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.

  • Letter
  • Published:

Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3

Abstract

Oncogenic tyrosine kinases have proved to be promising targets for the development of highly effective anticancer drugs. However, tyrosine kinase inhibitors (TKIs) against the human epidermal growth factor receptor (HER) family show only limited activity against HER2-driven breast cancers, despite effective inhibition of epidermal growth factor receptor (EGFR) and HER2 in vivo1,2,3,4,5,6,7,8. The reasons for this are unclear. Signalling in trans is a key feature of this multimember family and the critically important phosphatidylinositol-3-OH kinase (PI(3)K)/Akt pathway is driven predominantly through transphosphorylation of the kinase-inactive HER3 (refs 9, 10). Here we show that HER3 and consequently PI(3)K/Akt signalling evade inhibition by current HER-family TKIs in vitro and in tumours in vivo. This is due to a compensatory shift in the HER3 phosphorylation–dephosphorylation equilibrium, driven by increased membrane HER3 expression driving the phosphorylation reaction and by reduced HER3 phosphatase activity impeding the dephosphorylation reaction. These compensatory changes are driven by Akt-mediated negative-feedback signalling. Although HER3 is not a direct target of TKIs, HER3 substrate resistance undermines their efficacy and has thus far gone undetected. The experimental abrogation of HER3 resistance by small interfering RNA knockdown restores potent pro-apoptotic activity to otherwise cytostatic HER TKIs, re-affirming the oncogene-addicted nature of HER2-driven tumours and the therapeutic promise of this oncoprotein target. However, because HER3 signalling is buffered against an incomplete inhibition of HER2 kinase, much more potent TKIs or combination strategies are required to silence oncogenic HER2 signalling effectively. The biologic marker with which to assess the efficacy of HER TKIs should be the transphosphorylation of HER3 rather than autophosphorylation.

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

Figure 1: HER TKIs fail to induce sustained inhibition of HER3 signalling in HER2-driven breast cancer cells.
Figure 2: Forward shift in HER3 phosphorylation–dephosphorylation equilibrium following extended HER TKI treatment.
Figure 3: Mechanism of HER3 reactivation after extended HER TKI treatment.
Figure 4: Akt regulates HER3 signalling via negative feedback signalling.

Similar content being viewed by others

References

  1. Winer, E. P. et al. Phase II multicenter study to evaluate the efficacy and safety of Tarceva (Erlotinib HCl, OSI-774) in women with previously treated, locally advanced or metastatic breast cancer. Breast Cancer Res. Treatm. 76 (suppl. 1), abstr. 445. (2002)

  2. Blackwell, K. et al. A phase II, open-label, multicenter study of GW572016 in patients with trastuzumab-refractory metastatic breast cancer. Proc. Am. Soc. Clin. Oncol. 23, abstr. 3006. (2004)

    Article  Google Scholar 

  3. Dees, E. C. et al. Clinical summary of 67 heavily pre-treated patients with metastatic carcinomas treated with GW572016 in a phase Ib study. Proc. Am. Soc. Clin. Oncol. 23, abstr. 3188. (2004)

    Article  Google Scholar 

  4. Campos, S. M. et al. A phase 2, single agent study of CI-1033 administered at two doses in ovarian cancer patients who failed platinum therapy. Proc. Am. Soc. Clin. Oncol. 23, abstr. 5054. (2004)

    Article  Google Scholar 

  5. Baselga, J. et al. Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. J. Clin. Oncol. 23, 5323–5333 (2005)

    Article  CAS  Google Scholar 

  6. Bacus, S. S., Beresford, P. J., Yarden, Y., Spector, N. & Smith, B. The use of predicting factors and surrogate markers in patients' cancer biopsies treated with targeted antibodies to erbB receptors and erbB tyrosine kinase inhibitors. Proc. Am. Soc. Clin. Oncol. 22, abstr. 3408. (2003)

  7. Burris, H. A. et al. EGF10004: a randomized, multicenter, phase Ib study of the safety, biologic activity and clinical efficacy of the dual kinase inhibitor GW572016. Breast Cancer Res. Treatm. 82 (suppl. 1), abstr. 39. (2003)

  8. Spector, N. L. et al. Study of the biologic effects of lapatinib, a reversible inhibitor of ErbB1 and ErbB2 tyrosine kinases, on tumor growth and survival pathways in patients with advanced malignancies. J. Clin. Oncol. 23, 2502–2512 (2005)

    Article  CAS  Google Scholar 

  9. Soltoff, S. P., Carraway, K. L., Prigent, S. A., Gullick, W. G. & Cantley, L. C. ErbB3 is involved in activation of phosphatidylinositol 3-kinase by epidermal growth factor. Mol. Cell. Biol. 14, 3550–3558 (1994)

    Article  CAS  Google Scholar 

  10. Kim, H. H., Sierke, S. L. & Koland, J. G. Epidermal growth factor-dependent association of phosphatidylinositol 3-kinase with the erbB3 gene product. J. Biol. Chem. 269, 24747–24755 (1994)

    CAS  PubMed  Google Scholar 

  11. Kantarjian, H. et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N. Engl. J. Med. 346, 645–652 (2002)

    Article  CAS  Google Scholar 

  12. Muller, W. J., Sinn, E., Pattengale, P. K., Wallace, R. & Leder, P. Single-step induction of mammary adenocarcinoma in transgenic mice bearing the activated c-neu oncogene. Cell 54, 105–115 (1988)

    Article  CAS  Google Scholar 

  13. Slamon, D. J. et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235, 177–182 (1987)

    Article  ADS  CAS  Google Scholar 

  14. Moody, S. E. et al. Conditional activation of Neu in the mammary epithelium of transgenic mice results in reversible pulmonary metastasis. Cancer Cell 2, 451–461 (2002)

    Article  CAS  Google Scholar 

  15. Moasser, M. M., Basso, A., Averbuch, S. D. & Rosen, N. The tyrosine kinase inhibitor ZD1839 (“Iressa”) inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing tumor cells. Cancer Res. 61, 7184–7188 (2001)

    CAS  PubMed  Google Scholar 

  16. Moulder, S. L. et al. Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer Res. 61, 8887–8895 (2001)

    MathSciNet  CAS  PubMed  Google Scholar 

  17. Anderson, N. G., Ahmad, T., Chan, K., Dobson, R. & Bundred, N. J. ZD 1839 (Iressa), a novel epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, potently inhibits the growth of EGFR-positive cancer cell lines with or without erbB2 overexpression. Int. J. Cancer 94, 774–782 (2001)

    Article  CAS  Google Scholar 

  18. Campiglio, M. et al. Inhibition of proliferation and induction of apoptosis in breast cancer cells by the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor ZD1839 ('Iressa') is independent of EGFR expression level. J. Cell. Physiol. 198, 259–268 (2004)

    Article  CAS  Google Scholar 

  19. Akita, R. W. & Sliwkowski, M. X. Preclinical studies with Erlotinib (Tarceva). Semin. Oncol. 30, (suppl. 7)15–24 (2003)

    Article  CAS  Google Scholar 

  20. She, Q., Solit, D., Basso, A. & Moasser, M. M. Resistance to gefitinib (ZD1839, Iressa) in PTEN null HER overexpressing tumor cells can be overcome through restoration of PTEN function or pharmacologic modulation of constitutive PI3K/Akt pathway signaling. Clin. Cancer Res. 9, 4340–4346 (2003)

    CAS  PubMed  Google Scholar 

  21. Bianco, R. et al. Loss of PTEN/MMAC1/TEP in EGF receptor-expressing tumor cells counteracts the antitumor action of EGFR tyrosine kinase inhibitors. Oncogene 22, 2812–2822 (2003)

    Article  CAS  Google Scholar 

  22. Haas-Kogan, D. A. et al. Epidermal growth factor receptor, protein kinase B/Akt, and glioma response to erlotinib. J. Natl Cancer Inst. 97, 880–887 (2005)

    Article  CAS  Google Scholar 

  23. McKillop, D. et al. Tumor penetration of gefitinib (Iressa), an epidermal growth factor receptor tyrosine kinase inhibitor. Mol. Cancer Ther. 4, 641–649 (2005)

    Article  CAS  Google Scholar 

  24. Fry, D. W. et al. Specific, irreversible inactivation of the epidermal growth factor receptor and erbB2, by a new class of tyrosine kinase inhibitor. Proc. Natl Acad. Sci. USA 95, 12022–12027 (1998)

    Article  ADS  CAS  Google Scholar 

  25. Offterdinger, M., Schofer, C., Weipoltshammer, K. & Grunt, T. W. c-erbB-3: a nuclear protein in mammary epithelial cells. J. Cell Biol. 157, 929–939 (2002)

    Article  CAS  Google Scholar 

  26. Meng, T. C., Fukada, T. & Tonks, N. K. Reversible oxidation and inactivation of protein tyrosine phosphatases in vivo. Mol. Cell 9, 387–399 (2002)

    Article  CAS  Google Scholar 

  27. Tonks, N. K. Redox Redux: Revisiting PTPs and the control of cell signaling. Cell 121, 667–670 (2005)

    Article  CAS  Google Scholar 

  28. Manning, G., Whyte, D. B., Martinez, R., Hunter, T. & Sudarsanam, S. The protein kinase complement of the human genome. Science 298, 1912–1934 (2002)

    Article  ADS  CAS  Google Scholar 

  29. Tsou, H. R. et al. 6-Substituted-4-(3-bromophenylamino)quinazolines as putative irreversible inhibitors of the epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor (HER-2) tyrosine kinases with enhanced antitumor activity. J. Med. Chem. 44, 2719–2734 (2001)

    Article  CAS  Google Scholar 

  30. Huron, D. R. et al. A novel pyridopyrimidine inhibitor of abl kinase is a picomolar inhibitor of Bcr-abl-driven K562 cells and is effective against STI571-resistant Bcr-abl mutants. Clin. Cancer Res. 9, 1267–1273 (2003)

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Susan Komen Foundation (M.M.M.), the California Breast Cancer Research Program (M.M.M.), and an NIH grant (K.M.S.). We thank D. Stokoe and F. McCormick for review of the manuscript.

Author Contributions All authors contributed to the experiments in this work. The studies were conceived by M.M.M. with additional contributions from N.V.S. and K.M.S. The paper was written by N.V.S. and M.M.M.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mark M. Moasser.

Ethics declarations

Competing interests

Reprints and permissions information is available at www.nature.com/reprints. The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains supplementary figures 1-4, their legends, a detailed method and reagent section. (PDF 377 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sergina, N., Rausch, M., Wang, D. et al. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature 445, 437–441 (2007). https://doi.org/10.1038/nature05474

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature05474

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing