Inhibition of DNA double-strand break repair by the dual PI3K/mTOR inhibitor NVP-BEZ235 as a strategy for radiosensitization of glioblastoma

Clin Cancer Res. 2014 Mar 1;20(5):1235-48. doi: 10.1158/1078-0432.CCR-13-1607. Epub 2013 Dec 23.

Abstract

Purpose: Inhibitors of the DNA damage response (DDR) have great potential for radiosensitization of numerous cancers, including glioblastomas, which are extremely radio- and chemoresistant brain tumors. Currently, there are no DNA double-strand break (DSB) repair inhibitors that have been successful in treating glioblastoma. Our laboratory previously demonstrated that the dual phosphoinositide 3-kinase/mTOR inhibitor NVP-BEZ235 can potently inhibit the two central DDR kinases, DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and ataxia-telangiectasia mutated (ATM), in vitro. Here, we tested whether NVP-BEZ235 could also inhibit ATM and DNA-PKcs in tumors in vivo and assessed its potential as a radio- and chemosensitizer in preclinical mouse glioblastoma models.

Experimental design: The radiosensitizing effect of NVP-BEZ235 was tested by following tumor growth in subcutaneous and orthotopic glioblastoma models. Tumors were generated using the radioresistant U87-vIII glioma cell line and GBM9 neurospheres in nude mice. These tumors were then treated with ionizing radiation and/or NVP-BEZ235 and analyzed for DNA-PKcs and ATM activation, DSB repair inhibition, and attenuation of growth.

Results: NVP-BEZ235 potently inhibited both DNA-PKcs and ATM kinases and attenuated the repair of ionizing radiation-induced DNA damage in tumors. This resulted in striking tumor radiosensitization, which extended the survival of brain tumor-bearing mice. Notably, tumors displayed a higher DSB-load when compared with normal brain tissue. NVP-BEZ235 also sensitized a subset of subcutaneous tumors to temozolomide, a drug routinely used concurrently with ionizing radiation for the treatment of glioblastoma.

Conclusions: These results demonstrate that it may be possible to significantly improve glioblastoma therapy by combining ionizing radiation with potent and bioavailable DNA repair inhibitors such as NVP-BEZ235.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Ataxia Telangiectasia Mutated Proteins / antagonists & inhibitors
  • Ataxia Telangiectasia Mutated Proteins / metabolism
  • Blood-Brain Barrier / metabolism
  • Catalytic Domain
  • Cell Line, Tumor
  • DNA Breaks, Double-Stranded / drug effects*
  • DNA Repair / drug effects*
  • DNA-Activated Protein Kinase / antagonists & inhibitors
  • DNA-Activated Protein Kinase / chemistry
  • DNA-Activated Protein Kinase / metabolism
  • Dacarbazine / analogs & derivatives
  • Dacarbazine / pharmacology
  • Disease Models, Animal
  • Glioblastoma / drug therapy
  • Glioblastoma / genetics*
  • Glioblastoma / metabolism*
  • Glioblastoma / mortality
  • Glioblastoma / pathology
  • Glioblastoma / radiotherapy
  • Humans
  • Imidazoles / pharmacology*
  • Mice
  • Mice, Transgenic
  • Phosphoinositide-3 Kinase Inhibitors*
  • Protein Interaction Domains and Motifs / drug effects
  • Quinolines / pharmacology*
  • Radiation-Sensitizing Agents / pharmacology*
  • TOR Serine-Threonine Kinases / antagonists & inhibitors*
  • Temozolomide

Substances

  • Imidazoles
  • Phosphoinositide-3 Kinase Inhibitors
  • Quinolines
  • Radiation-Sensitizing Agents
  • Dacarbazine
  • Ataxia Telangiectasia Mutated Proteins
  • DNA-Activated Protein Kinase
  • TOR Serine-Threonine Kinases
  • dactolisib
  • Temozolomide