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.
Clin Cancer Res ; 16(1): 141-53, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20028765

ABSTRACT

PURPOSE: The mammalian target of rapamycin (mTOR) inhibitor temsirolimus has exhibited promising anticancer activity for the treatment of renal cell cancers (RCC). Survivin expression has been implicated in drug resistance and reducing its levels with the histone deacetylase (HDAC) inhibitor vorinostat may enhance the anticancer activity of temsirolimus. EXPERIMENTAL DESIGN: The sensitivity of RCC cell lines to the combination of temsirolimus and vorinostat was determined by measuring cell viability, clonogenic survival, and apoptosis. The effects of this combination on survivin levels were determined in vitro and in vivo. Survivin expression was silenced using small interfering RNA to evaluate its role in determining sensitivity to temsirolimus and vorinostat. The effect of the combination on angiogenesis was also determined in RCC xenograft models. RESULTS: Vorinostat synergistically improved the anticancer activity of temsirolimus in a panel of RCC cell lines in vitro and in two xenograft models in vivo. While each single agent led to a modest decrease in survivin levels, the combination dramatically reduced its expression, which correlated with an induction of apoptosis. Silencing survivin levels induced apoptosis and significantly improved the efficacy of temsirolimus and vorinostat. In addition, the temsirolimus/vorinostat combination led to a strong reduction in angiogenesis. CONCLUSIONS: Vorinostat augmented the anticancer activity of temsirolimus in both in vitro and in vivo models of RCC. The effectiveness of the combination was due to a decrease in survivin levels and corresponding induction of apoptosis, and enhanced inhibition of angiogenesis. Targeting survivin may be a promising therapeutic strategy to improve RCC therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/drug therapy , Hydroxamic Acids/pharmacology , Kidney Neoplasms/drug therapy , Microtubule-Associated Proteins/metabolism , Sirolimus/analogs & derivatives , Angiogenesis Inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Drug Synergism , Humans , Inhibitor of Apoptosis Proteins , Mice , Mice, Nude , Repressor Proteins , Sirolimus/therapeutic use , Survivin , Vorinostat , Xenograft Model Antitumor Assays
2.
J Cell Mol Med ; 14(10): 2448-59, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19583815

ABSTRACT

Autophagy is an evolutionarily conserved cell survival pathway that enables cells to recoup ATP and other critical biosynthetic molecules during nutrient deprivation or exposure to hypoxia, which are hallmarks of the tumour microenvironment. Autophagy has been implicated as a potential mechanism of resistance to anticancer agents as it can promote cell survival in the face of stress induced by chemotherapeutic agents by breaking down cellular components to generate alternative sources of energy. Disruption of autophagy with chloroquine (CQ) induces the accumulation of ubiquitin-conjugated proteins in a manner similar to the proteasome inhibitor bortezomib (BZ). However, CQ-induced protein accumulation occurs at a slower rate and is localized to lysosomes in contrast to BZ, which stimulates rapid buildup of ubiquitinated proteins and aggresome formation in the cytosol. The histone deacetylase (HDAC) inhibitor vorinostat (VOR) blocked BZ-induced aggresome formation, but promoted CQ-mediated ubiquitinated protein accumulation. Disruption of autophagy with CQ strongly enhanced VOR-mediated apoptosis in colon cancer cells. Accordingly, knockdown of the essential autophagy gene Atg7 also sensitized cells to VOR-induced apoptosis. Knockdown of HDAC6 greatly enhanced BZ-induced apoptosis, but only marginally sensitized cells to CQ. Subsequent studies determined that the CQ/VOR combination promoted a large increase in superoxide generation that was required for ubiquitinated protein accumulation and cell death. Finally, treatment with the CQ/VOR combination significantly reduced tumour burden and induced apoptosis in a colon cancer xenograft model. Collectively, our results establish that inhibition of autophagy with CQ induces ubiquitinated protein accumulation and VOR potentiates CQ-mediated aggregate formation, superoxide generation and apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Autophagy , Hydroxamic Acids/pharmacology , Ubiquitinated Proteins/metabolism , Animals , Antineoplastic Agents/metabolism , Boronic Acids/pharmacology , Bortezomib , Carcinoma/drug therapy , Carcinoma/metabolism , Cell Line, Tumor , Cell Survival , Chloroquine/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Female , HT29 Cells , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylases/metabolism , Humans , Lysosomes/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , Superoxides/metabolism , Vorinostat , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...