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










Publication year range
1.
Int J Cancer ; 124(6): 1449-56, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19065668

ABSTRACT

CDC25 phosphatases are key actors in cyclin-dependent kinases activation whose role is essential at various stages of the cell cycle. CDC25 expression is upregulated in a number of human cancers. CDC25 phosphatases are therefore thought to represent promising novel targets in cancer therapy. Here, we report the identification and the characterization of IRC-083864, an original bis-quinone moiety that is a potent and selective inhibitor of CDC25 phosphatases in the low nanomolar range. IRC-083864 inhibits cell proliferation of a number of cell lines, regardless of their resistance to other drugs. It irreversibly inhibits cell proliferation and cell cycle progression and prevents entry into mitosis. In addition, it inhibits the growth of HCT-116 tumor spheroids with induction of p21 and apoptosis. Finally, IRC-083864 reduced tumor growth in mice with established human prostatic and pancreatic tumor xenografts. This study describes a novel compound, which merits further study as a potential anticancer agent.


Subject(s)
Benzothiazoles/therapeutic use , Benzoxazoles/therapeutic use , Enzyme Inhibitors/therapeutic use , Quinones/therapeutic use , cdc25 Phosphatases/antagonists & inhibitors , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Cyclin-Dependent Kinases/metabolism , Flow Cytometry , Humans , Mice , Mice, Nude , Transplantation, Heterologous
2.
Mol Cancer Ther ; 6(1): 318-25, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17237290

ABSTRACT

The CDC25 cell cycle regulators are promising targets for new pharmacologic approaches in cancer therapy. Inhibitory compounds such as BN82685 have proven to be effective in specifically targeting CDC25 in cultured cells and in inhibiting tumor cell growth. Here, we report that BN82685 impairs microtubule dynamic instability and alters microtubule organization and assembly at the centrosome in interphase cells. Treatment of mitotic cells with BN82685 delays mitotic spindle assembly, chromosome capture, and metaphase plate formation. Furthermore, we show that combining low concentrations of both BN82685 and paclitaxel inhibits the proliferation of HT29 human colon cancer cells. Our results show a role for CDC25 phosphatases in regulating microtubule dynamics throughout the cell cycle and suggest that combinations of CDC25 inhibitors with microtubule-targeting agents may be of therapeutic value.


Subject(s)
Benzoquinones/pharmacology , Enzyme Inhibitors/pharmacology , Interphase/drug effects , Microtubules/drug effects , Spindle Apparatus/drug effects , Thiazoles/pharmacology , cdc25 Phosphatases/antagonists & inhibitors , Chromosomes, Human/drug effects , Drug Synergism , HT29 Cells , HeLa Cells , Humans , Metaphase/drug effects , Prometaphase/drug effects
3.
Mol Cancer Ther ; 5(6): 1446-51, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16818502

ABSTRACT

Cell cycle arrest at the G2-M checkpoint is an essential feature of the mechanisms that preserve genomic integrity. CDC25 phosphatases control cell cycle progression by dephosphorylating and activating cyclin-dependent kinase/cyclin complexes. Their activities are, therefore, tightly regulated to modulate cell cycle arrest in response to DNA damage exposure. Here, we report that overexpression of CDC25B affects viability, reduces clonogenic efficiency, and increases sensitivity of cancer cells to a genotoxic agent. We show that ectopic expression of CDC25B results in bypass of a genotoxic-induced G2-M checkpoint. In addition, cancer cells constitutively expressing high level of CDC25B are shown to be prone to exit prematurely from the G2-M checkpoint arrest and to enter mitosis. Finally, we show that this exit is dependent on CDC25B expression. Together with previous results, our data strongly support a model in which CDC25B is the key phosphatase that controls entry into mitosis after DNA damage, thus emphasizing the relevance of its overexpression in many human tumors.


Subject(s)
Cell Cycle Proteins/metabolism , cdc25 Phosphatases/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Cell Cycle Proteins/genetics , Cell Division/drug effects , Cell Survival/drug effects , Cell Survival/physiology , DNA Damage/drug effects , DNA Damage/physiology , Etoposide/pharmacology , Flow Cytometry , Fluorescent Antibody Technique, Indirect , G2 Phase/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Tetracycline/pharmacology , Tumor Cells, Cultured , Tumor Stem Cell Assay , cdc25 Phosphatases/genetics
4.
Bioorg Med Chem Lett ; 16(1): 171-5, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16216500

ABSTRACT

A focused set of heterocyclic quinones based on the benzothiazole, benzoxazole, benzimidazole, indazole and isoindole was prepared and screened with respect to the inhibition of the phosphatase activity of CDC25C. Benzoxazole- and benzothiazole-diones were at least 50 times more potent in inhibiting CDC25C than their benzimidazole-indazole- or isoindole-dione counterparts. These in vitro activities were in good correlation with the anti-proliferative effects observed with Mia PaCa-2 and DU-145 human tumor cell cultures. The IC(50) values obtained by WST-1 colorimetric assay ranged from 0.10 to 0.50 microM for the benzoxazole- or benzothiazole-diones and were above 10 microM for the other heterocyclic diones. This study further illustrates how the activity of the quinone pharmacophore can be selectively modulated by changing the type of five-membered heterocycle fused to the quinone ring.


Subject(s)
Cell Cycle Proteins/chemistry , Chemistry, Pharmaceutical/methods , cdc25 Phosphatases/chemistry , Benzothiazoles/chemistry , Benzoxazoles/chemistry , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Colorimetry , Drug Design , Ethylenediamines/chemistry , Humans , Inhibitory Concentration 50 , Models, Chemical , Quinones/chemistry , Recombinant Proteins/chemistry , Structure-Activity Relationship , Temperature , Time Factors , cdc25 Phosphatases/metabolism
5.
Mol Cancer Ther ; 4(9): 1378-87, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16170030

ABSTRACT

Cell cycle regulators, such as the CDC25 phosphatases, are potential targets for the development of new anticancer drugs. Here we report the identification and the characterization of BN82685, a quinone-based CDC25 inhibitor that is active in vitro and in vivo. BN82685 inhibits recombinant CDC25A, B, and C phosphatases in vitro. It inhibits the growth of human tumor cell lines with an IC(50) in the submicromolar range, independently of their resistance to chemotherapeutic agents. This inhibitory effect is irreversible on both the purified CDC25 enzyme in vitro and on tumor cell proliferation. The specificity of BN82685 towards the CDC25 phosphatases is shown by an increase in cyclin-dependent kinase 1 tyrosine 15 phosphorylation, by the reversion of the mitosis-inducing effect of CDC25B overexpression in HeLa cells, and by the lack of a growth inhibitory effect in an assay based on the use of a CDC25-independent fission yeast model. Finally, when administered p.o., BN82685 is shown to inhibit the growth of the human pancreatic tumor Mia PaCa-2 xenografted in athymic nude mice. BN82685 is therefore a promising new compound targeting CDC25, which confirms the interest of the inhibition of these enzymes as an anticancer therapeutic strategy.


Subject(s)
Benzoquinones/pharmacology , Enzyme Inhibitors/pharmacology , Pancreatic Neoplasms/pathology , Thiazoles/pharmacology , cdc25 Phosphatases/antagonists & inhibitors , Administration, Oral , Animals , Benzoquinones/administration & dosage , Benzoquinones/chemical synthesis , Biological Availability , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Female , HeLa Cells , Humans , Mice , Mice, Nude , Mitosis/drug effects , Pancreatic Neoplasms/enzymology , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces/metabolism , Thiazoles/administration & dosage , Thiazoles/chemical synthesis , Transplantation, Heterologous , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem Lett ; 14(23): 5809-12, 2004 Dec 06.
Article in English | MEDLINE | ID: mdl-15501045

ABSTRACT

A targeted library of small molecules has been prepared to optimize the biological activity of BN82002, our initial lead compound, recently described as an original inhibitor of CDC25 phosphatases. Some of these compounds inhibit CDC25 in the micromolar range and therefore reinforce the interest of CDC25 as an anticancer target.


Subject(s)
Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , cdc25 Phosphatases/antagonists & inhibitors , Humans , cdc25 Phosphatases/metabolism
7.
Cancer Res ; 64(9): 3320-5, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15126376

ABSTRACT

CDC25 dual-specificity phosphatases are essential regulators that dephosphorylate and activate cyclin-dependent kinase/cyclin complexes at key transitions of the cell cycle. CDC25 activity is currently considered to be an interesting target for the development of new antiproliferative agents. Here we report the identification of a new CDC25 inhibitor and the characterization of its effects at the molecular and cellular levels, and in animal models. BN82002 inhibits the phosphatase activity of recombinant human CDC25A, B, and C in vitro. It impairs the proliferation of tumoral cell lines and increases cyclin-dependent kinase 1 inhibitory tyrosine phosphorylation. In synchronized HeLa cells, BN82002 delays cell cycle progression at G1-S, in S phase and at the G2-M transition. In contrast, BN82002 arrests U2OS cell cycle mostly in the G1 phase. Selectivity of this inhibitor is demonstrated: (a) by the reversion of the mitotic-inducing effect observed in HeLa cells upon CDC25B overexpression; and (b) by the partial reversion of cell cycle arrest in U2OS expressing CDC25. We also show that BN82002 reduces growth rate of human tumor xenografts in athymic nude mice. BN82002 is a original CDC25 inhibitor that is active both in cell and animal models. This greatly reinforces the interest in CDC25 as an anticancer target.


Subject(s)
Enzyme Inhibitors/pharmacology , cdc25 Phosphatases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Division/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Ethylamines , Female , HeLa Cells , Humans , Mice , Mice, Nude , Mitosis/drug effects , Nitro Compounds , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays , cdc25 Phosphatases/biosynthesis , cdc25 Phosphatases/genetics
8.
Curr Genet ; 45(5): 283-8, 2004 May.
Article in English | MEDLINE | ID: mdl-14727060

ABSTRACT

Fission yeast is a simple eukaryotic model organism in which many aspects of cell cycle control can be explored. We examined by homologous recombination whether the human CDC25A phosphatase could substitute for the function of the fission yeast Cdc25. We first show: (a). that CDC25A efficiently replaces the endogenous Cdc25 mitotic inducer for vegetative growth and (b). that CDC25A is able to partially restore a functional checkpoint in response to both ionising and UV irradiation, but not a DNA replication checkpoint. We then describe a simple assay in which we demonstrate that growth of the humanised CDC25A strain is strongly repressed in a CDC25-dependent manner by BN2003, a potent chemical inhibitor of CDC25 belonging to the benzothiazoledione family. The ease of manipulation of fission yeast humanised CDC25 cells and the simplicity of the above assay offer a powerful tool with which to investigate the specificity of pharmacological inhibitors of CDC25.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fungal Proteins/antagonists & inhibitors , Schizosaccharomyces/metabolism , cdc25 Phosphatases/chemistry , cdc25 Phosphatases/metabolism , ras-GRF1/antagonists & inhibitors , Benzothiazoles , Cell Proliferation , DNA/metabolism , DNA Damage , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Genotype , Humans , Hydroxyurea/pharmacology , Infrared Rays , Inhibitory Concentration 50 , Mitosis , Models, Genetic , Phosphoric Monoester Hydrolases/chemistry , Plasmids/metabolism , Protein Isoforms , Temperature , Thiazoles/chemistry , Time Factors , Ultraviolet Rays
9.
Prog Cell Cycle Res ; 5: 225-34, 2003.
Article in English | MEDLINE | ID: mdl-14593717

ABSTRACT

As essential cell cycle regulators, the CDC25 phosphatases are currently considered as potential targets for the development of novel therapeutic approaches. Here, we review the function and regulation of CDC25 phosphatases, their involvement in cancer and Alzheimer's disease, and the properties of several recently identified inhibitors.


Subject(s)
Alzheimer Disease/enzymology , Cell Cycle/physiology , Enzyme Inhibitors/pharmacology , Neoplasms/enzymology , cdc25 Phosphatases/antagonists & inhibitors , Alzheimer Disease/drug therapy , Animals , Cell Cycle/drug effects , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/trends , Enzyme Inhibitors/therapeutic use , Humans , Molecular Structure , Neoplasms/drug therapy , cdc25 Phosphatases/metabolism
10.
Biochem Pharmacol ; 65(3): 423-33, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12527335

ABSTRACT

Epstein-Barr virus (EBV)-associated nasopharyngeal carcinomas (NPC) are much more sensitive to chemotherapy than other head and neck carcinomas. Spectacular regressions are frequently observed after induction chemotherapy. However, these favorable responses are difficult to predict and often of short duration. So far there have been only few experiments to investigate the mechanisms which underline the cytotoxic effects of anti-neoplastic drugs against NPC cells. In addition, these studies were performed almost entirely on EBV-negative cell lines therefore not truly representative of NPC cells. For the first time, we have used two EBV-positive NPC tumor lines derived from a North African (C15) and a Chinese (C666-1) patient as in vitro targets for a panel of anti-neoplastic agents. Doxorubicin, taxol and in a lesser extent cis-platinum efficiently inhibited NPC cell proliferation at clinically relevant concentrations, but all three agents failed to induce apoptosis. However, massive apoptosis of C15 cells was achieved when doxorubicin (1 microM) was combined with a farnesyl-transferase inhibitor, BIM 2001 (5 microM). Moreover, this apoptotic process was associated with a caspase-dependent early cleavage of the TNF-receptor associated factor 1 (TRAF-1) molecule, a signaling adaptor which is specifically expressed in latently EBV-infected cells. TRAF-1 cleavage might become a useful indicator of chemo-induced apoptosis in EBV-associated NPCs.


Subject(s)
Apoptosis , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Nasopharyngeal Neoplasms/pathology , Nitriles/pharmacology , Proteins/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Cell Division/drug effects , Drug Combinations , Farnesyltranstransferase , Female , Herpesvirus 4, Human/isolation & purification , Humans , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/virology , TNF Receptor-Associated Factor 1 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...