Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Reversible lysine-targeted probes reveal residence time-based kinase selectivity

Abstract

The expansion of the target landscape of covalent inhibitors requires the engagement of nucleophiles beyond cysteine. Although the conserved catalytic lysine in protein kinases is an attractive candidate for a covalent approach, selectivity remains an obvious challenge. Moreover, few covalent inhibitors have been shown to engage the kinase catalytic lysine in animals. We hypothesized that reversible, lysine-targeted inhibitors could provide sustained kinase engagement in vivo, with selectivity driven in part by differences in residence time. By strategically linking benzaldehydes to a promiscuous kinase binding scaffold, we developed chemoproteomic probes that reversibly and covalently engage >200 protein kinases in cells and mice. Probe–kinase residence time was dramatically enhanced by a hydroxyl group ortho to the aldehyde. Remarkably, only a few kinases, including Aurora A, showed sustained, quasi-irreversible occupancy in vivo, the structural basis for which was revealed by X-ray crystallography. We anticipate broad application of salicylaldehyde-based probes to proteins that lack a druggable cysteine.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Benzaldehyde probe 1 reversibly labels cellular proteins.
Fig. 2: Salicylaldehyde probes exhibit prolonged kinase residence time in cells.
Fig. 3: Quasi-irreversible engagement of AURKA in cells.
Fig. 4: Cocrystal structure of AURKA bound to salicylaldehyde 3.
Fig. 5: Quantifying kinase occupancy by salicylaldehyde 3 in mice.
Fig. 6: Salicylaldehyde probe 3 reveals PF-06873600 target engagement in vivo.

Similar content being viewed by others

Data availability

All mass spectrometry raw files have been deposited into the MassIVE database (http://massive.ucsd.edu) and can be downloaded by the identifier MSV000088924, as well as in ProteomeXchange (http://www.proteomexchange.org) with accession number PXD031899. Source data are provided in a source data file or in the Supplementary Datasets. Coordinates and structure factors have been deposited in the PDB under accession code 7FIC. Source data are provided with this paper.

Code availability

The script used for LFQ quantification is available upon request.

References

  1. Honigberg, L. A. et al. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc. Natl Acad. Sci. USA 107, 13075–13080 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Byrd, J. C. et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 369, 32–42 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Finlay, M. R. V. et al. Discovery of a potent and selective EGFR inhibitor (AZD9291) of both sensitizing and T790M resistance mutations that spares the wild type form of the receptor. J. Med. Chem. 57, 8249–8267 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Jänne, P. A. et al. AZD9291 in EGFR inhibitor–resistant non–small-cell lung cancer. N. Engl. J. Med. 372, 1689–1699 (2015).

    Article  PubMed  Google Scholar 

  5. Lanman, B. A. et al. Discovery of a covalent inhibitor of KRASG12C (AMG 510) for the treatment of solid tumors. J. Med. Chem. 63, 52–65 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Hong, D. S. et al. KRASG12C inhibition with sotorasib in advanced solid tumors. N. Engl. J. Med. 383, 1207–1217 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Singh, J., Petter, R. C., Baillie, T. A. & Whitty, A. The resurgence of covalent drugs. Nat. Rev. Drug Discov. 10, 307–317 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Liu, Q. et al. Developing irreversible inhibitors of the protein kinase cysteinome. Chem. Biol. 20, 146–159 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Chaikuad, A., Koch, P., Laufer, S. A. & Knapp, S. The cysteinome of protein kinases as a target in drug development. Angew. Chem. Int. Ed. 57, 4372–4385 (2018).

