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:

Kinome rewiring reveals AURKA limits PI3K-pathway inhibitor efficacy in breast cancer

Abstract

Dysregulation of the PI3K-AKT-mTOR signaling network is a prominent feature of breast cancers. However, clinical responses to drugs targeting this pathway have been modest, possibly because of dynamic changes in cellular signaling that drive resistance and limit drug efficacy. Using a quantitative chemoproteomics approach, we mapped kinome dynamics in response to inhibitors of this pathway and identified signaling changes that correlate with drug sensitivity. Maintenance of AURKA after drug treatment was associated with resistance in breast cancer models. Incomplete inhibition of AURKA was a common source of therapy failure, and combinations of PI3K, AKT or mTOR inhibitors with the AURKA inhibitor MLN8237 were highly synergistic and durably suppressed mTOR signaling, resulting in apoptosis and tumor regression in vivo. This signaling map identifies survival factors whose presence limits the efficacy of targeted therapies and reveals new drug combinations that may unlock the full potential of PI3K–AKT–mTOR pathway inhibitors in breast 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: Measurement of kinome dynamics to identify correlates of drug sensitivity.
Fig. 2: Maintenance of AURKA is associated with resistance to PI3K inhibition.
Fig. 3: AURKA suppression enhances sensitivity and drives cell death in response to PI3K-pathway inhibitors in breast cancer cell lines.
Fig. 4: The Aurora kinase inhibitor MLN8237 enhances sensitivity to everolimus (RAD001) and induces cell death in vivo.
Fig. 5: Aurora kinase co-inhibition durably suppresses mTORC1 signaling and alters the BAX/BCL2 ratio.
Fig. 6: AURKA transcription is regulated by MYC downstream of the PI3K pathway.

Similar content being viewed by others

References

  1. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 490, 61–70 (2012).

    Article  CAS  Google Scholar 

  2. Janku, F. et al. Assessing PIK3CA and PTEN in early-phase trials with PI3K/AKT/mTOR inhibitors. Cell Rep 6, 377–387 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Dey, N., De, P. & Leyland-Jones, B. PI3K-AKT-mTOR inhibitors in breast cancers: from tumor cell signaling to clinical trials. Pharmacol. Ther. 175, 91–106 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. O’Reilly, K. E. et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 66, 1500–1508 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Klempner, S. J., Myers, A. P. & Cantley, L. C. What a tangled web we weave: emerging resistance mechanisms to inhibition of the phosphoinositide 3-kinase pathway. Cancer Discov. 3, 1345–1354 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Shah, P.D. & Chandarlapaty, S. in PI3K-mTOR in Cancer and Cancer Therapy (eds. Dey, N., De, P. & Leyland-Jones, B.) 125–147 (Springer International Publishing, 2016).

  7. Duncan, J. S. et al. Dynamic reprogramming of the kinome in response to targeted MEK inhibition in triple-negative breast cancer. Cell 149, 307–321 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Corcoran, R. B. et al. TORC1 suppression predicts responsiveness to RAF and MEK inhibition in BRAF-mutant melanoma. Sci. Transl. Med. 5, 196ra98 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Anderson, G. R. et al. PIK3CA mutations enable targeting of a breast tumor dependency through mTOR-mediated MCL-1 translation. Sci. Transl. Med. 8, 369ra175 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Boussemart, L. et al. eIF4F is a nexus of resistance to anti-BRAF and anti-MEK cancer therapies. Nature 513, 105–109 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Elkabets, M. et al. mTORC1 inhibition is required for sensitivity to PI3K p110α inhibitors in PIK3CA-mutant breast cancer. Sci. Transl. Med. 5, 196ra99 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Vora, S. R. et al. CDK 4/6 inhibitors sensitize PIK3CA mutant breast cancer to PI3K inhibitors. Cancer Cell 26, 136–149 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bantscheff, M. et al. Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat. Biotechnol. 25, 1035–1044 (2007).

