenic factors that modulate diseases such as neovascular age-related macular degeneration. The evidence implicating vascular endothelial growth factor (VEGF) in particular has led to the development of a number of approved anti-VEGF therapies. Additional proangiogenic targets that have emerged as potential mediators of ocular neovascularization include hypoxia-inducible factor-1, angiopoietin-2, platelet-derived growth factor-B and components of the alternative complement pathway. As for VEGF, knowledge of these factors has led to a product pipeline of many more novel agents that are in various stages of clinical development in the setting of ocular neovascularization. These agents are represented by a range of drug classes and, in addition to novel small- and large-molecule VEGF inhibitors, include gene therapies, small interfering RNA agents and tyrosine kinase inhibitors. In addition, combination therapy is beginning to emerge as a strategy to improve the efficacy of individual therapies. Thus, a variety of agents, whether administered alone or as adjunctive therapy with agents targeting VEGF, offer the promise of expanding the range of treatments for ocular neovascular diseases.


Subject(s)
Angiopoietin-2/physiology , Complement Pathway, Alternative/physiology , Eye/blood supply , Hypoxia-Inducible Factor 1/physiology , Neovascularization, Pathologic/physiopathology , Proto-Oncogene Proteins c-sis/physiology , Vascular Endothelial Growth Factor A/physiology , Humans
11.
Invest Ophthalmol Vis Sci ; 55(6): 3709-19, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24845632

ABSTRACT

PURPOSE: Characterization of a mouse model of spontaneous choroidal neovascularization (sCNV) and its effect on retinal architecture and function. METHODS: The sCNV mouse phenotype was characterized by using fundus photography, fluorescein angiography, confocal scanning laser ophthalmoscopy (SLO), optical coherence tomography (OCT), ERG, immunostaining, biochemistry, and electron microscopy. A role for VEGF-A signaling in sCNV was investigated by using neutralizing antibodies and a role for macrophages explored by cell-depletion studies. RESULTS: The sCNV starts between postnatal day 10 and 15 (P10-P15), increasing in number and severity causing RPE disruption and dysfunction. Various morphological methods confirmed the choroidal origin and subretinal position of the angiogenic vessels. At approximately P25, vessels were present in the outer retina with instances of anastomosis of some sCNV lesions with the retinal vasculature. The number of CNV lesions was significantly decreased by systemic blockade of the VEGF-A pathway. Choroidal neovascularization size also was significantly modulated by reducing the number of lesion-associated macrophages. Later stages of sCNV were associated with edema, neuronal loss, and dysfunction. CONCLUSIONS: The sCNV mouse is a new model for the study of both early and late events associated with choroidal neovascularization. Pharmacological reduction in sCNV with VEGF-A antagonists and an anti-inflammatory strategy suggests the model may be useful for investigating novel targets for treating human ocular neovascular disease.


Subject(s)
Choroid/blood supply , Choroidal Neovascularization/metabolism , Edema/metabolism , Retinal Pigment Epithelium/ultrastructure , Vascular Endothelial Growth Factor A/metabolism , Animals , Choroid/ultrastructure , Choroidal Neovascularization/pathology , Disease Models, Animal , Edema/pathology , Electroretinography , Enzyme-Linked Immunosorbent Assay , Fluorescein Angiography , Fundus Oculi , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Confocal , Microscopy, Electron , Ophthalmoscopy , Phenotype , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/physiopathology , Retinal Vessels/metabolism , Retinal Vessels/ultrastructure , Signal Transduction , Tomography, Optical Coherence
12.
Ophthalmology ; 120(10): 2013-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23706949

