Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(10): eadl3576, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38457512

ABSTRACT

Suprachoroidal nonviral gene therapy with biodegradable poly(ß-amino ester) nanoparticles (NPs) provides widespread expression in photoreceptors and retinal pigmented epithelial (RPE) cells and therapeutic benefits in rodents. Here, we show in a human-sized minipig eye that suprachoroidal injection of 50 µl of NPs containing 19.2 µg of GFP expression plasmid caused GFP expression in photoreceptors and RPE throughout the entire eye with no toxicity. Two weeks after injection of 50, 100, or 200 µl, there was considerable within-eye and between-eye variability in expression that was reduced 3 months after injection of 200 µl and markedly reduced after three suprachoroidal injections at different locations around the eye. Reduction of bacterial CpG sequences in the expression plasmid resulted in a trend toward higher expression. These data indicate that nonviral suprachoroidal gene therapy with optimized polymer, expression plasmid, and injection approach has potential for treating photoreceptors throughout the entire retina of a human-sized eye.


Subject(s)
Nanoparticles , Retina , Animals , Humans , Swine , Swine, Miniature , Retina/metabolism , Plasmids/genetics , Genetic Therapy/methods
2.
iScience ; 26(2): 106078, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36844452

ABSTRACT

AXT107, a collagen-derived peptide that binds integrins αvß3 and α5ß1 with high affinity, suppresses vascular endothelial growth factor (VEGF) signaling, promotes angiopoietin 2-induced Tie2 activation, and suppresses neovascularization (NV) and vascular leakage. Immunohistochemical staining for αvß3 and α5ß1 was markedly increased in NV compared with normal retinal vessels. After intravitreous injection of AXT107, there was no staining with an anti-AXT107 antibody on normal vessels but robust staining of NV that co-localized with αvß3 and α5ß1. Likewise, after intravitreous injection, fluorescein amidite-labeled AXT107 co-localized with αvß3 and α5ß1 on NV but not normal vessels. AXT107 also co-localized with αv and α5 at cell-cell junctions of human umbilical vein endothelial cells (HUVECs). AXT107-integrin binding was demonstrated by ex vivo cross-linking/pull-down experiments. These data support the hypothesis that AXT107 therapeutic activity is mediated through binding αvß3 and α5ß1 which are markedly upregulated on endothelial cells in NV providing selective targeting of diseased vessels which has therapeutic and safety benefits.

3.
Nat Biomed Eng ; 4(11): 1053-1062, 2020 11.
Article in English | MEDLINE | ID: mdl-32895514

ABSTRACT

Eye-drop formulations should hold as high a concentration of soluble drug in contact with ocular epithelium for as long as possible. However, eye tears and frequent blinking limit drug retention on the ocular surface, and gelling drops typically form clumps that blur vision. Here, we describe a gelling hypotonic solution containing a low concentration of a thermosensitive triblock copolymer for extended ocular drug delivery. On topical application, the hypotonic formulation forms a highly uniform and clear thin layer that conforms to the ocular surface and resists clearance from blinking, increasing the intraocular absorption of hydrophilic and hydrophobic drugs and extending the drug-ocular-epithelium contact time with respect to conventional thermosensitive gelling formulations and commercial eye drops. We also show that the conformal gel layer allows for therapeutically relevant drug delivery to the posterior segment of the eyeball in pigs. Our findings highlight the importance of formulations that conform to the ocular surface before viscosity enhancement for increased and prolonged ocular surface contact and drug absorption.


Subject(s)
Drug Delivery Systems/methods , Eye/drug effects , Ophthalmic Solutions/administration & dosage , Ophthalmic Solutions/chemical synthesis , Administration, Topical , Animals , Eye/diagnostic imaging , Female , Gels/administration & dosage , Gels/chemistry , Hypotonic Solutions/administration & dosage , Hypotonic Solutions/chemistry , Male , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Polymers/administration & dosage , Polymers/chemistry , Rabbits , Rats, Sprague-Dawley , Swine
4.
Int J Mol Sci ; 21(14)2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32708100

ABSTRACT

Persistent inflammation is a complication associated with many ocular diseases. Changes in ocular vessels can amplify disease responses and contribute to vision loss by influencing the delivery of leukocytes to the eye, vascular leakage, and perfusion. Here, we report the anti-inflammatory activity for AXT107, a non-RGD, 20-mer αvß3 and α5ß1 integrin-binding peptide that blocks vascular endothelial growth factor (VEGF)-signaling and activates tyrosine kinase with immunoglobulin and EGF-like domains 2 (Tie2) using the normally inhibitory ligand angiopoietin 2 (Ang2). Tumor necrosis factor α (TNFα), a central inflammation mediator, induces Ang2 release from endothelial cells to enhance its stimulation of inflammation and vascular leakage. AXT107 resolves TNFα-induced vascular inflammation in endothelial cells by converting the endogenously released Ang2 into an agonist of Tie2 signaling, thereby disrupting both the synergism between TNFα and Ang2 while also preventing inhibitor of nuclear factor-κB α (IκBα) degradation directly through Tie2 signaling. This recovery of IκBα prevents nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) nuclear localization, thereby blocking NF-κB-induced inflammatory responses, including the production of VCAM-1 and ICAM-1, leukostasis, and vascular leakage in cell and mouse models. AXT107 also decreased the levels of pro-inflammatory TNF receptor 1 (TNFR1) without affecting levels of the more protective TNFR2. These data suggest that AXT107 may provide multiple benefits in the treatment of retinal/choroidal and other vascular diseases by suppressing inflammation and promoting vascular stabilization.


Subject(s)
Angiopoietin-2/metabolism , Collagen Type IV/pharmacology , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , I-kappa B Kinase/metabolism , Inflammation/drug therapy , Peptide Fragments/pharmacology , Receptor, TIE-2/metabolism , Angiopoietin-1/metabolism , Animals , Capillary Permeability/drug effects , Choroid Diseases/drug therapy , Collagen Type IV/therapeutic use , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Intercellular Adhesion Molecule-1/metabolism , Leukostasis/drug therapy , Leukostasis/metabolism , Mice , Mice, Inbred C57BL , Peptide Fragments/therapeutic use , Receptor, TIE-2/agonists , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Retinal Diseases/drug therapy , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
FASEB Bioadv ; 2(4): 219-233, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32259049

ABSTRACT

In patients with macular edema due to ischemic retinopathy, aqueous levels of hepatocyte growth factor (HGF) correlate with edema severity. We tested whether HGF expression and activity in mice with oxygen-induced ischemic retinopathy supports a role in macular edema. In ischemic retina, HGF was increased in endogenous cells and macrophages associated with retinal neovascularization (NV). HGF activator was increased in and around retinal vessels potentially providing vascular targeting. One day after intravitreous injection of HGF, VE-cadherin was reduced and albumin levels in retina and vitreous were significantly increased indicating vascular leakage. Injection of VEGF caused higher levels of vitreous albumin than HGF, and co-injection of both growth factors caused significantly higher levels than either alone. HGF increased the number of macrophages on the retinal surface, which was blocked by anti-c-Met and abrogated in chemokine (C-C motif) ligand 2 (CCL2)-/- mice. Injection of anti-c-Met significantly decreased leakage within 24 hours and after 5 days it reduced retinal NV in mice with ischemic retinopathy, but had no effect on choroidal NV. These data indicate that HGF is a pro-permeability, pro-inflammatory, and pro-angiogenic factor and along with its activator is increased in ischemic retina providing support for a potential role of HGF in macular edema in ischemic retinopathies.

6.
Biomaterials ; 243: 119935, 2020 06.
Article in English | MEDLINE | ID: mdl-32172031

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) has been implicated in the pathogenesis of choroidal neovascularization (NV) and is an appealing target because it increases multiple pro-angiogenic proteins and their receptors. Acriflavine (ACF) binds HIF-1α and HIF-2α preventing binding to HIF-1ß and inhibiting transcriptional activity of HIF-1 and HIF-2. Delivery of ACF to the eye by multiple routes strongly, but transiently, suppresses choroidal NV. We overcame design challenges and loaded highly water soluble ACF into poly(lactic-co-glycolic acid) (PLGA) microparticles (PLGA-ACF MPs) that release ACF in vitro for up to 60 days. Intravitreous injection of PLGA-ACF MPs in mice suppressed choroidal NV for at least 9 weeks and suprachoroidal injection of PLGA-ACF in rats suppressed choroidal NV for at least 18 weeks. Intravitreous, but not suprachoroidal injection, of PLGA-ACF MPs containing 38 µg of ACF in rabbits resulted in modest reduction of full-field electroretinogram (ERG) function. Over the span of 28 days after suprachoroidal injection of PLGA-ACF MP, rabbits had normal appearing retinas on fundus photographs, normal electroretinogram scotopic a- and b-wave amplitudes, no increase in intraocular pressure, and normal retinal histology. The active component of ACF, trypaflavine, had steady-state levels in the low nM range in RPE/choroid > retina for at least 16 weeks with a gradient from the side of the eye where the injection was done to the opposite side. These data suggest that suprachoroidal injection of PLGA-ACF MPs has the potential to provide a durable new treatment for retinal and choroidal vascular diseases.


Subject(s)
Choroidal Effusions , Choroidal Neovascularization , Acriflavine , Animals , Choroidal Neovascularization/drug therapy , Mice , Rabbits , Rats , Retina
7.
Cell Rep ; 29(1): 62-75.e7, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31577956

ABSTRACT

Id helix-loop-helix (HLH) proteins (Id1-4) bind E protein bHLH transcription factors, preventing them from forming active transcription complexes that drive changes in cell states. Id proteins are primarily expressed during development to inhibit differentiation, but they become re-expressed in adult tissues in diseases of the vasculature and cancer. We show that the genetic loss of Id1/Id3 reduces ocular neovascularization in mouse models of wet age-related macular degeneration (AMD) and retinopathy of prematurity (ROP). An in silico screen identifies AGX51, a small-molecule Id antagonist. AGX51 inhibits the Id1-E47 interaction, leading to ubiquitin-mediated degradation of Ids, cell growth arrest, and reduced viability. AGX51 is well-tolerated in mice and phenocopies the genetic loss of Id expression in AMD and ROP models by inhibiting retinal neovascularization. Thus, AGX51 is a first-in-class compound that antagonizes an interaction formerly considered undruggable and that may have utility in the management of multiple diseases.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Neovascularization, Pathologic/drug therapy , Small Molecule Libraries/pharmacology , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HCT116 Cells , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Inhibitor of Differentiation Protein 1/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Pathologic/metabolism
8.
Acta Biomater ; 97: 451-460, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31374338

ABSTRACT

Leading causes of vision loss include neovascular age-related macular degeneration (NVAMD) and macular edema (ME), which both require frequent intravitreal injections for treatment. A safe, poly(lactic-co-glycolic acid) (PLGA)-based biodegradable polymeric microparticle (MP) delivery system was developed that encapsulates and protects a biomimetic peptide from degradation, allows sustained intraocular release through polymer hydrolysis, and demonstrates a prolonged anti-angiogenic effect in vivo in three different NVAMD animal models (a laser-induced choroidal neovascularization mouse model, a rhoVEGF transgenic mouse model, and a Tet/opsin/VEGF transgenic mouse model) following intravitreal administration. The role of copolymer composition and microparticle shape was explored and 85:15 lactide-to-glycolide PLGA formed into ellipsoidal microparticles was found to be effective at inhibiting neovascularization for at least 16 weeks in vivo. Treatments were found to not only inhibit the growth of neovascularization, but also to cause regression of the neovasculature, reduce vascular leakage, and prevent exudative retinal detachment. These particulate devices are promising for the sustained release of biologics in the eye and may be useful for treating retinal diseases. STATEMENT OF SIGNIFICANCE: Devastating retinal diseases cause blindness in millions of people around the world. Current protein-based treatments have insufficient efficacy for many patients and also necessitate frequent intravitreal injections. Here, we demonstrate a new treatment consisting of a peptide encapsulated in biodegradable microparticles. We explore the effects of copolymer composition and physical shape of polymeric microparticles and find that both modulate peptide release. Efficacy of the treatment was validated in three different mouse models and the lead formulation was determined to be effective long-term, for at least 16 weeks in vivo, following a single injection. Treatments inhibited and regressed neovascularization as well as reduced vascular leakage. Anisotropic polymeric microparticles are promising for the sustained release of biologics in the eye.


Subject(s)
Biomimetic Materials , Choroidal Neovascularization/prevention & control , Peptides , Polylactic Acid-Polyglycolic Acid Copolymer , Animals , Anisotropy , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Intravitreal Injections , Mice , Mice, Transgenic , Peptides/chemistry , Peptides/pharmacokinetics , Peptides/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , RAW 264.7 Cells
9.
Invest Ophthalmol Vis Sci ; 59(12): 5266-5276, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30383198

ABSTRACT

Purpose: Quantitative understanding of the transport of therapeutic macromolecules following intraocular injections is critical for the design of efficient strategies in treating eye diseases, such as neovascular (wet) age-related macular degeneration (AMD) and macular edema (ME). Antiangiogenic treatments, such as neutralizing antibodies against VEGF or recently characterized antiangiogenic peptides, have shown promise in slowing disease progression. Methods: We developed a comprehensive three-dimensional (3D) transport model for intraocular injections using published data on drug distribution in rabbit eyes following intravitreal and suprachoroidal (SC) injection of sodium fluorescein (SF), bevacizumab, and ranibizumab. The model then was applied to evaluate the distribution of small molecules and antiangiogenic proteins following intravitreal and SC injections in human eyes. Results: The model predicts that intravitreally administered molecules are substantially mixed within the vitreous following injection, and that the long-term behavior of the injected drug does not depend on the initial mixing. Ocular pharmacokinetics of different drugs is sensitive to different clearance mechanisms. Effective retinal drug delivery is impacted by RPE permeability. For VEGF antibody, intravitreal injection provides sustained delivery to the retina, whereas SC injection provides more efficient, but short-lived, retinal delivery for smaller-sized molecules. Long-term suppression of neovascularization through SC administration of antiangiogenic drugs necessitates frequent injection or sustained delivery, such as microparticle-based delivery of antiangiogenic peptides. Conclusions: A comprehensive 3D model for intravitreal and SC drug injection is developed to provide a framework and platform for testing drug delivery routes and sustained delivery devices for new and existing drugs.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Choroid/drug effects , Fluorescein/pharmacokinetics , Models, Biological , Animals , Basement Membrane/metabolism , Bevacizumab/pharmacokinetics , Biological Transport , Drug Delivery Systems , Imaging, Three-Dimensional , Injections, Intraocular , Intravitreal Injections , Rabbits , Ranibizumab/pharmacokinetics , Retinal Pigment Epithelium/metabolism , Tissue Distribution , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vitreous Body/metabolism
10.
J Clin Invest ; 124(10): 4564-76, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25180601

ABSTRACT

Retinal and choroidal neovascularization (NV) and vascular leakage contribute to visual impairment in several common ocular diseases. The angiopoietin/TIE2 (ANG/TIE2) pathway maintains vascular integrity, and negative regulators of this pathway are potential therapeutic targets for these diseases. Here, we demonstrated that vascular endothelial-protein tyrosine phosphatase (VE-PTP), which negatively regulates TIE2 activation, is upregulated in hypoxic vascular endothelial cells, particularly in retinal NV. Intraocular injection of an anti-VE-PTP antibody previously shown to activate TIE2 suppressed ocular NV. Furthermore, a small-molecule inhibitor of VE-PTP catalytic activity (AKB-9778) activated TIE2, enhanced ANG1-induced TIE2 activation, and stimulated phosphorylation of signaling molecules in the TIE2 pathway, including AKT, eNOS, and ERK. In mouse models of neovascular age-related macular degeneration, AKB-9778 induced phosphorylation of TIE2 and strongly suppressed NV. Ischemia-induced retinal NV, which is relevant to diabetic retinopathy, was accentuated by the induction of ANG2 but inhibited by AKB-9778, even in the presence of high levels of ANG2. AKB-9778 also blocked VEGF-induced leakage from dermal and retinal vessels and prevented exudative retinal detachments in double-transgenic mice with high expression of VEGF in photoreceptors. These data support targeting VE-PTP to stabilize retinal and choroidal blood vessels and suggest that this strategy has potential for patients with a wide variety of retinal and choroidal vascular diseases.


Subject(s)
Aniline Compounds/pharmacology , Eye/blood supply , Receptor, TIE-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Retinal Vessels/pathology , Sulfonic Acids/pharmacology , Animals , Catalysis , Cell Hypoxia , Choroid/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia , Macular Degeneration , Mice , Mice, Transgenic , Oxygen/metabolism , Phosphorylation , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
11.
J Control Release ; 172(3): 625-33, 2013 Dec 28.
Article in English | MEDLINE | ID: mdl-24126220

ABSTRACT

Doxorubicin (DXR) and daunorubicin (DNR) inhibit hypoxia-inducible factor-1 (HIF-1) transcriptional activity by blocking its binding to DNA. Intraocular injections of DXR or DNR suppressed choroidal and retinal neovascularization (NV), but also perturbed retinal function as demonstrated by electroretinograms (ERGs). DXR was conjugated to novel copolymers of branched polyethylene glycol and poly(sebacic acid) (DXR-PSA-PEG3) and formulated into nanoparticles that when placed in aqueous buffer, slowly released small DXR-conjugates. Intraocular injection of DXR-PSA-PEG3 nanoparticles (1 or 10 µg DXR content) reduced HIF-1-responsive gene products, strongly suppressed choroidal and retinal NV, and did not cause retinal toxicity. In transgenic mice that express VEGF in photoreceptors, intraocular injection of DXR-PSA-PEG3 nanoparticles (10 µg DXR content) suppressed NV for at least 35 days. Intraocular injection of DXR-PSA-PEG3 nanoparticles (2.7 mg DXR content) in rabbits resulted in sustained DXR-conjugate release with detectable levels in aqueous humor and vitreous for at least 105 days. This study demonstrates a novel HIF-1-inhibitor-polymer conjugate formulated into controlled-release particles that maximizes efficacy and duration of activity, minimizes toxicity, and provides a promising new chemical entity for treatment of ocular NV.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Daunorubicin/administration & dosage , Delayed-Action Preparations/chemistry , Doxorubicin/administration & dosage , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Retina/drug effects , Retinal Neovascularization/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Daunorubicin/pharmacokinetics , Daunorubicin/therapeutic use , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Hypoxia-Inducible Factor 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Rabbits , Retina/metabolism , Retina/pathology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology
12.
Biomaterials ; 34(30): 7544-51, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23849876

