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:

A drug discovery platform to identify compounds that inhibit EGFR triple mutants

Abstract

Receptor tyrosine kinases (RTKs) are transmembrane receptors of great clinical interest due to their role in disease. Historically, therapeutics targeting RTKs have been identified using in vitro kinase assays. Due to frequent development of drug resistance, however, there is a need to identify more diverse compounds that inhibit mutated but not wild-type RTKs. Here, we describe MaMTH-DS (mammalian membrane two-hybrid drug screening), a live-cell platform for high-throughput identification of small molecules targeting functional protein–protein interactions of RTKs. We applied MaMTH-DS to an oncogenic epidermal growth factor receptor (EGFR) mutant resistant to the latest generation of clinically approved tyrosine kinase inhibitors (TKIs). We identified four mutant-specific compounds, including two that would not have been detected by conventional in vitro kinase assays. One of these targets mutant EGFR via a new mechanism of action, distinct from classical TKI inhibition. Our results demonstrate how MaMTH-DS is a powerful complement to traditional drug screening approaches.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Overview of MaMTH-DS platform and top candidates identified in pilot screen.
Fig. 2: Validation of midostaurin and gilteritinib as EGFR ex19del/T790M/C797S and EGFR L858R/T790M/C797S activating mutant inhibitors.
Fig. 3: Validation of EMI1 as an EGFR ex19del/T790M/C797S and EGFR L858R/T790M/C797S activating mutant inhibitor.
Fig. 4: Investigating effect of EMI1 on activated EGFR L858R/T790M/C797S endosomal trafficking.
Fig. 5: Generation and testing of EMI1 chemical analogs.

Similar content being viewed by others

Data availability

All data supporting the findings presented in this study are available in the paper and its Supplementary Information files.

Code availability

All R code used in the analysis of the presented drug screening data is available from the authors on request.

References

  1. Lemmon, M. A. & Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 141, 1117–1134 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Volinsky, N. & Kholodenko, B. N. Complexity of receptor tyrosine kinase signal processing. Cold Spring Harb. Perspect. Biol. 5, a009043–a009043 (2013).

    PubMed  PubMed Central  Google Scholar 

  3. Jia, Y., Quinn, C. M., Kwak, S. & Talanian, R. V. Current in vitro kinase assay technologies: the quest for a universal format. Curr. Drug Discov. Technol. 5, 59–69 (2008).

    CAS  PubMed  Google Scholar 

  4. Petschnigg, J. et al. The mammalian-membrane two-hybrid assay (MaMTH) for probing membrane-protein interactions in human cells. Nat. Methods 11, 585–592 (2014).

    CAS  PubMed  Google Scholar 

  5. Sokolina, K. et al. Systematic protein–protein interaction mapping for clinically relevant human GPCRs. Mol. Syst. Biol. 13, 918 (2017).

    PubMed  PubMed Central  Google Scholar 

  6. Snider, J. et al. Mapping the functional yeast ABC transporter interactome. Nat. Chem. Biol. 9, 565–572 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Stagljar, I., Korostensky, C., Johnsson, N. & te Heesen, S. A genetic system based on split-ubiquitin for the analysis of interactions between membrane proteins in vivo. Proc. Natl Acad. Sci. USA 95, 5187–5192 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Yao, Z. et al. A global analysis of the receptor tyrosine kinase-protein phosphatase interactome. Mol. Cell 65, 347–360 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Petschnigg, J. et al. Systematic identification of oncogenic EGFR interaction partners. J. Mol. Biol. 429, 280–294 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Robbins, A. K. & Horlick, R. A. Macrophage scavenger receptor confers an adherent phenotype to cells in culture. Biotechniques 25, 240–244 (1998).

    CAS  PubMed  Google Scholar 

  11. Zheng, Y. et al. Temporal regulation of EGF signalling networks by the scaffold protein Shc1. Nature 499, 166–171 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Saucier, C. et al. The Shc adaptor protein is critical for VEGF induction by Met/HGF and ErbB2 receptors and for early onset of tumor angiogenesis. Proc. Natl Acad. Sci. USA 101, 2345–2350 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Goździk-Spychalska, J. et al. C-MET inhibitors in the treatment of lung cancer. Curr. Treat. Options Oncol. 15, 670–682 (2014).

    PubMed  Google Scholar 

  14. Hagel, M. et al. First selective small molecule inhibitor of FGFR4 for the treatment of hepatocellular carcinomas with an activated FGFR4 signaling pathway. Cancer Discov. 5, 424–437 (2015).

    CAS  PubMed  Google Scholar 

  15. Eder, J. P. et al. A phase I study of foretinib, a multi-targeted inhibitor of c-Met and vascular endothelial growth factor receptor 2. Clin. Cancer Res. 16, 3507–3516 (2010).

    CAS  PubMed  Google Scholar 

  16. Qian, F. et al. Inhibition of tumor cell growth, invasion, and metastasis by EXEL-2880 (XL880, GSK1363089), a novel inhibitor of HGF and VEGF receptor tyrosine kinases. Cancer Res. 69, 8009–8016 (2009).

    CAS  PubMed  Google Scholar 

  17. Zhang, S. et al. The potent ALK inhibitor brigatinib (AP26113) overcomes mechanisms of resistance to first- and second-generation ALK inhibitors in preclinical models. Clin. Cancer Res. 22, 5527–5538 (2016).

    CAS  PubMed  Google Scholar 

  18. Walter, A. O. et al. Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC. Cancer Discov. 3, 1404–1415 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Jiang, T. & Zhou, C. Clinical activity of the mutant-selective EGFR inhibitor AZD9291 in patients with EGFR inhibitor-resistant non-small cell lung cancer. Transl. Lung Cancer Res. 3, 370–372 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Thress, K. S. et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat. Med. 21, 560–562 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhang, J.-H., Chung, T. D. & Oldenburg, K. R. A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J. Biomol. Screen. 4, 67–73 (1999).

    CAS  PubMed  Google Scholar 

  22. Brideau, C., Gunter, B., Pikounis, B. & Liaw, A. Improved statistical methods for hit selection in high-throughput screening. J. Biomol. Screen. 8, 634–647 (2003).

    PubMed  Google Scholar 

  23. Lee, H. J. et al. Noncovalent wild-type-sparing inhibitors of EGFR T790M. Cancer Discov. 3, 168–181 (2013).

    CAS  PubMed  Google Scholar 

  24. M., L. Midostaurin approved for FLT3-mutated AML. Blood 129, 3403–3406 (2017).

    Google Scholar 

  25. Galanis, A. et al. Crenolanib is a potent inhibitor of FLT3 with activity against resistance-conferring point mutants. Blood 123, 94–100 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Antar, A. et al. Inhibition of FLT3 in AML: a focus on sorafenib. Bone Marrow Transplant. 52, 344–351 (2017).

    CAS  PubMed  Google Scholar 

  27. Cortes, J. et al. Quizartinib, an FLT3 inhibitor, as monotherapy in patients with relapsed or refractory acute myeloid leukaemia: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 19, 889–903 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Perl, A. E. et al. Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1–2 study. Lancet Oncol. 18, 1061–1075 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Zabludoff, S. D. et al. AZD7762, a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies. Mol. Cancer Ther. 7, 2955–2966 (2008).

    CAS  PubMed  Google Scholar 

  30. Sausville, E. et al. Phase I dose-escalation study of AZD7762, a checkpoint kinase inhibitor, in combination with gemcitabine in US patients with advanced solid tumors. Cancer Chemother. Pharmacol. 73, 539–549 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Kim, S. N. et al. 7-Diethylamino-3(2′-benzoxazolyl)-coumarin is a novel microtubule inhibitor with antimitotic activity in multidrug resistant cancer cells. Biochem. Pharmacol. 77, 1773–1779 (2009).

    CAS  PubMed  Google Scholar 

  32. Mellman, I. & Yarden, Y. Endocytosis and cancer. Cold Spring Harb. Perspect. Biol. 5, a016949 (2013).

    PubMed  PubMed Central  Google Scholar 

  33. Villaseñor, R., Nonaka, H., Del Conte-Zerial, P., Kalaidzidis, Y. & Zerial, M. Regulation of EGFR signal transduction by analogue-to-digital conversion in endosomes. eLife 4, e06156 (2015).

    PubMed Central  Google Scholar 

  34. Collinet, C. et al. Systems survey of endocytosis by multiparametric image analysis. Nature 464, 243–249 (2010).

    CAS  PubMed  Google Scholar 

  35. Rink, J., Ghigo, E., Kalaidzidis, Y. & Zerial, M. Rab conversion as a mechanism of progression from early to late endosomes. Cell 122, 735–749 (2005).

    CAS  PubMed  Google Scholar 

  36. Gibson, D. G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods 6, 343–345 (2009).

    CAS  PubMed  Google Scholar 

  37. Yang, X. et al. A public genome-scale lentiviral expression library of human ORFs. Nat. Methods 8, 659–661 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. R: a language and environment for statistical computing (R Core Team, 2017).

  39. Box, G. E. P. & Cox, D. R. An analysis of transformations. R. Stat. Soc. 26, 211–252 (1964).

    Google Scholar 

  40. Boutros, M., Brás, L. P. & Huber, W. Analysis of cell-based RNAi screens. Genome Biol. 7, R66 (2006).

    PubMed  PubMed Central  Google Scholar 

  41. Aher, A. et al. CLASP suppresses microtubule catastrophes through a single TOG domain. Dev. Cell 46, 40–58.e8 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Mohan, R. et al. End-binding proteins sensitize microtubules to the action of microtubule-targeting agents. Proc. Natl Acad. Sci. USA 110, 8900–8905 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Jost, M. et al. Combined CRISPRi/a-based chemical genetic screens reveal that rigosertib is a microtubule-destabilizing agent. Mol. Cell 68, 210–223 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank L. Riley for valuable discussions and editing of this manuscript. We also thank P.A. Jänne and M.J. Eck (both at Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA) for providing us with the PC9-T790M and PC9-C797S cells used in our study, and the Center for Information Services and High Performance Computing of the TU Dresden for providing support and computational resources. This research was supported by funding from Consortium Québécois sur la Découverte du Médicament (CQDM) (Explore) and OCE (no. 23929). In addition, work in the Stagljar laboratory is supported by the Canadian Cancer Society Research Institute (no. 703889), Genome Canada via Ontario Genomics (grant nos, 9427 and 9428), Ontario Research fund (grant nos. ORF/DIG-501411 and RE08-009), CQDM (Quantum Leap) and Brain Canada (Quantum Leap, and Cancer Research Society (grant no. 23235)). J.H.P.’s work in the laboratory of Mark Lemmon at Yale was supported by National Institutes of Health grant nos. R01-CA198164 and R35-GM122485. G.P. acknowledges the support of the Ontario Institute for Cancer Research and its funding from the Government of Ontario.

Author information

Authors and Affiliations

Authors

Contributions

P.S. performed preliminary MaMTH testing and validation/functional analysis of identified compounds, managed the project and wrote the manuscript. J.S. generated MaMTH reporter constructs and cells lines, performed MaMTH-DS screening and data analysis, managed the project and wrote the manuscript. Y.K. and M.Z. performed and analyzed the high-throughput EGFR microscopy data. K.W., L.D., A.L., N.V., A.V., S.P., F.A., Z.Y. and V.W. performed construct generation, preliminary MaMTH validation and were involved in the validation/functional analysis of compounds. N.R., N.A.P., Y.W., A.S. and M.S.T. generated and validated organoids and provided cell lines. A.R. and A.A. performed and analyzed microtubule dynamics data. L.E.W.G., A.D. and J.W. performed MaMTH-DS screening. B.T., A.A., M.P., B.J., R.M., D.U. and R.A. performed medicinal chemistry and assisted technically throughout the whole project. G.P. performed computational chemistry, molecular docking and hit expansion. J.H.P. assisted with computational modeling and undertook some biochemical studies. R.M.S., M.J., M.S., M.F.M., N.L. and F.A.S. provided clinical expertise and support. All authors discussed the results and commented on the manuscript. I.S. initiated and supervised the project and wrote the manuscript.

Corresponding author

Correspondence to Igor Stagljar.

Ethics declarations

Competing interests

I.S., P.S. and J.S. (in conjunction with the University of Toronto) are listed as inventors on a patent (publication no. 20190091205) for the use of EMI1 (and structurally related analogs), midostaurin, gilteritinib and AZD7762 (and structurally related analogs) in the treatment of mutant EGFR-mediated NSCLC.

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 Figs. 1–32, Tables 1 and 2 and Notes 1 and 2

Reporting Summary

Supplementary Dataset 1

Collection of compounds used in MaMTH-DS pilot screening.

Supplementary Dataset 2

Results of MaMTH-DS screening of EGFR L858R/T790M/C797S in the presence of ShcI (round 1).

Supplementary Dataset 3

Results of MaMTH-DS screening of EGFR L858R/T790M/C797S in the presence of ShcI (round 2).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Saraon, P., Snider, J., Kalaidzidis, Y. et al. A drug discovery platform to identify compounds that inhibit EGFR triple mutants. Nat Chem Biol 16, 577–586 (2020). https://doi.org/10.1038/s41589-020-0484-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0484-2

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