1. Academic Validation
  2. Stereospecific targeting of MTH1 by (S)-crizotinib as an anticancer strategy

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

  • Nature. 2014 Apr 10;508(7495):222-7. doi: 10.1038/nature13194.
Kilian V M Huber 1 Eidarus Salah 2 Branka Radic 1 Manuela Gridling 1 Jonathan M Elkins 2 Alexey Stukalov 1 Ann-Sofie Jemth 3 Camilla Göktürk 3 Kumar Sanjiv 3 Kia Strömberg 3 Therese Pham 3 Ulrika Warpman Berglund 3 Jacques Colinge 1 Keiryn L Bennett 1 Joanna I Loizou 1 Thomas Helleday 3 Stefan Knapp 2 Giulio Superti-Furga 1
Affiliations

Affiliations

  • 1 CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria.
  • 2 Nuffield Department of Clinical Medicine, Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK.
  • 3 Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17121 Stockholm, Sweden.
Abstract

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.

Figures