Your browser doesn't support javascript.
loading
Radiation Drives the Evolution of Orthotopic Xenografts Initiated from Glioblastoma Stem-like Cells.
McAbee, Joseph H; Rath, Barbara H; Valdez, Kristin; Young, Dejauwne L; Wu, Xiaolin; Shankavaram, Uma T; Camphausen, Kevin; Tofilon, Philip J.
Afiliação
  • McAbee JH; Radiation Oncology Branch, NCI, Bethesda, Maryland.
  • Rath BH; Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.
  • Valdez K; Wake Forest University School of Medicine, Winston-Salem, North Carolina.
  • Young DL; Radiation Oncology Branch, NCI, Bethesda, Maryland.
  • Wu X; Radiation Oncology Branch, NCI, Bethesda, Maryland.
  • Shankavaram UT; Radiation Oncology Branch, NCI, Bethesda, Maryland.
  • Camphausen K; Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland.
  • Tofilon PJ; Radiation Oncology Branch, NCI, Bethesda, Maryland.
Cancer Res ; 79(23): 6032-6043, 2019 12 01.
Article em En | MEDLINE | ID: mdl-31615806
ABSTRACT
A consequence of the intratumor heterogeneity (ITH) of glioblastoma (GBM) is the susceptibility to treatment-driven evolution. To determine the potential of radiotherapy to influence GBM evolution, we used orthotopic xenografts initiated from CD133+ GBM stem-like cells (GSC). Toward this end, orthotopic xenografts grown in nude mice were exposed to a fractionated radiation protocol, which resulted in a significant increase in animal survival. Brain tumors from control and irradiated mice were then collected at morbidity and compared in terms of growth pattern, clonal diversity, and genomic architecture. In mice that received fractionated radiation, tumors were less invasive, with more clearly demarcated borders and tumor core hypercellularity as compared with controls, suggesting a fundamental change in tumor biology. Viral integration site analysis indicated a reduction in clonal diversity in the irradiated tumors, implying a decrease in ITH. Changes in clonal diversity were not detected after irradiation of GSCs in vitro, suggesting that the radiation-induced reduction in ITH was dependent on the brain microenvironment. Whole-exome sequencing revealed differences in mutation patterns between control and irradiated tumors, which included modifications in the presence and clonality of driver mutations associated with GBM. Moreover, changes in the distribution of mutations as a function of subpopulation size between control and irradiated tumors were consistent with subclone expansion and contraction, that is, subpopulation evolution. Taken together, these results indicate that radiation drives the evolution of the GSC-initiated orthotopic xenografts and suggest that radiation-driven evolution may have therapeutic implications for recurrent GBM.

SIGNIFICANCE:

Radiation drives the evolution of glioblastoma orthotopic xenografts; when translated to the clinic, this may have therapeutic implications for recurrent tumors.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Neoplasias Encefálicas / Glioblastoma / Heterogeneidade Genética / Evolução Molecular Tipo de estudo: Guideline Limite: Animals / Female / Humans Idioma: En Revista: Cancer Res Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Neoplasias Encefálicas / Glioblastoma / Heterogeneidade Genética / Evolução Molecular Tipo de estudo: Guideline Limite: Animals / Female / Humans Idioma: En Revista: Cancer Res Ano de publicação: 2019 Tipo de documento: Article