Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nat Med ; 13(7): 868-873, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17589522

ABSTRACT

Many sight-threatening diseases have two critical phases, vessel loss followed by hypoxia-driven destructive neovascularization. These diseases include retinopathy of prematurity and diabetic retinopathy, leading causes of blindness in childhood and middle age affecting over 4 million people in the United States. We studied the influence of omega-3- and omega-6-polyunsaturated fatty acids (PUFAs) on vascular loss, vascular regrowth after injury, and hypoxia-induced pathological neovascularization in a mouse model of oxygen-induced retinopathy. We show that increasing omega-3-PUFA tissue levels by dietary or genetic means decreased the avascular area of the retina by increasing vessel regrowth after injury, thereby reducing the hypoxic stimulus for neovascularization. The bioactive omega-3-PUFA-derived mediators neuroprotectinD1, resolvinD1 and resolvinE1 also potently protected against neovascularization. The protective effect of omega-3-PUFAs and their bioactive metabolites was mediated, in part, through suppression of tumor necrosis factor-alpha. This inflammatory cytokine was found in a subset of microglia that was closely associated with retinal vessels. These findings indicate that increasing the sources of omega-3-PUFA or their bioactive products reduces pathological angiogenesis. Western diets are often deficient in omega-3-PUFA, and premature infants lack the important transfer from the mother to the infant of omega-3-PUFA that normally occurs in the third trimester of pregnancy. Supplementing omega-3-PUFA intake may be of benefit in preventing retinopathy.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Neovascularization, Pathologic/chemically induced , Retinal Vessels/drug effects , Animals , Diet , Dietary Fats , Fatty Acids, Omega-6/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Retinal Diseases/chemically induced , Retinal Diseases/drug therapy
2.
Angiogenesis ; 16(2): 405-16, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23150059

ABSTRACT

Prominin-1, a pentaspan transmembrane protein, is a unique cell surface marker commonly used to identify stem cells, including endothelial progenitor cells and cancer stem cells. However, recent studies have shown that prominin-1 expression is not restricted to stem cells but also occurs in modified forms in many mature adult human cells. Although prominin-1 has been studied extensively as a stem cell marker, its physiological function of the protein has not been elucidated. We investigated prominin-1 function in two cell lines, primary human endothelial cells and B16-F10 melanoma cells, both of which express high levels of prominin-1. We found that prominin-1 directly interacts with the angiogenic and tumor survival factor vascular endothelial growth factor (VEGF) in both the primary endothelial cells and the melanoma cells. Knocking down prominin-1 in the endothelial cells disrupted capillary formation in vitro and decreased angiogenesis in vivo. Similarly, tumors derived from prominin-1 knockdown melanoma cells had a reduced growth rate in vivo. Further, melanoma cells with knocked down prominin-1 had diminished ability to interact with VEGF, which was associated with decreased bcl-2 protein levels and increased apoptosis. In vitro studies with soluble prominin-1 showed that it stabilized dimer formation of VEGF164, but not VEGF121. Taken together, our findings support the notion that prominin-1 plays an active role in cell growth through its ability to interact and potentiate the anti-apoptotic and pro-angiogenic activities of VEGF. Additionally, prominin-1 promotes tumor growth by supporting angiogenesis and inhibiting tumor cell apoptosis.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Peptides/metabolism , Vascular Endothelial Growth Factor A/metabolism , AC133 Antigen , Apoptosis , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Melanoma/pathology , Protein Binding , Real-Time Polymerase Chain Reaction
3.
Blood ; 114(9): 1987-98, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19465692