    Article  CAS  PubMed  Google Scholar 

  14. Sos, M. L. et al. Oncogene mimicry as a mechanism of primary resistance to BRAF inhibitors. Cell Rep. 8, 1037–1048 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Stuhlmiller, T. J. et al. Inhibition of lapatinib-induced kinome reprogramming in ERBB2-positive breast cancer by targeting BET family bromodomains. Cell Rep 11, 390–404 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lens, S. M. A., Voest, E. E. & Medema, R. H. Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nat. Rev. Cancer 10, 825–841 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Bosch, A. et al. PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor–positive breast cancer. Sci. Transl. Med. 7, 283ra51 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Leroy, C. et al. Activation of IGF1R/p110β/AKT/mTOR confers resistance to α-specific PI3K inhibition. Breast Cancer Res. 18, 41 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Foucquier, J. & Guedj, M. Analysis of drug combinations: current methodological landscape. Pharmacol. Res. Perspect. 3, e00149 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Fruman, D. A. & Rommel, C. PI3K and cancer: lessons, challenges and opportunities. Nat. Rev. Drug Discov. 13, 140–156 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Nikonova, A. S., Astsaturov, I., Serebriiskii, I. G., Dunbrack, R. L. Jr. & Golemis, E. A. Aurora A kinase (AURKA) in normal and pathological cell division. Cell. Mol. Life Sci. 70, 661–687 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Piccart, M. et al. Everolimus plus exemestane for hormone-receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: overall survival results from BOLERO-2†. Ann. Oncol 25, 2357–2362 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Choo, A. Y., Yoon, S.-O., Kim, S. G., Roux, P. P. & Blenis, J. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc. Natl. Acad. Sci. USA 105, 17414–17419 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Vadlakonda, L., Dash, A., Pasupuleti, M., Anil Kumar, K. & Reddanna, P. The paradox of Akt-mTOR interactions. Front. Oncol. 3, 165 (2013).

    PubMed  PubMed Central  Google Scholar 

  26. Laplante, M. & Sabatini, D. M. mTOR signaling in growth control and disease. Cell 149, 274–293 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Volkmann, N., Marassi, F. M., Newmeyer, D. D. & Hanein, D. The rheostat in the membrane: BCL-2 family proteins and apoptosis. Cell Death Differ. 21, 206–215 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. West, M. J., Stoneley, M. & Willis, A. E. Translational induction of the c-myc oncogene via activation of the FRAP/TOR signalling pathway. Oncogene 17, 769–780 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Sears, R. et al. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev 14, 2501–2514 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Diehl, J. A., Cheng, M., Roussel, M. F. & Sherr, C. J. Glycogen synthase kinase-3β regulates cyclin D1 proteolysis and subcellular localization. Genes Dev. 12, 3499–3511 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yang, H., He, L., Kruk, P., Nicosia, S. V. & Cheng, J. Q. Aurora-A induces cell survival and chemoresistance by activation of Akt through a p53-dependent manner in ovarian cancer cells. Int. J. Cancer 119, 2304–2312 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Dar, A. A., Belkhiri, A. & El-Rifai, W. The aurora kinase A regulates GSK-3β in gastric cancer cells. Oncogene 28, 866–875 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Martins, M. M. et al. Linking tumor mutations to drug responses via a quantitative chemical-genetic interaction map. Cancer Discov. 5, 154–167 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Muellner, M. K. et al. A chemical-genetic screen reveals a mechanism of resistance to PI3K inhibitors in cancer. Nat. Chem. Biol. 7, 787–793 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ilic, N., Utermark, T., Widlund, H. R. & Roberts, T. M. PI3K-targeted therapy can be evaded by gene amplification along the MYC-eukaryotic translation initiation factor 4E (eIF4E) axis. Proc. Natl. Acad. Sci. USA 108, E699–E708 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Liu, P. et al. Oncogenic PIK3CA-driven mammary tumors frequently recur via PI3K pathway-dependent and PI3K pathway-independent mechanisms. Nat. Med. 17, 1116–1120 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Nomanbhoy, T. K. et al. Chemoproteomic evaluation of target engagement by the cyclin-dependent kinase 4 and 6 inhibitor palbociclib correlates with cancer cell response. Biochemistry 55, 5434–5441 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Roux, P. P. et al. RAS/ERK signaling promotes site-specific ribosomal protein S6 phosphorylation via RSK and stimulates cap-dependent translation. J. Biol. Chem. 282, 14056–14064 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. She, Q.-B. et al. 4E-BP1 is a key effector of the oncogenic activation of the AKT and ERK signaling pathways that integrates their function in tumors. Cancer Cell 18, 39–51 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. den Hollander, J. et al. Aurora kinases A and B are up-regulated by Myc and are essential for maintenance of the malignant state. Blood 116, 1498–1505 (2010).

    Article  CAS  Google Scholar 

  41. Lu, L. et al. Aurora kinase A mediates c-Myc’s oncogenic effects in hepatocellular carcinoma. Mol. Carcinog. 54, 1467–1479 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Zheng, F. et al. Nuclear AURKA acquires kinase-independent transactivating function to enhance breast cancer stem cell phenotype. Nat. Commun. 7, 10180 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ferrell, J. E. Jr. Self-perpetuating states in signal transduction: positive feedback, double-negative feedback and bistability. Curr. Opin. Cell Biol. 14, 140–148 (2002).

    Article  CAS  PubMed  Google Scholar 

  44. Katsha, A. et al. Activation of EIF4E by aurora kinase A depicts a novel druggable axis in everolimus-resistant cancer cells. Clin. Cancer Res. 23, 3756–3768 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Brewer Savannah, K. J. et al. Dual targeting of mTOR and aurora-A kinase for the treatment of uterine Leiomyosarcoma. Clin. Cancer Res. 18, 4633–4645 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Liu, L.-L. et al. Inhibition of mTOR pathway sensitizes acute myeloid leukemia cells to aurora inhibitors by suppression of glycolytic metabolism. Mol. Cancer Res. 11, 1326–1336 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Baselga, J. et al. Buparlisib plus fulvestrant versus placebo plus fulvestrant in postmenopausal, hormone receptor-positive, HER2-negative, advanced breast cancer (BELLE-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 18, 904–916 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zwang, Y. et al. Synergistic interactions with PI3K inhibition that induce apoptosis. eLife 6, e24523 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Barr, P. M. et al. Phase II intergroup trial of alisertib in relapsed and refractory peripheral T-cell lymphoma and transformed mycosis fungoides: SWOG 1108. J. Clin. Oncol. Clin. Oncol. 33, 2399–2404 (2015).

    Article  CAS  Google Scholar 

  50. Neve, R. M. et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10, 515–527 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Barvian, M. et al. Pyrido[2,3-d]pyrimidin-7-one inhibitors of cyclin-dependent kinases. J. Med. Chem. 43, 4606–4616 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Daub, H. et al. Kinase-selective enrichment enables quantitative phosphoproteomics of the kinome across the cell cycle. Mol. Cell 31, 438–448 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Blake, J. F. et al. Discovery of pyrrolopyrimidine inhibitors of Akt. Bioorg. Med. Chem. Lett. 20, 5607–5612 (2010).

    Article  CAS  PubMed  Google Scholar 

  54. Tanaka, M. et al. An unbiased cell morphology-based screen for new, biologically active small molecules. PLoS Biol. 3, e128 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bankston, D. et al. A scaleable synthesis of BAY 43–9006: a potent Raf kinase inhibitor for the treatment of cancer. Org. Process Res. Dev. 6, 777–781 (2002).

    Article  CAS  Google Scholar 

  56. Statsuk, A. V. et al. Tuning a three-component reaction for trapping kinase substrate complexes. J. Am. Chem. Soc. 130, 17568–17574 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Choi, M. et al. MSstats: an R package for statistical analysis of quantitative mass spectrometry-based proteomic experiments. Bioinformatics 30, 2524–2526 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Lehár, J. et al. Synergistic drug combinations tend to improve therapeutically relevant selectivity. Nat. Biotechnol. 27, 659–666 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank members of the Bandyopadhyay laboratory for helpful discussions and technical assistance. We also thank A. Beardsley, E. Markegard, D. Ruggero and W. Weiss for helpful discussions and reagents. This work was supported in part by NCI U01CA168370 (S.B.), NIGMS R01GM107671 (S.B.), NCI R01CA170447 (A.G.), Prospect Creek Foundation (S.B., A.G.), OHSU Pilot Project Funding (S.B., J.K.), American Cancer Society Postdoctoral Fellowship (J.D.G.), the Gazarian Foundation (A.G.), DOD W81XWH-12-1-0272 and DOD W81XWH-16-1-0603 (A.G.).

Author information

Authors and Affiliations

Authors

Contributions

H.J.D., J.T.W., K.M.S, J.K., J.D.G., and S.B. contributed toward study conceptualization. H.J.D., J.T.W., and J.D.G. performed data analyses supporting the study. H.J.D. designed and performed the majority of experiments. N.B. assisted with samples for initial MIBs/MS profiling. R.S.L. J.T.W., and J.D.G. provided technical advice and guided the interpretation of mass spectrometry data from MIB/MS profiling. R.C. and O.M. assisted with animal studies, and K.N.S. helped with additional experiments. H.J.D. and S.B. composed the original draft, and all authors contributed to manuscript finalization. S.B. and A.G. supervised the study.

Corresponding authors

Correspondence to John D. Gordan or Sourav Bandyopadhyay.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–19

Reporting Summary

Supplementary Dataset 1

Kinase activity scores and P values from MIBs/MS profiling.

Supplementary Dataset 2

Gene set enrichment analysis (GSEA) of BYL719-treated cells.

Supplementary Dataset 3

Receptor status, synergy scores (Loewe and Bliss) and assessment of increase in apoptosis over single-agent or combinations tested in this study.

Supplementary Dataset 4

MYC signature derived from isogenic MCF10A cell lines.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Donnella, H.J., Webber, J.T., Levin, R.S. et al. Kinome rewiring reveals AURKA limits PI3K-pathway inhibitor efficacy in breast cancer. Nat Chem Biol 14, 768–777 (2018). https://doi.org/10.1038/s41589-018-0081-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-018-0081-9

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