Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 109(28): 11306-11, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733742

ABSTRACT

Antiangiogenesis therapy has become a vital part of the armamentarium against cancer. Hypertension is a dose-limiting toxicity for VEGF inhibitors. Thus, there is a pressing need to address the associated adverse events so these agents can be better used. The hypertension may be mediated by reduced NO bioavailability resulting from VEGF inhibition. We proposed that the hypertension may be prevented by coadministration with endostatin (ES), an endogenous angiogenesis inhibitor with antitumor effects shown to increase endothelial NO production in vitro. We determined that Fc-conjugated ES promoted NO production in endothelial and smooth muscle cells. ES also lowered blood pressure in normotensive mice and prevented hypertension induced by anti-VEGF antibodies. This effect was associated with higher circulating nitrate levels and was absent in eNOS-knockout mice, implicating a NO-mediated mechanism. Retrospective study of patients treated with ES in a clinical trial revealed a small but significant reduction in blood pressure, suggesting that the findings may translate to the clinic. Coadministration of ES with VEGF inhibitors may offer a unique strategy to prevent drug-related hypertension and enhance antiangiogenic tumor suppression.


Subject(s)
Blood Pressure/physiology , Endostatins/metabolism , Hypertension/metabolism , Hypertension/prevention & control , Nitric Oxide/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies/chemistry , Clinical Trials, Phase II as Topic , Female , Heart/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/prevention & control
2.
Hypertension ; 58(1): 85-92, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21482957

ABSTRACT

Vascular endothelial growth factor regulates neoplastic angiogenesis through production of endothelium-derived NO. We performed a prospective evaluation of vascular function during treatment with vandetanib, a vascular endothelial growth receptor 2 and 3 receptor tyrosine kinase inhibitor, to determine the effects of vascular endothelial growth receptor signal interruption on endothelial function in humans. Seventeen patients with stage IV breast cancer received dose-escalated vandetanib in combination with low-dose oral chemotherapy. We measured blood pressure, systemic nitrate/nitrite levels, and brachial artery vascular function. In vitro analyses of cultured endothelial cells were performed to determine the effect of vandetanib on NO production, akt(473) phosphorylation, and endothelial NO synthase protein content and membrane localization. Vandetanib treatment for 6 weeks significantly increased blood pressure, decreased resting brachial artery diameter, and decreased plasma systemic nitrate/nitrite levels compared with baseline. Flow-mediated vasodilation was preserved, and no change was noted in nitroglycerin-mediated vasodilation. In vitro, endothelial cell nitrite levels and akt(473) phosphorylation were reduced and vascular endothelial growth receptor 2 levels did not change, but endothelial NO synthase membrane concentration doubled. Vandetanib reduces constitutive NO production and increases blood pressure, yet flow-stimulated NO bioavailability was preserved. Changes in vascular function with tyrosine kinase inhibition are complex and require further study in humans.


Subject(s)
Breast Neoplasms/drug therapy , Endothelium, Vascular/metabolism , Neovascularization, Pathologic/prevention & control , Nitric Oxide/biosynthesis , Piperidines/administration & dosage , Quinazolines/administration & dosage , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Vasodilation/drug effects , Administration, Oral , Adult , Aged , Blood Pressure/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Middle Aged , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Prospective Studies , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
3.
Cancer Res ; 70(19): 7630-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20709759

ABSTRACT

Dysregulated fibroblast growth factor (FGF) signaling has been implicated in the pathogenesis of human cancers. Aberrant activation of FGF receptor 2 (FGFR2) signaling, through overexpression of FGFR2 and/or its ligands, mutations, and receptor amplification, has been found in a variety of human tumors. We generated monoclonal antibodies against the extracellular ligand-binding domain of FGFR2 to address the role of FGFR2 in tumorigenesis and to explore the potential of FGFR2 as a novel therapeutic target. We surveyed a broad panel of human cancer cell lines for the dysregulation of FGFR2 signaling and discovered that breast and gastric cancer cell lines harboring FGFR2 amplification predominantly express the IIIb isoform of the receptor. Therefore, we used an FGFR2-IIIb-specific antibody, GP369, to investigate the importance of FGFR2 signaling in vitro and in vivo. GP369 specifically and potently suppressed ligand-induced phosphorylation of FGFR2-IIIb and downstream signaling, as well as FGFR2-driven proliferation in vitro. The administration of GP369 in mice significantly inhibited the growth of human cancer xenografts harboring activated FGFR2 signaling. Our findings support the hypothesis that dysregulated FGFR2 signaling is one of the critical oncogenic pathways involved in the initiation and/or maintenance of tumors. Cancer patients with aberrantly activated/amplified FGFR2 signaling could potentially benefit from therapeutic intervention with FGFR2-targeting antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Neoplasms/therapy , Receptor, Fibroblast Growth Factor, Type 2/immunology , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Gene Amplification , Humans , Mice , Mice, SCID , Molecular Sequence Data , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/immunology , Phosphorylation/drug effects , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction , Xenograft Model Antitumor Assays
4.
J Biomed Mater Res A ; 95(1): 105-17, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20540095

ABSTRACT

Myocardial infarction (MI) results in the death of cardiomyocytes (CM), which causes scar formation and pathological remodeling of the heart. The delivery of healthy myocytes or bone marrow cells reduces pathological remodeling after MI, however, current cell injection methods have low cell survival rates and high cell loss. The main objective of this work was to develop a novel hydrogel that can promote survival of CMs. Photocrosslinkable azidobenzoic acid modified chitosan (Az-chitosan) was conjugated with the angiopoietin-1-derived peptide, QHREDGS. This novel peptide is thought to mediate attachment and survival responses of CM to angiopoietin-1 via integrin binding. Thin layers of Az-chitosan, Az-chitosan-QHREDGS, and Az-chitosan-DGQESHR (scrambled peptide control) were spin coated on glass slides and photocrosslinked with application of UV light (365 nm). Neonatal rat heart cells cultured up to 5 days, demonstrated significantly higher attachment and viability on Az-chitosan-QHREDGS compared to cells on other hydrogel controls. Surfaces were also stained for the CM-specific marker troponin I, demonstrating significantly higher percentage of CMs on Az-chitosan-QHREDGS compared to Az-chitosan. The cells cultivated on Az-chitosan-QHREDGS demonstrated significantly lower levels of caspase 3/7 activation after taxol treatment in comparison to cells cultivated on the control hydrogels, glass substrate, or Az-chitosan linked to RGD, an established integrin binding peptide that did not protect against apoptosis. Thus, Az-chitosan-QHREDGS supports attachment and survival of neonatal rat heart cells.


Subject(s)
Angiopoietin-1/pharmacology , Chitosan/pharmacology , Cross-Linking Reagents/pharmacology , Myocardium/cytology , Myocytes, Cardiac/cytology , Peptides/pharmacology , Ultraviolet Rays , Amino Acid Sequence , Angiopoietin-1/chemistry , Animals , Animals, Newborn , Apoptosis/drug effects , Azides/pharmacology , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Microscopy, Electron, Scanning , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Peptides/chemistry , Rats , Rats, Sprague-Dawley , Surface Properties/drug effects , Troponin I/metabolism
5.
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
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.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...