4.6 Article

Atovaquone is active against AML by upregulating the integrated stress pathway and suppressing oxidative phosphorylation

Journal

BLOOD ADVANCES
Volume 3, Issue 24, Pages 4215-4227

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/bloodadvances.2019000499

Keywords

-

Categories

Funding

  1. National Institutes of Health (NIH
  2. National Institute of Allergy and Infectious Diseases) [P30 AI036211]
  3. National Institutes of Health (NIH
  4. National Cancer Institute [NCI]) [P30 CA125123]
  5. National Institutes of Health (NIH
  6. National Center for Research Resources) [S10 RR024574]
  7. NIH (National Institute of Diabetes and Digestive and Kidney Diseases) [RO1DK114356]
  8. NIH (National Human Genome Research Institute) [UM1HG006348]
  9. NCI, NIH [U24CA114766, R01CA17026, R01CA160979]
  10. Texas Children's Hospital
  11. CURE Childhood Cancer
  12. Brent Leahey Fund
  13. European Union [826121]
  14. Robert A. Welch Foundation [C-1680]
  15. National Institute of General Medical Sciences, NIH [T32GM007753, F30 CA165740-01]
  16. Turn it Gold Fund
  17. National Science Foundation [CHE1055569]

Ask authors/readers for more resources

Atovaquone, a US Food and Drug Administration-approved antiparasitic drug previously shown to reduce interleukin-6/STAT3 signaling in myeloma cells, is well tolerated, and plasma concentrations of 40 to 80 mu M have been achieved with pediatric and adult dosing. We conducted preclinical testing of atovaquone with acute myeloid leukemia (AML) cell lines and pediatric patient samples. Atovaquone induced apoptosis with an EC50<30 mu M for most AML lines and primary pediatric AML specimens. In NSG mice xenografted with luciferase-expressing THP-1 cells and in those receiving a patient-derived xenograft, atovaquone-treated mice demonstrated decreased disease burden and prolonged survival. To gain a better understanding of the mechanism of atovaquone, we performed an integrated analysis of gene expression changes occurring in cancer cell lines after atovaquone exposure. Atovaquone promoted phosphorylation of eIF2 alpha, a key component of the integrated stress response and master regulator of protein translation. Increased levels of phosphorylated eIF2 alpha led to greater abundance of the transcription factor ATF4 and its target genes, including proapoptotic CHOP and CHAC1. Furthermore, atovaquone upregulated REDD1, an ATF4 target gene and negative regulator of the mechanistic target of rapamycin (mTOR), and caused REDD1-mediated inhibition of mTOR activity with similar efficacy as rapamycin. Additionally, atovaquone suppressed the oxygen consumption rate of AML cells, which has specific implications for chemotherapy-resistant AML blasts that rely on oxidative phosphorylation for survival. Our results provide insight into the complex a biological effects of atovaquone, highlighting its potential as an anticancer therapy with novel and diverse mechanisms of action, and support further clinical evaluation of atovaquone for pediatric and adult AML.

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