Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Sci Adv ; 9(46): eadi1394, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37976351

ABSTRACT

Vascular dysfunction resulting from endothelial hyperpermeability is a common and important feature of critical illness due to sepsis, trauma, and other conditions associated with acute systemic inflammation. Clarkson disease [monoclonal gammopathy-associated idiopathic systemic capillary leak syndrome (ISCLS)] is a rare, orphan disorder marked by spontaneous and recurrent episodes of hypotensive shock and peripheral edema due to widespread vascular leakage in peripheral tissues. Mortality from acute flares approaches 30% due to lack of effective therapies. We evaluated a monoclonal antibody (4E2) specific for the endothelial receptor tyrosine kinase Tie2 in ISCLS models. 4E2 activated Tie2 in ISCLS patient-derived endothelial cells and reduced baseline and proinflammatory mediator-induced barrier dysfunction. 4E2 also reduced mortality and/or vascular leakage associated with systemic histamine challenge or influenza infection in the SJL/J mouse model of ISCLS. These findings support a critical role for Tie2 dysregulation in ISCLS and highlight a viable therapeutic approach to this catastrophic disorder.


Subject(s)
Capillary Leak Syndrome , Sepsis , Mice , Animals , Humans , Capillary Leak Syndrome/complications , Endothelial Cells , Ligands , Antibodies , Receptor, TIE-2
2.
Drug Des Devel Ther ; 12: 495-504, 2018.
Article in English | MEDLINE | ID: mdl-29563774

ABSTRACT

BACKGROUND: VEGF is a highly selective mitogen that serves as the central regulator of tumor angiogenesis by mediating endothelial proliferation, permeability, and survival. Tanibirumab (TTAC-0001) is a fully human IgG1 monoclonal antibody derived from a fully human naïve single-chain variable fragment (ScFv) phage library that was developed to inhibit the effects of VEGF in the treatment of solid tumors, especially those of the brain. METHODS: In the present study, we conducted intravenous pharmacokinetic studies of TTAC-0001 in mice, rats, and cynomolgus monkeys. At the doses studied (3 mg/kg, 10 mg/kg, 30 mg/kg), TTAC-0001 exhibited dose proportionality in mice and monkeys. At a dose of ~10 mg/kg, the clearance of TTAC-0001 from serum was 0.017 mL/h in mice, 0.35 mL/h in rats, and 2.19 mL/h in cynomolgus monkeys, and the terminal half-life ranged from 20-30 h among the three species. Pharmacokinetic data in mice, rats, and cynomolgus monkeys were used to predict the pharmacokinetics of TTAC-0001 in humans using allometric scaling. The predicted serum clearance of TTAC-0001 in humans was 102.45 mL/h and the terminal half-life was 27.52 h. RESULTS: The maximum life span-corrected clearance value was 72.92 mL/h. The observed clearance in humans was more similar to the predicted scaled clearance. CONCLUSION: We investigated the pharmacokinetics of TTAC-0001 in mice, rats, and cynomolgus monkeys after intravenous administration. At the doses studied, TTAC-0001 exhibited dose proportionality in mice and monkeys. The scaled pharmacokinetics of TTAC-0001 reported here was useful for designing first-in-human studies. Allometric scaling in the therapeutic antibody is feasible.


Subject(s)
Antibodies, Monoclonal/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Female , Humans , K562 Cells , Macaca fascicularis , Mice , Mice, Inbred BALB C , Molecular Structure , Rats , Structure-Activity Relationship , Tissue Distribution
3.
MAbs ; 7(6): 1195-204, 2015.
Article in English | MEDLINE | ID: mdl-26325365

ABSTRACT

Vascular endothelial growth factor (VEGF) and its receptors are considered the primary cause of tumor-induced angiogenesis. Specifically, VEGFR-2/kinase insert domain receptor (KDR) is part of the major signaling pathway that plays a significant role in tumor angiogenesis, which is associated with the development of various types of tumor and metastasis. In particular, KDR is involved in tumor angiogenesis as well as cancer cell growth and survival. In this study, we evaluated the therapeutic potential of TTAC-0001, a fully human antibody against VEGFR-2/KDR. To assess the efficacy of the antibody and pharmacokinetic (PK) relationship in vivo, we tested the potency of TTAC-0001 in glioblastoma and colorectal cancer xenograft models. Antitumor activity of TTAC-0001 in preclinical models correlated with tumor growth arrest, induction of tumor cell apoptosis, and inhibition of angiogenesis. We also evaluated the combination effect of TTAC-0001 with a chemotherapeutic agent in xenograft models. We were able to determine the relationship between PK and the efficacy of TTAC-0001 through in vivo single-dose PK study. Taken together, our data suggest that targeting VEGFR-2 with TTAC-0001 could be a promising approach for cancer treatment.


Subject(s)
Antibodies, Monoclonal/immunology , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , Area Under Curve , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/immunology , Glioblastoma/metabolism , HCT116 Cells , HT29 Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/immunology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
4.
MAbs ; 7(5): 957-68, 2015.
Article in English | MEDLINE | ID: mdl-25942475

ABSTRACT

Angiogenesis is one of the most important processes for cancer cell survival, tumor growth and metastasis. Vascular endothelial growth factor (VEGF) and its receptor, particularly VEGF receptor-2 (VEGFR-2, or kinase insert domain-containing receptor, KDR), play critical roles in tumor-associated angiogenesis. We developed TTAC-0001, a human monoclonal antibody against VEGFR-2/KDR from a fully human naïve single-chain variable fragment phage library. TTAC-0001 was selected as a lead candidate based on its affinity, ligand binding inhibition and inhibition of VEGFR-2 signal in human umbilical vein endothelial cells (HUVEC). TTAC-0001 inhibited binding of VEGF-C and VEGF-D to VEGFR-2 in addition to VEGF-A. It binds on the N-terminal regions of domain 2 and domain 3 of VEGFR-2. It could inhibit the phosphorylation of VEGFR-2/KDR and ERK induced by VEGF in HUVEC. TTAC-0001 also inhibited VEGF-mediated endothelial cell proliferation, migration and tube formation in vitro, as well as ex vivo vessel sprouting from rat aortic rings and neovascularization in mouse matrigel model in vivo. Our data indicates that TTAC-0001 blocks the binding of VEGFs to VEGFR-2/KDR and inhibits VEGFR-induced signaling pathways and angiogenesis. Therefore, these data strongly support the further development of TTAC-0001 as an anti-cancer agent in the clinic.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Pathologic , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Humans , Immunohistochemistry , Mice , Rats , Surface Plasmon Resonance
5.
J Ocul Pharmacol Ther ; 30(10): 847-53, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25188901

ABSTRACT

PURPOSE: The study investigated the effect of intravitreally administered tanibirumab, a fully human monoclonal antibody against vascular endothelial growth factor receptor 2, in a rat model of laser-induced choroidal neovascularization (CNV). METHODS: CNV was induced by laser photocoagulation on day 0 in the eyes of Brown Norway rats. Intravitreal injection of tanibirumab or phosphate-buffered saline (PBS) was done on day 0 (prevention arm) or day 7 (treatment arm). Seven days after injection, the eyes were enucleated and retinal pigment epithelium-choroid-sclera flat mounts were prepared. Areas of CNV were determined in the flat mounts using tetramethylrhodamine isothiocyanate Bandeiraea simplicifolia (BS) isolectin labeling and intravenously administered fluorescein isothiocyanate-dextran and quantified using an image analysis program. RESULTS: In the prevention arm, the mean area of CNV measured by BS isolectin labeling was reduced by 28.2% and 53.9% in tanibirumab-treated eyes (20 and 60 µg, respectively) compared with PBS-treated control eyes on day 7 (P=0.038 and P<0.001, respectively). In the treatment arm, the mean area of CNV measured by BS isolectin labeling was reduced by 28.7% and 46.0% in tanibirumab-treated eyes (20 and 60 µg, respectively) compared with PBS-treated control eyes on day 14 (P=0.048 and P<0.001, respectively). CONCLUSIONS: Intravitreally administered tanibirumab partially suppressed the formation of new CNV and partially regressed preformed laser-induced CNV in the rat model. Tanibirumab may be a feasible treatment for CNV associated with age-related macular degeneration or other causes.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Choroidal Neovascularization/drug therapy , Animals , Antibodies, Monoclonal, Humanized , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Disease Models, Animal , Humans , Intravitreal Injections , Laser Coagulation , Plant Lectins/chemistry , Random Allocation , Rats , Staining and Labeling/methods , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
6.
Biochem Biophys Res Commun ; 303(1): 37-45, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12646163

ABSTRACT

Members of both calpain and caspase protease families can degrade several components of focal adhesions, leading to disassembly of these complexes. In this report, we investigated the disappearance of tensin from cell adhesion sites of chicken embryonic fibroblast cells (CEFs) exposed to etoposide and demonstrated that loss of tensin from cell adhesions during etoposide-induced apoptosis may be due to degradation of tensin by caspase-3. Tensin cleavage by caspase-3 at the sequence DYPD(1226)G separates the amino-terminal region containing the actin binding domain and the carboxyl-terminal region containing the SH2 domain. The resultant carboxyl-terminal fragment of tensin is unable to bind phosphoinositide 3-kinase (PI3-kinase) transducing cell survival signaling. We also demonstrated that overexpression of the amino-terminal tensin fragment induced disruption of actin cytoskeleton in chicken embryonic fibroblasts. Therefore, caspase-mediated cleavage of tensin contributes to the disruption of actin organization and interrupts ECM-mediated survival signals through phosphatidylinositol 3-kinase.


Subject(s)
Actins/metabolism , Apoptosis , Caspases/metabolism , Cytoskeleton/metabolism , Microfilament Proteins/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Calpain/metabolism , Caspase 3 , Cell Adhesion , Cell Culture Techniques , Cell Survival , Chick Embryo , DNA/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Etoposide/pharmacology , Fibroblasts/metabolism , Green Fluorescent Proteins , Immunoblotting , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Tensins , Time Factors , Transfection , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL