Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
FASEB J ; 31(12): 5453-5465, 2017 12.
Article in English | MEDLINE | ID: mdl-28821631

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading form of cancer worldwide, and its incidence is increasing rapidly in the United States, tripling over the past 3 decades. The current chemotherapeutic strategies against localized and metastatic HCC are ineffective. Here we report that 6-methoxyethylamino-numonafide (MEAN) is a potent growth inhibitor of murine xenografts of 2 human HCC cell lines. At the same dose and with the same treatment strategies, MEAN was more efficacious in inhibiting tumor growth in mice than sorafenib, the only approved drug for HCC. Treatment by MEAN at an effective dose for 6 wk was well tolerated by animals. Combined therapy using both sorafenib and MEAN enhanced tumor growth inhibition over monotherapy with either agent. Additional experiments revealed that MEAN inhibited tumor growth through mechanisms distinct from those of either its parent compound, amonafide, or sorafenib. MEAN suppressed C-MYC expression and increased expression of several tumor suppressor genes, including Src homology region 2 domain-containing phosphatase-1 (SHP-1) and TXNIP (thioredoxin-interacting protein). As an encouraging feature for envisioned clinical application, the IC50 of MEAN was not significantly changed in several drug-resistant cell lines with activated P-glycoprotein drug efflux pumps compared to drug-sensitive parent cells, demonstrating the ability of MEAN to be effective in cells resistant to existing chemotherapy regimens. MEAN is a promising candidate for clinical development as a single-agent therapy or in combination with sorafenib for the management of HCC.-Liu, Y., Lou, G., Norton, J. T., Wang, C., Kandela, I., Tang, S., Shank, N. I., Gupta, P., Huang, M., Avram, M. J., Green, R., Mazar, A., Appella, D., Chen, Z., Huang, S. 6-Methoxyethylamino-numonafide inhibits hepatocellular carcinoma xenograft growth as a single agent and in combination with sorafenib.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Naphthalimides/therapeutic use , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Alanine Transaminase/metabolism , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/metabolism , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Male , Mice , Niacinamide/therapeutic use , Sorafenib , Xenograft Model Antitumor Assays
2.
J Med Chem ; 58(14): 5637-48, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26098096

ABSTRACT

Activin belongs to the TGFß superfamily, which is associated with several disease conditions, including cancer-related cachexia, preterm labor with delivery, and osteoporosis. Targeting activin and its related signaling pathways holds promise as a therapeutic approach to these diseases. A small-molecule ligand-binding groove was identified in the interface between the two activin ßA subunits and was used for a virtual high-throughput in silico screening of the ZINC database to identify hits. Thirty-nine compounds without significant toxicity were tested in two well-established activin assays: FSHß transcription and HepG2 cell apoptosis. This screening workflow resulted in two lead compounds: NUCC-474 and NUCC-555. These potential activin antagonists were then shown to inhibit activin A-mediated cell proliferation in ex vivo ovary cultures. In vivo testing showed that our most potent compound (NUCC-555) caused a dose-dependent decrease in FSH levels in ovariectomized mice. The Blitz competition binding assay confirmed target binding of NUCC-555 to the activin A:ActRII that disrupts the activin A:ActRII complex's binding with ALK4-ECD-Fc in a dose-dependent manner. The NUCC-555 also specifically binds to activin A compared with other TGFß superfamily member myostatin (GDF8). These data demonstrate a new in silico-based strategy for identifying small-molecule activin antagonists. Our approach is the first to identify a first-in-class small-molecule antagonist of activin binding to ALK4, which opens a completely new approach to inhibiting the activity of TGFß receptor superfamily members. in addition, the lead compound can serve as a starting point for lead optimization toward the goal of a compound that may be effective in activin-mediated diseases.


Subject(s)
Activins/antagonists & inhibitors , High-Throughput Screening Assays , User-Computer Interface , Activins/chemistry , Activins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Evaluation, Preclinical , Female , Follicle Stimulating Hormone/antagonists & inhibitors , Hep G2 Cells , Humans , Mice , Molecular Docking Simulation , Ovary/cytology , Ovary/drug effects , Protein Conformation , Signal Transduction/drug effects
3.
Free Radic Biol Med ; 60: 157-67, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23416365

ABSTRACT

Chemoresistance due to oxidative stress resistance or upregulation of Bcl-2 contributes to poor outcome in the treatment of hematological malignancies. In this study, we utilize the copper-chelator drug ATN-224 (choline tetrathiomolybdate) to induce cell death in oxidative stress-resistant cells and cells overexpressing Bcl-2 by modulating the cellular redox environment and causing mitochondrial dysfunction. ATN-224 treatment decreases superoxide dismutase 1 (SOD1) activity, increases intracellular oxidants, and induces peroxynitrite-dependent cell death. ATN-224 also targets the mitochondria, decreasing both cytochrome c oxidase (CcOX) activity and mitochondrial membrane potential. The concentration of ATN-224 required to induce cell death is proportional to SOD1 levels, but independent of Bcl-2 status. In combination with doxorubicin, ATN-224 enhances cell death. In primary B-cell acute lymphoblastic leukemia patient samples, ATN-224 decreases the viable cell number. Our findings suggest that ATN-224's dual targeting of SOD1 and CcOX is a promising approach for treatment of hematological malignancies either as an adjuvant or as a single agent.


Subject(s)
Chelating Agents/administration & dosage , Hematologic Neoplasms/drug therapy , Molybdenum/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , B-Lymphocytes/cytology , Cell Death/drug effects , Cell Survival/drug effects , Copper/chemistry , Copper/metabolism , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/physiopathology , Humans , Mice , Oxidative Stress/drug effects , Peroxynitrous Acid/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Primary Cell Culture , Proto-Oncogene Proteins c-bcl-2 , Stress, Physiological/drug effects , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , U937 Cells
4.
Clin Cancer Res ; 14(22): 7526-34, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010871

ABSTRACT

PURPOSE: Copper chelation reduces the secretion of many angiogenic factors and reduces tumor growth and microvascular density in animal models. ATN-224 is a second-generation analogue of ammonium tetrathiomolybdate. The aim of our phase I study was to reduce serum copper levels, as measured by ceruloplasmin, to 5 to 15 mg/dL (normal 16-60) in 14 to 21 days, to determine the pharmacokinetic profile of ATN-224 and to evaluate dose-limiting toxicities. PATIENTS AND METHODS: Cohorts of patients were treated with escalating oral doses of ATN-224 until copper depletion followed by a titrated maintenance dose. RESULTS: Eighteen patients received 78 cycles of ATN-224. Mean baseline ceruloplasmin was 39.6 mg/dL. The maximum administered dose was 330 mg/d where grade 3 fatigue was dose-limiting. At the maximum tolerated dose of 300 mg/d, the median time to achieve target ceruloplasmin was 21 days, and toxicities included grade 3 anemia, grade 3 neutropenia, fatigue, and sulfur eructation. ATN-224 treatment caused a significant reduction (> 90%) in RBC superoxide dismutase 1 activity and circulating endothelial cells. Pharmacokinetic data indicate greater absorption of ATN-224 and more rapid ceruloplasmin reduction when administered with a proton pump inhibitor. Stable disease of > 6 months was observed in 2 patients. CONCLUSIONS: Oral ATN-224 is a well-tolerated therapy and at a loading dose of 300 mg/d leads to a reduction of serum ceruloplasmin levels in 80% patients within 21 days. A loading dose of 300 mg/d for 2 weeks followed by a titrated maintenance dose will be the recommended starting dose for phase II study.


Subject(s)
Chelating Agents/adverse effects , Chelating Agents/pharmacokinetics , Chelating Agents/therapeutic use , Chelation Therapy , Choline/adverse effects , Choline/pharmacokinetics , Choline/therapeutic use , Copper/blood , Molybdenum/adverse effects , Molybdenum/pharmacokinetics , Molybdenum/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Ceruloplasmin/drug effects , Cytokines/blood , Cytokines/drug effects , Endothelial Cells/drug effects , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Superoxide Dismutase/blood , Superoxide Dismutase/drug effects
5.
Proc Natl Acad Sci U S A ; 99(19): 12224-9, 2002 Sep 17.
Article in English | MEDLINE | ID: mdl-12196635

ABSTRACT

Conformationally altered proteins and protein fragments derived from the extracellular matrix and hemostatic system may function as naturally occurring angiogenesis inhibitors. One example of such a protein is cleaved high molecular weight kininogen (HKa). HKa inhibits angiogenesis by inducing apoptosis of proliferating endothelial cells, effects mediated largely by HKa domain 5. However, the mechanisms underlying the antiangiogenic activity of HKa have not been characterized, and its binding site on proliferating endothelial cells has not been defined. Here, we report that the induction of endothelial cell apoptosis by HKa, as well as the antiangiogenic activity of HKa in the chick chorioallantoic membrane, was inhibited completely by antitropomyosin monoclonal antibody TM-311. TM-311 also blocked the high-affinity Zn2+-dependent binding of HKa to both purified tropomyosin and proliferating endothelial cells. Confocal microscopic analysis of endothelial cells stained with monoclonal antibody TM-311, as well as biotin labeling of cell surface proteins on intact endothelial cells, revealed that tropomyosin exposure was enhanced on the surface of proliferating cells. These studies demonstrate that the antiangiogenic effects of HKa depend on high-affinity binding to endothelial cell tropomyosin.


Subject(s)
Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Kininogen, High-Molecular-Weight/metabolism , Kininogen, High-Molecular-Weight/pharmacology , Tropomyosin/metabolism , Allantois/blood supply , Allantois/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Base Sequence , Cells, Cultured , Chick Embryo , Chorion/blood supply , Chorion/drug effects , DNA, Complementary/genetics , Endothelium, Vascular/cytology , Humans , Kininogen, High-Molecular-Weight/genetics , Neovascularization, Physiologic/drug effects , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tropomyosin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL