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.
Bioorg Med Chem ; 21(7): 2145-55, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23411397

ABSTRACT

The uPAR·uPA protein-protein interaction (PPI) is involved in signaling and proteolytic events that promote tumor invasion and metastasis. A previous study had identified 4 (IPR-803) from computational screening of a commercial chemical library and shown that the compound inhibited uPAR·uPA PPI in competition biochemical assays and invasion cellular studies. Here, we synthesize 4 to evaluate in vivo pharmacokinetic (PK) and efficacy studies in a murine breast cancer metastasis model. First, we show, using fluorescence polarization and saturation transfer difference (STD) NMR, that 4 binds directly to uPAR with sub-micromolar affinity of 0.2 µM. We show that 4 blocks invasion of breast MDA-MB-231, and inhibits matrix metalloproteinase (MMP) breakdown of the extracellular matrix (ECM). Derivatives of 4 also inhibited MMP activity and blocked invasion in a concentration-dependent manner. Compound 4 also impaired MDA-MB-231 cell adhesion and migration. Extensive in vivo PK studies in NOD-SCID mice revealed a half-life of nearly 5h and peak concentration of 5 µM. Similar levels of the inhibitor were detected in tumor tissue up to 10h. Female NSG mice inoculated with highly malignant TMD-MDA-MB-231 in their mammary fat pads showed that 4 impaired metastasis to the lungs with only four of the treated mice showing severe or marked metastasis compared to ten for the untreated mice. Compound 4 is a promising template for the development of compounds with enhanced PK parameters and greater efficacy.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , Protein Interaction Maps/drug effects , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Small Molecule Libraries/therapeutic use , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Receptors, Urokinase Plasminogen Activator/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/pharmacology , Urokinase-Type Plasminogen Activator/metabolism
2.
Bioorg Med Chem ; 20(15): 4760-73, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22771232

ABSTRACT

The urokinase receptor (uPAR) serves as a docking site to the serine protease urokinase-type plasminogen activator (uPA) to promote extracellular matrix (ECM) degradation and tumor invasion and metastasis. Previously, we had reported a small molecule inhibitor of the uPAR·uPA interaction that emerged from structure-based virtual screening. Here, we measure the affinity of a large number of derivatives from commercial sources. Synthesis of additional compounds was carried out to probe the role of various groups on the parent compound. Extensive structure-based computational studies suggested a binding mode for these compounds that led to a structure-activity relationship study. Cellular studies in non-small cell lung cancer (NSCLC) cell lines that include A549, H460 and H1299 showed that compounds blocked invasion, migration and adhesion. The effects on invasion of active compounds were consistent with their inhibition of uPA and MMP proteolytic activity. These compounds showed weak cytotoxicity consistent with the confined role of uPAR to metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Drug Design , Mannose-Binding Lectins/antagonists & inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Molecular Dynamics Simulation , Piperidines/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzoates/chemical synthesis , Benzoates/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mannose-Binding Lectins/isolation & purification , Mannose-Binding Lectins/metabolism , Membrane Glycoproteins/isolation & purification , Membrane Glycoproteins/metabolism , Molecular Structure , Molecular Weight , Piperidines/chemical synthesis , Piperidines/chemistry , Receptors, Cell Surface/isolation & purification , Receptors, Cell Surface/metabolism , Structure-Activity Relationship
3.
ACS Chem Biol ; 6(11): 1232-43, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21875078

ABSTRACT

Interaction of the urokinase receptor (uPAR) with its binding partners such as the urokinase-type plasminogen activator (uPA) at the cell surface triggers a series of proteolytic and signaling events that promote invasion and metastasis. Here, we report the discovery of a small molecule (IPR-456) and its derivatives that inhibit the tight uPAR·uPA protein-protein interaction. IPR-456 was discovered by virtual screening against multiple conformations of uPAR sampled from explicit-solvent molecular dynamics simulations. Biochemical characterization reveal that the compound binds to uPAR with submicromolar affinity (K(d) = 310 nM) and inhibits the tight protein-protein interaction with an IC(50) of 10 µM. Free energy calculations based on explicit-solvent molecular dynamics simulations suggested the importance of a carboxylate moiety on IPR-456, which was confirmed by the activity of several derivatives including IPR-803. Immunofluorescence imaging showed that IPR-456 inhibited uPA binding to uPAR of breast MDA-MB-231 tumor cells with an IC(50) of 8 µM. The compounds blocked MDA-MB-231 cell invasion, but IPR-456 showed little effect on MDA-MB-231 migration and no effect on adhesion, suggesting that uPAR mediates these processes through its other binding partners.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Neoplasm Invasiveness/prevention & control , Piperidines/pharmacology , Receptors, Urokinase Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Antineoplastic Agents/chemistry , Benzoates/chemistry , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Drug Discovery , Drug Screening Assays, Antitumor , Female , Humans , Molecular Conformation/drug effects , Molecular Weight , Neoplasm Invasiveness/pathology , Piperidines/chemistry , Protein Binding/drug effects , Receptors, Urokinase Plasminogen Activator/chemistry , Structure-Activity Relationship , Urokinase-Type Plasminogen Activator/chemistry
4.
J Med Chem ; 54(20): 7193-205, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21851064

ABSTRACT

Virtual screening targeting the urokinase receptor (uPAR) led to (±)-3-(benzo[d][1,3]dioxol-5-yl)-N-(benzo[d][1,3]dioxol-5-ylmethyl)-4-phenylbutan-1-amine 1 (IPR-1) and N-(3,5-dimethylphenyl)-1-(4-isopropylphenyl)-5-(piperidin-4-yl)-1H-pyrazole-4-carboxamide 3 (IPR-69). Synthesis of an analogue of 1, namely, 2 (IPR-9), and 3 led to breast MDA-MB-231 invasion, migration and adhesion assays with IC(50) near 30 µM. Both compounds blocked angiogenesis with IC(50) of 3 µM. Compounds 2 and 3 inhibited cell growth with IC(50) of 6 and 18 µM and induced apoptosis. Biochemical assays revealed leadlike properties for 3, but not 2. Compound 3 administered orally reached peak concentration of nearly 40 µM with a half-life of about 2 h. In NOD-SCID mice inoculated with breast TMD-231 cells in their mammary fat pads, compound 3 showed a 20% reduction in tumor volumes and less extensive metastasis was observed for the treated mice. The suitable pharmacokinetic properties of 3 and the encouraging preliminary results in metastasis make it an ideal starting point for next generation compounds.


Subject(s)
Anilides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/pathology , Pyrazoles/chemical synthesis , Receptors, Urokinase Plasminogen Activator/metabolism , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Anilides/pharmacokinetics , Anilides/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Databases, Factual , Drug Screening Assays, Antitumor , Female , Human Umbilical Vein Endothelial Cells , Humans , Matrix Metalloproteinase Inhibitors , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Signal Transduction , Structure-Activity Relationship , Transplantation, Heterologous
5.
J Chem Inf Model ; 51(4): 755-9, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21438548

ABSTRACT

We assess the performance of our previously reported structure-based support vector machine target-specific scoring function across 41 targets, 40 among them from the Directory of Useful Decoys (DUD). The area under the curve of receiver operating characteristic plots (ROC-AUC) revealed that scoring with SVM-SP resulted in consistently better enrichment over all target families, outperforming Glide and other scoring functions, most notably among kinases. In addition, SVM-SP performance showed little variation among protein classes, exhibited excellent performance in a test case using a homology model, and in some cases showed high enrichment even with few structures used to train a model. We put SVM-SP to the test by virtual screening 1125 compounds against two kinases, EGFR and CaMKII. Among the top 25 EGFR compounds, three compounds (1-3) inhibited kinase activity in vitro with IC50 of 58, 2, and 10 µM. In cell cultures, compounds 1-3 inhibited nonsmall cell lung carcinoma (H1299) cancer cell proliferation with similar IC50 values for compound 3. For CaMKII, one compound inhibited kinase activity in a dose-dependent manner among 20 tested with an IC50 of 48 µM. These results are encouraging given that our in-house library consists of compounds that emerged from virtual screening of other targets with pockets that are different from typical ATP binding sites found in kinases. In light of the importance of kinases in chemical biology, these findings could have implications in future efforts to identify chemical probes of kinases within the human kinome.


Subject(s)
Artificial Intelligence , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Drug Discovery/methods , ErbB Receptors/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Algorithms , Binding Sites , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Lung Neoplasms/enzymology , Models, Molecular , Quantitative Structure-Activity Relationship , ROC Curve
6.
ACS Med Chem Lett ; 1(5): 229-233, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20824148

ABSTRACT

In an effort to develop a rational approach to identify anti-cancer agents with selective polypharmacology, we mine millions of docked protein-ligand complexes involving more than a thousand cancer targets from multiple signaling pathways to identify new structural templates for proven pharmacophores. Our method combines Support Vector Machine-based scoring to enrich the initial library of 1,592 molecules, with a fingerprint-based search for molecules that have the same binding profile as the EGFR kinase inhibitor erlotinib. Twelve new compounds were identified. In vitro activity assays revealed that three inhibited EGFR with IC(50) values ranging from 250 nM to 200 µM. Additional in vitro studies with hERG, CYP450, DNA and cell culture-based assays further compared their properties to erlotinib. One compound combined suitable pharmacokinetic properties while closely mimicking the binding profile of erlotinib. The compound also inhibited H1299 and H460 tumor cell proliferation. The other two compounds shared some of the binding profile of erlotinib, and one gave the most potent inhibition of tumor cell growth. Interestingly, among the compounds that had not shown inhibition of EGFR, four blocked H1299 and H460 proliferation, one potently with IC(50) values near 1 µM. This compound was from the menogaril family, which reached Phase II clinical trial for the treatment of lymphomas. This suggests that our computational approach comparing binding profile may have favored molecules with anti-cancer properties like erlotinib.

SELECTION OF CITATIONS
SEARCH DETAIL
...