Your browser doesn't support javascript.
loading
RAD51 inhibition in triple negative breast cancer cells is challenged by compensatory survival signaling and requires rational combination therapy.
Wiegmans, Adrian P; Miranda, Mariska; Wen, Shu Wen; Al-Ejeh, Fares; Möller, Andreas.
Afiliação
  • Wiegmans AP; Tumor Microenvironment Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia.
  • Miranda M; Personalized Medicine Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia.
  • Wen SW; Tumor Microenvironment Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia.
  • Al-Ejeh F; Personalized Medicine Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia.
  • Möller A; Tumor Microenvironment Laboratory, QIMR Berghofer, Herston Rd, Herston QLD 4006, Australia.
Oncotarget ; 7(37): 60087-60100, 2016 09 13.
Article em En | MEDLINE | ID: mdl-27507046
ABSTRACT
The molecular rationale to induce synthetic lethality, by targeting defective homologous recombination repair in triple negative breast cancer (TNBC), has proven to have several shortcomings. Not meeting the expected minimal outcomes in clinical trials has highlighted common clinical resistance mechanisms including; increased expression of the target gene PARP1, increased expression or reversion mutation of BRCA1, or up-regulation of the compensatory homologous recombination protein RAD51. Indeed, RAD51 has been demonstrated to be an alternative synthetic lethal target in BRCA1-mutated cancers. To overcome selective pressure on DNA repair pathways, we examined new potential targets within TNBC that demonstrate synthetic lethality in association with RAD51 depletion. We confirmed complementary targets of PARP1/2 and DNA-PK as well as a new synthetic lethality combination with p38. p38 is considered a relevant target in breast cancer, as it has been implicated in resistance to chemotherapy, including tamoxifen. We show that the combination of targeting RAD51 and p38 inhibits cell proliferation both in vitro and in vivo, which was further enhanced by targeting of PARP1. Analysis of the molecular mechanisms revealed that depletion of RAD51 increased ERK1/2 and p38 signaling. Our results highlight a potential compensatory mechanism via p38 that limits DNA targeted therapy.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transdução de Sinais / Proteína Quinase 14 Ativada por Mitógeno / Rad51 Recombinase / Neoplasias de Mama Triplo Negativas Limite: Animals / Humans Idioma: En Revista: Oncotarget Ano de publicação: 2016 Tipo de documento: Article País de afiliação: Austrália

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transdução de Sinais / Proteína Quinase 14 Ativada por Mitógeno / Rad51 Recombinase / Neoplasias de Mama Triplo Negativas Limite: Animals / Humans Idioma: En Revista: Oncotarget Ano de publicação: 2016 Tipo de documento: Article País de afiliação: Austrália