ABSTRACT

Aberrant angiogenesis can cause or contribute to a number of diseases such as neovascular age-related macular degeneration (NVAMD). While current NVAMD treatments target angiogenesis, these treatments are not effective for all patients and also require frequent intravitreal injections. New agents and delivery systems to treat NVAMD could be beneficial to many patients. We have recently developed a serpin-derived peptide as an anti-angiogenic agent. Here, this peptide is investigated for activity in human retinal endothelial cells in vitro and for reducing angiogenesis in a laser-induced choroidal neovascularization mouse model of NVAMD in vivo. While frequent intravitreal injections can be tolerated clinically, reducing the number of injections can improve patient compliance, safety, and outcomes. To achieve this goal, and to maximize the in vivo activity of injected peptide, we have developed biodegradable polymers and controlled release particle formulations to extend anti-angiogenic therapy. To create these devices, the anionic peptides are first self-assembled into nanoparticles using a biodegradable cationic polymer and then as a second step, these nanoparticles are encapsulated into biodegradable poly(lactic-co-glycolic acid) (PLGA) microparticles. In situ, these particles show approximately zero-order, linear release of the anionic peptide over 200 days. These particles are made of safe, hydrolytically degradable polymers and have low endotoxin. Long-term in vivo experiments in the laser-induced neovascularization model for NVAMD show that these peptide-releasing particles decrease angiogenesis for at least fourteen weeks in vivo following a single particle dose and therefore are a promising treatment strategy for NVAMD.


Subject(s)
Biocompatible Materials/chemistry , Choroidal Neovascularization/drug therapy , Nanoparticles/chemistry , Peptides/administration & dosage , Peptides/therapeutic use , Polymers/chemistry , Serpins/therapeutic use , Animals , Biodegradation, Environmental , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Humans , Intravitreal Injections , Lactic Acid/chemistry , Mice , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Retina/pathology , Serpins/pharmacology
13.
Invest Ophthalmol Vis Sci ; 54(1): 503-11, 2013 Jan 21.
Article in English | MEDLINE | ID: mdl-23169884

ABSTRACT

PURPOSE: To test the effect of pazopanib, a tyrosine kinase inhibitor that blocks VEGF and platelet-derived growth factor (PDGF) receptors and c-Kit, on vascular leakage and neovascularization (NV) in the retina. METHODS: Pazopanib was tested to determine its effect on VEGF-induced vascular permeability via measurement of [(3)H]mannitol retina to lung (RLLR) and retina to renal leakage ratios (RRLR) and in rho/VEGF mice with subretinal NV. In rabbits, the effect of intravitreal, topical, and systemic pazopanib on VEGF-induced leakage was tested by vitreous fluorophotometry. RESULTS: In mice, oral pazopanib (40 mg/kg twice a day [bid]) reduced RLLR (0.84 to 0.58, P = 0.0014) and RRLR (0.55 to 0.30, P = 0.0018) in VEGF-injected eyes. After intraocular injection of VEGF into both eyes, topical pazopanib (10 mg/mL three times a day [tid] for 14 days) reduced RLLR (0.85 vs. 0.56, P = 0.001), RRLR (0.44 vs. 0.28, P = 0.0075), and immunoreactive albumin in the retina compared to values in fellow eye controls. Treatment of one eye of rho/VEGF mice with 10 mg/mL, but not 5 mg/mL, pazopanib tid reduced the mean area of subretinal NV compared to that in fellow eyes (0.0055 vs. 0.0025 mm(2), P = 0.020). In rabbits, intravitreal pazopanib suppressed VEGF-induced fluorescein leakage, but topical (10 mg/mL four times a day [qid] or 12 mg/mL bid) had no significant effect. Systemic administration of pazopanib by osmotic pump with or without 10 mg/mL drops tid also failed to suppress VEGF-induced leakage. CONCLUSIONS: Administration of pazopanib topically or systemically suppressed retinal vascular leakage in mice, but not rabbits. These data suggest differences in the blood-retinal barrier (BRB) of mice and rabbits and indicate that penetration through the outer BRB may be needed for topically administered drugs to exert effects in the retina.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Blood-Retinal Barrier/drug effects , Capillary Permeability/drug effects , Pyrimidines/therapeutic use , Retinal Neovascularization/prevention & control , Sulfonamides/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Administration, Oral , Administration, Topical , Angiogenesis Inhibitors/administration & dosage , Animals , Female , Fluorophotometry , Indazoles , Intravitreal Injections , Kidney/metabolism , Lung , Mannitol/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Pyrimidines/administration & dosage , Rabbits , Species Specificity , Sulfonamides/administration & dosage , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/toxicity
14.
J Cell Physiol ; 225(3): 855-64, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20607799

ABSTRACT

TM601 is a synthetic polypeptide with sequence derived from the venom of the scorpion Leiurus quinquestriatus that has anti-neoplastic activity. It has recently been demonstrated to bind annexin A2 on cultured tumor and vascular endothelial cells and to suppress blood vessel growth on chick chorioallantoic membrane. In this study, we investigated the effects of TM601 in models of ocular neovascularization (NV). When administered by intraocular injection, intravenous injections, or periocular injections, TM601 significantly suppressed the development of choroidal NV at rupture sites in Bruch's membrane. Treatment of established choroidal NV with TM601 caused apoptosis of endothelial cells and regression of the NV. TM601 suppressed ischemia-induced and vascular endothelial growth factor-induced retinal NV and reduced excess vascular permeability induced by vascular endothelial growth factor. Immunostaining with an antibody directed against TM601 showed that after intraocular or periocular injection, TM601 selectively bound to choroidal or retinal NV and co-localized with annexin A2, which is undetectable in normal retinal and choroidal vessels, but is upregulated in endothelial cells participating in choroidal or retinal NV. Intraocular injection of plasminogen or tissue plasminogen activator, which like TM601 bind to annexin A2, also suppressed retinal NV. This study supports the hypothesis that annexin A2 is an important target for treatment of neovascular diseases and suggests that TM601, through its interaction with annexin A2, causes suppression and regression of ocular NV and reduces vascular leakage and thus may provide a new treatment for blinding diseases such as neovascular age-related macular degeneration and diabetic retinopathy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Annexin A2/metabolism , Bruch Membrane/blood supply , Choroidal Neovascularization/prevention & control , Retinal Neovascularization/prevention & control , Retinal Vessels/drug effects , Retinopathy of Prematurity/prevention & control , Scorpion Venoms/pharmacology , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Capillary Permeability/drug effects , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/physiopathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fibrinolysin/administration & dosage , Humans , Infant, Newborn , Injections, Intravenous , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/physiopathology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Retinal Vessels/physiopathology , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/physiopathology , Rhodopsin/genetics , Scorpion Venoms/administration & dosage , Scorpion Venoms/metabolism , Tissue Plasminogen Activator/administration & dosage , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
15.
Invest Ophthalmol Vis Sci ; 49(4): 1705-11, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18385094

ABSTRACT

PURPOSE: Nicotinic acetylcholine receptors (nAChR) are best known for their role in neurotransmission, but they have recently been demonstrated on vascular endothelial cells. Acetylcholine is their endogenous ligand, but they are also stimulated by nicotine. By stimulating nAChR, nicotine promotes tumor angiogenesis as well as atherosclerotic plaque neovascularization. In this study, the authors investigated the role of nAChR in the pathogenesis of choroidal neovascularization (CNV). METHODS: The effect of the nonselective nAChR antagonist mecamylamine was tested on human retinal and choroidal endothelial cells in vitro and in a murine model of CNV. RESULTS: Several nAChR isoforms were identified in retinal and choroidal microvascular endothelial cells, and the ability of these cells to form tubules when grown in growth factor-reduced basement membrane matrix and supplemented with VEGF was suppressed by the nAChR antagonist mecamylamine. Supplementation of the drinking water of mice with nicotine increased the size of CNV lesions at Bruch membrane rupture sites, an effect that was blocked by subcutaneous administration of mecamylamine (50 mg/kg/d) by an osmotic pump. In the absence of nicotine, CNV formation was suppressed by the infusion of 50 mg/kg/d mecamylamine or by topical application 0.1 or 1% mecamylamine to the cornea. CONCLUSIONS: These data suggest that endogenous activation of nAChR promotes CNV and that activation of nAChR by nicotine may contribute to the increased incidence of CNV seen in smokers with age-related macular degeneration (AMD). Topically administered mecamylamine could provide an appealing new treatment approach for CNV.


Subject(s)
Choroidal Neovascularization/prevention & control , Disease Models, Animal , Endothelium, Vascular/drug effects , Mecamylamine/pharmacology , Nicotine/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/metabolism , Animals , Cells, Cultured , Choroid/blood supply , Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Endothelium, Vascular/metabolism , Female , Humans , Immunoblotting , Mecamylamine/administration & dosage , Mice , Mice, Inbred C57BL , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptors, Nicotinic/genetics , Retinal Vessels/cytology
16.
FASEB J ; 21(12): 3219-30, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17522382

ABSTRACT

Hypoxia causes increased expression of several proteins that have the potential to promote neovascularization. Vascular endothelial growth factor (VEGF) is up-regulated by hypoxia in the retina and plays a central role in the development of several types of ocular neovascularization, but the effects of other hypoxia-regulated proteins are less clear. Stromal-derived factor-1 (SDF-1) and its receptor, CXCR4, have hypoxia response elements in the promoter regions of their genes and are increased in hypoxic liver and heart. In this study, we found that SDF-1 and CXCR4 are increased in hypoxic retina, with SDF-1 localized in glial cells primarily near the surface of the retina and CXCR4 localized in bone marrow-derived cells. Glial cells also expressed CXCR4, which suggested the possibility of autocrine stimulation, but influx of bone marrow-derived cells is the major source of increased levels of CXCR4. High levels of VEGF in the retina in the absence of hypoxia also increased levels of Cxcr4 and Sdf1 mRNA. CXCR4 antagonists reduced influx of bone marrow-derived cells into ischemic retina and strongly suppressed retinal neovascularization, VEGF-induced subretinal neovascularization, and choroidal neovascularization. These data suggest that SDF-1 and CXCR4 contribute to the involvement of bone marrow-derived cells and collaborate with VEGF in the development of several types of ocular neovascularization. They provide new targets for therapeutic intervention that may help to bolster and supplement effects obtained with VEGF antagonists.


Subject(s)
Chemokine CXCL12/metabolism , Corneal Neovascularization , Hypoxia , Receptors, CXCR4/metabolism , Retina/anatomy & histology , Retina/physiology , Retinal Neovascularization , Animals , Antigens, Differentiation/metabolism , Bone Marrow Cells , Chemokine CXCL12/genetics , Humans , Ischemia/metabolism , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pyridines/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Retina/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
17.
Exp Eye Res ; 83(5): 1260-7, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16950258

ABSTRACT

Periocular injections of the polyamine analog CGC-11144 three times a week causes regression of choroidal neovascularization. This regimen was selected to maximize chances of success for proof of concept, but is not ideal for clinical application. In this study we explored other regimens for periocular delivery of CGC-11144, and 2 other polyamine analogs, CGC-11047 and CGC-11093. A single periocular injection of 200 microg of CGC-11144, 2 mg of CGC-11047, or 1.5 mg of CGC-11093 caused significant suppression and regression of laser-induced choroidal neovascularization. An injection of 2 mg of CGC-11047 or 1.5 mg of CGC-11093 one or two weeks before, but not 3 weeks before, rupture of Bruch's membrane also caused significant suppression. Periocular injection of polyamine analogs also caused strong inhibition of retinal or subretinal neovascularization in mice with oxygen-induced ischemic retinopathy or Rhodopsin promoter/VEGF transgenic mice, respectively. These data suggest that periocular injection of one of 3 different polyamine analogs inhibits retinal or choroidal neovascularization and a single injection provides inhibitory activity for at least 2 to 3 weeks, which could provide the basis for a feasible treatment regimen for clinical trials.


Subject(s)
Biogenic Polyamines/administration & dosage , Choroidal Neovascularization/drug therapy , Ophthalmic Solutions/administration & dosage , Animals , Bruch Membrane/injuries , Disease Models, Animal , Drug Administration Schedule , Female , Injections, Intraperitoneal , Ischemia/drug therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen , Photoreceptor Cells, Vertebrate/chemistry , Polyamines/pharmacology , Retinal Neovascularization/drug therapy , Retinal Vessels/drug effects , Rupture , Time Factors , Treatment Outcome , Vascular Endothelial Growth Factor A/analysis
18.
J Cell Physiol ; 208(1): 161-6, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16557520

ABSTRACT

Vascular endothelial cells receive proangiogenic or antiangiogenic signals from components of extracellular matrix (ECM) depending upon the situation and many molecular signals can have opposite effects in different vascular beds. Tissue inhibitor of metalloproteinase 1 is antiangiogenic in several tissues, but promotes retinal neovascularization. When cleaved from native collagens, several of the non-collagenous domains (NC1) of basement membrane collagens have antiangiogenic effects in some tissues, but this is context dependent for the NC1 of the alpha 1 chain of collagen IV. It is critical to examine effects in several well-defined model systems before assuming that an ECM component is universally antiangiogenic. In this study, we examined the effects of a recombinant fragment of NC1 of the alpha 2 chain of type IV collagen (alpha2(IV)NC1) in a well-characterized model of ocular neovascularization. Intravitreous or periocular injections of alpha2(IV)NC1 caused selective apoptosis of endothelial cells participating in neovascularization resulting in suppression of neovascularization when the peptide was given prior to onset of new vessel sprouting. Importantly, when the peptide was given after neovascularization had already developed, it caused the new vessels to regress. This suggests that alpha2(IV)NC1, which has previously been shown to suppress tumor angiogenesis in xenograft models, is also a strong antiangiogenic agent in the choroid and is a therapeutic candidate for treatment of neovascular age-related macular degeneration.


Subject(s)
Apoptosis/drug effects , Choroidal Neovascularization/prevention & control , Collagen Type IV/pharmacology , Neovascularization, Physiologic/drug effects , Recombinant Proteins/pharmacology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Apoptosis/physiology , Choroid/blood supply , Choroid/drug effects , Choroid/pathology , Choroid/physiopathology , Choroidal Neovascularization/etiology , Choroidal Neovascularization/physiopathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/physiology , Extracellular Matrix/physiology , Female , In Situ Nick-End Labeling , Injections , Lasers , Macular Degeneration/drug therapy , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Protein Structure, Tertiary
19.
Invest Ophthalmol Vis Sci ; 46(9): 3323-30, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123436

ABSTRACT

PURPOSE: Polyamine analogues inhibit tumor growth in vitro and in vivo, and oligoamines with a chain length of 10, 12, or 14 are particularly potent. This study was conducted to investigate the effect of the decamines CGC-11144 and CGC-11150 in a mouse model of choroidal neovascularization (CNV). METHODS: Mice with laser-induced rupture of Bruch's membrane were given intraperitoneal, intravitreous, or periocular injection of CGC-11144, CGC-11150, or vehicle, and after 14 days, they were perfused with fluorescein-labeled dextran, and the area of CNV was measured on choroidal flatmounts by image analysis. In some groups of mice, treatments were started 7 days after rupture of Bruch's membrane to determine the effect of the agent on established CNV. Electroretinograms (ERGs) were performed to assess the effects on retinal function, and histopathology was used to evaluate retinal structure. RESULTS: Intraperitoneal injection of 10 or 20 mg/kg CGC-11144 or CGC-11150 resulted in small but significant reductions in the area of CNV. Intravitreous injection of 20 microg CGC-11144 or CGC-11150 on days 0 and 7 after rupture of Bruch's membrane resulted in a approximately 40% reduction in the area of CNV, with a similar reduction after periocular injections of 0.2 mg CGC-11144 three times a week for 2 weeks. Both intravitreous and periocular delivery of CGC-11144 also caused significant regression of established CNV. Within 2 days of periocular injection of CGC-11144, there was apoptosis in CNV lesions, but not in normal blood vessels or other retinal cells. Periocular injections of d,l-alpha-difluoromethyl-ornithine (DFMO), which decreases polyamine levels by a different mechanism, also inhibited CNV. There was no decline in ERG amplitudes or abnormal retinal morphology after daily injections of 0.2 mg CGC-11144 for 2 weeks, but a single intravitreous injection compromised retinal structure and function. CONCLUSIONS: Periocular delivery of the polyamine analogues may be a useful approach for the treatment of CNV.


Subject(s)
Biogenic Polyamines/therapeutic use , Choroidal Neovascularization/drug therapy , Animals , Apoptosis , Biogenic Polyamines/administration & dosage , Bruch Membrane/surgery , Choroidal Neovascularization/diagnosis , Choroidal Neovascularization/physiopathology , Dextrans , Disease Models, Animal , Drug Administration Routes , Eflornithine/therapeutic use , Electroretinography , Female , Fluorescein Angiography , Fluoresceins , In Situ Nick-End Labeling , Injections, Intraperitoneal , Laser Therapy , Mice , Mice, Inbred C57BL , Orbit , Retina/physiology , Vitreous Body
20.
J Cell Physiol ; 195(2): 241-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12652651

ABSTRACT

Vascular endothelial growth factor (VEGF) plays a central role in the development of retinal neovascularization and diabetic macular edema. There is also evidence suggesting that VEGF is an important stimulator for choroidal neovascularization. In this study, we investigated the effect of a specific inhibitor of VEGF, VEGF-TRAP(R1R2), in models for these disease processes. VEGF-TRAP(R1R2) is a fusion protein, which combines ligand binding elements taken from the extracellular domains of VEGF receptors 1 and 2 fused to the Fc portion of IgG1. Subcutaneous injections or a single intravitreous injection of VEGF-TRAP(R1R2) strongly suppressed choroidal neovascularization in mice with laser-induced rupture of Bruch's membrane. Subcutaneous injection of VEGF-TRAP(R1R2) also significantly inhibited subretinal neovascularization in transgenic mice that express VEGF in photoreceptors. In two models of VEGF-induced breakdown of the blood-retinal barrier (BRB), one in which recombinant VEGF is injected into the vitreous cavity and one in which VEGF expression is induced in the retina in transgenic mice, VEGF-TRAP(R1R2) significantly reduced breakdown of the BRB. These data confirm that VEGF is a critical stimulus for the development of choroidal neovascularization and indicate that VEGF-TRAP(R1R2) may provide a new agent for consideration for treatment of patients with choroidal neovascularization and diabetic macular edema.


Subject(s)
Choroid Diseases/drug therapy , Diabetic Retinopathy/drug therapy , Endothelial Growth Factors/antagonists & inhibitors , Lymphokines/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Recombinant Fusion Proteins/pharmacology , Animals , Blood-Retinal Barrier/drug effects , Blood-Retinal Barrier/physiology , Choroid/drug effects , Choroid/metabolism , Choroid/physiopathology , Choroid Diseases/metabolism , Choroid Diseases/physiopathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/physiopathology , Disease Models, Animal , Endothelial Growth Factors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Receptors, Growth Factor/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Retina/drug effects , Retina/metabolism , Retina/physiopathology , Retinal Artery/drug effects , Retinal Artery/pathology , Retinal Artery/physiopathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...