4.8 Article

Inhibition of acetyl-CoA carboxylase suppresses fatty acid synthesis and tumor growth of non-small-cell lung cancer in preclinical models

Journal

NATURE MEDICINE
Volume 22, Issue 10, Pages 1108-1119

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/nm.4181

Keywords

-

Funding

  1. US National Institutes of Health (NIH) [R01CA172229, P01CA120964, R01CA188652]
  2. Samuel Waxman Cancer Research Foundation
  3. Leona M. and Harry B. Helmsley Charitable Trust [2012-PG- MED002]
  4. US Department of Defense [W81-XWH-13-1-01-5]
  5. American Cancer Society (ACS) [124183-PF-13-023-01-CSM]
  6. T32 postdoctoral training grant [5 T32 CA009370, T32: 5T32CA009370-33]
  7. American Cancer Society [PF-15-037-01-DMC]
  8. F31 NIH predoctoral training fellowship [F31CA196066-01]
  9. UCSD Medical Scientist Training Program fellowship [2 T32 GM007198]
  10. T32 predoctoral training grant [T32 GM 007240]
  11. Damon Runyon Cancer Research Foundation fellowship [DRG-2219-15]
  12. Salk [CCSG P30 CA014195]
  13. next-generation sequencing (NGS) core facility at the Salk Institute - NIH-NCI [CCSG P30 014195]
  14. Chapman Foundation
  15. Glenn Foundation
  16. Helmsley Charitable Trust

Ask authors/readers for more resources

Continuous de novo fatty acid synthesis is a common feature of cancer that is required to meet the biosynthetic demands of a growing tumor. This process is controlled by the rate-limiting enzyme acetyl-CoA carboxylase (ACC), an attractive but traditionally intractable drug target. Here we provide genetic and pharmacological evidence that in preclinical models ACC is required to maintain the de novo fatty acid synthesis needed for growth and viability of non-small-cell lung cancer (NSCLC) cells. We describe the ability of ND-646-an allosteric inhibitor of the ACC enzymes ACC1 and ACC2 that prevents ACC subunit dimerization to suppress fatty acid synthesis in vitro and in vivo. Chronic ND-646 treatment of xenograft and genetically engineered mouse models of NSCLC inhibited tumor growth. When administered as a single agent or in combination with the standard-of-care drug carboplatin, ND-646 markedly suppressed lung tumor growth in the Kras;Trp53(-/-) (also known as KRAS p53) and Kras;Stk11(-/-) (also known as KRAS Lkb1) mouse models of NSCLC. These findings demonstrate that ACC mediates a metabolic liability of NSCLC and that ACC inhibition by ND-646 is detrimental to NSCLC growth, supporting further examination of the use of ACC inhibitors in oncology.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available