Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Atherosclerosis ; 250: 158-65, 2016 07.
Article in English | MEDLINE | ID: mdl-27236706

ABSTRACT

BACKGROUND AND AIMS: Besides LDL-cholesterol, local vascular inflammation plays a key role in atherogenesis. Efficient therapies to treat the inflammatory component of the disease have not been established. The discovery of specialized inflammation-resolving mediators, such as resolvins may provide new opportunities for treatment. This study examines whether the ω-3 fatty acid eicosapentaenoic acid-derived resolvin E1 (RvE1), can reduce atherosclerosis, when administered alone or in combination with a cholesterol-lowering statin. METHODS: ApoE*3Leiden mice were fed a hypercholesterolemic diet for 9 weeks and subsequently treated with RvE1-low (1 mg/kg/day), RvE1-high (5 mg/kg/day), atorvastatin (1.5 mg/kg/day) or the combination of atorvastatin and RvE1-low for the following 16 weeks. RESULTS: RvE1-low and RvE1-high reduced atherosclerotic lesion size to the same extent (-35%; p < 0.05), attenuated the formation of severe lesions, also seen as a proportional increase in the presence of mild lesions, but did not alter plasma cholesterol levels. Cholesterol-lowering atorvastatin reduced atherosclerosis (-27%, p < 0.05), and the combination of RvE1 and atorvastatin further attenuated lesion size (-51%, p < 0.01) and increased the content of mild lesions. RvE1 did not affect plasma SAA, E-selectin, VCAM-1 or MCP-1 but did reduce plasma EPHX4 and down-regulated the local expression of pro-atherogenic genes in the aortae, (e.g. Cd74, Cd44, Ccl2, Ccr5 and Adam17) and significantly inactivated IFN-γ (p < 0.001) and TNF-α (p < 0.001) signalling pathways. CONCLUSIONS: RvE1 attenuates atherogenesis both alone and on top of a statin. The local effects of RvE1 are demonstrated by the modulated aortic expression of genes involved in inflammatory and immune responses, without altering plasma cholesterol or circulating SAA.


Subject(s)
Aorta/pathology , Atorvastatin/pharmacology , Cholesterol/blood , Eicosapentaenoic Acid/analogs & derivatives , Lipids/blood , Animals , Atherosclerosis/blood , Cholesterol, LDL/blood , E-Selectin/blood , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Omega-3/metabolism , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation , Mice , Mice, Knockout, ApoE , Oligonucleotide Array Sequence Analysis , Serum Amyloid A Protein/metabolism , Treatment Outcome , Vascular Cell Adhesion Molecule-1/blood
2.
Invest Ophthalmol Vis Sci ; 56(4): 2728-36, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25758817

ABSTRACT

PURPOSE: To investigate the role of the lipid mediator, resolvin E1 (RvE1), in corneal inflammation. METHODS: The effect of RvE1 on stimulated human corneal epithelial cells (HCECs) and neutrophils, and mouse macrophage was assessed. C57BL/6 mouse corneas were abraded and treated with RvE1 either before or after stimulation with lipopolysaccharide (LPS) and antibiotic-killed Pseudomonas aeruginosa and Staphylococcus aureus. The levels of CXC chemokines in the cornea were quantified, and the presence of neutrophils in corneal infiltrates was detected by immunohistochemistry and by in vivo confocal microscopy. The effect of RvE1 on apoptosis in the corneal epithelium was assessed using the TUNEL assay. RESULTS: RvE1 significantly inhibited cytokine production in HCECs and neutrophils, and mouse macrophages and cornea. The development of corneal infiltrates, specifically neutrophils, in response to stimulation with LPS, P. aeruginosa, and S. aureus was also significantly reduced. There was no apoptotic effect of RvE1 on mouse corneal epithelial cells. CONCLUSIONS: RvE1 inhibits corneal inflammation induced by LPS, Gram negative (P. aeruginosa) and Gram positive (S. aureus) bacteria. These findings indicate that RvE1 as a potential anti-inflammatory therapy for patients with corneal inflammation and also, when given together with antibiotics, for bacterial keratitis.


Subject(s)
Cornea/pathology , Eicosapentaenoic Acid/analogs & derivatives , Eye Infections, Bacterial/drug therapy , Keratitis/drug therapy , Animals , Apoptosis/drug effects , Cornea/drug effects , Cornea/microbiology , Disease Models, Animal , Eicosapentaenoic Acid/therapeutic use , Eye Infections, Bacterial/microbiology , Eye Infections, Bacterial/pathology , Humans , Keratitis/microbiology , Keratitis/pathology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Treatment Outcome
3.
Invest Ophthalmol Vis Sci ; 54(9): 6269-79, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23942967

ABSTRACT

PURPOSE: Neuroprotectin D1 (NPD1) is an anti-inflammatory and proresolving lipid mediator biosynthesized from the omega-3-polyunsaturated fatty acid docosahexaenoic acid (DHA). The purpose of this study is to test the therapeutic potential of NPD1 for the treatment of herpes simplex virus (HSV)-induced stromal keratitis (SK) using a mouse model. METHODS: C57BL/6 mice were infected ocularly with HSV-1 strain RE. Infected animals were treated topically with methyl ester prodrug NPD1 (300 ng/eye, 5-µL drop). Development of SK lesions, infiltration of inflammatory cells into the cornea, and production of proinflammatory cytokines, chemokines, and angiogenic factors were compared to untreated animals using slit-lamp biomicroscopy, flow cytometry, ELISA, and quantitative PCR (qPCR). RESULTS: Topical administration of NPD1 resulted in a significant reduction in the severity and incidence of SK, as well as the extent of corneal neovascularization in the NPD1-treated animals compared to their untreated counterparts. Infiltration of fewer neutrophils and pathogenic CD4⁺ T cells into the cornea, along with a lower number of cells that could be induced ex vivo to produce IFN-γ and IL-17, occurred with NPD1 treatment. Additionally, treatment with NPD1 diminished the production of proinflammatory cytokines, chemokines, and angiogenic factors, such as IL-6, CXCL1, CXCL-10, CCL-20, VEGF-A, MMP-2, and MMP-9 in the corneas of infected animals. Importantly, treatment with NPD1 increased the production of the anti-inflammatory cytokine, IL-10. CONCLUSIONS: Our novel findings demonstrate that NPD1 treatment could represent a valuable therapeutic approach to control SK lesions.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Docosahexaenoic Acids/therapeutic use , Keratitis, Herpetic/drug therapy , Administration, Topical , Animals , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/metabolism , Chemokines/metabolism , Corneal Neovascularization/drug therapy , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunohistochemistry , Keratitis, Herpetic/immunology , Mice , Mice, Inbred C57BL , Neutrophils/metabolism
4.
Proc Natl Acad Sci U S A ; 109(37): 14983-8, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22927428

ABSTRACT

Inappropriate neutrophil activation contributes to the pathogenesis of acute lung injury (ALI). Apoptosis is essential for removal of neutrophils from inflamed tissues and timely resolution of inflammation. Resolvin E1 (RvE1) is an endogenous lipid mediator derived from the ω-3 polyunsaturated fatty acid eicosapentaenoic acid that displays proresolving actions. Because the balance of prosurvival and proapoptosis signals determines the fate of neutrophils, we investigated the impact of RvE1 on neutrophil apoptosis and the outcome of neutrophil-mediated pulmonary inflammation in mice. Culture of human neutrophils with RvE1 accelerated apoptosis evoked by phagocytosis of opsonized Escherichia coli or yeast. RvE1 through the leukotriene B(4) receptor BLT1 enhanced NADPH oxidase-derived reactive oxygen species generation and subsequent activation of caspase-8 and caspase-3. RvE1 also attenuated ERK and Akt-mediated apoptosis-suppressing signals from myeloperoxidase, serum amyloid A, and bacterial DNA, shifting the balance of pro- and anti-survival signals toward apoptosis via induction of mitochondrial dysfunction. In mice, RvE1 treatment enhanced the resolution of established neutrophil-mediated pulmonary injury evoked by intratracheal instillation or i.p. administration of live E. coli or intratracheal instillation of carrageenan plus myeloperoxidase via facilitating neutrophil apoptosis and their removal by macrophages. The actions of RvE1 were prevented by the pan-caspase inhibitor zVAD-fmk. These results identify a mechanism, promotion of phagocytosis-induced neutrophil apoptosis and mitigation of potent anti-apoptosis signals, by which RvE1 could enhance resolution of acute lung inflammation.


Subject(s)
Apoptosis/drug effects , Eicosapentaenoic Acid/analogs & derivatives , Neutrophils/drug effects , Phagocytosis/drug effects , Pneumonia/drug therapy , Amino Acid Chloromethyl Ketones , Analysis of Variance , Animals , Caspase 3/metabolism , Caspase 8/metabolism , Cells, Cultured , Eicosapentaenoic Acid/antagonists & inhibitors , Eicosapentaenoic Acid/pharmacology , Escherichia coli , Humans , Mice , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Receptors, Leukotriene B4/metabolism , Statistics, Nonparametric , Yeasts
5.
Ocul Surf ; 10(2): 108-16, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22482471

ABSTRACT

In September 2010, a Symposium in Florence, Italy, was held to address the unmet need for global treatments for dry eye disease (DED). It was sponsored by The Tear Film & Ocular Surface Society (TFOS; www.TearFilm.org) and co-sponsored by the Association for Research in Vision & Ophthalmology (www.arvo.org). The Symposium objectives were two-fold: first, to discuss accepted and emerging clinical endpoints of DED with regulatory experts from around the world; and second, to consider how to improve clinical trials of treatments for DED. The Symposium focused on the personal and collective burden of DED, as well as the developmental and regulatory challenges associated with generating new DED therapeutics. This article provides a synopsis of many of the presentations, discussions and recommendations of this Symposium.


Subject(s)
Dry Eye Syndromes/therapy , Health Services Needs and Demand , Needs Assessment , Global Health , Humans
6.
Cornea ; 31(11): 1299-303, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22257864

ABSTRACT

PURPOSE: Resolvin E1 (RvE1; RX-10001) belongs to a new class of endogenous immunoregulating mediators, originally identified as a metabolite of the omega-3 polyunsaturated fatty acid, eicosapentaenoic acid. Based on its proven efficacy in models of chronic inflammation, this study investigated the efficacy of resolvin E1 in a murine model of dry eye. METHODS: C57/B6 mice, aged 6 to 8 weeks, were treated with systemic scopolamine and exposed to air draft and low humidity for 16 hours/day for 5 days and allocated to the following groups: unexposed controls, disease controls, treatment with vehicle or RvE1 delivered topically as its methyl ester prodrug, RX-10005, to enhance corneal surface penetration. Treatment was initiated at the time of desiccating stress induction. Treatment efficacy was assessed by corneal permeability using Oregon Green Dextran and by conjunctival goblet cell density using periodic acid-Schiff reagent. RESULTS: RvE1 reduced the increase in corneal staining by 80% compared with untreated disease controls. Goblet cell density was reduced by 20% in disease controls but fully maintained in the group receiving RvE1. CONCLUSIONS: RvE1, delivered as its methyl ester prodrug, improved the outcome measures of corneal staining and goblet cell density in this murine model of dry eye, indicating the potential utility of endogenous resolvins and resolvin analogues in the treatment of dry eye.


Subject(s)
Disease Models, Animal , Dry Eye Syndromes/prevention & control , Eicosapentaenoic Acid/analogs & derivatives , Epithelium, Corneal/drug effects , Goblet Cells/cytology , Prodrugs/pharmacology , Administration, Topical , Animals , Biological Transport , Cell Count , Cell Membrane Permeability/physiology , Cell Survival , Dry Eye Syndromes/metabolism , Eicosapentaenoic Acid/pharmacology , Epithelium, Corneal/metabolism , Female , Fluorescent Dyes/metabolism , Male , Mice , Mice, Inbred C57BL
7.
Epilepsia ; 52(9): 1601-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21569016

ABSTRACT

PURPOSE: Temporal lobe epilepsy, one of the most common epilepsy syndromes, is characterized by hippocampal hyperexcitability and progressive seizure susceptibility. Omega-3 fatty acids are involved in neuronal excitability and have anticonvulsant properties. We studied the effect of docosahexaenoic acid (DHA) or its derived lipid mediator, neuroprotectin D1 (NPD1, 10R,17S-dihydroxy-docosa-4Z,7Z,11E,13E,15Z,19Z-hexaenoic acid), in evoked seizures using a rapid kindling model of temporal lobe epilepsy. METHODS: DHA or NPD1 was administered in rodents with or without kindling acquisition. Locomotor seizures and evoked epileptiform hippocampal activity immediately after hippocampal stimulations were analyzed. KEY FINDINGS: DHA or NPD1 limits hippocampal electrically induced hyperexcitability. Seizures induced by kindling triggered NPD1 synthesis in the hippocampus. Supplying its precursor, DHA, or direct injection of NPD1 into the third ventricle resulted in attenuation of kindling progression and hippocampal hyperexcitability. SIGNIFICANCE: The significance of NPD1 in temporal lobe epilepsy could open new pathways for understanding the initiation and propagation of seizures and the role this lipid mediator plays in the neuronal network.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/pathology , Hippocampus/drug effects , Neuroprotective Agents/administration & dosage , Animals , Biophysics , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Drug Administration Routes , Electric Stimulation/adverse effects , Electroencephalography/methods , Epilepsy, Temporal Lobe/etiology , Hippocampus/physiology , Kindling, Neurologic/drug effects , Kindling, Neurologic/physiology , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/metabolism , Rats , Rats, Wistar
8.
J Immunol ; 186(3): 1735-46, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21187448

ABSTRACT

Stromal keratitis (SK) is a chronic immunopathological lesion of the eye caused by HSV-1 infection and a common cause of blindness in humans. The inflammatory lesions are primarily perpetuated by neutrophils with the active participation of CD4(+) T cells. Therefore, targeting these immune cell types represents a potentially valuable form of therapy to reduce the severity of disease. Resolvin E1 (RvE1), an endogenous lipid mediator, was shown to promote resolution in several inflammatory disease models. In the current report, we determined whether RvE1 administration begun at different times after ocular infection of mice with HSV could influence the severity of SK lesions. Treatment with RvE1 significantly reduced the extent of angiogenesis and SK lesions that occurred. RvE1-treated mice had fewer numbers of inflammatory cells that included Th1 and Th17 cells as well as neutrophils in the cornea. The mechanisms by which RvE1 acts appear to be multiple. These included reducing the influx of neutrophils and pathogenic CD4(+) T cells, increasing production of the anti-inflammatory cytokine IL-10, and inhibitory effects on the production of proinflammatory mediators and molecules, such as IL-6, IFN-γ, IL-17, KC, VEGF-A, MMP-2, and MMP-9, that are involved in corneal neovascularization and SK pathogenesis. These findings are, to our knowledge, the first to show that RvE1 treatment could represent a novel approach to control lesion severity in a virally induced immunopathological disease.


Subject(s)
Antiviral Agents/administration & dosage , Eicosapentaenoic Acid/analogs & derivatives , Herpesvirus 1, Human/immunology , Inflammation Mediators/administration & dosage , Keratitis, Herpetic/immunology , Keratitis, Herpetic/prevention & control , Administration, Topical , Angiogenesis Inhibitors/administration & dosage , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Movement/drug effects , Cell Movement/immunology , Cornea/blood supply , Cornea/drug effects , Disease Models, Animal , Eicosapentaenoic Acid/administration & dosage , Female , Herpesvirus 1, Human/drug effects , Humans , Keratitis, Herpetic/virology , Mice , Mice, Inbred BALB C , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology
9.
J Ocul Pharmacol Ther ; 26(5): 431-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20874497

ABSTRACT

PURPOSE: Dry eye (DE) is a common ocular surface disease, particularly among women and the elderly, with chronic symptoms of eye irritation and, in severe cases, blurred vision. Several studies have shown that there is an inflammatory component in DE, although the pathogenesis is not thoroughly understood. Resolvin E1 (RvE1; RX-10001) is an endogenous mediator derived from the omega-3 polyunsaturated fatty acid eicosapentaenoic acid and is involved in inflammation resolution and tissue protection. Here we investigated the role of RvE1 in a DE mouse model. METHODS: Thirteen- to 14-week-old female BALB/C mice were exposed to desiccating conditions. One week after DE exposure, animals were treated topically with drug or vehicle 4 times per day for an additional week. Controls were nontreated animals placed in a normal environment. Schirmer's test was performed before treatment initiation and at days 2 and 4 after treatment. Density of corneal epithelial cells was analyzed in vivo using the Rostock Cornea Module of the Heidelberg Retina Tomograph (HRT-II). Corneas were processed using Western blot analysis and immunofluorescence examination. RESULTS: Schirmer's test showed a significant decrease in tear production in DE compared with controls. There was no change at 2 and 4 days after treatment with the vehicle, but a significant increase was observed at 2 and 4 days in the RvE1-treated group. The density of the superficial epithelial cells showed a significant decrease after DE compared with controls, which increased after 7 days of RvE1 treatment. Western blot analysis showed that α-smooth muscle actin and cyclooxygenase-2 (COX-2) expression were strongly upregulated after DE and decreased after 7 days of RvE1 treatment. Immunofluorescence confirmed strong positive staining of α-smooth muscle actin and COX-2 in stroma and/or in epithelia after DE, which decreased with RvE1 treatment. The percentage of infiltrating CD4+ T cells and CD11b+ cells decreased after RvE1 treatment when compared with DE. CONCLUSION: RvE1 promotes tear production, corneal epithelial integrity, and a decrease in inflammatory inducible COX-2. In the stroma, RvE1 inhibits keratocyte transformation to myofibroblasts and lowers the number of monocytes/macrophages in this DE mouse model. These results suggest that RvE1 and similar resolvin analogs have therapeutic potential in the treatment of DE.


Subject(s)
Dry Eye Syndromes/drug therapy , Eicosapentaenoic Acid/analogs & derivatives , Inflammation/drug therapy , Actins/metabolism , Animals , CD11b Antigen/metabolism , CD4 Antigens/metabolism , Cornea/drug effects , Cornea/pathology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Dry Eye Syndromes/etiology , Dry Eye Syndromes/pathology , Eicosapentaenoic Acid/pharmacology , Epithelial Cells/metabolism , Female , Inflammation/chemically induced , Inflammation/complications , Inflammation/prevention & control , Mice , Mice, Inbred BALB C , Myofibroblasts/metabolism , Tears/drug effects
10.
Invest Ophthalmol Vis Sci ; 51(11): 5601-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20538990

ABSTRACT

PURPOSE: To determine whether resolvin E1 (RvE1), an endogenous oxygenation product of eicosapentaenoic acid (EPA), induces increases in migration in human corneal epithelial cells (HCECs) and to identify signal pathways mediating this response. METHODS: Migration was measured with the scratch wound assay. Western blot analysis identified changes in the phosphorylation status of prospective intracellular signal transduction mediators. Immunocytochemistry probed for intracellular paxillin localization and actin reorganization. RESULTS: RvE1 enhanced HCEC migratory rates to levels comparable to those induced by epidermal growth factor (EGF). These increases were accompanied by increases in the phosphorylation status of epidermal growth factor receptor (EGFR), Akt, p38 MAPK, GSK-3α/ß, and paxillin, which essentially persisted for up to 60 minutes. The EGFR inhibitor AG1478 blocked the subsequent effects of RvE1 to induce increases in phosphorylation status and cell migration. The PI3-K inhibitor LY294002 or wortmannin or the p38 inhibitor BIRB796 blocked resolvin-induced increases in cell migration. Either the matrix metalloproteinase (MMP) inhibitor GM6001 or the specific heparin-bound EGF-like growth factor inhibitor CRM197 suppressed RvE1-induced stimulation of EGFR/PI3-K/Akt phosphorylation and cell migration. CONCLUSIONS: RvE1 enhances HCEC migration through MMP and sheddase-mediated EGFR transactivation. This response is dependent on PI3-K and p38-linked signaling eliciting paxillin (Tyr118) phosphorylation.


Subject(s)
Cell Movement/drug effects , Eicosapentaenoic Acid/analogs & derivatives , Epithelium, Corneal/cytology , ErbB Receptors/genetics , Transcriptional Activation , Actins/metabolism , Blotting, Western , Cell Movement/physiology , Dose-Response Relationship, Drug , Eicosapentaenoic Acid/antagonists & inhibitors , Eicosapentaenoic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Epithelium, Corneal/metabolism , ErbB Receptors/metabolism , Fluorescent Antibody Technique, Indirect , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Paxillin/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Wound Healing/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Mol Vis ; 16: 320-9, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20216940

ABSTRACT

PURPOSE: To examine the effects of neuroprotectin D1 (NPD1), a stereospecific derivative of docosahexaenoic acid, on choroidal neovascularization (CNV) in a laser-induced mouse model. Specifically, this was assessed by clinically grading laser-induced lesions, measuring leakage area, and volumetrically quantifying vascular endothelial cell proliferation. METHODS: C57Bl/6 mice were treated with vehicle control or NPD1, and choroidal neovascularization was induced by laser rupture of Bruch's membrane; treatment was administered throughout the first week of recovery. One and two weeks after CNV induction, fundus fluorescein angiography was performed. Angiograms were clinically graded to assess leakage severity, while leakage area was measured by image analysis of angiograms. Proliferation of vascular endothelial cells was evaluated volumetrically by three-dimensional laser confocal immunofluorescent microscopy of cytoskeletal, nuclear, and endothelial cell markers. RESULTS: At seven days after CNV induction, NPD1-treated mice had 60% fewer clinically relevant lesions than controls, dropping to 80% fewer by 14 days. NPD1 mice exhibited 25% smaller leakage area than controls at 7 days and 44% smaller area at 14 days. Volumetric immunofluorescence revealed 46% less vascular endothelial cell volume in 7-day NPD1-treated mice than in 7-day controls, and by 14 days NPD1 treatment was 68% lower than controls. Furthermore, comparison of 7- and 14-day volumes of NPD1-treated mice revealed a 50% reduction at 14 days. CONCLUSIONS: NPD1 significantly inhibits choroidal neovascularization. There are at least two possible mechanisms that could explain the neuroprotective action of NPD1. Ultimately, nuclear factor-kappaB could be inhibited with a reduction in cyclooxygenase-2 (COX-2) to reduce vascular endothelial growth factor (VEGF) expression, and/or activation of the resolution phase of the inflammatory response/survival pathways could be upregulated. Moreover, NPD1 continues to be effective after treatment is concluded, suggesting sustained protection and highlighting the potential applicability of this lipid mediator in preventing or ameliorating endothelial cell growth in pathoangiogenesis.


Subject(s)
Choroidal Neovascularization/prevention & control , Docosahexaenoic Acids/pharmacology , Animals , Cell Proliferation/drug effects , Cell Size/drug effects , Choroidal Neovascularization/chemically induced , Endothelial Cells/drug effects , Endothelial Cells/pathology , Fluorescein/metabolism , Fluorescein Angiography , Fundus Oculi , Lasers , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Retina/drug effects , Retina/pathology
12.
J Immunol ; 170(8): 4397-402, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682277

ABSTRACT

We have recently demonstrated that a recombinant single-chain bispecific Ab construct, bscCD19xCD3, in vitro induces rapid B lymphoma-directed cytotoxicity at picomolar concentrations with unstimulated peripheral T cells. In this study, we show that treatment of nonobese diabetic SCID mice with submicrogram doses of bscCD19xCD3 could prevent growth of s.c. human B lymphoma xenografts and essentially cured animals when given at an early tumor stage. The effect was dose dependent, dependent on E:T ratio and the time between tumor inoculation and administration of bscCD19xCD3. No therapeutic effect was seen in the presence of human lymphocytes alone, a vehicle control, or with a bispecific single-chain construct of identical T cell-binding activity but different target specificity. In a leukemic nonobese diabetic SCID mouse model, treatment with bscCD19xCD3 prolonged survival of mice in a dose-dependent fashion. The human lymphocytes used as effector cells in both animal models did not express detectable T cell activation markers at the time of coinoculation with tumor cells. The bispecific Ab therefore showed an in vivo activity comparable to that observed in cell culture with respect to high potency and T cell costimulus independence. These properties make bscCD19xCD3 superior to previously investigated CD19 bispecific Ab-based therapies.


Subject(s)
Antibodies, Bispecific/pharmacology , Antigens, CD19/immunology , Antineoplastic Agents/pharmacology , CD3 Complex/immunology , Growth Inhibitors/pharmacology , Leukemia, B-Cell/prevention & control , Lymphoma, B-Cell/prevention & control , T-Lymphocyte Subsets/immunology , Transplantation, Heterologous , Animals , Antibodies, Bispecific/genetics , Antineoplastic Agents/chemical synthesis , Cells, Cultured , Graft Survival/genetics , Graft Survival/immunology , Growth Inhibitors/chemical synthesis , Humans , Injections, Intravenous , Injections, Subcutaneous , Leukemia, B-Cell/genetics , Leukemia, B-Cell/immunology , Leukemia, B-Cell/pathology , Lymphocyte Activation/genetics , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/prevention & control , Time Factors , Transplantation, Heterologous/methods , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL