4.6 Article

Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance

Journal

MOLECULAR CANCER THERAPEUTICS
Volume 19, Issue 5, Pages 1110-1122

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/1535-7163.MCT-19-0745

Keywords

-

Categories

Funding

  1. Cancer Research Society
  2. Institute of Cancer Research of the Canadian Institutes of Health Research (CIHR) [22185]
  3. Natural Sciences and Engineering Research Council of Canada [RGPIN-2013-436204]
  4. CGS-D award from the CIHR
  5. Nova Scotia Health Research Foundation studentship
  6. NS graduate scholarship
  7. Killam Laureate scholarship
  8. Nova Scotia Research and Innovation Graduate and Killam Laureate scholarships
  9. CGS-D award from CIHR
  10. DeWolfe Graduate Award from the Dalhousie Medical Research Foundation
  11. Beatrice Hunter Cancer Research Institute
  12. Canadian Imperial Bank of Commerce

Ask authors/readers for more resources

Dysregulation of DNA methylation is an established feature of breast cancers. DNA demethylating therapies like decitabine are proposed for the treatment of triple-negative breast cancers (TNBC) and indicators of response need to be identified. For this purpose, we characterized the effects of decitabine in a panel of 10 breast cancer cell lines and observed a range of sensitivity to decitabine that was not subtype specific. Knockdown of potential key effectors demonstrated the requirement of deoxycytidine kinase (DCK) for decitabine response in breast cancer cells. In treatment-naive breast tumors, DCK was higher in TNBCs, and DCK levels were sustained or increased post chemotherapy treatment. This suggests that limited DCK levels will not be a barrier to response in patients with TNBC treated with decitabine as a second-line treatment or in a clinical trial. Methylome analysis revealed that genome-wide, region-specific, tumor suppressor gene-specific methylation, and decitabine-induced demethylation did not predict response to decitabine. Gene set enrichment analysis of transcriptome data demonstrated that decitabine induced genes within apoptosis, cell cycle, stress, and immune pathways. Induced genes included those characterized by the viral mimicry response; however, knockdown of key effectors of the pathway did not affect decitabine sensitivity suggesting that breast cancer growth suppression by decitabine is independent of viral mimicry. Finally, taxol-resistant breast cancer cells expressing high levels of multidrug resistance transporter ABCB1 remained sensitive to decitabine, suggesting that the drug could be used as second-line treatment for chemoresistant patients.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available