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:

Profiling of residual breast cancers after neoadjuvant chemotherapy identifies DUSP4 deficiency as a mechanism of drug resistance

Abstract

Neoadjuvant chemotherapy (NAC) induces a pathological complete response (pCR) in 30% of patients with breast cancer. However, many patients have residual cancer after chemotherapy, which correlates with a higher risk of metastatic recurrence and poorer outcome than those who achieve a pCR. We hypothesized that molecular profiling of tumors after NAC would identify genes associated with drug resistance. Digital transcript counting was used to profile surgically resected breast cancers after NAC. Low concentrations of dual specificity protein phosphatase 4 (DUSP4), an ERK phosphatase, correlated with high post-NAC tumor cell proliferation and with basal-like breast cancer (BLBC) status. BLBC had higher DUSP4 promoter methylation and gene expression patterns of Ras-ERK pathway activation relative to other breast cancer subtypes. DUSP4 overexpression increased chemotherapy-induced apoptosis, whereas DUSP4 depletion dampened the response to chemotherapy. Reduced DUSP4 expression in primary tumors after NAC was associated with treatment-refractory high Ki-67 scores and shorter recurrence-free survival. Finally, inhibition of mitogen-activated protein kinase kinase (MEK) synergized with docetaxel treatment in BLBC xenografts. Thus, DUSP4 downregulation activates the Ras-ERK pathway in BLBC, resulting in an attenuated response to anti-cancer chemotherapy.

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: Ki-67–associated gene expression in chemotherapy-refractory breast cancers.
Figure 2: DUSP4 expression is deficient in BLBC and associates with Ras-ERK pathway activation.
Figure 3: DUSP4 expression is inversely associated with Ras-ERK pathway activation.
Figure 4: DUSP4 regulates MAPK signaling and cell viability.
Figure 5: Reduction of DUSP4 expression after NAC correlates with enhanced tumor Ki-67 score and shorter RFS.
Figure 6: Pharmacological or genetic inhibition of MEK improves chemotherapy-induced apoptosis in vitro and in vivo.

Similar content being viewed by others

Accession codes

Accessions

ArrayExpress

Gene Expression Omnibus

NCBI Reference Sequence

References

  1. Levina, V., Marrangoni, A.M., DeMarco, R., Gorelik, E. & Lokshin, A.E. Drug-selected human lung cancer stem cells: cytokine network, tumorigenic and metastatic properties. PLoS ONE 3, e3077 (2008).

    Article  Google Scholar 

  2. Morel, A.P. et al. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS ONE 3, e2888 (2008).

    Article  Google Scholar 

  3. Creighton, C.J. et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc. Natl. Acad. Sci. USA 106, 13820–13825 (2009).

    Article  CAS  Google Scholar 

  4. Calcagno, A.M. et al. Prolonged drug selection of breast cancer cells and enrichment of cancer stem cell characteristics. J. Natl. Cancer Inst. 102, 1637–1652 (2010).

    Article  CAS  Google Scholar 

  5. Geiss, G.K. et al. Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat. Biotechnol. 26, 317–325 (2008).

    Article  CAS  Google Scholar 

  6. Guarneri, V. et al. A prognostic model based on nodal status and Ki-67 predicts the risk of recurrence and death in breast cancer patients with residual disease after preoperative chemotherapy. Ann. Oncol. 20, 1193–1198 (2009).

    Article  CAS  Google Scholar 

  7. Jones, R.L. et al. The prognostic significance of Ki67 before and after neoadjuvant chemotherapy in breast cancer. Breast Cancer Res. Treat. 116, 53–68 (2009).

    Article  CAS  Google Scholar 

  8. Paik, S. et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N. Engl. J. Med. 351, 2817–2826 (2004).

    Article  CAS  Google Scholar 

  9. Hess, K.R. et al. Pharmacogenomic predictor of sensitivity to preoperative chemotherapy with paclitaxel and fluorouracil, doxorubicin, and cyclophosphamide in breast cancer. J. Clin. Oncol. 24, 4236–4244 (2006).

    Article  CAS  Google Scholar 

  10. Farmer, P. et al. A stroma-related gene signature predicts resistance to neoadjuvant chemotherapy in breast cancer. Nat. Med. 15, 68–74 (2009).

    Article  CAS  Google Scholar 

  11. DiMeo, T.A. et al. A novel lung metastasis signature links Wnt signaling with cancer cell self-renewal and epithelial-mesenchymal transition in basal-like breast cancer. Cancer Res. 69, 5364–5373 (2009).

    Article  CAS  Google Scholar 

  12. Cheang, M.C. et al. Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J. Natl. Cancer Inst. 101, 736–750 (2009).

    Article  CAS  Google Scholar 

  13. Parker, J.S. et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J. Clin. Oncol. 27, 1160–1167 (2009).

    Article  Google Scholar 

  14. Sweet-Cordero, A. et al. An oncogenic KRAS2 expression signature identified by cross-species gene-expression analysis. Nat. Genet. 37, 48–55 (2005).

    Article  CAS  Google Scholar 

  15. Chiaradonna, F. et al. Ras-dependent carbon metabolism and transformation in mouse fibroblasts. Oncogene 25, 5391–5404 (2006).

    Article  CAS  Google Scholar 

  16. Sánchez-Munoz, A. et al. Lack of evidence for KRAS oncogenic mutations in triple-negative breast cancer. BMC Cancer 10, 136 (2010).

    Article  Google Scholar 

  17. Forbes, S.A. et al. The Catalogue of Somatic Mutations in Cancer (COSMIC). Curr. Protoc. Hum. Genet. Chapter 10 Unit 10.11 (2008).

  18. Cadalbert, L.C. et al. Differential regulation of MAP kinase activation by a novel splice variant of human MAP kinase phosphatase-2. Cell. Signal. 22, 357–365 (2010).

    Article  CAS  Google Scholar 

  19. Mirzoeva, O.K. et al. Basal subtype and MAPK/ERK kinase (MEK)-phosphoinositide 3-kinase feedback signaling determine susceptibility of breast cancer cells to MEK inhibition. Cancer Res. 69, 565–572 (2009).

    Article  CAS  Google Scholar 

  20. 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  Google Scholar 

  21. Popovici, V. et al. Effect of training-sample size and classification difficulty on the accuracy of genomic predictors. Breast Cancer Res. 12, R5 (2010).

    Article  Google Scholar 

  22. Holm, K. et al. Molecular subtypes of breast cancer are associated with characteristic DNA methylation patterns. Breast Cancer Res. 12, R36 (2010).

    Article  Google Scholar 

  23. Bediaga, N.G. et al. DNA methylation epigenotypes in breast cancer molecular subtypes. Breast Cancer Res. 12, R77 (2010).

    Article  Google Scholar 

  24. Pratilas, C.A. et al. (V600E)BRAF is associated with disabled feedback inhibition of RAF-MEK signaling and elevated transcriptional output of the pathway. Proc. Natl. Acad. Sci. USA 106, 4519–4524 (2009).

    Article  CAS  Google Scholar 

  25. Wang, Y. et al. Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 365, 671–679 (2005).

    Article  CAS  Google Scholar 

  26. Stickeler, E. et al. Basal-like molecular subtype and HER4 up-regulation and response to neoadjuvant chemotherapy in breast cancer. Oncol. Rep. 26, 1037–1045 (2011).

    CAS  Google Scholar 

  27. Korde, L.A. et al. Gene expression pathway analysis to predict response to neoadjuvant docetaxel and capecitabine for breast cancer. Breast Cancer Res. Treat. 119, 685–699 (2010).

    Article  CAS  Google Scholar 

  28. Yacoub, A. et al. Sequence dependent exposure of mammary carcinoma cells to Taxotere and the MEK1/2 inhibitor U0126 causes enhanced cell killing in vitro. Cancer Biol. Ther. 2, 670–676 (2003).

    CAS  Google Scholar 

  29. Dowsett, M. et al. Short-term changes in Ki-67 during neoadjuvant treatment of primary breast cancer with anastrozole or tamoxifen alone or combined correlate with recurrence-free survival. Clin. Cancer Res. 11, 951s–958s (2005).

    CAS  Google Scholar 

  30. Dowsett, M. et al. Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. J. Natl. Cancer Inst. 99, 167–170 (2007).

    Article  CAS  Google Scholar 

  31. Miller, W.R. et al. Gene expression profiles differentiating between breast cancers clinically responsive or resistant to letrozole. J. Clin. Oncol. 27, 1382–1387 (2009).

    Article  CAS  Google Scholar 

  32. Britson, J.S., Barton, F., Balko, J.M. & Black, E.P. Deregulation of DUSP activity in EGFR-mutant lung cancer cell lines contributes to sustained ERK1/2 signaling. Biochem. Biophys. Res. Commun. 390, 849–854 (2009).

    Article  CAS  Google Scholar 

  33. Chitale, D. et al. An integrated genomic analysis of lung cancer reveals loss of DUSP4 in EGFR-mutant tumors. Oncogene 28, 2773–2783 (2009).

    Article  CAS  Google Scholar 

  34. Waha, A. et al. Epigenetic downregulation of mitogen-activated protein kinase phosphatase MKP-2 relieves its growth suppressive activity in glioma cells. Cancer Res. 70, 1689–1699 (2010).

    Article  CAS  Google Scholar 

  35. Armes, J.E. et al. Candidate tumor-suppressor genes on chromosome arm 8p in early-onset and high-grade breast cancers. Oncogene 23, 5697–5702 (2004).

    Article  CAS  Google Scholar 

  36. Cadalbert, L., Sloss, C.M., Cameron, P. & Plevin, R. Conditional expression of MAP kinase phosphatase-2 protects against genotoxic stress-induced apoptosis by binding and selective dephosphorylation of nuclear activated c-jun N-terminal kinase. Cell. Signal. 17, 1254–1264 (2005).

    Article  CAS  Google Scholar 

  37. Brumby, A.M. et al. Identification of novel Ras-cooperating oncogenes in Drosophila melanogaster: a RhoGEF/Rho-family/JNK pathway is a central driver of tumorigenesis. Genetics 105–125 (2011).

    Article  CAS  Google Scholar 

  38. Kan, Z. et al. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 466, 869–873 (2010).

    Article  CAS  Google Scholar 

  39. Wang, J. et al. Sustained c-Jun-NH2-kinase activity promotes epithelial-mesenchymal transition, invasion, and survival of breast cancer cells by regulating extracellular signal-regulated kinase activation. Mol. Cancer Res. 8, 266–277 (2010).

    Article  Google Scholar 

  40. Creighton, C.J., Chang, J.C. & Rosen, J.M. Epithelial-mesenchymal transition (EMT) in tumor-initiating cells and its clinical implications in breast cancer. J. Mammary Gland Biol. Neoplasia 15, 253–260 (2010).

    Article  Google Scholar 

  41. Haibe-Kains, B. genefu R package: Relevant Functions for Gene Expression Analysis, Especially in Breast Cancer <http://www.bioconductor.org/packages/release/bioc/html/genefu.html> (2009).

  42. Balko, J.M., Jones, B.R., Coakley, V.L. & Black, E.P. Combined MEK and EGFR inhibition demonstrates synergistic activity in EGFR-dependent NSCLC. Cancer Biol. Ther. 8, 522–530 (2009).

    Article  CAS  Google Scholar 

  43. Pusztai, L. Gene expression profiling of breast cancer. Breast Cancer Res. 11 (suppl .3), S11 (2009).

    Article  Google Scholar 

  44. Boersma, B.J. et al. A stromal gene signature associated with inflammatory breast cancer. Int. J. Cancer 122, 1324–1332 (2008).

    Article  CAS  Google Scholar 

  45. Stickeler, E. et al. Basal-like molecular subtype and HER4 up-regulation and response to neoadjuvant chemotherapy in breast cancer. Oncol. Rep. 26, 1037–1045 (2011).

    CAS  Google Scholar 

  46. Hennessy, B.T. et al. Pharmacodynamic markers of perifosine efficacy. Clin. Cancer Res. 13, 7421–7431 (2007).

    Article  CAS  Google Scholar 

  47. Hu, J. et al. Non-parametric quantification of protein lysate arrays. Bioinformatics 23, 1986–1994 (2007).

    Article  CAS  Google Scholar 

  48. Liang, J. et al. The energy sensing LKB1-AMPK pathway regulates p27(kip1) phosphorylation mediating the decision to enter autophagy or apoptosis. Nat. Cell Biol. 9, 218–224 (2007).

    Article  CAS  Google Scholar 

  49. Stemke-Hale, K. et al. An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res. 68, 6084–6091 (2008).

    Article  CAS  Google Scholar 

  50. Tibes, R. et al. Reverse phase protein array: validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells. Mol. Cancer Ther. 5, 2512–2521 (2006).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank E. Penni Black (University of Kentucky), who provided the AdMEK1ca adenoviral construct. The authors would also like to thank P.D. Smith and AstraZeneca for supplying selumetinib utilized in the in vivo experiments. This work was supported by Breast Cancer Specialized Program of Research Excellence (SPORE) grant P50CA98131, Vanderbilt-Ingram Cancer Center Support grant P30CA68485, a grant from the Breast Cancer Research Foundation, the American Cancer Society Clinical Research Professorship grant CRP-07-234 and the Lee Jeans Translational Breast Cancer Research Program (to C.L.A.). G.B.M. was supported by the SU2C Dream Team award, PO1CA099031 and the Komen Promise grant KG 081694. J.S. and M.D. were funded by the Breakthrough Breast Cancer and the Royal Marsden National Institutes of Health Research Biomedical Research Centre.

Author information

Authors and Affiliations

Authors

Contributions

J.M.B. designed and performed the experiments, and authored the manuscript. R.S.C. provided expert guidance on in vivo models and performed immunofluorescence. M.G.K., N.M.G.-I. and M.E.S. provided pathology support and performed IHC staining and scoring. D.B.V., T.W.M. and N.E.B. provided scientific advice. K.S.-H., A.M.G.-A. and G.B.M. performed RPPA analysis. J.S., M.D., J.A.P. and H.L.G. provided post-NAC patient tumor samples. J.J.S. and I.M.M. identified the Vanderbilt cohort and managed the clinical database, including performing retrospective chart review. M.E.S. and I.M.M. functioned as honest brokers to maintain patient privacy. C.L.A. provided scientific direction, established collaborations, prepared the manuscript with J.M.B. and allocated funding for the work. All the authors participated in the preparation of the manuscript.

Corresponding author

Correspondence to Carlos L Arteaga.

Ethics declarations

Competing interests

M.D. receives research grant funds and has received honoraria for consulting and advisory board work from AstraZeneca.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 2788 kb)

Supplementary Tables

Supplementary Tables 1–5 (XLS 411 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Balko, J., Cook, R., Vaught, D. et al. Profiling of residual breast cancers after neoadjuvant chemotherapy identifies DUSP4 deficiency as a mechanism of drug resistance. Nat Med 18, 1052–1059 (2012). https://doi.org/10.1038/nm.2795

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2795

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