4.5 Article

KDM1A inhibition is effective in reducing stemness and treating triple negative breast cancer

Journal

BREAST CANCER RESEARCH AND TREATMENT
Volume 185, Issue 2, Pages 343-357

Publisher

SPRINGER
DOI: 10.1007/s10549-020-05963-1

Keywords

KDM1A; LSD1; Triple negative breast cancer; Cancer stem cells; STAT3; Epithelial mesenchymal transition

Categories

Funding

  1. Bank of America Foundation, Max and Minnie Tomerlin Voelcker Fund
  2. Elsa U. Pardee Foundation Grant
  3. Mays Cancer Center Support Grant [P30CA054174-17]
  4. NIH Shared Instrument Grant [1S10OD021805-01]
  5. CPRIT Core Facility Award [RP160732]
  6. UT Health San Antonio
  7. Mays Cancer Center P30 Cancer Center Support Grant [NIH-NCI P30 CA054174]
  8. Mays Cancer Center P30 Cancer Center Support Grant (National Center for Advancing Translational Sciences, NIH) [UL1 TR002645]
  9. [NIH-CA223828]
  10. [NIH-T32GM113896]

Ask authors/readers for more resources

Cancer stem cells (CSCs) in TNBC express high levels of KDM1A, and inhibition of KDM1A reduces CSC enrichment and tumor growth. KDM1A inhibitor NCD38 shows potential as a novel drug for treating TNBC by targeting STAT3 and EMT signaling pathways.
Purpose Cancer stem cells (CSCs) are highly tumorigenic, spared by chemotherapy, sustain tumor growth, and are implicated in tumor recurrence after conventional therapies in triple negative breast cancer (TNBC). Lysine-specific histone demethylase 1A (KDM1A) is highly expressed in several human malignancies and CSCs including TNBC. However, the precise mechanistic role of KDM1A in CSC functions and therapeutic utility of KDM1A inhibitor for treating TNBC is poorly understood. Methods The effect of KDM1A inhibition on cell viability, apoptosis, and invasion were examined by Cell Titer Glo, Caspase 3/7 Glo, and matrigel invasion assays, respectively. Stemness and self-renewal of CSCs were examined using mammosphere formation and extreme limiting dilution assays. Mechanistic studies were conducted using RNA-sequencing, RT-qPCR, Western blotting and reporter gene assays. Mouse xenograft and patient derived xenograft models were used for preclinical evaluation of KDM1A inhibitor. Results TCGA data sets indicated that KDM1A is highly expressed in TNBC. CSCs express high levels of KDM1A and inhibition of KDM1A reduced the CSCs enrichment in TNBC cells. KDM1A inhibition reduced cell viability, mammosphere formation, self-renewal and promoted apoptosis of CSCs. Mechanistic studies suggested that IL6-JAK-STAT3 and EMT pathways were downregulated in KDM1A knockdown and KDM1A inhibitor treated cells. Importantly, doxycycline inducible knockout of KDM1A reduced tumor progression in orthotopic xenograft models and KDM1A inhibitor NCD38 treatment significantly reduced tumor growth in patient derived xenograft (PDX) models. Conclusions Our results establish that KDM1A inhibition mitigates CSCs functions via inhibition of STAT3 and EMT signaling, and KDM1A inhibitor NCD38 may represent a novel class of drug for treating TNBC.

Authors

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

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available