Your browser doesn't support javascript.
loading
ATR inhibition preferentially targets homologous recombination-deficient tumor cells.
Krajewska, M; Fehrmann, R S N; Schoonen, P M; Labib, S; de Vries, E G E; Franke, L; van Vugt, M A T M.
Afiliação
  • Krajewska M; Department of Medical Onclology, University Medical Center Groningen, University of Groningen, The Netherlands.
  • Fehrmann RS; Department of Medical Onclology, University Medical Center Groningen, University of Groningen, The Netherlands.
  • Schoonen PM; Department of Medical Onclology, University Medical Center Groningen, University of Groningen, The Netherlands.
  • Labib S; Department of Medical Onclology, University Medical Center Groningen, University of Groningen, The Netherlands.
  • de Vries EG; Department of Medical Onclology, University Medical Center Groningen, University of Groningen, The Netherlands.
  • Franke L; Department of Medical Genetics, University Medical Center Groningen, University of Groningen, The Netherlands.
  • van Vugt MA; Department of Medical Onclology, University Medical Center Groningen, University of Groningen, The Netherlands.
Oncogene ; 34(26): 3474-81, 2015 Jun.
Article em En | MEDLINE | ID: mdl-25174396
ABSTRACT
Homologous recombination (HR) is required for faithful repair of double-strand DNA breaks. Defects in HR repair cause severe genomic instability and challenge cellular viability. Paradoxically, various cancers are HR defective and have apparently acquired characteristics to survive genomic instability. We aimed to identify these characteristics to uncover therapeutic targets for HR-deficient cancers. Cytogenetic analysis of 1143 ovarian cancers showed that the degree of genomic instability was correlated to amplification of replication checkpoint genes ataxia telangiectasia and Rad3-related kinase (ATR) and CHEK1. To test whether genomic instability leads to increased reliance on replication checkpoint signaling, we inactivated Rad51 to model HR-related genomic instability. Rad51 inactivation caused defective HR repair and induced aberrant replication dynamics. Notably, inhibition of Rad51 led to increased ATR/checkpoint kinase-1 (Chk1)-mediated replication stress signaling. Importantly, inhibition of ATR or Chk1 preferentially killed HR-deficient cancer cells. Combined, our data show that defective HR caused by Rad51 inhibition results in differential sensitivity for ATR and Chk1 inhibitors, implicating replication checkpoint kinases as potential drug targets for HR-defective cancers.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Pirazinas / Sulfonas / Recombinação Homóloga / Neoplasias / Antineoplásicos Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Oncogene Assunto da revista: BIOLOGIA MOLECULAR / NEOPLASIAS Ano de publicação: 2015 Tipo de documento: Article País de afiliação: Holanda

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Pirazinas / Sulfonas / Recombinação Homóloga / Neoplasias / Antineoplásicos Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Oncogene Assunto da revista: BIOLOGIA MOLECULAR / NEOPLASIAS Ano de publicação: 2015 Tipo de documento: Article País de afiliação: Holanda