ABSTRACT

PURPOSE: To report 36-month outcomes of RIDE (NCT00473382) and RISE (NCT00473330), trials of ranibizumab in diabetic macular edema (DME). DESIGN: Phase III, randomized, multicenter, double-masked, 3-year trials, sham injection-controlled for 2 years. PARTICIPANTS: Adults with DME (n=759), baseline best-corrected visual acuity (BCVA) 20/40 to 20/320 Snellen equivalent, and central foveal thickness (CFT) ≥ 275 µm on optical coherence tomography. METHODS: Patients were randomized equally (1 eye per patient) to monthly 0.5 mg or 0.3 mg ranibizumab or sham injection. In the third year, sham patients, while still masked, were eligible to cross over to monthly 0.5 mg ranibizumab. Macular laser was available to all patients starting at month 3; panretinal laser was available as necessary. MAIN OUTCOME MEASURES: The proportion of patients gaining ≥15 Early Treatment Diabetic Retinopathy Study letters in BCVA from baseline at month 24. RESULTS: Visual acuity (VA) outcomes seen at month 24 in ranibizumab groups were consistent through month 36; the proportions of patients who gained ≥15 letters from baseline at month 36 in the sham/0.5 mg, 0.3 mg, and 0.5 mg ranibizumab groups were 19.2%, 36.8%, and 40.2%, respectively, in RIDE and 22.0%, 51.2%, and 41.6%, respectively, in RISE. In the ranibizumab arms, reductions in CFT seen at 24 months were, on average, sustained through month 36. After crossover to 1 year of treatment with ranibizumab, average VA gains in the sham/0.5 mg group were lower compared with gains seen in the ranibizumab patients after 1 year of treatment (2.8 vs. 10.6 and 11.1 letters). Per-injection rates of endophthalmitis remained low over time (∼0.06% per injection). The incidence of serious adverse events potentially related to systemic vascular endothelial growth factor inhibition was 19.7% in patients who received 0.5 mg ranibizumab compared with 16.8% in the 0.3 mg group. CONCLUSIONS: The strong VA gains and improvement in retinal anatomy achieved with ranibizumab at month 24 were sustained through month 36. Delayed treatment in patients receiving sham treatment did not seem to result in the same extent of VA improvement observed in patients originally randomized to ranibizumab. Ocular and systemic safety was generally consistent with the results seen at month 24. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found after the references.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Diabetic Retinopathy/drug therapy , Macular Edema/drug therapy , Adult , Diabetic Retinopathy/physiopathology , Female , Humans , Intravitreal Injections , Macular Edema/physiopathology , Male , Ranibizumab , Visual Acuity/physiology
13.
Ophthalmology ; 118(8): 1594-602, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21684606

ABSTRACT

PURPOSE: Assess 12-month efficacy and safety of intraocular injections of 0.3 mg or 0.5 mg ranibizumab in patients with macular edema after branch retinal vein occlusion (BRVO). DESIGN: Prospective, randomized, sham injection-controlled, double-masked, multicenter trial. PARTICIPANTS: A total of 397 patients with macular edema after BRVO. METHODS: Eligible patients were randomized 1:1:1 to 6 monthly injections of 0.3 mg or 0.5 mg ranibizumab or sham injections. After 6 months, all patients with study eye best-corrected visual acuity (BCVA) ≤20/40 or central subfield thickness ≥250 µm were to receive ranibizumab. Patients could receive rescue laser treatment once during the treatment period and once during the observation period if criteria were met. MAIN OUTCOME MEASURES: The main efficacy outcome reported is mean change from baseline BCVA letter score at month 12. Additional visual and anatomic parameters were assessed. RESULTS: Mean (95% confidence interval) change from baseline BCVA letter score at month 12 was 16.4 (14.5-18.4) and 18.3 (15.8-20.9) in the 0.3 mg and 0.5 mg groups, respectively, and 12.1 (9.6-14.6) in the sham/0.5 mg group (P<0.01, each ranibizumab group vs. sham/0.5 mg). The percentage of patients who gained ≥15 letters from baseline BCVA at month 12 was 56.0% and 60.3% in the 0.3 mg and 0.5 mg groups, respectively, and 43.9% in the sham/0.5 mg group. On average, there was a marked reduction in central foveal thickness (CFT) after the first as-needed injection of 0.5 mg ranibizumab in the sham/0.5 mg group, which was sustained through month 12. No new ocular or nonocular safety events were identified. CONCLUSIONS: At month 12, treatment with ranibizumab as needed during months 6-11 maintained, on average, the benefits achieved by 6 monthly ranibizumab injections in patients with macular edema after BRVO, with low rates of ocular and nonocular safety events. In the sham/0.5 mg group, treatment with ranibizumab as needed for 6 months resulted in rapid reduction in CFT to a similar level as that in the 0.3 mg ranibizumab treatment group and an improvement in BCVA, but not to the extent of that in the 2 ranibizumab groups. Intraocular injections of ranibizumab provide an effective treatment for macular edema after BRVO. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found after the references.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal/administration & dosage , Macular Edema/drug therapy , Retinal Vein Occlusion/complications , Activities of Daily Living , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Double-Blind Method , Female , Fluorescein Angiography , Humans , Intravitreal Injections , Macular Edema/etiology , Macular Edema/physiopathology , Male , Middle Aged , Prospective Studies , Ranibizumab , Sickness Impact Profile , Surveys and Questionnaires , Tomography, Optical Coherence , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Visual Acuity/drug effects , Visual Acuity/physiology
14.
Adv Exp Med Biol ; 703: 1-7, 2010.
Article in English | MEDLINE | ID: mdl-20711703

ABSTRACT

The complement cascade has been identified as a key factor in the pathogenesis of age-related macular degeneration (AMD). As a result, pharmacological modulation of the complement cascade is being investigated as a therapeutic strategy for AMD. The genetic data point to a triggering of the complement cascade, which subsequently cannot be damped down. Despite promising genetic, preclinical and immunolabeling data, important questions remain to be answered regarding the role of complement in the pathogenesis of AMD. The involvement of the complement cascade in the vision threatening stages of AMD, e.g. geographic atrophy and choroidal neovascularization, remain unknown. Additionally, the optimal component(s) of the complement cascade to be targeted for modulation still need to be identified. Answering these and other questions will provide investigators with a clear framework with which to evaluate progress in the field and help guide the development of future clinical therapeutics.


Subject(s)
Complement System Proteins/metabolism , Macular Degeneration/etiology , Macular Degeneration/immunology , Choroidal Neovascularization/immunology , Complement Activation , Complement Inactivating Agents/therapeutic use , Geographic Atrophy/immunology , Humans , Inflammation/immunology , Macular Degeneration/drug therapy , Models, Immunological , Retinal Drusen/immunology
15.
Invest Ophthalmol Vis Sci ; 51(2): 1028-35, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19815730

ABSTRACT

PURPOSE: To explore the role of integrin alpha5beta1 in proliferative vitreoretinopathy (PVR) pathogenesis by evaluating the expression alpha5beta1 on ARPE-19 cells and patient proliferative membranes, quantifying the inhibitory effects of JSM6427 (a small molecule alpha5beta1 inhibitor) on ARPE-19 cell adhesion and migration, and assessing the therapeutic potential of JSM6427 in a rabbit retinal detachment model. METHODS: Expression of alpha5beta1 was evaluated on activated ARPE-19 cells by flow cytometry and on PVR membranes by immunohistochemistry. ARPE-19 cells were used in fibronectin-dependent adhesion and migration assays with various concentrations of JSM6427; IC(50) was calculated. In the rabbit model, eyes were intravitreally injected with vehicle or JSM6427 on day 0 or 1 after retinal detachment; BrdU was administered intravitreally on day 3, and retinal tissues were harvested on day 3 (4 hours later) or 7. Retinal scarring, cellular proliferation, and inflammatory responses were quantified, and retinal morphology was analyzed in retinal sections. RESULTS: Activated ARPE-19 cells and PVR membranes expressed high levels of alpha5beta1; expression was low in control eyes. JSM6427 provided a dose-dependent blockade of ARPE-19 cell adhesion to fibronectin (IC(50), 7.1 +/- 2.5 microM) and inhibition of migration (IC(50), 6.0 +/- 4.5 microM). In the rabbit model, intravitreal injection of JSM6427 provided significant inhibition of proliferation of retinal cells (Müller cells, microglia, and macrophages) on days 3 and 7 after detachment and inhibition of inflammatory response and retinal scarring on day 7 after detachment. CONCLUSIONS: JSM6427 is a promising treatment for PVR, with data suggesting that inhibition of alpha5beta1-fibronectin interactions addresses multiple pathways involving retinal pigment epithelial, glial, and inflammatory cells.


Subject(s)
Disease Models, Animal , Integrin alpha5beta1/antagonists & inhibitors , Propionates/pharmacology , Pyridines/pharmacology , Pyrrolidines/pharmacology , Retinal Detachment/prevention & control , Retinal Pigment Epithelium/drug effects , Vitreoretinopathy, Proliferative/prevention & control , Animals , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Epiretinal Membrane/metabolism , Female , Fibronectins/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Integrin alpha5beta1/metabolism , Male , Rabbits , Retinal Detachment/metabolism , Retinal Detachment/pathology , Retinal Pigment Epithelium/metabolism , Vitreoretinopathy, Proliferative/etiology , Vitreoretinopathy, Proliferative/metabolism
16.
Arch Ophthalmol ; 127(10): 1329-35, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19822850

ABSTRACT

OBJECTIVE: To evaluate the pharmacologic activity and tolerability of JSM6427, a potent and first selective small-molecule inhibitor of integrin alpha5beta1, in monkey and rabbit models of choroidal neovascularization (CNV). METHODS: JSM6427 selectivity for alpha5beta1 was evaluated by in vitro binding assays while the ability of JSM6427 to inhibit CNV was investigated in a laser-induced monkey model and a growth factor-induced rabbit model. Intravitreal injections of JSM6427 (100, 300, or 1000 microg) or vehicle were administered immediately after the CNV induction procedure and at weekly intervals for 4 weeks. Fluorescein angiography was performed weekly. Ocular tolerability was evaluated ophthalmoscopically and histologically in both models; additional assessments in monkeys included electroretinography, biomicroscopy, pathological examination, and analysis of JSM6427 pharmacokinetics. RESULTS: JSM6427 was highly selective for the alpha5beta1-fibronectin interaction. Weekly intravitreal injections of JSM6427 resulted in a statistically significant dose-dependent inhibition of CNV in laser-induced and growth factor-induced models without any ocular JSM6427-related adverse effects. JSM6427 was cleared through the systemic circulation with no evidence of systemic accumulation. CONCLUSIONS: Intravitreal JSM6427 provided dose-dependent inhibition of CNV in monkey and rabbit experimental models. CLINICAL RELEVANCE: JSM6427 may provide a new approach for the treatment of ocular neovascular diseases such as age-related macular degeneration in humans.


Subject(s)
Aminopyridines/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/drug therapy , Disease Models, Animal , Integrin alpha5beta1/antagonists & inhibitors , beta-Alanine/analogs & derivatives , Aminopyridines/pharmacokinetics , Angiogenesis Inhibitors/pharmacokinetics , Animals , Choroidal Neovascularization/diagnosis , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Electroretinography , Female , Fibroblast Growth Factor 2/antagonists & inhibitors , Fluorescein Angiography , Injections , Macaca fascicularis , Male , Rabbits , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vitreous Body , beta-Alanine/pharmacokinetics , beta-Alanine/therapeutic use
17.
Retina ; 29(6 Suppl): S42-4, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19553800

ABSTRACT

The strategies for improving control of age-related macular degeneration with combination therapies are evolving. The focus on more effective blockade of choroidal neovascularization has shifted to include control of additional processes implicated in disease progression, such as neural death, inflammation, or fibrosis. Although there is likely to be a strong interrelationship between choroidal neovascularization and inflammation that contributes to advanced stages of disease progression, including fibrosis, combining treatment strategies may enlarge the opportunity to prevent both early and late vision loss.


Subject(s)
Choroidal Neovascularization/therapy , Drug Therapy, Combination , Macular Degeneration/therapy , Animals , Combined Modality Therapy , Fibrosis/prevention & control , Humans , Inflammation/prevention & control , Neuroprotective Agents/therapeutic use , Retina/pathology
19.
Arch Ophthalmol ; 127(4): 374-80, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19365011

ABSTRACT

OBJECTIVES: To assess the safety and efficacy of intravitreous pegaptanib sodium for the treatment of macular edema following central retinal vein occlusion (CRVO). DESIGN: This dose-ranging, double-masked, multicenter, phase 2 trial included subjects with CRVO for 6 months' or less duration randomly assigned (1:1:1) to receive pegaptanib sodium or sham injections every 6 weeks for 24 weeks (0.3 mg and 1 mg, n=33; sham, n=32). MAIN OUTCOME MEASURE: Visual acuity at week 30. RESULTS: In the primary analysis at week 30, 12 of 33 (36%) subjects treated with 0.3 mg of pegaptanib sodium and 13 of 33 (39%) treated with 1 mg gained 15 or more letters from baseline vs 9 of 32 (28%) sham-treated subjects (P= .48 for 0.3 mg and P= .35 for 1 mg of pegaptanib sodium vs sham). In secondary analyses, subjects treated with pegaptanib sodium were less likely to lose 15 or more letters (9% and 6%; 0.3-mg and 1-mg pegaptanib sodium groups, respectively) compared with sham-treated eyes (31%; P= .03 for 0.3 mg and P= .01 for 1 mg of pegaptanib sodium vs sham) and showed greater improvement in mean visual acuity (+7.1 and +9.9, respectively, vs -3.2 letters with sham; P= .09 for 0.3 mg and P= .02 for 1 mg of pegaptanib sodium vs sham). By week 1, the mean central retinal thickness decreased in the 0.3-mg and 1-mg pegaptanib sodium groups by 269 microm and 210 microm, respectively, vs 5 microm with sham (P< .001). CONCLUSIONS: Based on this 30-week study, intravitreous pegaptanib sodium appears to provide visual and anatomical benefits in the treatment of macular edema following CRVO. APPLICATION TO CLINICAL PRACTICE: Benefits accrued with intravitreous pegaptanib sodium treatment of macular edema following CRVO suggest a role for vascular endothelial growth factor in the pathogenesis of this condition. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00088283.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Aptamers, Nucleotide/administration & dosage , Macular Edema/drug therapy , Retinal Vein Occlusion/drug therapy , Angiogenesis Inhibitors/adverse effects , Aptamers, Nucleotide/adverse effects , Double-Blind Method , Female , Fluorescein Angiography , Humans , Injections , Macular Edema/etiology , Male , Middle Aged , Retinal Vein Occlusion/complications , Tonometry, Ocular , Vascular Endothelial Growth Factor A/metabolism , Visual Acuity , Vitreous Body
20.
Semin Immunopathol ; 30(2): 65-84, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18340447

ABSTRACT

There is an accumulating body of evidence that immunological mechanisms play a prominent role in the pathogenesis of diabetic retinopathy (DR), which is characterized by many features typical of inflammation. The upregulation of cytokines and other inflammatory mediators leading to persistent low-grade inflammation and an influx of leukocytes, is believed to contribute actively to DR-associated damage to the retinal vasculature and retinal neovascularization. This review will describe preclinical and clinical studies that document an inflammatory basis for DR and that support the use of nonsteroidal anti-inflammatory drugs, corticosteroids, and anti-vascular endothelial growth factor agents in its treatment. In addition, emerging therapeutic approaches based on ongoing investigations will be discussed, including those involving blockade of angiotensin receptors and other molecular targets such as tumor necrosis factor-alpha.


Subject(s)
Diabetic Retinopathy/immunology , Adrenal Cortex Hormones/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Clinical Trials as Topic , Diabetic Retinopathy/drug therapy , Humans , Inflammation/immunology , Neovascularization, Pathologic/immunology , Retina/immunology , Retinal Vessels/immunology , Retinal Vessels/physiopathology , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...