4.8 Article

Stereospecific targeting of MTH1 by (S)-crizotinib as an anticancer strategy

Journal

NATURE
Volume 508, Issue 7495, Pages 222-+

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/nature13194

Keywords

-

Funding

  1. Austrian Academy of Sciences
  2. GEN-AU initiative of the Austrian Federal Ministry for Science and Research
  3. European Union
  4. SGC, a registered charity [1097737]
  5. Canadian Institutes for Health Research
  6. Canada Foundation for Innovation
  7. Genome Canada
  8. GlaxoSmithKline
  9. Pfizer
  10. Eli Lilly
  11. Takeda
  12. AbbVie
  13. Novartis Research Foundation
  14. Boehringer Ingelheim
  15. Ontario Ministry of Research and Innovation
  16. Wellcome Trust [092809/Z/10/Z]
  17. European Union [278568]
  18. Torsten and Ragnar Soderberg Foundation
  19. Knut and Alice Wallenberg Foundation
  20. Swedish Research Council
  21. European Research Council
  22. Swedish Cancer Society
  23. European Union FP7 Career Integration Grant [PCIG11-GA-2012-321602]
  24. FWF Grant [P24766-B20]
  25. Austrian Science Fund (FWF) [P24766] Funding Source: Austrian Science Fund (FWF)

Ask authors/readers for more resources

Activated RAS GTPase signalling is a critical driver of oncogenic transformation and malignant disease. Cellular models of RAS-dependent cancers have been used to identify experimental small molecules, such as SCH51344, but their molecular mechanism of action remains generally unknown. Here, using a chemical proteomic approach, we identify the target of SCH51344 as the human mutT homologue MTH1 (also known as NUDT1), a nucleotide pool sanitizing enzyme. Loss-of-function of MTH1 impaired growth of KRAS tumour cells, whereas MTH1 overexpression mitigated sensitivity towards SCH51344. Searching for more drug-like inhibitors, we identified the kinase inhibitor crizotinib as a nanomolar suppressor of MTH1 activity. Surprisingly, the clinically used (R)-enantiomer of the drug was inactive, whereas the (S)-enantiomer selectively inhibited MTH1 catalytic activity. Enzymatic assays, chemical proteomic profiling, kinome-wide activity surveys and MTH1 co-crystal structures of both enantiomers provide a rationale for this remarkable stereospecificity. Disruption of nucleotide pool homeostasis via MTH1 inhibition by (S)-crizotinib induced an increase in DNA single-strand breaks, activated DNA repair in human colon carcinoma cells, and effectively suppressed tumour growth in animal models. Our results propose (S)-crizotinib as an attractive chemical entity for further pre-clinical evaluation, and small-molecule inhibitors of MTH1 in general as a promising novel class of anticancer agents.

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