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 ; 12(6): 925-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536719

ABSTRACT

CUDC-101 is a novel, small-molecule, anticancer agent targeting histone deacetylase (HDAC), EGF receptor (EGFR), and HER2. It is currently in phase I clinical development in patients with solid tumors. Previously, we reported that CUDC-101 has potent antiproliferative and proapoptotic activity in cultured tumor cells and in vivo xenograft models. We now show that cancer cells that have acquired resistance to single-target EGFR inhibitors through upregulation of AXL or loss of E-cadherin remain sensitive to CUDC-101, which inhibits MET- and AXL-mediated signaling, restores E-cadherin expression, and reduces cell migration. CUDC-101 also efficiently inhibited the proliferation of MET-overexpressing non-small cell lung cancer and gastric cancer cell lines and inhibited the migration and invasion of invasive tumor cells. Taken together, these results suggest that coupling HDAC and HER2 inhibitory activities to an EGFR inhibitor may potentially be effective in overcoming drug resistance and preventing cancer cell migration.


Subject(s)
ErbB Receptors/metabolism , Histone Deacetylases/metabolism , Hydroxamic Acids/administration & dosage , Quinazolines/administration & dosage , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/administration & dosage , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-2/antagonists & inhibitors , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
2.
Clin Cancer Res ; 18(15): 4104-13, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22693356

ABSTRACT

PURPOSE: Given that histone deacetylase (HDAC) inhibitors are known to induce multiple epigenetic modifications affecting signaling networks and act synergistically with phosphatidylinositol 3-kinase (PI3K) inhibitors, we developed a strategy to simultaneously inhibit HDACs and PI3K in cancer cells. EXPERIMENTAL DESIGN: We constructed dual-acting inhibitors by incorporating HDAC inhibitory functionality into a PI3K inhibitor pharmacophore. CUDC-907, a development candidate selected from these dual inhibitors, was evaluated in vitro and in vivo to determine its pharmacologic properties, anticancer activity, and mechanism of action. RESULTS: CUDC-907 potently inhibits class I PI3Ks as well as classes I and II HDAC enzymes. Through its integrated HDAC inhibitory activity, CUDC-907 durably inhibits the PI3K-AKT-mTOR pathway and compensatory signaling molecules such as RAF, MEK, MAPK, and STAT-3, as well as upstream receptor tyrosine kinases. CUDC-907 shows greater growth inhibition and proapoptotic activity than single-target PI3K or HDAC inhibitors in both cultured and implanted cancer cells. CONCLUSIONS: CUDC-907 may offer improved therapeutic benefits through simultaneous, sustained disruption of multiple oncogenic signaling networks.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylases/metabolism , Morpholines/pharmacology , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinase/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Female , HCT116 Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Mice , Mice, Nude , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , Quinazolines/pharmacology , Sf9 Cells , Tumor Burden/drug effects , Vorinostat , Xenograft Model Antitumor Assays
3.
Cancer Res ; 70(9): 3647-56, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20388807

ABSTRACT

Receptor tyrosine kinase inhibitors have recently become important therapeutics for a variety of cancers. However, due to the heterogeneous and dynamic nature of tumors, the effectiveness of these agents is often hindered by poor response rates and acquired drug resistance. To overcome these limitations, we created a novel small molecule, CUDC-101, which simultaneously inhibits histone deacetylase and the receptor kinases epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) in cancer cells. Because of its integrated histone deacetylase inhibition, CUDC-101 synergistically blocked key regulators of EGFR/HER2 signaling pathways, also attenuating multiple compensatory pathways, such as AKT, HER3, and MET, which enable cancer cells to escape the effects of conventional EGFR/HER2 inhibitors. CUDC-101 displayed potent antiproliferative and proapoptotic activities against cultured and implanted tumor cells that are sensitive or resistant to several approved single-targeted drugs. Our results show that CUDC-101 has the potential to dramatically improve the treatment of heterogeneous and drug-resistant tumors that cannot be controlled with single-target agents. Further, they provide a framework to create individual small molecules that simultaneously antagonize multiple biochemically distinct oncogenic targets, suggesting a general paradigm to surpass conventional, single-target cancer therapeutics. Cancer Res; 70(9); 3647-56. (c)2010 AACR.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , ErbB Receptors/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Mice , Mice, Nude , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
4.
J Med Chem ; 53(5): 2000-9, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20143778

ABSTRACT

By incorporating histone deacetylase (HDAC) inhibitory functionality into the pharmacophore of the epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) inhibitors, we synthesized a novel series of compounds with potent, multiacting HDAC, EGFR, and HER2 inhibition and identified 7-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yloxy)-N-hydroxyheptanamide 8 (CUDC-101) as a drug candidate, which is now in clinical development. 8 displays potent in vitro inhibitory activity against HDAC, EGFR, and HER2 with an IC(50) of 4.4, 2.4, and 15.7 nM, respectively. In most tumor cell lines tested, 8 exhibits efficient antiproliferative activity with greater potency than vorinostat (SAHA), erlotinib, lapatinib, and combinations of vorinostat/erlotinib and vorinostat/lapatinib. In vivo, 8 promotes tumor regression or inhibition in various cancer xenograft models including nonsmall cell lung cancer (NSCLC), liver, breast, head and neck, colon, and pancreatic cancers. These results suggest that a single compound that simultaneously inhibits HDAC, EGFR, and HER2 may offer greater therapeutic benefits in cancer over single-acting agents through the interference with multiple pathways and potential synergy among HDAC and EGFR/HER2 inhibitors.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , ErbB Receptors/antagonists & inhibitors , Histone Deacetylases/metabolism , Quinazolines/chemistry , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Growth Processes/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Female , HeLa Cells , Hep G2 Cells , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/enzymology , Quinazolines/chemical synthesis , Receptor, ErbB-2/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
5.
Mol Cancer Ther ; 8(12): 3296-306, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952121

ABSTRACT

CUDC-305 is a heat shock protein 90 (HSP90) inhibitor of the novel imidazopyridine class. Here, we report its activities in non-small cell lung cancer (NSCLC) cell lines with gene deregulations conferring primary or secondary resistance to epidermal growth factor receptor (EGFR) inhibitors. We show that CUDC-305 binds strongly to HSP90 extracted from erlotinib-resistant NSCLC cells (IC50 70 nmol/L). This result correlates well with the potent antiproliferative activity in erlotinib-resistant NSCLC cell lines (IC50 120-700 nmol/L) reported previously. Furthermore, it exhibits durable inhibition of multiple oncoproteins and induction of apoptosis in erlotinib-resistant NSCLC cells. CUDC-305 potently inhibits tumor growth in subcutaneous xenograft models of H1975 and A549, which harbor EGFR T790M mutation or K-ras mutations conferring acquired and primary erlotinib resistance, respectively. In addition, CUDC-305 significantly prolongs animal survival in orthotopic lung tumor models of H1975 and A549, which may be partially attributed to its preferential exposure in lung tissue. Furthermore, CUDC-305 is able to extend animal survival in a brain metastatic model of H1975, further confirming its ability to cross the blood-brain barrier. Correlating with its effects in various tumor models, CUDC-305 induces degradation of receptor tyrosine kinases and downstream signaling molecules of the PI3K/AKT and RAF/MEK/ERK pathways simultaneously, with concurrent induction of apoptosis in vivo. In a combination study, CUDC-305 enhanced the antitumor activity of a standard-of-care agent in the H1975 tumor model. These results suggest that CUDC-305 holds promise for the treatment of NSCLC with primary or acquired resistance to EGFR inhibitor therapy.


Subject(s)
Benzodioxoles/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Imidazoles/pharmacology , Lung Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Benzodioxoles/metabolism , Benzodioxoles/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Erlotinib Hydrochloride , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Imidazoles/metabolism , Imidazoles/pharmacokinetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Signal Transduction/drug effects , Survival Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 15(12): 4046-57, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19509149

ABSTRACT

PURPOSE: We designed and synthesized CUDC-305, an HSP90 inhibitor of the novel imidazopyridine class. Here, we report its unique pharmacologic properties and antitumor activities in a variety of tumor types. EXPERIMENTAL DESIGN: The potency of the compound was analyzed by fluorescence polarization competition binding assay. Its antiproliferative activities were assessed in 40 human cancer cell lines. Its pharmacologic properties and antitumor activities were evaluated in a variety of tumor xenograft models. RESULTS: CUDC-305 shows high affinity for HSP90alpha/beta (IC(50), approximately 100 nmol/L) and HSP90 complex derived from cancer cells (IC(50), 48.8 nmol/L). It displays potent antiproliferative activity against a broad range of cancer cell lines (mean IC(50), 220 nmol/L). CUDC-305 exhibits high oral bioavailability (96.0%) and selective retention in tumor (half-life, 20.4 hours) compared with normal tissues. Furthermore, CUDC-305 can cross blood-brain barrier and reach therapeutic levels in brain tissue. CUDC-305 exhibits dose-dependent antitumor activity in an s.c. xenograft model of U87MG glioblastoma and significantly prolongs animal survival in U87MG orthotopic model. CUDC-305 also displays potent antitumor activity in animal models of erlotinib-resistant non-small cell lung cancer and induces tumor regression in animal models of MDA-MB-468 breast cancer and MV4-11 acute myelogenous leukemia. Correlating with its efficacy in these various tumor models, CUDC-305 robustly inhibits multiple signaling pathways, including PI3K/AKT and RAF/MEK/ERK, and induces apoptosis. In combination studies, CUDC-305 enhances the antitumor activity of standard-of-care agents in breast and colorectal tumor models. CONCLUSION: CUDC-305 is a promising drug candidate for the treatment of a variety of cancers, including brain malignancies.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzodioxoles/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Imidazoles/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Blood-Brain Barrier/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Nude , Mice, SCID , Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/physiology , Xenograft Model Antitumor Assays
7.
Genes Cells ; 10(6): 503-16, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15938710

ABSTRACT

Embryonic stem (ES) cells have the capacity to differentiate to every cell type that constitutes fetal or adult tissues. To trace and quantitatively assess the differentiation of ES cells into gut endodermal cells, we used an ES cell line with the lacZ gene inserted into the pdx-1 locus. Targeted mutations of pdx-1 in mice demonstrate that pdx-1 is required for pancreatic and rostral duodenal development; therefore, pdx-1 serves as an excellent early gut regional specific marker. When these ES cells were differentiated by removal of leukemia inhibitory factor (LIF), only fractional cells turned into lacZ positive, which indicates pancreatic-duodenal differentiation. Co-cultivation of ES cells with pancreatic rudiments induced a significant increase in the proportion of lacZ positive cell numbers and this increase was further enhanced by forced expression of a chick putative endoderm inducer gene, cmix. Transforming growth factor (TGF)-beta2 mimicked the effects of pancreatic rudiments and this effect was enhanced by cmix expression. Expression analysis showed over-expression of cmix induced endodermal marker genes. These data indicate that one can make use of this knowledge on molecular events of embryonic development to drive ES cells to differentiate into pdx-1 expressing endodermal cells in vitro.


Subject(s)
Endoderm/metabolism , Homeodomain Proteins/metabolism , Signal Transduction , Stem Cells/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Biomarkers , Cell Differentiation/physiology , Cell Line , Chickens , Coculture Techniques , Duodenum/cytology , Duodenum/embryology , Duodenum/metabolism , Embryo, Nonmammalian , Endoderm/cytology , Fibroblast Growth Factors/metabolism , Genetic Engineering , Homeodomain Proteins/genetics , Lac Operon , Mice , Microinjections , Models, Biological , Pancreas/cytology , Pancreas/embryology , Pancreas/metabolism , RNA/metabolism , Stem Cells/cytology , Trans-Activators/genetics , Transforming Growth Factor beta/genetics , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL