Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 4: e878, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24157872

ABSTRACT

Patients with advanced prostate cancer (PCa) and multiple myeloma (MM) have limited long-term responses to available therapies. The histone deacetylase inhibitor panobinostat has shown significant preclinical and clinical anticancer activity in both hematological and solid malignancies and is currently in phase III trials for relapsed MM. Bisphosphonates (BPs), such as zoledronic acid (ZOL), inhibit osteoclast-mediated bone resorption and are indicated for the treatment of bone metastasis. BPs, including ZOL, have also shown anticancer activity in several preclinical and clinical studies. In the present report, we found a potent synergistic antiproliferative effect of panobinostat/ZOL treatment in three PCa and three MM cell lines as well as in a PCa ZOL-resistant subline, independently of p53/KRAS status, androgen dependency, or the schedule of administration. The synergistic effect was also observed in an anchorage-independent agar assay in both ZOL-sensitive and ZOL-resistant cells and was confirmed in vivo in a PCa xenograft model. The co-administration of the antioxidant N-acetyl-L-cysteine blocked the increased reactive oxygen species generation and apoptosis observed in the combination setting compared with control or single-agent treatments, suggesting that oxidative injury plays a functional role in the synergism. Proapoptotic synergy was also partially antagonized by the addition of geranyl-geraniol, which bypasses the inhibition of farnesylpyrophosphate synthase by ZOL in the mevalonate pathway, supporting the involvement of this pathway in the synergy. Finally, at the molecular level, the inhibition of basal and ZOL-induced activation of p38-MAPK by panobinostat in sensitive and ZOL-resistant cells and in tumor xenografts could explain, at least in part, the observed synergism.


Subject(s)
Diphosphonates/therapeutic use , Hydroxamic Acids/therapeutic use , Imidazoles/therapeutic use , Indoles/therapeutic use , Mevalonic Acid/metabolism , Multiple Myeloma/drug therapy , Prostatic Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Diphosphonates/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Activation/drug effects , Humans , Hydroxamic Acids/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Male , Mice , Mice, Nude , Models, Biological , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Panobinostat , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Zoledronic Acid
2.
Br J Cancer ; 103(11): 1680-91, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21045833

ABSTRACT

BACKGROUND: Potentiation of anticancer activity of capecitabine is required to improve its therapeutic index. In colorectal cancer (CRC) cells, we evaluated whether the histone deacetylase-inhibitor vorinostat may induce synergistic antitumour effects in combination with capecitabine by modulating the expression of thymidine phosphorylase (TP), a key enzyme in the conversion of capecitabine to 5-florouracil (5-FU), and thymidylate synthase (TS), the target of 5-FU. METHODS: Expression of TP and TS was measured by real-time PCR, western blotting and immunohistochemistry. Knockdown of TP was performed by specific small interfering RNA. Antitumour activity of vorinostat was assessed in vitro in combination with the capecitabine active metabolite deoxy-5-fluorouridine (5'-DFUR) according to the Chou and Talay method and by evaluating apoptosis as well as in xenografts-bearing nude mice in combination with capecitabine. RESULTS: Vorinostat induced both in vitro and in vivo upregulation of TP as well as downregulation of TS in cancer cells, but not in ex vivo treated peripheral blood lymphocytes. Combined treatment with vorinostat and 5'-DFUR resulted in a synergistic antiproliferative effect and increased apoptotic cell death in vitro. This latter effect was impaired in cells where TP was knocked. In vivo, vorinostat plus capecitabine potently inhibited tumour growth, increased apoptosis and prolonged survival compared with control or single-agent treatments. CONCLUSIONS: Overall, this study suggests that the combination of vorinostat and capecitabine is an innovative antitumour strategy and warrants further clinical evaluation for the treatment of CRC.


Subject(s)
Antineoplastic Agents/pharmacology , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Thymidine Phosphorylase/genetics , Animals , Apoptosis/drug effects , Capecitabine , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Drug Synergism , Female , Floxuridine/pharmacology , Fluorouracil/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Mice , Mice, Inbred BALB C , Thymidylate Synthase/genetics , Up-Regulation , Vorinostat , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...