    Article  CAS  Google Scholar 

  10. Cuesta, A. & Taunton, J. Lysine-targeted inhibitors and chemoproteomic probes. Annu. Rev. Biochem. 88, 365–381 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Dalton, S. E. et al. Selectively targeting the kinome-conserved lysine of PI3Kδ as a general approach to covalent kinase inhibition. J. Am. Chem. Soc. 140, 932–939 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Gambini, L. et al. Covalent inhibitors of protein–protein interactions targeting lysine, tyrosine, or histidine residues. J. Med. Chem. 62, 5616–5627 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tamura, T. et al. Rapid labelling and covalent inhibition of intracellular native proteins using ligand-directed N-acyl-N-alkyl sulfonamide. Nat. Commun. 9, 1870 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Cuesta, A., Wan, X., Burlingame, A. L. & Taunton, J. Ligand conformational bias drives enantioselective modification of a surface-exposed lysine on Hsp90. J. Am. Chem. Soc. 142, 3392–3400 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Wan, X. et al. Discovery of lysine-targeted eIF4E inhibitors through covalent docking. J. Am. Chem. Soc. 142, 4960–4964 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Pettinger, J., Jones, K. & Cheeseman, M. D. Lysine-targeting covalent inhibitors. Angew. Chem. Int. Ed. 56, 15200–15209 (2017).

    Article  CAS  Google Scholar 

  17. Hacker, S. M. et al. Global profiling of lysine reactivity and ligandability in the human proteome. Nat. Chem. 9, 1181–1190 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Copeland, R. A. The drug–target residence time model: a 10-year retrospective. Nat. Rev. Drug Discov. 15, 87–95 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Bradshaw, J. M. et al. Prolonged and tunable residence time using reversible covalent kinase inhibitors. Nat. Chem. Biol. 11, 525–531 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Eliot, A. C. & Kirsch, J. F. Pyridoxal phosphate enzymes: mechanistic, structural, and evolutionary considerations. Annu. Rev. Biochem. 73, 383–415 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Oksenberg, D. et al. GBT440 increases haemoglobin oxygen affinity, reduces sickling and prolongs RBC half-life in a murine model of sickle cell disease. Br. J. Haematol. 175, 141–153 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Gampe, C. & Verma, V. A. Curse or cure? A perspective on the developability of aldehydes as active pharmaceutical ingredients. J. Med. Chem. 63, 14357–14381 (2020).

    Article  CAS  PubMed  Google Scholar 

  23. Gushwa, N. N., Kang, S., Chen, J. & Taunton, J. Selective targeting of distinct active site nucleophiles by irreversible Src-family kinase inhibitors. J. Am. Chem. Soc. 134, 20214–20217 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Patricelli, M. P. et al. Functional interrogation of the kinome using nucleotide acyl phosphates. Biochemistry 46, 350–358 (2007).

    Article  CAS  PubMed  Google Scholar 

  25. Zhao, Q. et al. Broad-spectrum kinase profiling in live cells with lysine-targeted sulfonyl fluoride probes. J. Am. Chem. Soc. 139, 680–685 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bell, I. M. et al. Biochemical and structural characterization of a novel class of inhibitors of the type 1 insulin-like growth factor and insulin receptor kinases. Biochemistry 44, 9430–9440 (2005).

    Article  CAS  PubMed  Google Scholar 

  27. Quach, D. et al. Strategic design of catalytic lysine-targeting reversible covalent BCR-ABL inhibitors. Angew. Chem. Int. Ed. 60, 17131–17137 (2021).

    Article  CAS  Google Scholar 

  28. Freeman-Cook, K. D. et al. Discovery of PF-06873600, a CDK2/4/6 inhibitor for the treatment of cancer. J. Med. Chem. 64, 9056–9077 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Freeman-Cook, K. et al. Expanding control of the tumor cell cycle with a CDK2/4/6 inhibitor. Cancer Cell 39, 1404–1421.e11 (2021).

    Article  CAS  PubMed  Google Scholar 

  30. Bruyneel, W., Charette, J. J. & De Hoffmann, E. Kinetics of hydrolysis of hydroxy and methoxy derivatives of N-benzylidene-2-aminopropane. J. Am. Chem. Soc. 88, 3808–3813 (1966).

