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:

Discovery of a selective inhibitor of doublecortin like kinase 1

Abstract

Doublecortin like kinase 1 (DCLK1) is an understudied kinase that is upregulated in a wide range of cancers, including pancreatic ductal adenocarcinoma (PDAC). However, little is known about its potential as a therapeutic target. We used chemoproteomic profiling and structure-based design to develop a selective, in vivo-compatible chemical probe of the DCLK1 kinase domain, DCLK1-IN-1. We demonstrate activity of DCLK1-IN-1 against clinically relevant patient-derived PDAC organoid models and use a combination of RNA-sequencing, proteomics and phosphoproteomics analysis to reveal that DCLK1 inhibition modulates proteins and pathways associated with cell motility in this context. DCLK1-IN-1 will serve as a versatile tool to investigate DCLK1 biology and establish its role in cancer.

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: DCLK1-IN-1 is a potent DCLK1/2 inhibitor.
Fig. 2: DCLK1-IN-1 engages DCLK1 potently and selectively in cells.
Fig. 3: DCLK1 is a target of aberrant KRAS-ERK signaling and is dispensable in DCLK1+ PDAC cell lines.
Fig. 4: DCLK1 is a vulnerability in DCLK1+ patient-derived organoids.

Similar content being viewed by others

Data availability

KINOMEscan and KiNativ data are provided in Supplementary Datasets 1 and 2. Cell line RNA-sequencing data has been deposited to the NCBI GEO (accession number GSE140490). Cell line and deidentified patient-derived organoid RNA-sequencing analyzed data files are provided in Supplementary Dataset 3. Cell line and deidentified patient-derived organoid mass spectrometry-based proteomics and phosphoproteomics analyzed data files are provided in Supplementary Datasets 4 and 5.

References

  1. Fedorov, O., Muller, S. & Knapp, S. The (un)targeted cancer kinome. Nat. Chem. Biol. 6, 166–169 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Westphalen, C. B., Quante, M. & Wang, T. C. Functional implication of Dclk1 and Dclk1-expressing cells in cancer. Small GTPases 8, 164–171 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Nishio, K. et al. Doublecortin and CaM kinase-like-1 as an independent prognostic factor in patients with resected pancreatic carcinoma. World J. Gastroenterol. 23, 5764–5772 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Adamska, A., Domenichini, A. & Falasca, M. Pancreatic ductal adenocarcinoma: current and evolving therapies. Int. J. Mol. Sci. 18, E1338 (2017).

    Article  PubMed  CAS  Google Scholar 

  5. Westphalen, C. B. et al. Dclk1 defines quiescent pancreatic progenitors that promote injury-induced regeneration and tumorigenesis. Cell Stem Cell 18, 441–455 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Brubaker, D. K. et al. Proteogenomic network analysis of context-specific KRAS signaling in mouse-to-human cross-species translation. Cell Syst. 9, 258–270.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Antolin, A. A. et al. Objective, quantitative, data-driven assessment of chemical probes. Cell Chem. Biol. 25, 194–205.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Deng, X. et al. Characterization of a selective inhibitor of the Parkinson’s disease kinase LRRK2. Nat. Chem. Biol. 7, 203–205 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yang, Q. et al. Pharmacological inhibition of BMK1 suppresses tumor growth through PML. Cancer Cell 18, 258–267 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Deng, X. et al. Discovery of a benzo(e)pyrimido-(5,4-b)(1,4)diazepin-6(11H)-one as a potent and selective inhibitor of Big MAP Kinase 1. ACS Med. Chem. Lett. 2, 195–200 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lin, E. C. et al. ERK5 kinase activity is dispensable for cellular immune response and proliferation. Proc. Natl Acad. Sci. USA 113, 11865–11870 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang, J. et al. Structural and atropisomeric factors governing the selectivity of pyrimido-benzodiazipinones as inhibitors of kinases and bromodomains. ACS Chem. Biol. 13, 2438–2448 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Weygant, N. et al. Small molecule kinase inhibitor LRRK2-IN-1 demonstrates potent activity against colorectal and pancreatic cancer through inhibition of doublecortin-like kinase 1. Mol. Cancer 13, 103 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Xu, Y. & Vakoc, C. R. Targeting cancer cells with BET bromodomain inhibitors. Cold Spring Harb. Perspect. Med. 7, a026674 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Hoang, V. T. et al. Oncogenic signaling of MEK5-ERK5. Cancer Lett. 392, 51–59 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lin, A. et al. Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci. Transl. Med. 11, eaaw8412 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Miduturu, C. V. et al. High-throughput kinase profiling: a more efficient approach toward the discovery of new kinase inhibitors. Chem. Biol. 18, 868–879 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Patricelli, M. P. et al. In situ kinase profiling reveals functionally relevant properties of native kinases. Chem. Biol. 18, 699–710 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Patel, O. et al. Biochemical and structural insights into doublecortin-like kinase domain 1. Structure 24, 1550–1561 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Davis, M. I. et al. Comprehensive analysis of kinase inhibitor selectivity. Nat. Biotechnol. 29, 1046–1051 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Uhlen, M. et al. Proteomics. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  PubMed  CAS  Google Scholar 

  22. Vasta, J. D. et al. Quantitative, wide-spectrum kinase profiling in live cells for assessing the effect of cellular ATP on target engagement. Cell Chem. Biol. 25, 206–214.e11 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Beno, B. R., Yeung, K. S., Bartberger, M. D., Pennington, L. D. & Meanwell, N. A. A survey of the role of noncovalent sulfur interactions in drug design. J. Med. Chem. 58, 4383–4438 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Chen, H. et al. Discovery of a novel allosteric inhibitor-binding site in ERK5: comparison with the canonical kinase hinge ATP-binding site. Acta Crystallogr. D: Biol. Crystallogr. 72, 682–693 (2016).

    Article  CAS  Google Scholar 

  25. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nabet, B. et al. The dTAG system for immediate and target-specific protein degradation. Nat. Chem. Biol. 14, 431–441 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Janes, M. R. et al. Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell 172, 578–589.e17 (2018).

    Article  CAS  PubMed  Google Scholar 

  28. Muzumdar, M. D. et al. Survival of pancreatic cancer cells lacking KRAS function. Nat. Commun. 8, 1090 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Gilmartin, A. G. et al. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin. Cancer Res. 17, 989–1000 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Filippakopoulos, P. et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. McDonald, E. R.3rd et al. Project DRIVE: a compendium of cancer dependencies and synthetic lethal relationships uncovered by large-scale, deep RNAi screening. Cell 170, 577–592.e10 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Tsherniak, A. et al. Defining a cancer dependency map. Cell 170, 564–576.e16 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gendoo, D. M. A. et al. Whole genomes define concordance of matched primary, xenograft, and organoid models of pancreas cancer. PLoS Comput. Biol. 15, e1006596 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Tiriac, H. et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 8, 1112–1129 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Foster, S. A. et al. Activation mechanism of oncogenic deletion mutations in BRAF, EGFR, and HER2. Cancer Cell 29, 477–493 (2016).

    Article  CAS  PubMed  Google Scholar 

  36. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Drost, J. & Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 18, 407–418 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Ting, H. A. & von Moltke, J. The immune function of tuft cells at gut mucosal surfaces and beyond. J. Immunol. 202, 1321–1329 (2019).

    Article  CAS  PubMed  Google Scholar 

  39. Keller, S. et al. High-precision isothermal titration calorimetry with automated peak-shape analysis. Anal. Chem. 84, 5066–5073 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Erazo, T. et al. Canonical and kinase activity-independent mechanisms for extracellular signal-regulated kinase 5 (ERK5) nuclear translocation require dissociation of Hsp90 from the ERK5-Cdc37 complex. Mol. Cell. Biol. 33, 1671–1686 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wang, B. et al. ATXN1L, CIC, and ETS transcription factors modulate sensitivity to MAPK pathway inhibition. Cell Rep. 18, 1543–1557 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Brinkman, E. K., Chen, T., Amendola, M. & van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Banker, G. & Goslin, K. Culturing Nerve Cells. (The MIT Press: 1998).

  44. Boj, S. F. et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324–338 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Loven, J. et al. Revisiting global gene expression analysis. Cell 151, 476–482 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Paulo, J. A. Nicotine alters the proteome of two human pancreatic duct cell lines. JOP 15, 465–474 (2014).

    PubMed  Google Scholar 

  48. Paulo, J. A. & Gygi, S. P. Nicotine-induced protein expression profiling reveals mutually altered proteins across four human cell lines. Proteomics 17, 1–2 (2017).

    Article  CAS  Google Scholar 

  49. Navarrete-Perea, J., Yu, Q., Gygi, S. P. & Paulo, J. A. Streamlined tandem mass tag (SL-TMT) protocol: an efficient strategy for quantitative (phospho)proteome profiling using tandem mass tag-synchronous precursor selection-MS3. J. Proteome Res. 17, 2226–2236 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. McAlister, G. C. et al. MultiNotch MS3 enables accurate, sensitive, and multiplexed detection of differential expression across cancer cell line proteomes. Anal. Chem. 86, 7150–7158 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Eng, J. K., McCormack, A. L. & Yates, J. R. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J. Am. Soc. Mass. Spectrom. 5, 976–989 (1994).

    Article  CAS  PubMed  Google Scholar 

  52. Elias, J. E. & Gygi, S. P. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat. Methods 4, 207–214 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Huttlin, E. L. et al. A tissue-specific atlas of mouse protein phosphorylation and expression. Cell 143, 1174–1189 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Beausoleil, S. A., Villen, J., Gerber, S. A., Rush, J. & Gygi, S. P. A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nat. Biotechnol. 24, 1285–1292 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Lyons, J. et al. Integrated in vivo multiomics analysis identifies p21-activated kinase signaling as a driver of colitis. Sci. Signal 11, eaan3580 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Kostic for critical reading of the manuscript and S. Nabet and members of the Gray laboratory for helpful discussions. We gratefully acknowledge S. Gygi for use of CORE for mass spectrometry data analysis software. This work was supported by an American Cancer Society Postdoctoral Fellowship PF-17-010-01-CDD (B.N.), Claudia Adams Barr Program in Innovative Basic Cancer Research Award (B.N.), Katherine L. and Steven C. Pinard Research Fund (N.S.G. and B.N.), Hope Funds for Cancer Research Postdoctoral Fellowship (S.R.), Harvard Catalyst KL2/CMeRIT Fellowship (S.R.), Perry Levy Fellowship (S.R.), the Lustgarten Foundation (S.R., B.M.W., A.J.A. and W.C.H.), NCI HCMI program (A.J.A. and S.R.), American Cancer Society 129089-PF-16-088-01-TBG (E.J.P.), KU-KIST Graduate School of Converging Science and Technology Program (T.S.), Spanish Ministerio de Economia y Competitividad grant no. SAF2015-60268R, cofunded by Fondo Europeo de Desarrollo Regional funds (J.M.L.), Pancreatic Cancer Action Network Catalyst Award (A.J.A.), Doris Duke Charitable Foundation Clinician Scientist Development Award (A.J.A.), NCI K08 CA218420 (A.J.A.), NCI U01 CA176058 (W.C.H.), NCI U01 CA199253 (W.C.H.) and NCI U01 CA224146 (W.C.H.), American Cancer Society Award 132205-RSG-18-039-01-DMC (K.D.W.), Welch Foundation grant no. I1829 (K.D.W.), 2017 AACR-Bayer Innovation and Discovery grant no. 17-80-44-GRAY (N.S.G.), DF/HCC GI SPORE Developmental Research Project Award P50CA127003 (N.S.G. and K.M.H.) and Hale Center for Pancreatic Research (J.D.M., B.M.W., A.J.A., W.C.H. and N.S.G.).

Author information

Authors and Affiliations

Authors

Contributions

F.M.F., B.N. and N.S.G. conceived and led the study. F.M.F. performed medicinal chemistry optimization, with input from J.W. B.N. developed and characterized dTAG model systems, performed cellular PDAC studies and performed DCLK1 nanoBRET assays. B.N., S.R., R.L.K., R.W.S.N. and R.S. performed patient-derived organoid culture assays and prepared organoid samples for RNA-sequencing, proteomics and phosphoproteomics. B.N. and A.L.L. performed pulldown experiments and prepared cellular samples for KiNativ, RNA-sequencing, proteomics and phosphoproteomics. Y.L., W.H. and L.L. performed DCLK1 protein purification, ITC and molecular shift assay. M.K. performed proteomics and phosphoproteomics experiments. A.Y. performed the maximum tolerated dose studies. S.H. performed zebrafish toxicity assays. E.J.P. performed western blot and immunohistochemistry analysis of mouse model tissues. L.H. performed validation of organoid sensitivity. J.K. performed RNA-sequencing data analysis. N.D.-M. and S.E performed ERK5 biochemical and cellular assays. Z.Z., C.R.C., J.D.V. and M.B.R. developed the DCLK1 nanoBRET assay reagents. R.O. performed viability and morphology testing on rat cortical neurons. T.S. and N.D.K. performed molecular modeling. J.M.L., S.M., C.Y.L., A.T.L., K.M.H., J.D.M., B.M.W., A.J.A., W.C.H., K.D.W. and N.S.G. supervised the study. F.M.F., B.N. and S.R. wrote the manuscript, with edits from N.S.G. All authors read and approved the manuscript.

Corresponding author

Correspondence to Nathanael S. Gray.

Ethics declarations

Competing interests

F.M.F. and N.S.G. are inventors on a patent application related to the DCLK1 inhibitors described in this manuscript (WO/2018/075608). B.N. is an inventor on patent applications related to the dTAG system described in this manuscript (WO/2017/024318, WO/2017/024319, WO/2018/148443, WO/2018/148440). Z.Z., C.R.C., J.D.V. and M.B.R. are employees of Promega Corporation. B.M.W. receives research funding from Celgene, Inc, and is a consultant for G1 Therapeutics, BioLineRx and GRAIL. A.J.A. has consulted for Oncorus, Inc. W.C.H. is a consultant for Thermo Fisher, AjuIB, MPM Capital, iTeos and Paraxel and is a Scientific Founder and serves on the Scientific Advisory Board (SAB) for KSQ Therapeutics. K.D.W. is a member of the SAB for Vibliome Therapeutics. N.S.G. is a Scientific Founder, member of the SAB and equity holder in C4 Therapeutics, Syros, Soltego, B2S, Gatekeeper and Petra Pharmaceuticals. The Gray laboratory receives or has received research funding from Novartis, Takeda, Astellas, Taiho, Janssen, Kinogen, Voroni, Her2llc, Deerfield and Sanofi.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2, Figs. 1–22 and Note.

Reporting Summary

Supplementary Dataset 1. KinomeSCAN.

Full KINOMEscan dataset.

Supplementary Dataset 2. KiNativ.

Full KiNativ dataset from lysate and live cell format.

Supplementary Dataset 3. RNA-sequencing.

RNA-sequencing dataset for PANFR0172_T2, PANFR0172_T3 and PATU-8988T on comparison of DMSO and 2.5 µM DCLK1-IN-1 treatments for 24 h.

Supplementary Dataset 4. Proteomics.

Mass spectrometry-based proteomics dataset for PANFR0172_T2, PANFR0172_T3 and PATU-8988T on comparison of DMSO and 2.5 µM DCLK1-IN-1 treatments for 24 h.

Supplementary Dataset 5. Phosphoproteomics.

Mass spectrometry-based phosphoproteomics dataset for PANFR0172_T2, PANFR0172_T3 and PATU-8988T on comparison of DMSO and 2.5 µM DCLK1-IN-1 treatments for 24 h.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ferguson, F.M., Nabet, B., Raghavan, S. et al. Discovery of a selective inhibitor of doublecortin like kinase 1. Nat Chem Biol 16, 635–643 (2020). https://doi.org/10.1038/s41589-020-0506-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0506-0

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer