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 ; 8(1): 45-54, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19139112

ABSTRACT

The protein kinase checkpoint kinase 1 (Chk1) has been implicated as a key regulator of cell cycle progression and DNA repair, and inhibitors of Chk1 (e.g., UCN-01 and EXEL-9844) potentiate the cytotoxic actions of chemotherapeutic drugs in tumor cells. We have examined the ability of PD-321852, a small-molecule Chk1 inhibitor, to potentiate gemcitabine-induced clonogenic death in a panel of pancreatic cancer cell lines and evaluated the relationship between endpoints associated with Chk1 inhibition and chemosensitization. Gemcitabine chemosensitization by minimally toxic concentrations of PD-321852 ranged from minimal (<3-fold change in survival) in Panc1 cells to >30-fold in MiaPaCa2 cells. PD-321852 inhibited Chk1 in all cell lines as evidenced by stabilization of Cdc25A; in combination with gemcitabine, a synergistic loss of Chk1 protein was observed in the more sensitized cell lines. Gemcitabine chemosensitization, however, did not correlate with abrogation of the S-M or G2-M checkpoint; PD-321852 did not induce premature mitotic entry in gemcitabine-treated BxPC3 or M-Panc96 cells, which were sensitized to gemcitabine 6.2- and 4.6-fold, respectively. In the more sensitized cells lines, PD-321852 not only inhibited gemcitabine-induced Rad51 focus formation and the recovery from gemcitabine-induced replication stress, as evidenced by persistence of gamma-H2AX, but also depleted these cells of Rad51 protein. Our data suggest the inhibition of this Chk1-mediated Rad51 response to gemcitabine-induced replication stress is an important factor in determining gemcitabine chemosensitization by Chk1 inhibition in pancreatic cancer cells.


Subject(s)
Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Biocatalysis , Carbazoles/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Checkpoint Kinase 1 , DNA Damage , Deoxycytidine/pharmacology , Humans , Pancreatic Neoplasms/genetics , Phosphorylation/drug effects , Gemcitabine
2.
Eur J Med Chem ; 43(6): 1276-96, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17869387

ABSTRACT

A series of N-6 substituted 9-hydroxy-4-phenylpyrrolo[3,4-c]carbazole-1,3(2H,6H)-diones were prepared from N-substituted (5-methoxyphenyl)ethenylindoles. The target compounds were tested for their ability to inhibit the G2/M cell cycle checkpoint kinases, Wee1 and Chk1. Analogues with neutral or cationic N-6 side chains were potent dual inhibitors. Acidic side chains provided potent (average IC(50) 0.057 microM) and selective (average ratio 223-fold) Wee1 inhibition. Co-crystal structures of inhibitors bound to Wee1 show that the pyrrolo[3,4-c]carbazole scaffold binds in the ATP-binding site, with N-6 substituents involved in H-bonding to conserved water molecules. HT-29 cells treated with doxorubicin and then target compounds demonstrate an active Cdc2/cyclin B complex, inhibition of the doxorubicin-induced phosphorylation of tyrosine 15 of Cdc2 and abrogation of the G2 checkpoint.


Subject(s)
Carbazoles/chemical synthesis , Carbazoles/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Carbazoles/chemistry , HT29 Cells , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
3.
J Med Chem ; 49(16): 4896-911, 2006 Aug 10.
Article in English | MEDLINE | ID: mdl-16884302

ABSTRACT

High-throughput screening has identified a novel class of inhibitors of the checkpoint kinase Wee1, which have potential for use in cancer chemotherapy. These inhibitors are based on a 4-phenylpyrrolo[3,4-c]carbazole-1,3(2H,6H)-dione template and have been shown by X-ray crystallography to bind at the ATP site of the enzyme. An extensive study of the effects of substitution around this template has been carried out, which has identified substituents which lead to improvements in potency and selectivity for Wee1. While retention of the maleimide ring and pendant 4-phenyl group is necessary for potency, replacement of the carbazole nitrogen by oxygen is well tolerated and results in improved Wee1 selectivity against the related checkpoint kinase Chk1. Wee1 potency and selectivity are also enhanced by the incorporation of lipophilic functionality at the 2'-position of the 4-phenyl ring, and Wee1 selectivity against Chk1 is favored by C3-C5 alkyl substitution of the carbazole nitrogen. These studies provide a basis for the design of active analogues of the pyrrolocarbazole lead with improved physical properties.


Subject(s)
Benzene Derivatives/chemical synthesis , Carbazoles/chemical synthesis , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Pyrroles/chemical synthesis , Benzene Derivatives/chemistry , Carbazoles/chemistry , Checkpoint Kinase 1 , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Kinases/chemistry , Pyrroles/chemistry , Structure-Activity Relationship
4.
Mol Cancer Ther ; 2(4): 401-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12700284

ABSTRACT

CI-994 or N-acetyldinaline [4-(acetylamino)-N-(2-amino-phenyl) benzamide] is an antitumor cytostatic agent currently undergoing clinical trial. Although several changes in cellular metabolism induced by the drug have been characterized, the primary molecular mechanism of its antitumor activity has been previously unknown. Here, we show that CI-994 is a histone deacetylase (HDAC) inhibitor that causes histone hyperacetylation in living cells. In assays of isolated enzymes, CI-994 inhibited HDAC-1 and HDAC-2 in a concentration-dependent fashion but had no effect on the activity of the prototypical histone acetyltransferase GCN5. Acetylated histone H3-specific Western blots were used to monitor histone acetylation in HCT-8 colon carcinoma cells treated with CI-994 in vitro. CI-994 induced hyperacetylation of H3 in a time- and dose-dependent fashion. H3 hyperacetylation was detectable as early as 30 min after the addition of CI-994 to cells. These data demonstrate that inhibition of HDAC is an early event in cells treated with CI-994 and suggest that this inhibition is mechanistically related to the antitumor activity of this compound.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Histones/chemistry , Phenylenediamines/pharmacology , Acetylation , Benzamides , Blotting, Western , Cell Line, Tumor , Cell Nucleus/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , Inhibitory Concentration 50 , Models, Chemical , Precipitin Tests , Time Factors
5.
Clin Cancer Res ; 9(4): 1267-73, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12684394

ABSTRACT

Inhibition of the constitutively active Bcr-abl tyrosine kinase(TK) by STI571 has proven to be a highly effective treatment for chronic myelogenous leukemia (CML). However, STI571 is only transiently effective in blast crisis, and drug resistance emerges by amplification of or development of mutational changes in Bcr-abl. We have screened a family of TK inhibitors of the pyrido [2,3-d]pyrimidine class, unrelated to STI571, and describe here a compound with substantial activity against STI-resistant mutant Bcr-abl proteins. This compound, PD166326, is a dual specificity TK inhibitor and inhibits src and abl in vitro with IC(50)s of 6 and 8 nM respectively. PD166326 inhibits the growth of K562 cells with IC(50) of 300 pM, leading to apoptotic G(1) arrest, whereas non-Bcr-abl cell types require >1000 times higher concentrations. We tested the effects of PD166326 on two of the clinically observed STI571-resistant Bcr-abl mutants. PD166326 potently inhibits the E255K mutant Bcr-abl protein and the growth of Bcr-ablE255K-driven cells. The T315I mutant Bcr-abl protein, which is mutated within the ATP-binding pocket, is resistant to PD166326; however, the growth of Bcr-ablT315I-driven cells is partially sensitive to this compound, likely through the inhibition of Bcr-abl effector pathways. These findings show that TK drug resistance is a structure-specific phenomenon and can be overcome by TK inhibitors of other structural classes, suggesting new approaches for future anticancer drug development. PD166326 is a prototype of a new generation of anti-Bcr-abl compounds with picomolar potency and substantial activity against STI571-resistant mutants.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/metabolism , Mutation , Piperazines/therapeutic use , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrimidines/therapeutic use , Benzamides , Blotting, Western , Cell Cycle , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , G1 Phase/drug effects , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , K562 Cells , Pyridines/pharmacology , Pyrimidines/pharmacology
6.
Oncogene ; 21(51): 7797-807, 2002 Nov 07.
Article in English | MEDLINE | ID: mdl-12420216

ABSTRACT

Src is a non-receptor protein tyrosine kinase, the expression and activity of which is increased in >80% of human colon cancers with respect to normal colonic epithelium. Previous studies from this and other laboratories have demonstrated that Src activity contributes to tumorigenicity of established colon adenocarcinoma cell lines. Src participates in the regulation of many signal transduction pathways, among which are those leading to cellular survival. In this study, we addressed the potential role of Src activation to a specific aspect of tumor cell survival, resistance to detachment-induced apoptosis (anoikis). Using five colon tumor cell lines with different biologic properties and genetic alterations, we demonstrate that expression and activity of Src corresponds with resistance to anoikis. Enforced expression of activated Src in subclones of SW480 cells (of low intrinsic Src expression and activity) increases resistance to anoikis; whereas decreased Src expression in HT29 cells (of high Src expression and activity) by transfection with anti-sense Src expression vectors increases susceptibility to anoikis. In contrast, increasing or decreasing Src expression had no effect on susceptibility to staurosporine-induced apoptosis in attached cells. PD173955, a Src family-specific tyrosine kinase inhibitor, increases the susceptibility of HT29 cells to anoikis in a dose- and time-dependent manner. Increasing Src expression and activity led to increased phosphorylation of Akt, a mediator of cellular survival pathways, whereas decreasing Src activity led to decreased Akt phosphorylation. In colon tumor cells with high Src activity, the PI3 kinase inhibitor LY 294002 sensitized cells to anoikis. These results suggest that Src activation may contribute to colon tumor progression and metastasis in part by activating Akt-mediated survival pathways that decrease sensitivity of detached cells to anoikis.


Subject(s)
Adenocarcinoma/pathology , Anoikis/physiology , Colonic Neoplasms/pathology , Neoplasm Proteins/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins pp60(c-src)/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Amino Acid Substitution , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Adhesion , Cell Cycle , Chromones/pharmacology , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Disease Progression , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Genes, src , Humans , Morpholines/pharmacology , Mutation, Missense , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Pyridones/pharmacology , Pyrimidines/pharmacology , Recombinant Fusion Proteins/physiology , Staurosporine/pharmacology , Transfection , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology , bcl-X Protein
7.
J Biol Chem ; 277(14): 12318-23, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11812791

ABSTRACT

Protein kinase C delta (PKC delta) is tyrosine-phosphorylated and catalytically inactive in mouse keratinocytes transformed by a ras oncogene. In several other model systems, Src kinases are upstream regulators of PKC delta. To examine this relationship in epidermal carcinogenesis, v-ras transformed mouse keratinocytes were treated with a selective Src kinase inhibitor (PD 173958). PD 173958 decreased autophosphorylation of Src, Fyn, and Lyn kinases and prevented tyrosine phosphorylation of the Src kinase substrate p120. PD 173958 also prevented PKC delta tyrosine phosphorylation and activated PKC delta as detected by membrane translocation. Expression of keratinocyte differentiation markers increased in PD 173958-treated v-ras-keratinocytes, and fluid-filled domes emerged, indicative of tight junction formation. Antisense PKC delta or bryostatin 1 inhibited dome formation, while overexpression of PKC delta in the presence of PD 173958 enhanced the formation of domes. Plasmids encoding phenylalanine mutants of PKC delta tyrosine residues 64 and 565 induced domes in the absence of PD 173958, while phenylalanine mutants of tyrosine residues 52, 155, and 187 were inactive. Thus, Src kinase mediated post-translational modification of PKC delta on specific tyrosine residues in ras-transformed mouse keratinocytes inactivates PKC delta and contributes to alterations in the differentiated phenotype and tight junction formation associated with neoplasia.


Subject(s)
Isoenzymes/metabolism , Keratinocytes/enzymology , Protein Kinase C/metabolism , Skin Neoplasms/enzymology , Tyrosine/metabolism , src-Family Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Bryostatins , Cell Differentiation , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Immunoblotting , Lactones/pharmacology , Macrolides , Mice , Mice, Inbred BALB C , Oligonucleotides, Antisense/pharmacology , Phenotype , Phenylalanine/chemistry , Phosphorylation , Precipitin Tests , Protein Binding , Protein Kinase C-delta , Pyridines/pharmacology , Pyrimidines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...