Your browser doesn't support javascript.
loading
p53 Loss in MYC-Driven Neuroblastoma Leads to Metabolic Adaptations Supporting Radioresistance.
Yogev, Orli; Barker, Karen; Sikka, Arti; Almeida, Gilberto S; Hallsworth, Albert; Smith, Laura M; Jamin, Yann; Ruddle, Ruth; Koers, Alexander; Webber, Hannah T; Raynaud, Florence I; Popov, Sergey; Jones, Chris; Petrie, Kevin; Robinson, Simon P; Keun, Hector C; Chesler, Louis.
Afiliación
  • Yogev O; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.
  • Barker K; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.
  • Sikka A; Department of Surgery and Cancer, Imperial College, London, United Kingdom.
  • Almeida GS; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom. Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom.
  • Hallsworth A; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.
  • Smith LM; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.
  • Jamin Y; Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom.
  • Ruddle R; Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom.
  • Koers A; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.
  • Webber HT; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.
  • Raynaud FI; Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom.
  • Popov S; Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom. Department of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
  • Jones C; Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom. Department of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
  • Petrie K; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.
  • Robinson SP; Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom.
  • Keun HC; Department of Surgery and Cancer, Imperial College, London, United Kingdom.
  • Chesler L; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom. louis.chesler@icr.ac.uk.
Cancer Res ; 76(10): 3025-35, 2016 05 15.
Article en En | MEDLINE | ID: mdl-27197232
ABSTRACT
Neuroblastoma is the most common childhood extracranial solid tumor. In high-risk cases, many of which are characterized by amplification of MYCN, outcome remains poor. Mutations in the p53 (TP53) tumor suppressor are rare at diagnosis, but evidence suggests that p53 function is often impaired in relapsed, treatment-resistant disease. To address the role of p53 loss of function in the development and pathogenesis of high-risk neuroblastoma, we generated a MYCN-driven genetically engineered mouse model in which the tamoxifen-inducible p53ER(TAM) fusion protein was expressed from a knock-in allele (Th-MYCN/Trp53(KI)). We observed no significant differences in tumor-free survival between Th-MYCN mice heterozygous for Trp53(KI) (n = 188) and Th-MYCN mice with wild-type p53 (n = 101). Conversely, the survival of Th-MYCN/Trp53(KI/KI) mice lacking functional p53 (n = 60) was greatly reduced. We found that Th-MYCN/Trp53(KI/KI) tumors were resistant to ionizing radiation (IR), as expected. However, restoration of functional p53ER(TAM) reinstated sensitivity to IR in only 50% of Th-MYCN/Trp53(KI/KI) tumors, indicating the acquisition of additional resistance mechanisms. Gene expression and metabolic analyses indicated that the principal acquired mechanism of resistance to IR in the absence of functional p53 was metabolic adaptation in response to chronic oxidative stress. Tumors exhibited increased antioxidant metabolites and upregulation of glutathione S-transferase pathway genes, including Gstp1 and Gstz1, which are associated with poor outcome in human neuroblastoma. Accordingly, glutathione depletion by buthionine sulfoximine together with restoration of p53 activity resensitized tumors to IR. Our findings highlight the complex pathways operating in relapsed neuroblastomas and the need for combination therapies that target the diverse resistance mechanisms at play. Cancer Res; 76(10); 3025-35. ©2016 AACR.
Asunto(s)

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Tolerancia a Radiación / Adaptación Fisiológica / Proteína p53 Supresora de Tumor / Apoptosis / Proteína Proto-Oncogénica N-Myc / Neuroblastoma Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans / Male Idioma: En Revista: Cancer Res Año: 2016 Tipo del documento: Article País de afiliación: Reino Unido

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Tolerancia a Radiación / Adaptación Fisiológica / Proteína p53 Supresora de Tumor / Apoptosis / Proteína Proto-Oncogénica N-Myc / Neuroblastoma Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans / Male Idioma: En Revista: Cancer Res Año: 2016 Tipo del documento: Article País de afiliación: Reino Unido