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.
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
2.
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
3.
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
4.
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
5.
J Med Chem ; 50(25): 6367-82, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-17994684

ABSTRACT

Indomethacin, a nonselective cyclooxygenase (COX) inhibitor, was modified in three distinct regions in an attempt both to increase cyclooxygenase-2 (COX-2) selectivity and to enhance drug safety by covalent attachment of an organic nitrate moiety as a nitric oxide donor. A human whole-blood COX assay shows the modifications on the 3-acetic acid part of the indomethacin yielding an amide-nitrate derivative 32 and a sulfonamide-nitrate derivative 61 conferred COX-2 selectivity. Along with their respective des-nitrate analogs, for example, 31 and 62, the nitrates 32 and 61 were effective antiinflammatory agents in the rat air-pouch model. After oral dosing, though, only 32 increased nitrate and nitrite levels in rat plasma, indicating that its nitrate tether served as a nitric oxide donor in vivo. In a rat gastric injury model, examples 31 and 32 both show a 98% reduction in gastric lesion score compared to that of indomethacin. In addition, the nitrated derivative 32 inducing 85% fewer gastric lesions when coadministered with aspirin as compared to the combination of aspirin and valdecoxib.


Subject(s)
Cyclooxygenase 2 Inhibitors/chemical synthesis , Indomethacin/analogs & derivatives , Indomethacin/chemical synthesis , Nitric Oxide Donors/chemical synthesis , Animals , Aspirin/adverse effects , Celecoxib , Cyclooxygenase 2 Inhibitors/adverse effects , Cyclooxygenase 2 Inhibitors/pharmacology , Drug Design , Drug Synergism , Female , Gastric Mucosa/pathology , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , Indomethacin/adverse effects , Indomethacin/pharmacology , Male , Nitric Oxide Donors/adverse effects , Nitric Oxide Donors/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Structure-Activity Relationship , Sulfonamides/pharmacology
6.
Free Radic Biol Med ; 39(9): 1191-207, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16214035

ABSTRACT

Nitric oxide (NO) enhances anti-inflammatory drug action. Through a metabonomics approach termed "NObonomics," the effects of a prototypic NO donor (organic nitrate)-cyclooxygenase-2 inhibitor hybrid (NO-coxib), NMI-1093, on the NO metabolite status of the circulation and major organs have been profiled in vivo in the rat. An oral anti-inflammatory NMI-1093 bolus elicited acute tissue-, time-, and dose-dependent changes in oxidative and nitroso/nitrosyl NO metabolites. Gastric N-nitrosation and hepatic S-nitrosation and heme nitrosylation emerged as sensitive indices of this NO-coxib's metabolism. Acute NMI-1093-induced nitros(yl)ation correlated positively as a function of nitrate plus nitrite formation across all organs examined, suggesting a unifying in vivo mechanism consequent to NMI-1093 biotransformation that links oxidative and nitros(yl)ative routes of NO chemical biology and thereby may support downstream NO signaling. NMI-1093 depressed erythrocyte nitros(yl)ation, likely by inhibiting cellular carbonic anhydrase and shifting the intracellular balance between nitrogen oxides and carbonates. Glutathione-S-transferase or cytochrome P450 inhibitors also attenuated NMI-1093's NO metabolism in a compartment-selective fashion. Although not itself a NO donor, the des-nitro coxib analog of NMI-1093 influenced basal NO metabolite profiles, implicating a cyclooxygenase-NO synthase interaction in physiological NO regulation. By detailing the global NO metrics of a unique coxib bearing a popular NO-donor pharmacophore (i.e., a nitrate moiety) and defining some critical mechanistic determinants, this study demonstrates how NObonomics can serve as valuable tool in helping elucidate NO systems biology and the effect of NO-donor and non-NO-donating therapeutics thereon.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Nitrates/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Nitrites/metabolism , Animals , Brain/drug effects , Brain/metabolism , Carbonic Anhydrases/drug effects , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/chemistry , Dose-Response Relationship, Drug , Erythrocytes/enzymology , Gastric Mucosa/metabolism , Heme/metabolism , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/chemistry , Oxazoles/administration & dosage , Oxazoles/chemistry , Oxazoles/pharmacology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Stomach/drug effects , Sulfonamides/administration & dosage , Sulfonamides/chemistry , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...