ABSTRACT

Angiostatin, a proteolytic fragment of plasminogen, is a potent endogenous antiangiogenic agent. The molecular mechanisms governing angiostatin's antiangiogenic and antitumor effects are not well understood. Here, we report the identification of mitochondrial compartment as the ultimate target of angiostatin. After internalization of angiostatin into the cell, at least 2 proteins within the mitochondria bind this molecule: malate dehydrogenase, a member of Krebs cycle, and adenosine triphosphate synthase. In vitro and in vivo studies revealed differential regulation of key prosurvival and angiogenesis-related proteins in angiostatin-treated tumors and tumor-endothelium. Angiostatin induced apoptosis via down-regulation of mitochondrial BCL-2. Angiostatin treatment led to down-regulation of c-Myc and elevated levels of another key antiangiogenic protein, thrombospondin-1, reinforcing its antitumor and antiangiogenic effects. Further evidence is provided for reduced recruitment and infiltration of bone marrow-derived macrophages in angiostatin-treated tumors. The observed effects of angiostatin were restricted to the tumor site and were not observed in other major organs of the mice, indicating unique tumor specific bioavailability. Together, our data suggest mitochondria as a novel target for antiangiogenic therapy and provide mechanistic insights to the antiangiogenic and antitumor effects of angiostatin.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiostatins/physiology , Apoptosis , Gene Expression Regulation , Mitochondria/metabolism , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Male , Mice , Mice, SCID , Models, Biological , Neoplasms/therapy , Proto-Oncogene Proteins c-bcl-2/metabolism
4.
FASEB J ; 22(10): 3728-35, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18606869

ABSTRACT

The endothelium lining blood vessels serves as a barrier against vascular hyperpermeability, and its maintenance is critical to organ health. Inflammatory mediators evoke tissue edema by disrupting the expression of membrane junctional proteins, which mediate binding between endothelial cell membranes. Endothelial cell-cell junctions form a diffusion barrier between the intravascular and interstitial space. To prevent the morbidity and mortality caused by exaggerated vascular permeability associated with pathological states (e.g., inflammatory and hypersensitivity disorders, pulmonary edema, traumatic lung injury, cerebral edema resulting from stroke, and others), it is important to develop therapeutic approaches to stabilize these interendothelial junctions. Vascular endothelial growth factor (VEGF), a potent proangiogenic cytokine, was first described as vascular permeability factor (VPF). Doxycycline, a tetracycline derivative, has been shown to inhibit angiogenesis in both humans and animal models. We now report that oral doxycycline prevents VPF/VEGF-induced vascular permeability, interleukin-2-induced pulmonary edema, and delayed-type hypersensitivity (DTH) in mice. Remarkably, doxycycline also inhibits tumor growth and tumor-associated vascular hyperpermeability. Finally, we show that doxycycline targets the adherens junction in vascular endothelial cells by inducing the total amount of VE-cadherin expression while decreasing the degree of its phosphorylation. The potential of doxycyline as a therapeutic inhibitor of vascular hyperpermeability in human clinical conditions is promising and warrants further studies.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antigens, CD/biosynthesis , Cadherins/biosynthesis , Capillary Permeability/drug effects , Cell Membrane/drug effects , Doxycycline/administration & dosage , Endothelium, Vascular/drug effects , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Administration, Oral , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/drug therapy , Cell Membrane/metabolism , Cell Proliferation/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Hypersensitivity, Delayed/pathology , Hypersensitivity, Delayed/prevention & control , Interleukin-2/toxicity , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Phosphorylation/drug effects , Pulmonary Edema/chemically induced , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control
5.
FASEB J ; 22(2): 522-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17873101

ABSTRACT

Endometriosis affects 10-15% of women and is associated with pelvic pain and infertility. Angiogenesis plays an essential role in its pathogenesis. Dendritic cells (DCs) were recently implicated in supporting tumor angiogenesis. As both tumors and endometriosis lesions depend on angiogenesis, we investigated the possibility that DCs may also play a role in endometriosis. We induced endometriosis in 8-wk-old female C57BL/6 mice by implantation of autologous endometrium into the peritoneal cavity. We observed an abundance of CD11c(+) DCs infiltrating sites of angiogenesis in endometriosis lesions. We noticed a similar pattern of infiltrating DCs at sites of angiogenesis in the peritoneal Lewis lung carcinoma tumor model. These DCs were immature (major histocompatability complex class II(low)) and expressed vascular endothelial growth factor receptor 2. Peritoneal implanted bone marrow-derived DCs (BMDCs) incorporated into both endometriosis lesions and into B16 melanoma tumors and enhanced their growth at 8 days compared with controls (5.1+/-2.5 vs. 1.5+/-0.5 mm(2), n=4 and 4, P<0.0001 for endometriosis; 67.6+/-15.1 vs. 22.7+/-14.6 mm(2), n=5 and 7, P=0.0004 for mouse melanoma). Finally, immature BMDCs but not mature BMDCs enhanced microvascular endothelial cell migration in vitro (219+/-51 vs. 93+/-32 cells, P=0.02). Based on these findings, we suggest a novel role for DCs in supporting angiogenesis and promoting lesion growth both in endometriosis and in tumors.


Subject(s)
Dendritic Cells , Endometriosis/pathology , Neovascularization, Pathologic/pathology , Animals , Cell Differentiation , Cell Movement , Cell Transplantation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Disease Models, Animal , Endothelial Cells/pathology , Female , Lung Neoplasms/pathology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
FASEB J ; 22(8): 3010-23, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18502941

ABSTRACT

Angiopoietins were thought to be endothelial cell-specific via the tie2 receptor. We showed that angiopoietin-1 (ang1) also interacts with integrins on cardiac myocytes (CMs) to increase survival. Because ang1 monomers bind and activate integrins (not tie2), we determined their function in vivo. We examined monomer and multimer expressions during physiological and pathological cardiac remodeling and overexpressed ang1 monomers in phenylephrine-induced cardiac hypertrophy. Cardiac ang1 levels (mRNA, protein) increased during postnatal development and decreased with phenylephrine-induced cardiac hypertrophy, whereas tie2 phosphorylations were unchanged. We found that most or all of the changes during cardiac remodeling were in monomers, offering an explanation for unchanged tie2 activity. Heart tissue contains abundant ang1 monomers and few multimers (Western blotting). We generated plasmids that produce ang1 monomers (ang1-256), injected them into mice, and confirmed cardiac expression (immunohistochemistry, RT-PCR). Ang1 monomers localize to CMs, smooth muscle cells, and endothelial cells. In phenylephrine-induced cardiac hypertrophy, ang1-256 reduced left ventricle (LV)/tibia ratios, fetal gene expressions (atrial and brain natriuretic peptides, skeletal actin, beta-myosin heavy chain), and fibrosis (collagen III), and increased LV prosurvival signaling (akt, MAPK(p42/44)), and AMPK(T172). However, tie2 phosphorylations were unchanged. Ang1-256 increased integrin-linked kinase, a key regulator of integrin signaling and cardiac health. Collectively, these results suggest a role for ang1 monomers in cardiac remodeling.


Subject(s)
Angiopoietin-1/chemistry , Angiopoietin-1/metabolism , Cardiomegaly/prevention & control , Integrins/metabolism , Angiopoietin-1/genetics , Animals , Base Sequence , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cell Line , DNA Primers/genetics , Endothelial Cells/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Phenylephrine/toxicity , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Quaternary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, TIE-2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ventricular Remodeling/genetics , Ventricular Remodeling/physiology
7.
Clin Cancer Res ; 14(5): 1487-93, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316573

