Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 53(4): 2170-80, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22410553

ABSTRACT

PURPOSE: To determine whether systemic treatment with AMG 386, a selective angiopoietin 1/2-neutralizing peptibody, inhibits neovascular processes in animal models of ocular disease. METHODS: AMG 386 was tested in a laser-induced choroidal neovascularization (CNV) model in monkeys using fluorescein angiography. The biodistribution of (125)I-AMG 386 was determined in cynomolgus monkeys by whole-body autoradiography and radioanalysis of ocular tissues. A murine retinopathy of prematurity (ROP) model was used to examine the effect of AMG 386 on established and newly formed retinal vessels, either as a single agent or when combined with VEGF inhibition.AMG 386 pharmacokinetics were evaluated in each model. RESULTS: In the CNV model, AMG 386 significantly decreased fluorescent angiographic leakage and reduced fibroplasia, indicating an impaired healing response consistent with angiogenesis blockade. Radiolabeled AMG 386 was widely distributed across ocular tissues, with highest concentrations in the choroid, cornea, retinal pigmented epithelium, iris/ciliary body, and sclera. In the ROP model, AMG 386 prevented pathologic retinal angiogenesis when administered from P8 to P16 but transiently impeded regression of these abnormal vessels when administered from P17 to P23. Combining AMG 386 with VEGF inhibition led to cooperative prevention of retinal angiogenesis in this model. No AMG 386-related ocular toxicities occurred, and no treatment-related clinical observations were made in any of the studies. CONCLUSIONS: In this study, AMG 386 inhibited angiogenesis in animal models of CNV and ROP, supporting investigation of AMG 386 for the treatment of ocular neovascular diseases in the clinical setting.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Choroidal Neovascularization/prevention & control , Disease Models, Animal , Recombinant Fusion Proteins/pharmacology , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/prevention & control , Angiogenesis Inhibitors/pharmacokinetics , Angiopoietin-1/antagonists & inhibitors , Angiopoietin-2/antagonists & inhibitors , Animals , Animals, Newborn , Autoradiography , Capillary Permeability/drug effects , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Eye/metabolism , Female , Fluorescein Angiography , Humans , In Situ Hybridization , Infant, Newborn , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/pharmacokinetics , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Vessels/drug effects , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Tissue Distribution
2.
Mol Cancer Ther ; 9(10): 2641-51, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20937592

ABSTRACT

AMG 386 is an investigational first-in-class peptide-Fc fusion protein (peptibody) that inhibits angiogenesis by preventing the interaction of angiopoietin-1 (Ang1) and Ang2 with their receptor, Tie2. Although the therapeutic value of blocking Ang2 has been shown in several models of tumorigenesis and angiogenesis, the potential benefit of Ang1 antagonism is less clear. To investigate the consequences of Ang1 neutralization, we have developed potent and selective peptibodies that inhibit the interaction between Ang1 and its receptor, Tie2. Although selective Ang1 antagonism has no independent effect in models of angiogenesis-associated diseases (cancer and diabetic retinopathy), it induces ovarian atrophy in normal juvenile rats and inhibits ovarian follicular angiogenesis in a hormone-induced ovulation model. Surprisingly, the activity of Ang1 inhibitors seems to be unmasked in some disease models when combined with Ang2 inhibitors, even in the context of concurrent vascular endothelial growth factor inhibition. Dual inhibition of Ang1 and Ang2 using AMG 386 or a combination of Ang1- and Ang2-selective peptibodies cooperatively suppresses tumor xenograft growth and ovarian follicular angiogenesis; however, Ang1 inhibition fails to augment the suppressive effect of Ang2 inhibition on tumor endothelial cell proliferation, corneal angiogenesis, and oxygen-induced retinal angiogenesis. In no case was Ang1 inhibition shown to (a) confer superior activity to Ang2 inhibition or dual Ang1/2 inhibition or (b) antagonize the efficacy of Ang2 inhibition. These results imply that Ang1 plays a context-dependent role in promoting postnatal angiogenesis and that dual Ang1/2 inhibition is superior to selective Ang2 inhibition for suppression of angiogenesis in some postnatal settings.


