Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 21(6): 1815-8, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21316232

ABSTRACT

A series of 4-aminopyrimidine-5-carbaldehyde oxime was discovered to have potent VEGFR-2 inhibitory activity. Described here are the chemistry for analogue synthesis and SAR study results. The PK properties, kinase profiling, and in vivo efficacy study for compound 4b are also discussed.


Subject(s)
Oximes/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Mice , Oximes/chemistry , Rats , Structure-Activity Relationship
2.
Angiogenesis ; 12(3): 287-96, 2009.
Article in English | MEDLINE | ID: mdl-19544081

ABSTRACT

Angiogenesis is a complex process that relies on a variety of growth factors and signaling pathways to stimulate endothelial cell responses and establish functional blood vessels. Signaling through the vascular endothelial growth factor (VEGF) receptors is an important mediator of angiogenesis, a hallmark of tumor growth and metastasis. Inhibition of signaling through VEGF has been clinically validated with FDA-approvals of bevacizumab, sorafenib, and suntinib. Our goal was to discover an orally available, selective VEGFR-2 inhibitor. A novel oxime, 1-{4-[6-amino-5-(methoxyimino-methyl)-pyrimidin-4-yloxy]-2-chloro-phenyl}-3-ethyl-urea (JNJ-38158471), was identified as a potent and selective inhibitor of VEGFR-2. While JNJ-38158471 shares some structure features with sorafenib, unlike sorafenib, it lacks Raf kinase activity. JNJ-38158471 inhibits VEGFR-2 (IC50 = 40 nM) and closely related tyrosine kinases, Ret (180 nM) and Kit (500 nM); it has no significant activity (>1 microM) against VEGFR-1 and VEGFR-3. At nanomolar levels, it inhibits VEGF-stimulated autophosphorylation of VEGFR-2 in a whole cell assay and inhibits VEGF-dependent endothelial migration. Once-daily oral dosing of JNJ-3815871 to nude mice bearing human A431, HCT116, and A375 tumors resulted in up to 90% tumor growth inhibition. Strikingly, after termination of JNJ-38158471 monotherapy-treatment of A375 xenografts, tumor growth delay was significantly prolonged up to 4 weeks. Anti-tumor efficacy correlated well with the observed dose concentrations (on a mg/kg basis) necessary to inhibit VEGF-induced corneal angiogenesis in C57BL/6J mice. In addition, the compound inhibited spontaneous polyp formation in the APC min-mouse model. These data demonstrate that JNJ-38158471 is a well tolerated, orally available, highly selective VEGFR-2 inhibitor that may have therapeutic benefit in human malignancies.


Subject(s)
Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/physiology , Female , HCT116 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Models, Biological , Neoplasms/pathology , Oximes/administration & dosage , Oximes/pharmacology , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/pharmacology , Substrate Specificity , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem Lett ; 16(23): 6063-6, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16979339

ABSTRACT

A novel 4-aminopyrimidine-5-carboxaldehyde oxime scaffold with inhibitory activity against VEGFR-2 kinase has been identified. With a 4-fluoro-2-methylindol-5-yloxy group at the 6-position and alkyl groups as the oxime side chains, many analogues showed good potency for VEGFR-2. This series also exhibited antiproliferative activity against cancer cells, causing cell accumulation at the G2/M phase of the cell cycle and preventing cells from entering mitosis. Described here are the chemistry, structure-activity relationships (SAR), and biological testing for this series.


Subject(s)
Oximes/chemistry , Oximes/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Cell Proliferation/drug effects , HeLa Cells , Humans , Molecular Structure , Oximes/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
Mol Cancer Ther ; 5(2): 347-55, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16505108

ABSTRACT

We investigated the significance of erythropoietin receptor (EPOR) expression following treatment with recombinant human erythropoietin (rHuEPO; epoetin alpha) and the effect of recombinant epoetins (epoetin alpha, epoetin beta, and darbepoetin alpha) alone or in combination with anticancer therapy on tumor growth in two well-established preclinical models of breast carcinoma (MDA-MB-231 and MCF-7 cell lines). Expression and localization of EPOR under hypoxic and normoxic conditions in MDA-MB-231 and MCF-7 cells were evaluated by immunoblotting, flow cytometry, and immunohistochemistry. EPOR binding was evaluated using [125I]rHuEPO. Proliferation, migration, and signaling in MDA-MB-231 and MCF-7 cells following treatment with rHuEPO were evaluated. Tumor growth was assessed following administration of recombinant epoetins alone and in combination with paclitaxel (anticancer therapy) in orthotopically implanted MDA-MB-231 and MCF-7 breast carcinoma xenograft models in athymic mice. EPOR expression was detected in both tumor cell lines. EPOR localization was found to be exclusively cytosolic and no specific [125I]rHuEPO binding was observed. There was no stimulated migration, proliferation, or activation of mitogen-activated protein kinase and AKT following rHuEPO treatment. In mice, treatment with recombinant epoetins alone and in combination with paclitaxel resulted in equivalent tumor burdens compared with vehicle-treated controls. Results from our study suggest that although EPOR expression was observed in two well-established breast carcinoma cell lines, it was localized to a cytosolic distribution and did not transduce a signaling cascade in tumors that leads to tumor growth. The addition of recombinant epoetins to paclitaxel did not affect the outcome of paclitaxel therapy in breast carcinoma xenograft models. These results show that recombinant epoetins do not evoke a physiologic response on EPOR-bearing tumor cells as assessed by numerous variables, including growth, migration, and cytotoxic challenge in preclinical in vivo tumor models.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Erythropoietin/therapeutic use , Receptors, Erythropoietin/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/chemistry , Breast Neoplasms/metabolism , Carcinoma/chemistry , Carcinoma/metabolism , Cell Hypoxia , Cell Movement/drug effects , Cell Proliferation/drug effects , Erythropoietin/adverse effects , Female , Humans , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Paclitaxel/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/analysis , Recombinant Proteins , Xenograft Model Antitumor Assays
5.
Cancer Res ; 66(1): 221-31, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16397235

ABSTRACT

FTY720, a potent immunomodulator, becomes phosphorylated in vivo (FTY-P) and interacts with sphingosine-1-phosphate (S1P) receptors. Recent studies showed that FTY-P affects vascular endothelial growth factor (VEGF)-induced vascular permeability, an important aspect of angiogenesis. We show here that FTY720 has antiangiogenic activity, potently abrogating VEGF- and S1P-induced angiogenesis in vivo in growth factor implant and corneal models. FTY720 administration tended to inhibit primary and significantly inhibited metastatic tumor growth in a mouse model of melanoma growth. In combination with a VEGFR tyrosine kinase inhibitor PTK787/ZK222584, FTY720 showed some additional benefit. FTY720 markedly inhibited tumor-associated angiogenesis, and this was accompanied by decreased tumor cell proliferation and increased apoptosis. In transfected HEK293 cells, FTY-P internalized S1P1 receptors, inhibited their recycling to the cell surface, and desensitized S1P receptor function. Both FTY720 and FTY-P apparently failed to impede VEGF-produced increases in mitogen-activated protein kinase activity in human umbilical vascular endothelial cells (HUVEC), and unlike its activity in causing S1PR internalization, FTY-P did not result in a decrease of surface VEGFR2 levels in HUVEC cells. Pretreatment with FTY720 or FTY-P prevented S1P-induced Ca2+ mobilization and migration in vascular endothelial cells. These data show that functional antagonism of vascular S1P receptors by FTY720 potently inhibits angiogenesis; therefore, this may provide a novel therapeutic approach for pathologic conditions with dysregulated angiogenesis.


Subject(s)
Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/antagonists & inhibitors , Animals , Calcium/metabolism , Cell Growth Processes/drug effects , Cell Movement/drug effects , Cornea/blood supply , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Female , Fingolimod Hydrochloride , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Phosphorylation , Phthalazines/pharmacology , Propylene Glycols/pharmacokinetics , Pyridines/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Cancer Res ; 64(9): 2984-7, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15126329

ABSTRACT

Inhibition of endothelial cell growth by fumagillin has been assumed to be mediated by inhibition of the molecular target methionine aminopeptidase 2 (MetAp2). New data show that depletion of MetAp2 by siRNA does not inhibit endothelial cell growth. Moreover, MetAp2-depleted endothelial cells remain responsive to inhibition by either fumagillin or a newly identified MetAp2 enzyme inhibitor. These data suggest that MetAp2 function is not required for endothelial cell proliferation.


Subject(s)
Aminopeptidases/deficiency , Azepines/pharmacology , Fatty Acids, Unsaturated/pharmacology , Metalloendopeptidases/deficiency , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/genetics , Aminopeptidases/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Division/drug effects , Cell Division/physiology , Cell Line, Tumor , Cyclohexanes , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacology , Humans , Lung Neoplasms/enzymology , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , RNA, Small Interfering/genetics , Sesquiterpenes , Substrate Specificity , Transfection
8.
Prostaglandins Other Lipid Mediat ; 73(1-2): 29-45, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15165029

ABSTRACT

Sphingosine 1-phosphate (S1P) is a pleiotropic lysophospholipid mediator involved in many cellular responses, including transient calcium mobilization, activation of MAP kinase signaling, inhibition of adenylyl cyclase and increased cell migration. S1P has been shown to be an effective activator of vascular endothelial cells via the interaction with cell surface G protein-coupled receptors (GPCRs), namely S1P-R (formerly EDG-R). The potent immunomodulator, FTY720, is phosphorylated by sphingosine kinase (SK) to FTY720-P. Recently it was shown that FTY720-P, not FTY720, can bind to four out of five of the S1P-R. In the present study, we evaluated the effects of FTY720, FTY720-P, and analogues of FTY720-P: an active (R)-enantiomer [AFD(R)] and an inactive (S)-enantiomer [AFD(S)], on endothelial cell functions. Treatment of HUVEC with FTY720-P, but not FTY720, lead to a robust transient increase in calcium mobilization, detected using the fluorometric imaging plate reader (FLIPR) assay. Additionally, only the phosphorylated derivative (FTY720-P) stimulated MAPK activation. We also observed complementary activities of S1P and FTY720-P in an established in vitro endothelial morphogenesis (Matrigel tube formation) assay and an in vitro endothelial cell migration assay. Using a potent inhibitor of sphingosine kinase, N,N-dimethylsphingosine (DMS), FTY720's effects were inhibited in the migration assay, suggesting that FTY720-P is the active mediator. The effects of FTY720-P in these assays were inhibited by pre-treatment with PTx (pertussis toxin), indicating the requirement of a Gi-coupled S1P receptor. These findings suggest that agonist of S1P-R are able to regulate important endothelial cell properties, which may lead to a greater insight into vascular functions.


Subject(s)
Endothelium, Vascular/metabolism , Immunosuppressive Agents/metabolism , Lysophospholipids/metabolism , Propylene Glycols/metabolism , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Fingolimod Hydrochloride , GTP-Binding Protein alpha Subunits, Gi-Go , Humans , Immunosuppressive Agents/pharmacology , Isomerism , Mitogen-Activated Protein Kinases/metabolism , Molecular Structure , Organogenesis/drug effects , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Propylene Glycols/pharmacology , Structure-Activity Relationship , Umbilical Veins/cytology , Umbilical Veins/metabolism
9.
J Biol Chem ; 277(47): 45211-8, 2002 Nov 22.
Article in English | MEDLINE | ID: mdl-12237301

ABSTRACT

We have shown previously that the oligomeric endostatin domain of collagen XVIII (NC1) functioned as a motility-inducing factor regulating the extracellular matrix-dependent morphogenesis of endothelial cells. This motogenic activity gave rise to structures resembling filipodia and lamellipodia and was dependent on Rac, Cdc42, and mitogen-activated protein kinase. Here, we demonstrate that these properties of endostatin are primarily mediated by laminin in the basement membrane and heparan sulfates on the cell surface. The sites of interaction between laminin and oligomeric endostain include the N-terminal regions of all three laminin chains (amino acids 204-1243 of the alpha chain, 932-1161 of the beta chain, and 150-965 of the gamma chain). A monoclonal antibody that blocks the interactions between endostatin and laminin was utilized to inhibit the motogenic activity of endostatin. In parallel, we have engineered selective point mutations and produced recombinant forms that lack binding to heparan sulfates on the cell surface. Our data are consistent with a model of endostatin with two binding sites: one mainly to laminin in the basement membrane and the other to heparan sulfates on the cell surface. The two binding domains on endostatin appear to be separate with the possibility of some overlap between the two sites.


Subject(s)
Angiogenesis Inhibitors/metabolism , Collagen/metabolism , Laminin/metabolism , Peptide Fragments/metabolism , Amino Acid Sequence , Angiogenesis Inhibitors/genetics , Binding Sites , Collagen/genetics , Collagen Type XVIII , Dimerization , Endostatins , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/genetics , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
10.
Mol Med ; 8(1): 1-8, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11984000

ABSTRACT

BACKGROUND: Implantation of foreign materials into mice and humans has been noted to result in the appearance of soft tissue sarcomas at the site of implantation. These materials include metal replacement joints and Dacron vascular grafts. In addition, occupational exposure to nickel has been shown to result in an increased risk of carcinogenesis. The molecular mechanisms of foreign body-induced carcinogenesis are not fully understood. MATERIALS AND METHODS: In order to gain insight into these mechanisms, we implanted nickel sulfide into wild type C57BL/6 mice as well as a mouse heterozygous for the tumor suppressor gene, p53. Malignant fibrous histiocytomas arose in all mice, and we have characterized the profile of tumor suppressor genes and signal transduction pathways altered in these cells. RESULTS: All tumors demonstrated hypermethylation of the tumor suppressor gene p16, as well as activation of the mitogen activated protein kinase (MAP kinase) signaling pathway. This knowledge may be beneficial in the prevention and treatment of tumors caused by foreign body implantation. CONCLUSIONS: Oxidative stress induced by nickel sulfide appears to cause loss of p16 and activation of MAP kinase signaling. These findings support the hypothesis of synergistic interactions between MAP kinase activation and p16 loss in carcinogenesis.


Subject(s)
Carcinogens/toxicity , DNA Methylation/drug effects , Genes, p16/drug effects , Histiocytoma, Benign Fibrous/chemically induced , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Muscle Neoplasms/chemically induced , Neoplasm Proteins/genetics , Nickel/toxicity , Reactive Oxygen Species/toxicity , Animals , Carcinogens/pharmacology , CpG Islands , Cyclin-Dependent Kinase Inhibitor p16 , Drug Implants , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Genes, p53 , Genes, ras , Hindlimb , Histiocytoma, Benign Fibrous/enzymology , Histiocytoma, Benign Fibrous/genetics , Mice , Mice, Inbred C57BL , Muscle Neoplasms/enzymology , Muscle Neoplasms/genetics , Mutagenesis , Neoplasm Proteins/metabolism , Nickel/pharmacology , Oxidative Stress , Polymerase Chain Reaction , Reactive Oxygen Species/pharmacology , Tumor Suppressor Protein p14ARF/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...