    Article  CAS  Google Scholar 

  31. Herscovitch, R., Charette, J. J. & De Hoffmann, E. Physicochemical properties of Schiff bases. III. Substituent effects on the kinetics of hydrolysis of N-salicylidene-2-aminopropane derivatives. J. Am. Chem. Soc. 96, 4954–4958 (1974).

    Article  CAS  Google Scholar 

  32. Schweppe, D. K. et al. Full-featured, real-time database searching platform enables fast and accurate multiplexed quantitative proteomics. J. Proteome Res. 19, 2026–2034 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Willems, E. et al. The functional diversity of Aurora kinases: a comprehensive review. Cell Div. 13, 7 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Otto, T. et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell 15, 67–78 (2009).

    Article  CAS  PubMed  Google Scholar 

  35. Gustafson, W. C. et al. Drugging MYCN through an allosteric transition in Aurora kinase A. Cancer Cell 26, 414–427 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Richards, M. W. et al. Structural basis of N-Myc binding by Aurora-A and its destabilization by kinase inhibitors. Proc. Natl Acad. Sci. USA 113, 13726–13731 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Donnella, H. J. et al. Kinome rewiring reveals AURKA limits PI3K-pathway inhibitor efficacy in breast cancer. Nat. Chem. Biol. 14, 768–777 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Musante, V. et al. Reciprocal regulation of ARPP-16 by PKA and MAST3 kinases provides a cAMP-regulated switch in protein phosphatase 2A inhibition. eLife 6, e24998 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Spinelli, E. et al. Pathogenic MAST3 variants in the STK domain are associated with epilepsy. Ann. Neurol. 90, 274–284 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gasser, J. A. et al. SGK3 mediates INPP4B-dependent PI3K signaling in breast cancer. Mol. Cell 56, 595–607 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bago, R. et al. The hVps34-SGK3 pathway alleviates sustained PI3K/Akt inhibition by stimulating mTORC1 and tumour growth. EMBO J. 35, 2263–2263 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Vasudevan, K. M. et al. AKT-independent signaling downstream of oncogenic PIK3CA mutations in human cancer. Cancer Cell 16, 21–32 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Burgess, S. G. & Bayliss, R. The structure of C290A:C393A Aurora A provides structural insights into kinase regulation. Acta Crystallogr. Sect. F 71, 315–319 (2015).

    Article  CAS  Google Scholar 

  44. Kabsch, W. Automatic processing of rotation diffraction data from crystals of initially unknown symmetry and cell constants. J. Appl. Crystallogr. 26, 795–800 (1993).

    Article  CAS  Google Scholar 

  45. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Afonine, P. V. et al. Towards automated crystallographic structure refinement with phenix.refine. Acta Crystallogr. Sect. D 68, 352–367 (2012).

    Article  CAS  Google Scholar 

  47. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. Sect. D 66, 486–501 (2010).

    Article  CAS  Google Scholar 

  48. Lebedev, A. A. et al. JLigand: a graphical tool for the CCP4 template-restraint library. Acta Crystallogr. Sect. D 68, 431–440 (2012).

    Article  CAS  Google Scholar 

  49. Murshudov, G. N., Vagin, A. A. & Dodson, E. J. Refinement of macromolecular structures by the maximum-likelihood method. Acta Crystallogr. Sect. D 53, 240–255 (1997).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Funding for this study was provided by the National Cancer Institute (NCI) (NIH NCI F31CA214028, A.C.), Ono Pharma Foundation (J.T.) and Pfizer. Mass spectrometry was supported in part by the University of California, San Francisco (UCSF) Program for Breakthrough Biomedical Research and the Adelson Medical Research Foundation (A.L.B.).

Author information

Authors and Affiliations

Authors

Contributions

T.Y. and J.T. conceived the project, designed the experiments and analyzed the data. T.Y. synthesized the aldehyde probes, evaluated the probes in biochemical and chemoproteomics experiments, and acquired and analyzed LFQ and TMT proteomics data. A.C. acquired and analyzed LFQ proteomics data. X.W. crystallized and determined the structure of AURKA–probe 3. G.B.C. refined the X-ray structure. B.H., P.K. and J.R.M. performed mouse pharmacology experiments. J.D.C. analyzed TMT proteomics data. T.Y. and J.T. wrote the manuscript with input from all of the authors. J.C.K., J.D.L., S.N. and A.L.B. edited the manuscript.

Corresponding author

Correspondence to Jack Taunton.

Ethics declarations

Competing interests

T.Y., B.H., P.K., J.R.M., J.C.K., J.D.L., S.N. and J.D.C. are current or former employees of Pfizer. J.T. is a founder of Global Blood Therapeutics, Principia Biopharma, Kezar Life Sciences, Cedilla Therapeutics and Terremoto Biosciences, and is a scientific advisor to Entos.

Peer review

Peer review information

Nature Chemical Biology thanks Benjamin Cravatt and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Reversible protein labeling by benzaldehyde probe 2.

(a) Chemical structures of 1 and 2. (b) Jurkat cells were treated with 1 or 2 (2 μM, 30 min), followed by compound washout for the indicated times. Cells were lysed in the presence of 25 mM sodium cyanoborohydride, except as indicated (#). After copper-promoted click conjugation with TAMRA-azide, samples were analyzed by in-gel fluorescence and Coomassie blue staining. Data are representative of two independent experiments.

Source data

Extended Data Fig. 2 Probe 3 modification of overexpressed Src.

(a) COS-7 cells were transfected with WT or K295Q (#) Flag-Src, or not transfected (*), and then treated with the indicated concentrations of probe 3 (30 min). After lysis in the presence of sodium borohydride and TAMRA-azide conjugation, samples were analyzed by in-gel fluorescence and western blotting. Data are representative of two independent experiments. (b) Concentration-dependent labeling of Flag-Src (n = 2, mean values from two independently performed experiments were plotted).

Source data

Extended Data Fig. 3 Rapid dissociation of probe 3 from overexpressed Src.

(a) COS-7 cells were transfected with Flag-Src, or not transfected (*), and treated with probe 3 (2 μM, 30 min), followed by washout for the indicated times. After lysis in the presence of sodium borohydride and TAMRA-azide conjugation, samples were analyzed by in-gel fluorescence and western blotting. (b) Normalized fluorescence intensity of ~65 kDa band corresponding to Flag-Src (mean ± SD, n = 3).

Source data

Extended Data Fig. 4 Dissociation of probe 3 from recombinant AURKA and Src.

AURKA or Src (5 μM) was treated with probe 3 (5.1 μM) in 50 mM HEPES, pH 8.0 at RT for 5 min, followed by 20-fold dilution into buffer containing 10 μM XO44. The percentage of XO44-modified and unmodified kinase was quantified by LC-MS at the indicated time points, and % unmodified kinase (corresponding to probe 3-bound kinase) was plotted vs. time (n = 2, mean values from two independently performed experiments were plotted).

Extended Data Fig. 5 Related to Fig. 5.

Volcano plot showing log2 fold-change and significance (−log10 p-value; two tailed t-test assuming unequal variance) of proteins enriched by probe 3 (t = 3 h post-dose) vs. vehicle. Red, kinases; gray, non-kinases.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2, Supplementary Tables 1 and 2, and Supplementary Note.

Reporting Summary

Supplementary Datasets 1–12

Chemoproteomics dataset.

Source data

Source Data Fig. 1

Uncropped gel.

Source Data Fig. 2

Uncropped gels.

Source Data Fig. 3

Uncropped gels and western blots.

Source Data Extended Data Fig. 1

Uncropped gel.

Source Data Extended Data Fig. 2

Uncropped gel and blots.

Source Data Extended Data Fig. 3

Uncropped gel and blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, T., Cuesta, A., Wan, X. et al. Reversible lysine-targeted probes reveal residence time-based kinase selectivity. Nat Chem Biol 18, 934–941 (2022). https://doi.org/10.1038/s41589-022-01019-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01019-1

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research