Item request has been placed! ×
Item request cannot be made. ×
loading  Processing Request

Active-site mTOR inhibitors augment HSV1-dICP0 infection in cancer cells via dysregulated eIF4E/4E-BP axis

Item request has been placed! ×
Item request cannot be made. ×
loading   Processing Request
  • معلومة اضافية
    • Contributors:
      McGill University = Université McGill [Montréal, Canada]; University of Ottawa [Ottawa]; Institut Armand Frappier (INRS-IAF); Réseau International des Instituts Pasteur (RIIP)-Institut National de la Recherche Scientifique [Québec] (INRS); McMaster University [Hamilton, Ontario]; Ottawa Hospital Research Institute [Ottawa] (OHRI); This research was funded by grants from the Terry Fox Research Institute to NS and TA, and the CHEO Foundation, the Cancer Research Society / Stephen E. Drabin Research Fund, the Brain Tumour Foundation of Canada, and the Canadian Breast Cancer Foundation to TA. The funders had no role in study design, data collection and analysis, Decision to publish, or preparation of the manuscript; We thank Isabelle Harvey and Lynn Kyte for technical assistance, Xu Zhang and Sebastian Morales for help with experiments, and William Muller (McGill) for the mouse mammary epithelial cells.
    • الموضوع:
      2017
    • نبذة مختصرة :
      International audience; Herpes Simplex Virus 1 (HSV1) is amongst the most clinically advanced oncolytic virus platforms. However, efficient and sustained viral replication within tumours is limiting. Rapamycin can stimulate HSV1 replication in cancer cells, but active-site dual mTORC1 and mTORC2 (mammalian target of rapamycin complex 1 and 2) inhibitors (asTORi) were shown to suppress the virus in normal cells. Surprisingly, using the infected cell protein 0 (ICP0)-deleted HSV1 (HSV1-dICP0), we found that asTORi markedly augment infection in cancer cells and a mouse mammary cancer xenograft. Mechanistically, asTORi repressed mRNA translation in normal cells, resulting in defective antiviral response but also inhibition of HSV1-dICP0 replication. asTORi also reduced antiviral response in cancer cells, however in contrast to normal cells, transformed cells and cells transduced to elevate the expression of eukaryotic initiation factor 4E (eIF4E) or to silence the repressors eIF4E binding proteins (4E-BPs), selectively maintained HSV1-dICP0 protein synthesis during asTORi treatment, ultimately supporting increased viral replication. Our data show that altered eIF4E/4E-BPs expression can act to promote HSV1-dICP0 infection under prolonged mTOR inhibition. Thus, pharmacoviral combination of asTORi and HSV1 can target cancer cells displaying dysregulated eIF4E/4E-BPs axis.
    • File Description:
      application/pdf
    • ISSN:
      1553-7374
      1553-7366
    • Rights:
      OPEN
    • الرقم المعرف:
      edsair.doi.dedup.....26c92dee5a0216e1929b4920ce8f0aa3