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.

Overcoming resistance to HER2 inhibitors through state-specific kinase binding

Abstract

The heterodimeric receptor tyrosine kinase complex formed by HER2 and HER3 can act as an oncogenic driver and is also responsible for rescuing a large number of cancers from a diverse set of targeted therapies. Inhibitors of these proteins, particularly HER2, have dramatically improved patient outcomes in the clinic, but recent studies have demonstrated that stimulating the heterodimeric complex, either via growth factors or by increasing the concentrations of HER2 and HER3 at the membrane, significantly diminishes the activity of the inhibitors. To identify an inhibitor of the active HER2–HER3 oncogenic complex, we developed a panel of Ba/F3 cell lines suitable for ultra-high-throughput screening. Medicinal chemistry on the hit scaffold resulted in a previously uncharacterized inhibitor that acts through preferential inhibition of the active state of HER2 and, as a result, is able to overcome cellular mechanisms of resistance such as growth factors or mutations that stabilize the active form of HER2.

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

Figure 1: NRG rescues HER2-overexpressing cancer cells from HER2 inhibitors.
Figure 2: Lapatinib is unable to bind to the active HER2–HER3 heterodimer.
Figure 3: Design and execution of a high-throughput screen identifies a novel HER2–HER3 inhibitor.
Figure 4: Compound 2 is a selective type I inhibitor of HER2.
Figure 5: A type I inhibitor of HER2 is insensitive to the presence of NRG.
Figure 6: Compound 3 inhibits the active HER2–HER3 heterodimer in multiple oncogenic settings.

Accession codes

Accessions

Protein Data Bank

References

  1. Lemmon, M.A., Schlessinger, J. & Ferguson, K.M. The EGFR family: not so prototypical receptor tyrosine kinases. Cold Spring Harb. Perspect. Biol. 6, a020768 (2014).

    Article  Google Scholar 

  2. Kovacs, E., Zorn, J.A., Huang, Y., Barros, T. & Kuriyan, J. A structural perspective on the regulation of the epidermal growth factor receptor. Annu. Rev. Biochem. 84, 739–764 (2015).

    Article  CAS  Google Scholar 

  3. Doerner, A., Scheck, R. & Schepartz, A. Growth factor identity is encoded by discrete coiled-coil rotamers in the EGFR juxtamembrane region. Chem. Biol. 22, 776–784 (2015).

    Article  CAS  Google Scholar 

  4. Zhang, X., Gureasko, J., Shen, K., Cole, P.A. & Kuriyan, J. An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor. Cell 125, 1137–1149 (2006).

    Article  CAS  Google Scholar 

  5. Schneider, M.R. & Yarden, Y. The EGFR-HER2 module: a stem cell approach to understanding a prime target and driver of solid tumors. Oncogene 35, 2949–2960 (2015).

    Article  Google Scholar 

  6. Yarden, Y. & Sliwkowski, M.X. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell Biol. 2, 127–137 (2001).

    Article  CAS  Google Scholar 

  7. Red Brewer, M. et al. Mechanism for activation of mutated epidermal growth factor receptors in lung cancer. Proc. Natl. Acad. Sci. USA 110, E3595–E3604 (2013).

    Article  Google Scholar 

  8. Wang, Z. et al. Mechanistic insights into the activation of oncogenic forms of EGF receptor. Nat. Struct. Mol. Biol. 18, 1388–1393 (2011).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  10. Lee-Hoeflich, S.T. et al. A central role for HER3 in HER2-amplified breast cancer: implications for targeted therapy. Cancer Res. 68, 5878–5887 (2008).

    Article  CAS  Google Scholar 

  11. Tzahar, E. et al. A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Mol. Cell. Biol. 16, 5276–5287 (1996).

    Article  CAS  Google Scholar 

  12. Vaught, D.B. et al. HER3 is required for HER2-induced preneoplastic changes to the breast epithelium and tumor formation. Cancer Res. 72, 2672–2682 (2012).

    Article  CAS  Google Scholar 

  13. Jaiswal, B.S. et al. Oncogenic ERBB3 mutations in human cancers. Cancer Cell 23, 603–617 (2013).

    Article  CAS  Google Scholar 

  14. Wilson, T.R., Lee, D.Y., Berry, L., Shames, D.S. & Settleman, J. Neuregulin-1-mediated autocrine signaling underlies sensitivity to HER2 kinase inhibitors in a subset of human cancers. Cancer Cell 20, 158–172 (2011).

    Article  CAS  Google Scholar 

  15. Bose, R. et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 3, 224–237 (2013).

    Article  CAS  Google Scholar 

  16. Wang, S.E. et al. HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer Cell 10, 25–38 (2006).

    Article  Google Scholar 

  17. Greulich, H. et al. Functional analysis of receptor tyrosine kinase mutations in lung cancer identifies oncogenic extracellular domain mutations of ERBB2. Proc. Natl. Acad. Sci. USA 109, 14476–14481 (2012).

    Article  CAS  Google Scholar 

  18. Geyer, C.E. et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N. Engl. J. Med. 355, 2733–2743 (2006).

    Article  CAS  Google Scholar 

  19. Verma, S. et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N. Engl. J. Med. 367, 1783–1791 (2012).

    Article  CAS  Google Scholar 

  20. Phillips, G.D. et al. Dual targeting of HER2-positive cancer with trastuzumab emtansine and pertuzumab: critical role for neuregulin blockade in antitumor response to combination therapy. Clin. Cancer Res. 20, 456–468 (2014).

    Article  CAS  Google Scholar 

  21. Wilson, T.R. et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 487, 505–509 (2012).

    Article  CAS  Google Scholar 

  22. Sergina, N.V. et al. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature 445, 437–441 (2007).

    Article  CAS  Google Scholar 

  23. Chakrabarty, A., Sánchez, V., Kuba, M.G., Rinehart, C. & Arteaga, C.L. Feedback upregulation of HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors. Proc. Natl. Acad. Sci. USA 109, 2718–2723 (2012).

    Article  CAS  Google Scholar 

  24. Chandarlapaty, S. et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell 19, 58–71 (2011).

    Article  CAS  Google Scholar 

  25. Amin, D.N. et al. Resiliency and vulnerability in the HER2-HER3 tumorigenic driver. Sci. Transl. Med. 2, 16ra7 (2010).

    Article  Google Scholar 

  26. Das, P.M. et al. Reactivation of epigenetically silenced HER4/ERBB4 results in apoptosis of breast tumor cells. Oncogene 29, 5214–5219 (2010).

    Article  CAS  Google Scholar 

  27. Sartor, C.I. et al. Her4 mediates ligand-dependent antiproliferative and differentiation responses in human breast cancer cells. Mol. Cell. Biol. 21, 4265–4275 (2001).

    Article  CAS  Google Scholar 

  28. Aertgeerts, K. et al. Structural analysis of the mechanism of inhibition and allosteric activation of the kinase domain of HER2 protein. J. Biol. Chem. 286, 18756–18765 (2011).

    Article  CAS  Google Scholar 

  29. Wood, E.R. et al. A unique structure for epidermal growth factor receptor bound to GW572016 (Lapatinib): relationships among protein conformation, inhibitor off-rate, and receptor activity in tumor cells. Cancer Res. 64, 6652–6659 (2004).

    Article  CAS  Google Scholar 

  30. Littlefield, P. et al. Structural analysis of the EGFR/HER3 heterodimer reveals the molecular basis for activating HER3 mutations. Sci. Signal. 7, ra114 (2014).

    Article  Google Scholar 

  31. Red Brewer, M. et al. The juxtamembrane region of the EGF receptor functions as an activation domain. Mol. Cell 34, 641–651 (2009).

    Article  Google Scholar 

  32. Warmuth, M., Kim, S., Gu, X.-J., Xia, G. & Adrián, F. Ba/F3 cells and their use in kinase drug discovery. Curr. Opin. Oncol. 19, 55–60 (2007).

    Article  CAS  Google Scholar 

  33. Jura, N. et al. Mechanism for activation of the EGF receptor catalytic domain by the juxtamembrane segment. Cell 137, 1293–1307 (2009).

    Article  Google Scholar 

  34. Park, J.H., Liu, Y., Lemmon, M.A. & Radhakrishnan, R. Erlotinib binds both inactive and active conformations of the EGFR tyrosine kinase domain. Biochem. J. 448, 417–423 (2012).

    Article  CAS  Google Scholar 

  35. Shi, F., Telesco, S.E., Liu, Y., Radhakrishnan, R. & Lemmon, M.A. ErbB3/HER3 intracellular domain is competent to bind ATP and catalyze autophosphorylation. Proc. Natl. Acad. Sci. USA 107, 7692–7697 (2010).

    Article  CAS  Google Scholar 

  36. Jura, N., Shan, Y., Cao, X., Shaw, D.E. & Kuriyan, J. Structural analysis of the catalytically inactive kinase domain of the human EGF receptor 3. Proc. Natl. Acad. Sci. USA 106, 21608–21613 (2009).

    Article  CAS  Google Scholar 

  37. Rexer, B.N. et al. Human breast cancer cells harboring a gatekeeper T798M mutation in HER2 overexpress EGFR ligands and are sensitive to dual inhibition of EGFR and HER2. Clin. Cancer Res. 19, 5390–5401 (2013).

    Article  CAS  Google Scholar 

  38. Yoshida, T. et al. Identification and characterization of a novel chemotype MEK inhibitor able to alter the phosphorylation state of MEK1/2. Oncotarget 3, 1533–1545 (2012).

    Article  Google Scholar 

  39. Adrián, F.J. et al. Allosteric inhibitors of Bcr-abl-dependent cell proliferation. Nat. Chem. Biol. 2, 95–102 (2006).

    Article  Google Scholar 

  40. Scaltriti, M. et al. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. J. Natl. Cancer Inst. 99, 628–638 (2007).

    Article  CAS  Google Scholar 

  41. Chandarlapaty, S. et al. Inhibitors of HSP90 block p95-HER2 signaling in Trastuzumab-resistant tumors and suppress their growth. Oncogene 29, 325–334 (2010).

    Article  CAS  Google Scholar 

  42. Sperinde, J. et al. Quantitation of p95HER2 in paraffin sections by using a p95-specific antibody and correlation with outcome in a cohort of trastuzumab-treated breast cancer patients. Clin. Cancer Res. 16, 4226–4235 (2010).

    Article  CAS  Google Scholar 

  43. Sáez, R. et al. p95HER-2 predicts worse outcome in patients with HER-2-positive breast cancer. Clin. Cancer Res. 12, 424–431 (2006).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  45. Schüttelkopf, A.W. & van Aalten, D.M.F. PRODRG: a tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr. D Biol. Crystallogr. 60, 1355–1363 (2004).

    Article  Google Scholar 

Download references

Acknowledgements

We thank M. Hull, H. Nguyen, M. Wogan, J. Janes, P.G. Schultz and J. Roland from Calibr for technical assistance and helpful discussions. SK-BR-3 cells were a gift from S. Bandyopadhyay, and parental Ba/F3 cells were a gift from N. Shah (both at the University of California, San Francisco, San Francisco, California, USA). This work was supported in part by the Samuel Waxman Cancer Research Foundation (C.J.N., M.A.L. and K.M.S.), the U.S. National Institutes of Health (grant R01 GM109176-01A1 to K.M.S.), the Great Rivers Affiliate of the American Heart Association (predoctoral fellowship 11PRE7670020 to J.H.P.), and NIGMS (grant R01-GM099891 to M.A.L.).

Author information

Authors and Affiliations

Authors

Contributions

C.J.N., M.A.L., W.S. and K.M.S. designed research; S.P. and W.S. performed the high-throughput screen and counter-screens; J.H.P. cocrystalized the 2–EGFR complex and performed the HER3 kinase assay; and C.J.N. conducted cell proliferation/growth and death experiments, western blots, chemical synthesis, creation of Ba/F3 cell lines, and in vitro HER2 and HER3 assays. All authors analyzed data and contributed to the writing of the manuscript.

Corresponding authors

Correspondence to Weijun Shen or Kevan M Shokat.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Tables 1–7 and Supplementary Figures 1–20. (PDF 7055 kb)

Supplementary Note

Synthetic Procedures (PDF 666 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Novotny, C., Pollari, S., Park, J. et al. Overcoming resistance to HER2 inhibitors through state-specific kinase binding. Nat Chem Biol 12, 923–930 (2016). https://doi.org/10.1038/nchembio.2171

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2171

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