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.
Mol Cancer Ther ; 14(4): 931-40, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637314

ABSTRACT

Dysregulation of PI3K/PTEN pathway components, resulting in hyperactivated PI3K signaling, is frequently observed in various cancers and correlates with tumor growth and survival. Resistance to a variety of anticancer therapies, including receptor tyrosine kinase (RTK) inhibitors and chemotherapeutic agents, has been attributed to the absence or attenuation of downregulating signals along the PI3K/PTEN pathway. Thus, PI3K inhibitors have therapeutic potential as single agents and in combination with other therapies for a variety of cancer indications. XL147 (SAR245408) is a potent and highly selective inhibitor of class I PI3Ks (α, ß, γ, and δ). Moreover, broad kinase selectivity profiling of >130 protein kinases revealed that XL147 is highly selective for class I PI3Ks over other kinases. In cellular assays, XL147 inhibits the formation of PIP3 in the membrane, and inhibits phosphorylation of AKT, p70S6K, and S6 in multiple tumor cell lines with diverse genetic alterations affecting the PI3K pathway. In a panel of tumor cell lines, XL147 inhibits proliferation with a wide range of potencies, with evidence of an impact of genotype on sensitivity. In mouse xenograft models, oral administration of XL147 results in dose-dependent inhibition of phosphorylation of AKT, p70S6K, and S6 with a duration of action of at least 24 hours. Repeat-dose administration of XL147 results in significant tumor growth inhibition in multiple human xenograft models in nude mice. Administration of XL147 in combination with chemotherapeutic agents results in antitumor activity in xenograft models that is enhanced over that observed with the corresponding single agents.


Subject(s)
Antineoplastic Agents/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Quinoxalines/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Male , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Quinoxalines/administration & dosage , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 13(5): 1078-91, 2014 May.
Article in English | MEDLINE | ID: mdl-24634413

ABSTRACT

Activation of the PI3K (phosphoinositide 3-kinase) pathway is a frequent occurrence in human tumors and is thought to promote growth, survival, and resistance to diverse therapies. Here, we report pharmacologic characterization of the pyridopyrimidinone derivative XL765 (SAR245409), a potent and highly selective pan inhibitor of class I PI3Ks (α, ß, γ, and δ) with activity against mTOR. Broad kinase selectivity profiling of >130 protein kinases revealed that XL765 is highly selective for class I PI3Ks and mTOR over other kinases. In cellular assays, XL765 inhibits the formation of PIP(3) in the membrane, and inhibits phosphorylation of AKT, p70S6K, and S6 phosphorylation in multiple tumor cell lines with different genetic alterations affecting the PI3K pathway. In a panel of tumor cell lines, XL765 inhibits proliferation with a wide range of potencies, with evidence of an impact of genotype on sensitivity. In mouse xenograft models, oral administration of XL765 results in dose-dependent inhibition of phosphorylation of AKT, p70S6K, and S6 with a duration of action of approximately 24 hours. Repeat dose administration of XL765 results in significant tumor growth inhibition in multiple human xenograft models in nude mice that is associated with antiproliferative, antiangiogenic, and proapoptotic effects.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/genetics , Neoplasms/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Quinoxalines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Humans , Inhibitory Concentration 50 , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Quinoxalines/administration & dosage , Ribosomal Protein S6 Kinases/metabolism , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem Lett ; 22(15): 4979-85, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22765894

ABSTRACT

Variously substituted indolin-2-ones were synthesized and evaluated for activity against KDR, Flt-1, FGFR-1 and PDGFR. Extension at the 5-position of the oxindole ring with ethyl piperidine (compound 7i) proved to be the most beneficial for attaining both biochemical and cellular potencies. Further optimization of 7i to balance biochemical and cellular potencies with favorable ADME/ PK properties led to the identification of 8h, a compound with a clean CYP profile, acceptable pharmacokinetic and toxicity profiles, and robust efficacy in multiple xenograft tumor models.


Subject(s)
Drug Design , Indoles/chemical synthesis , Piperidines/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Cytochrome P-450 CYP3A/metabolism , Female , Half-Life , Humans , Indoles/pharmacokinetics , Indoles/therapeutic use , Lung/drug effects , Lung/metabolism , Mice , Neoplasms/drug therapy , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , Transplantation, Heterologous
4.
J Med Chem ; 55(9): 4322-35, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22497444

ABSTRACT

Targeting glycosphingolipid synthesis has emerged as a novel approach for treating metabolic diseases. 32 (EXEL-0346) represents a new class of glucosylceramide synthase (GCS) inhibitors. This report details the elaboration of hit 8 with the goal of achieving and maintaining maximum GCS inhibition in vivo. 32 inhibited GCS with an IC(50) of 2 nM and achieved maximum hepatic GCS inhibition after four or five daily doses in rodents. Robust improvements in glucose tolerance in DIO mice and ZDF rats were observed after 2 weeks of q.d. dosing. Four weeks of dosing resulted in decreased plasma triglycerides and reduced hepatic fat deposition. Thus, 32 provides insight into the amount of metabolic regulation that can be restored following achievement of maximal target knockdown.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Glucosyltransferases/antagonists & inhibitors , Phenylalanine/analogs & derivatives , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , Gangliosides/metabolism , Glucose Tolerance Test , Glucosyltransferases/metabolism , Humans , Liver/drug effects , Liver/enzymology , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Nude , Phenylalanine/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Zucker , Structure-Activity Relationship , Triglycerides/blood
5.
Bioorg Med Chem Lett ; 21(22): 6773-7, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21974949

ABSTRACT

A novel series of potent inhibitors of glucosylceramide synthase are described. The optimization of biochemical and cellular potency as well as ADME properties led to compound 23c. Broad tissue distribution was obtained following oral administration to mice. Thus 23c could be another useful tool compound for studying the effects of GCS inhibition in vitro and in vivo.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Administration, Oral , Animals , Drug Discovery , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Humans , Mice , Mice, Inbred C57BL , Structure-Activity Relationship
6.
Mol Cancer Ther ; 10(12): 2298-308, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21926191

ABSTRACT

The signaling pathway of the receptor tyrosine kinase MET and its ligand hepatocyte growth factor (HGF) is important for cell growth, survival, and motility and is functionally linked to the signaling pathway of VEGF, which is widely recognized as a key effector in angiogenesis and cancer progression. Dysregulation of the MET/VEGF axis is found in a number of human malignancies and has been associated with tumorigenesis. Cabozantinib (XL184) is a small-molecule kinase inhibitor with potent activity toward MET and VEGF receptor 2 (VEGFR2), as well as a number of other receptor tyrosine kinases that have also been implicated in tumor pathobiology, including RET, KIT, AXL, and FLT3. Treatment with cabozantinib inhibited MET and VEGFR2 phosphorylation in vitro and in tumor models in vivo and led to significant reductions in cell invasion in vitro. In mouse models, cabozantinib dramatically altered tumor pathology, resulting in decreased tumor and endothelial cell proliferation coupled with increased apoptosis and dose-dependent inhibition of tumor growth in breast, lung, and glioma tumor models. Importantly, treatment with cabozantinib did not increase lung tumor burden in an experimental model of metastasis, which has been observed with inhibitors of VEGF signaling that do not target MET. Collectively, these data suggest that cabozantinib is a promising agent for inhibiting tumor angiogenesis and metastasis in cancers with dysregulated MET and VEGFR signaling.


Subject(s)
Anilides/therapeutic use , Cell Growth Processes/drug effects , Neoplasm Metastasis/prevention & control , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Pyridines/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Anilides/pharmacology , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Mice , Mice, Nude , Models, Biological , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
7.
Cancer Res ; 69(20): 8009-16, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19808973

ABSTRACT

The Met receptor tyrosine kinase and its ligand, hepatocyte growth factor (HGF), are overexpressed and/or activated in a wide variety of human malignancies. Vascular endothelial growth factor (VEGF) receptors are expressed on the surface of vascular endothelial cells and cooperate with Met to induce tumor invasion and vascularization. EXEL-2880 (XL880, GSK1363089) is a small-molecule kinase inhibitor that targets members of the HGF and VEGF receptor tyrosine kinase families, with additional inhibitory activity toward KIT, Flt-3, platelet-derived growth factor receptor beta, and Tie-2. Binding of EXEL-2880 to Met and VEGF receptor 2 (KDR) is characterized by a very slow off-rate, consistent with X-ray crystallographic data showing that the inhibitor is deeply bound in the Met kinase active site cleft. EXEL-2880 inhibits cellular HGF-induced Met phosphorylation and VEGF-induced extracellular signal-regulated kinase phosphorylation and prevents both HGF-induced responses of tumor cells and HGF/VEGF-induced responses of endothelial cells. In addition, EXEL-2880 prevents anchorage-independent proliferation of tumor cells under both normoxic and hypoxic conditions. In vivo, these effects produce significant dose-dependent inhibition of tumor burden in an experimental model of lung metastasis. Collectively, these data indicate that EXEL-2880 may prevent tumor growth through a direct effect on tumor cell proliferation and by inhibition of invasion and angiogenesis mediated by HGF and VEGF receptors.


Subject(s)
Anilides/pharmacology , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Dermis/cytology , Dermis/drug effects , Dermis/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Hepatocyte Growth Factor/pharmacology , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/prevention & control , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...