Your browser doesn't support javascript.
loading
ATR inhibition using gartisertib enhances cell death and synergises with temozolomide and radiation in patient-derived glioblastoma cell lines.
Lozinski, Mathew; Bowden, Nikola A; Graves, Moira C; Fay, Michael; Day, Bryan W; Stringer, Brett W; Tooney, Paul A.
Afiliación
  • Lozinski M; School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia.
  • Bowden NA; Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
  • Graves MC; Mark Hughes Foundation Centre for Brain Cancer Research, University of Newcastle, NSW, Australia.
  • Fay M; School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia.
  • Day BW; Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
  • Stringer BW; School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, NSW, Australia.
  • Tooney PA; Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
Oncotarget ; 15: 1-18, 2024 Jan 16.
Article en En | MEDLINE | ID: mdl-38227740
ABSTRACT
Glioblastoma cells can restrict the DNA-damaging effects of temozolomide (TMZ) and radiation therapy (RT) using the DNA damage response (DDR) mechanism which activates cell cycle arrest and DNA repair pathways. Ataxia-telangiectasia and Rad3-Related protein (ATR) plays a pivotal role in the recognition of DNA damage induced by chemotherapy and radiation causing downstream DDR activation. Here, we investigated the activity of gartisertib, a potent ATR inhibitor, alone and in combination with TMZ and/or RT in 12 patient-derived glioblastoma cell lines. We showed that gartisertib alone potently reduced the cell viability of glioblastoma cell lines, where sensitivity was associated with the frequency of DDR mutations and higher expression of the G2 cell cycle pathway. ATR inhibition significantly enhanced cell death in combination with TMZ and RT and was shown to have higher synergy than TMZ+RT treatment. MGMT promoter unmethylated and TMZ+RT resistant glioblastoma cells were also more sensitive to gartisertib. Analysis of gene expression from gartisertib treated glioblastoma cells identified the upregulation of innate immune-related pathways. Overall, this study identifies ATR inhibition as a strategy to enhance the DNA-damaging ability of glioblastoma standard treatment, while providing preliminary evidence that ATR inhibition induces an innate immune gene signature that warrants further investigation.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Neoplasias Encefálicas / Glioblastoma Límite: Humans Idioma: En Revista: Oncotarget Año: 2024 Tipo del documento: Article País de afiliación: Australia

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Neoplasias Encefálicas / Glioblastoma Límite: Humans Idioma: En Revista: Oncotarget Año: 2024 Tipo del documento: Article País de afiliación: Australia