ABSTRACT

PURPOSE: The half-life of the antiangiogenic molecule endostatin that has been used in clinical trial is short ( approximately 2 h). In addition, approximately 50% of the clinical grade endostatin molecules lack four amino acids at their NH(2) termini. Lack of these amino acids gives rise to a molecule that is devoid of zinc, resulting in no antitumor activity. Our goal was to develop a new version of endostatin that does not show such deficiency. EXPERIMENTAL DESIGN: A recombinant human endostatin conjugated to the Fc domain of IgG was constructed and expressed in mammalian cell culture. The presence of Fc has been shown by previous investigators to play a major role in increasing the half-life of the molecule. Fc-endostatin was tested in tumor-bearing mice, and its half-life was compared with the clinical grade endostatin. RESULTS: The antitumor dose of Fc-endostatin was found to be approximately 100 times less than the clinical grade endostatin. The half-life of Fc-endostatin in the circulation was found to be weeks rather than hours, as observed for endostatin alone. In addition, a U-shaped curve was observed for antitumor activity of endostatin as a function of endostatin concentration delivered to the animals. CONCLUSION: Fc-endostatin is a superior molecule to the original clinical endostatin. Due to its long half-life, the amount of protein required is substantially reduced compared with the clinically tested endostatin. Furthermore, in view of the U-shaped curve of efficacy observed for endostatin, we estimate that the requirement for Fc-endostatin is approximately 700-fold less than endostatin alone. The half-life of endostatin is similar to that of vascular endothelial growth factor-Trap and Avastin, two other antiangiogenic reagents. We conclude that a new clinical trial of endostatin, incorporating Fc, may benefit cancer patients.


Subject(s)
Endostatins/immunology , Endostatins/pharmacokinetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Melanoma, Experimental/metabolism , Pancreatic Neoplasms/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Animals , Apoptosis , Enzyme-Linked Immunosorbent Assay , Half-Life , Humans , Immunoenzyme Techniques , In Situ Nick-End Labeling , Male , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Mutation/genetics , Pancreatic Neoplasms/pathology , Recombinant Proteins/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Lancet ; 370(9604): 2011-9, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18083403

ABSTRACT

BACKGROUND: Sunitinib, a multitargeted tyrosine-kinase inhibitor, which is approved by both US and European Commission regulatory agencies for clinical use, extends survival of patients with metastatic renal-cell carcinoma and gastrointestinal stromal tumours, but concerns have arisen about its cardiac safety. We therefore assessed the cardiovascular risk associated with sunitinib in patients with metastatic gastrointestinal stromal tumours. METHODS: We retrospectively reviewed all cardiovascular events in 75 patients with imatinib-resistant, metastatic, gastrointestinal stromal tumours who had been enrolled in a phase I/II trial investigating the efficacy of sunitinib. The composite cardiovascular endpoint was cardiac death, myocardial infarction, and congestive heart failure. We also examined sunitinib's effects on left ventricular ejection fraction (LVEF) and blood pressure. We investigated potential mechanisms of sunitinib-associated cardiac effects by studies in isolated rat cardiomyocytes and in mice. FINDINGS: Eight of 75 (11%) patients given repeating cycles of sunitinib in the phase I/II trial had a cardiovascular event, with congestive heart failure recorded in six of 75 (8%). Ten of 36 (28%) patients treated at the approved sunitinib dose had absolute LVEF reductions in ejection fraction (EF) of at least 10%, and seven of 36 (19%) had LVEF reductions of 15 EF% or more. Sunitinib induced increases in mean systolic and diastolic blood pressure, and 35 of 75 (47%) individuals developed hypertension (>150/100 mm Hg). Congestive heart failure and left ventricular dysfunction generally responded to sunitinib being withheld and institution of medical management. Sunitinib caused mitochondrial injury and cardiomyocyte apoptosis in mice and in cultured rat cardiomyocytes. INTERPRETATION: Left ventricular dysfunction might be due, in part, to direct cardiomyocyte toxicity, exacerbated by hypertension. Patients treated with sunitinib should be closely monitored for hypertension and LVEF reduction, especially those with a history of coronary artery disease or cardiac risk factors.


Subject(s)
Antineoplastic Agents/adverse effects , Heart Failure/chemically induced , Indoles/adverse effects , Pyrroles/adverse effects , Animals , Antineoplastic Agents/therapeutic use , Blood Pressure/drug effects , Female , Gastrointestinal Stromal Tumors/drug therapy , Humans , Indoles/therapeutic use , Male , Mice , Middle Aged , Multicenter Studies as Topic , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrroles/therapeutic use , Rats , Retrospective Studies , Stroke Volume/drug effects , Sunitinib
9.
J Invest Dermatol ; 130(5): 1307-17, 2010 May.
Article in English | MEDLINE | ID: mdl-20072135

ABSTRACT

UV light-based damage to skin cells can cause photoaging and skin cancer. A major cause of UV light-induced damage to skin is increased free radicals, such as superoxides. Increased superoxides can cause oxidative and nitrative damage to cell components. Thus, agents that counteract these damages may have therapeutic value. Herein, we show that angiopoietin-1 (ang1) prevented and blocked H(2)O(2)-induced increases in superoxides in human spontaneously immortalized keratinocyte line, HaCaT, and primary melanocytes (HeMn). Ang1 prevented H(2)O(2)-induced increases in damage to DNA (8-hydroxy-2'-deoxyguanosine) and proteins (nitrotyrosinylation). Ang1 promoted skin cell metabolism/viability, adhesion, and akt and MAPK(p42/44) activations. Using multi-gene transcriptional profiling, we found that skin cells express integrin subunits {(beta(1), beta(4-6), beta(8), alpha(v), alpha(2), alpha(3), alpha(6) (HaCaT)), (beta(1), beta(3), beta(5), beta(8), alpha(v), alpha(3) (HeMn))} and lack tie2 receptor mRNA. Integrin antibodies (alpha(v), beta(1)) disrupted skin cell adhesion to ang1 and ang1-induced decreases in superoxides. Our findings show that ang1 blocks free radical damage to skin cells and may be clinically useful to prevent and/or reduce photoaging and skin cancer.


Subject(s)
Angiopoietin-1/pharmacology , Keratinocytes/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Skin Aging/drug effects , Angiopoietin-1/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/physiology , DNA Damage/drug effects , Gene Expression Profiling , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Integrins/genetics , Keratinocytes/cytology , Keratinocytes/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxidants/metabolism , Oxidants/pharmacology , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism , Skin/cytology , Skin/metabolism , Skin Aging/physiology , Skin Neoplasms/metabolism , Skin Neoplasms/prevention & control , Tyrosine/analogs & derivatives , Tyrosine/metabolism
10.
J Biol Chem ; 283(40): 27230-8, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18660502

ABSTRACT

Class three semaphorins (SEMAs) were originally shown to be mediators of axon guidance that repelled axons and collapsed growth cones, but it is now evident that SEMA3F, for example, has similar effects on tumor cells and endothelial cells (EC). In both human U87MG glioma cells and human umbilical vein EC, SEMA3F induced rapid cytoskeletal collapse, suppressed cell contractility, decreased phosphorylation of cofilin, and inhibited cell migration in culture. Analysis of the signaling pathways showed that SEMA3F formed a complex with NRP2 (neuropilin-2) and plexin A1. These interactions eventually led to inactivation of the small GTPase, RhoA, which is necessary for stress fiber formation and cytoskeleton integrity. A novel upstream RhoA mediator was shown to be ABL2, also known as ARG, a membrane-anchored nonreceptor tyrosine kinase. Within minutes after the addition of SEMA3F, ABL2 directly bound plexin A1 but not to a plexin A1 mutant lacking the cytoplasmic domain. In addition, ABL2 phosphorylated and thereby activated p190RhoGAP, which inactivated RhoA (GTP to GDP), resulting in cytoskeleton collapse and inhibition of cell migration. On the other hand, cells overexpressing an ABL2 inactive kinase mutant or treated with ABL2 small interfering RNA did not inactivate RhoA. Cells treated with p190RhoGAP small interfering RNA also did not inactivate RhoA. Together, these results suggested that ABL2/ARG is a novel mediator of SEMA3F-induced RhoA inactivation and collapsing activity.


Subject(s)
Cytoskeleton/metabolism , Glioma/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line, Tumor , Cytoskeleton/genetics , Endothelial Cells/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Glioma/genetics , Humans , Membrane Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Neuropilin-2/genetics , Neuropilin-2/metabolism , Phosphorylation , Proto-Oncogene Proteins c-abl/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction/genetics , Swine , rhoA GTP-Binding Protein/genetics
11.
Invest Ophthalmol Vis Sci ; 49(8): 3666-70, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18408184

ABSTRACT

PURPOSE: Dendritic cells (DCs) are innate immune cells that have recently been shown to support angiogenesis in tumors, endometriosis, and lymph nodes. A major cause of legal blindness is wet age-related macular degeneration (wet ARMD), wherein abnormal blood vessels grow under the retina, an abnormality also referred to as choroidal neovascularization (CNV). The purpose of the present study was to investigate the role of DCs in the development of CNV. METHODS: Laser photocoagulation was used to induce CNV in C57BL/6J mice. The authors analyzed CNV lesions for the presence of DCs using flow cytometry and immunostaining at designated times. They also analyzed the effects of intravenous DC transplantation on CNV development by measuring the lesion area using confocal microscopy 1 week after laser injury. RESULTS: The authors analyzed CNV lesions for the presence of DCs by flow cytometry and observed that CD11c(+) major histocompatibility complex (MHC) class II(+) DCs transiently infiltrated the CNV lesions, reaching a peak at 2 to 4 days after laser injury. These DCs were mostly immature (CD11c(+) MHCII(low)) and expressed vascular endothelial growth factor receptor 2. Immunostaining of laser-induced CNV lesions confirmed that DCs are located at the sites of newly formed blood vessels. Intravenously injected DCs incorporated into the CNV lesions. However, only immature DCs enhanced CNV size. CONCLUSIONS: These results suggest a role for DCs in promoting angiogenesis and lesion growth in laser-induced CNV. The present data suggest that DCs may represent potential cellular targets for therapeutic intervention in wet ARMD.


Subject(s)
Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Dendritic Cells/physiology , Animals , CD11c Antigen/metabolism , Cell Movement , Cell Transplantation , Cells, Cultured , Choroid/blood supply , Dendritic Cells/transplantation , Disease Models, Animal , Endothelium, Vascular/metabolism , Flow Cytometry , Histocompatibility Antigens Class II/metabolism , Laser Coagulation , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Nat Biotechnol ; 26(7): 799-807, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18587385

ABSTRACT

Targeting angiogenesis, the formation of blood vessels, is an important modality for cancer therapy. TNP-470, a fumagillin analog, is among the most potent and broad-spectrum angiogenesis inhibitors. However, a major clinical limitation is its poor oral availability and short half-life, necessitating frequent, continuous parenteral administration. We have addressed these issues and report an oral formulation of TNP-470, named Lodamin. TNP-470 was conjugated to monomethoxy-polyethylene glycol-polylactic acid to form nanopolymeric micelles. This conjugate can be absorbed by the intestine and selectively accumulates in tumors. Lodamin significantly inhibits tumor growth, without causing neurological impairment in tumor-bearing mice. Using the oral route of administration, it first reaches the liver, making it especially efficient in preventing the development of liver metastasis in mice. We show that Lodamin is an oral nontoxic antiangiogenic drug that can be chronically administered for cancer therapy or metastasis prevention.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Cyclohexanes/administration & dosage , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Polyesters/administration & dosage , Sesquiterpenes/administration & dosage , Administration, Oral , Animals , Cell Line, Tumor , Humans , Mice , O-(Chloroacetylcarbamoyl)fumagillol , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL