NVP-BEZ235, a novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor, elicits multifaceted antitumor activities in human gliomas

TJ Liu, D Koul, T LaFortune, N Tiao, RJ Shen… - Molecular cancer …, 2009 - AACR
TJ Liu, D Koul, T LaFortune, N Tiao, RJ Shen, SM Maira, C Garcia-Echevrria, WKA Yung
Molecular cancer therapeutics, 2009AACR
Aberrant genetic alternations in human gliomas, such as amplification of epidermal growth
factor receptor, mutation and/or deletion of tumor suppressor gene PTEN, and mutations of
PIK3CA, contribute to constitutive activation of the phosphatidylinositol 3-kinase (PI3K)
pathway. We investigated the potential antitumor activity of NVP-BEZ235, which is a novel
dual PI3K/mammalian target of rapamycin (mTOR) inhibitor in gliomas. The compound
suppressed glioma cell proliferation with IC50 values in the low nanomolar range by …
Abstract
Aberrant genetic alternations in human gliomas, such as amplification of epidermal growth factor receptor, mutation and/or deletion of tumor suppressor gene PTEN, and mutations of PIK3CA, contribute to constitutive activation of the phosphatidylinositol 3-kinase (PI3K) pathway. We investigated the potential antitumor activity of NVP-BEZ235, which is a novel dual PI3K/mammalian target of rapamycin (mTOR) inhibitor in gliomas. The compound suppressed glioma cell proliferation with IC50 values in the low nanomolar range by specifically inhibiting the activity of target proteins including Akt, S6K1, S6, and 4EBP1 in the PI3K/Akt/mTOR signaling pathway. NVP-BEZ235 treatment of glioma cell lines led to G1 cell cycle arrest and induced autophagy. Furthermore, expression of the vascular endothelial growth factor (VEGF), which is an important angiogenic modulator in glioma cells, was significantly decreased, suggesting that NVP-BEZ235 may also exert an antiangiogenic effect. Preclinical testing of the therapeutic efficacy of NVP-BEZ235 showed that it significantly prolonged the survival of tumor-bearing animals without causing any obvious toxicity. Tumor extracts harvested from animals after treatment showed that the compound inhibited the activity of target proteins in the PI3K/Akt/mTOR cascade. Immunohistochemical analyses also showed a significant reduction in staining for VEGF von Willebrand factor (factor VIII) in NVP-BEZ235–treated tumor sections compared with controls, further confirming that NVP-BEZ235 has an antiangiogenic effect in vivo. We conclude from these findings that NVP-BEZ235 antagonizes PI3K and mTOR signaling and induces cell cycle arrest, down-regulation of VEGF, and autophagy. These results warrant further development of NVP-BEZ235 for clinical trials for human gliomas or other advanced cancers with altered PI3K/Akt/mTOR signaling. [Mol Cancer Ther 2009;8(8):2204–10]
AACR