Subject(s)
Angiopoietin-1/antagonists & inhibitors , Cell Division/drug effects , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/prevention & control , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Cornea/blood supply , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms, Experimental/blood supply , Ovarian Follicle/blood supply , Rats , Rats, Sprague-Dawley
3.
Cancer Res ; 70(6): 2213-23, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20197469

ABSTRACT

Inhibition of angiopoietin-2 (Ang2) can slow tumor growth, but the underlying mechanism is not fully understood. Because Ang2 is expressed in growing blood vessels and promotes angiogenesis driven by vascular endothelial growth factor (VEGF), we asked whether the antitumor effect of Ang2 inhibition results from reduced sprouting angiogenesis and whether the effect is augmented by inhibition of VEGF from tumor cells. Using Colo205 human colon carcinomas in nude mice as a model, we found that selective inhibition of Ang2 by the peptide-Fc fusion protein L1-7(N) reduced the number of vascular sprouts by 46% and tumor growth by 62% over 26 days. Strikingly, when the Ang2 inhibitor was combined with a function-blocking anti-VEGF antibody, the number of sprouts was reduced by 82%, tumor vascularity was reduced by 67%, and tumor growth slowed by 91% compared with controls. The reduction in tumor growth was accompanied by decreased cell proliferation and increased apoptosis. We conclude that inhibition of Ang2 slows tumor growth by limiting the expansion of the tumor vasculature by sprouting angiogenesis, in a manner that is complemented by concurrent inhibition of VEGF and leads to reduced proliferation and increased apoptosis of tumor cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiopoietin-2/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/blood supply , Colonic Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiopoietin-2/biosynthesis , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Growth Processes/drug effects , Colonic Neoplasms/pathology , Drug Synergism , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Rats , Receptors, Fc/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/immunology , Xenograft Model Antitumor Assays
4.
Am J Pathol ; 175(5): 2159-70, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19815705

ABSTRACT

Angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) have complex actions in angiogenesis and vascular remodeling due to their effects on Tie2 receptor signaling. Ang2 blocks Ang1-mediated activation of Tie2 in endothelial cells under certain conditions but is a Tie2 receptor agonist in others. We examined the effects of selective inhibitors of Ang1 (mL4-3) or Ang2 (L1-7[N]), alone or in combination, on the vasculature of human Colo205 tumors in mice. The Ang2 inhibitor decreased the overall abundance of tumor blood vessels by reducing tumor growth and keeping vascular density constant. After inhibition of Ang2, tumor vessels had many features of normal blood vessels (normalization), as evidenced by junctional accumulation of vascular endothelial-cadherin, junctional adhesion molecule-A, and platelet/endothelial cell adhesion molecule-1 in endothelial cells, increased pericyte coverage, reduced endothelial sprouting, and remodeling into smaller, more uniform vessels. The Ang1 inhibitor by itself had little noticeable effect on the tumor vasculature. However, when administered with the Ang2 inhibitor, the Ang1 inhibitor prevented tumor vessel normalization, but not the reduction in tumor vascularity produced by the Ang2 inhibitor. These findings are consistent with a model whereby inhibition of Ang2 leads to normalization of tumor blood vessels by permitting the unopposed action of Ang1, but decreases tumor vascularity primarily by blocking Ang2 actions.


Subject(s)
Angiopoietin-1/antagonists & inhibitors , Angiopoietin-2/antagonists & inhibitors , Blood Vessels/anatomy & histology , Blood Vessels/metabolism , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Blood Vessels/pathology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/metabolism , Pericytes/cytology , Pericytes/metabolism , Phenotype , Signal Transduction/physiology
6.
Bioorg Med Chem Lett ; 18(17): 4844-8, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18682324

ABSTRACT

A novel class of potent and selective inhibitors of KDR incorporating an indazole moiety 1 is reported. The discovery, synthesis, and structure-activity relationships of this series of inhibitors have been investigated. The most promising compounds were also profiled to determine their pharmacokinetic properties and evaluated in a VEGF-induced vascular permeability assay.


Subject(s)
Indazoles/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Capillary Permeability/drug effects , Cells, Cultured , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , Indazoles/chemical synthesis , Indazoles/chemistry , Indazoles/metabolism , Male , Mice , Mice, Nude , Protein Binding/physiology , Protein Kinase Inhibitors/classification , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
J Med Chem ; 51(6): 1668-80, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18324759

ABSTRACT

We have previously shown N-arylnaphthamides can be potent inhibitors of vascular endothelial growth factor receptors (VEGFRs). N-Alkyl and N-unsubstituted naphthamides were prepared and found to yield nanomolar inhibitors of VEGFR-2 (KDR) with an improved selectivity profile against a panel of tyrosine and serine/threonine kinases. The inhibitory activity of this series was retained at the cellular level. Naphthamides 3, 20, and 22 exhibited good pharmacokinetics following oral dosing and showed potent inhibition of VEGF-induced angiogenesis in the rat corneal model. Once-daily oral administration of 22 for 14 days led to 85% inhibition of established HT29 colon cancer and Calu-6 lung cancer xenografts at doses of 10 and 20 mg/kg, respectively.


Subject(s)
Antineoplastic Agents/pharmacology , Endothelial Cells/drug effects , Naphthalenes/pharmacology , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Corneal Neovascularization/blood , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Design , Drug Evaluation, Preclinical , Female , Humans , Inhibitory Concentration 50 , Injections, Intravenous , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microsomes, Liver/drug effects , Models, Molecular , Molecular Structure , Naphthalenes/chemical synthesis , Naphthalenes/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Stereoisomerism , Structure-Activity Relationship
8.
J Med Chem ; 51(6): 1649-67, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18324761

ABSTRACT

A series of naphthyl-based compounds were synthesized as potential inhibitors of vascular endothelial growth factor (VEGF) receptors. Investigations of structure-activity relationships led to the identification of a series of naphthamides that are potent inhibitors of the VEGF receptor tyrosine kinase family. Numerous analogues demonstrated low nanomolar inhibition of VEGF-dependent human umbilical vein endothelial cell (HUVEC) proliferation, and of these several compounds possessed favorable pharmacokinetic (PK) profiles. In particular, compound 48 demonstrated significant antitumor efficacy against established HT29 human colon adenocarcinoma xenografts implanted in athymic mice. A full account of the preparation, structure-activity relationships, pharmacokinetic properties, and pharmacology of analogues within this series is presented.


Subject(s)
Antineoplastic Agents/pharmacology , Endothelial Cells/drug effects , Naphthalenes/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Corneal Neovascularization/blood , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Design , Drug Evaluation, Preclinical , Female , Humans , Inhibitory Concentration 50 , Injections, Intravenous , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microsomes, Liver/drug effects , Models, Molecular , Molecular Structure , Naphthalenes/chemical synthesis , Naphthalenes/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Stereoisomerism , Structure-Activity Relationship
9.
J Med Chem ; 51(6): 1695-705, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18311900

ABSTRACT

Angiogenesis is vital for solid tumor growth, and its prevention is a proven strategy for the treatment of disease states such as cancer. The vascular endothelial growth factor (VEGF) pathway provides several opportunities by which small molecules can act as inhibitors of endothelial proliferation and migration. Critical to these processes is signaling through VEGFR-2 or the kinase insert domain receptor (KDR) upon stimulation by its ligand VEGF. Herein, we report the discovery of 2,3-dihydro-1,4-benzoxazines as inhibitors of intrinsic KDR activity (IC 50 < 0.1 microM) and human umbilical vein endothelial cell (HUVEC) proliferation with IC 50 < 0.1 microM. More specifically, compound 16 was identified as a potent (KDR: < 1 nM and HUVEC: 4 nM) and selective inhibitor that exhibited efficacy in angiogenic in vivo models. In addition, this series of molecules is typically well-absorbed orally, further demonstrating the 2,3-dihydro-1,4-benzoxazine moiety as a promising platform for generating kinase-based antiangiogenic therapeutic agents.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Benzoxazines/administration & dosage , Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Animals , Benzoxazines/chemical synthesis , Benzoxazines/chemistry , Biological Availability , Cell Line , Cell Proliferation/drug effects , Corneal Neovascularization/blood , Crystallography, X-Ray , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Female , Humans , Injections, Subcutaneous , Ligands , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Animal , Models, Molecular , Molecular Structure , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Bioorg Med Chem Lett ; 17(21): 6003-8, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17869515

ABSTRACT

Inhibition of tumor-induced angiogenesis is a promising strategy in anticancer research. Neovascularization is a process required for both tumor growth and metastasis. Enhanced understanding of the underlying molecular mechanisms has led to the discovery of a variety of pharmaceutically attractive targets. Decades of investigation suggest that vascular endothelial growth factor (VEGF) and its receptors, in particular VEGFR2 or kinase insert-domain-containing receptor (Kdr), play a critical role in the growth and survival of endothelial cells in newly forming vasculature. The clinical utility of inhibitors of this receptor tyrosine kinase is currently under intense investigation. Herein we report our efforts in this arena.


Subject(s)
Niacinamide/pharmacology , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Cell Line, Tumor , Humans
11.
J Med Chem ; 50(18): 4351-73, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17696416

ABSTRACT

Inhibition of the VEGF signaling pathway has become a valuable approach in the treatment of cancers. Guided by X-ray crystallography and molecular modeling, a series of 2-aminobenzimidazoles and 2-aminobenzoxazoles were identified as potent inhibitors of VEGFR-2 (KDR) in both enzymatic and HUVEC cellular proliferation assays. In this report we describe the synthesis and structure-activity relationship of a series of 2-aminobenzimidazoles and benzoxazoles, culminating in the identification of benzoxazole 22 as a potent and selective VEGFR-2 inhibitor displaying a good pharmacokinetic profile. Compound 22 demonstrated efficacy in both the murine matrigel model for vascular permeability (79% inhibition observed at 100 mg/kg) and the rat corneal angiogenesis model (ED(50) = 16.3 mg/kg).


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Benzimidazoles/chemical synthesis , Benzoxazoles/chemical synthesis , Pyridines/chemical synthesis , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Benzoxazoles/pharmacokinetics , Benzoxazoles/pharmacology , Biological Availability , Capillary Permeability/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cornea/blood supply , Cornea/drug effects , Crystallography, X-Ray , Drug Design , Endothelial Cells/cytology , Endothelial Cells/drug effects , Female , Humans , Male , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Umbilical Veins/cytology , Vascular Endothelial Growth Factor Receptor-2/chemistry
12.
J Med Chem ; 50(4): 611-26, 2007 Feb 22.
Article in English | MEDLINE | ID: mdl-17253678

ABSTRACT

Inhibition of angiogenesis is a promising and clinically validated approach for limiting tumor growth and survival. The receptor tyrosine kinase Tie-2 is expressed almost exclusively in the vascular endothelium and is required for developmental angiogenesis and vessel maturation. However, the significance of Tie-2 signaling in tumor angiogenesis is not well understood. In order to evaluate the therapeutic utility of inhibiting Tie-2 signaling, we developed a series of potent and orally bioavailable small molecule Tie-2 kinase inhibitors with selectivity over other kinases, especially those that are believed to be important for tumor angiogenesis. Our earlier work provided pyridinyl pyrimidine 6 as a potent, nonselective Tie-2 inhibitor that was designed on the basis of X-ray cocrystal structures of KDR inhibitors 34 (triazine) and 35 (nicotinamide). Lead optimization resulted in pyridinyl triazine 63, which exhibited >30-fold selectivity over a panel of kinases, good oral exposure, and in vivo inhibition of Tie-2 phosphorylation.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Benzamides/chemical synthesis , Pyridines/chemical synthesis , Receptor, TIE-2/antagonists & inhibitors , Triazines/chemical synthesis , Administration, Oral , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Benzamides/pharmacokinetics , Benzamides/pharmacology , Binding Sites , Blood Proteins/metabolism , Crystallography, X-Ray , Female , Humans , Injections, Intraperitoneal , Injections, Intravenous , Male , Mice , Models, Molecular , Molecular Structure , Phosphorylation , Protein Binding , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, TIE-2/chemistry , Receptor, TIE-2/metabolism , Structure-Activity Relationship , Triazines/pharmacokinetics , Triazines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
13.
J Med Chem ; 50(4): 627-40, 2007 Feb 22.
Article in English | MEDLINE | ID: mdl-17253679

ABSTRACT

The recognition that aberrant angiogenesis contributes to the pathology of inflammatory diseases, cancer, and myocardial ischemia has generated considerable interest in the molecular mechanisms that regulate blood vessel growth. The receptor tyrosine kinase Tie-2 is expressed primarily by vascular endothelial cells and is critical for embryonic vasculogenesis. Interference with the Tie-2 pathway by diverse blocking agents such as soluble Tie-2 receptors, anti-Tie-2 intrabodies, anti-Ang-2 antibodies, and peptide-Fc conjugates has been shown to suppress tumor growth in xenograft studies. An alternative strategy for interfering with the Tie-2 signaling pathway involves direct inhibition of the kinase functions of the Tie-2 receptor. Herein we describe the development of alkynylpyrimidine amide derivatives as potent, selective, and orally available ATP-competitive inhibitors of Tie-2 autophosphorylation.


Subject(s)
Alkynes/chemical synthesis , Amides/chemical synthesis , Angiogenesis Inhibitors/chemical synthesis , Pyrimidines/chemical synthesis , Receptor, TIE-2/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Alkynes/pharmacokinetics , Alkynes/pharmacology , Amides/pharmacokinetics , Amides/pharmacology , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Binding Sites , Blood Proteins/metabolism , Cell Line , Female , Humans , In Vitro Techniques , Lung/drug effects , Lung/enzymology , Male , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Phosphorylation , Protein Binding , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, TIE-2/metabolism , Stereoisomerism , Structure-Activity Relationship
14.
Cancer Res ; 66(17): 8715-21, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951187

ABSTRACT

The growth of solid tumors is dependent on the continued stimulation of endothelial cell proliferation and migration resulting in angiogenesis. The angiogenic process is controlled by a variety of factors of which the vascular endothelial growth factor (VEGF) pathway and its receptors play a pivotal role. Small-molecule inhibitors of VEGF receptors (VEGFR) have been shown to inhibit angiogenesis and tumor growth in preclinical models and in clinical trials. A novel nicotinamide, AMG 706, was identified as a potent, orally bioavailable inhibitor of the VEGFR1/Flt1, VEGFR2/kinase domain receptor/Flk-1, VEGFR3/Flt4, platelet-derived growth factor receptor, and Kit receptors in preclinical models. AMG 706 inhibited human endothelial cell proliferation induced by VEGF, but not by basic fibroblast growth factor in vitro, as well as vascular permeability induced by VEGF in mice. Oral administration of AMG 706 potently inhibited VEGF-induced angiogenesis in the rat corneal model and induced regression of established A431 xenografts. AMG 706 was well tolerated and had no significant effects on body weight or on the general health of the animals. Histologic analysis of tumor xenografts from AMG 706-treated animals revealed an increase in endothelial apoptosis and a reduction in blood vessel area that preceded an increase in tumor cell apoptosis. In summary, AMG 706 is an orally bioavailable, well-tolerated multikinase inhibitor that is presently under clinical investigation for the treatment of human malignancies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Indoles/therapeutic use , Niacinamide/analogs & derivatives , Proto-Oncogene Proteins c-kit/drug effects , Animals , Carcinoma, Squamous Cell/drug therapy , Cell Line , Cell Line, Tumor , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Fibroblasts/drug effects , Humans , Indoles/chemical synthesis , Leukemia, Megakaryoblastic, Acute/drug therapy , Mice , Mice, Nude , Niacinamide/chemical synthesis , Niacinamide/therapeutic use , Oligonucleotides , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Skin/drug effects , Transplantation, Heterologous , Umbilical Veins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...