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:

Glucocorticoids promote breast cancer metastasis

Abstract

Diversity within or between tumours and metastases (known as intra-patient tumour heterogeneity) that develops during disease progression is a serious hurdle for therapy1,2,3. Metastasis is the fatal hallmark of cancer and the mechanisms of colonization, the most complex step in the metastatic cascade4, remain poorly defined. A clearer understanding of the cellular and molecular processes that underlie both intra-patient tumour heterogeneity and metastasis is crucial for the success of personalized cancer therapy. Here, using transcriptional profiling of tumours and matched metastases in patient-derived xenograft models in mice, we show cancer-site-specific phenotypes and increased glucocorticoid receptor activity in distant metastases. The glucocorticoid receptor mediates the effects of stress hormones, and of synthetic derivatives of these hormones that are used widely in the clinic as anti-inflammatory and immunosuppressive agents. We show that the increase in stress hormones during breast cancer progression results in the activation of the glucocorticoid receptor at distant metastatic sites, increased colonization and reduced survival. Our transcriptomics, proteomics and phospho-proteomics studies implicate the glucocorticoid receptor in the activation of multiple processes in metastasis and in the increased expression of kinase ROR1, both of which correlate with reduced survival. The ablation of ROR1 reduced metastatic outgrowth and prolonged survival in preclinical models. Our results indicate that the activation of the glucocorticoid receptor increases heterogeneity and metastasis, which suggests that caution is needed when using glucocorticoids to treat patients with breast cancer who have developed cancer-related complications.

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

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

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

Fig. 1: Increase in GR activation in distant metastases.
Fig. 2: GR activation increases metastatic colonization and reduces survival.
Fig. 3: GR activation induces signalling networks and protein kinases that are implicated in breast cancer progression.
Fig. 4: ROR1 mediates GR-induced lung metastatic colonization.

Similar content being viewed by others

Data availability

All mass spectrometry raw data files have been deposited to the ProteomeXchange Consortium- accession code PXD009102, http://proteomecentral.proteomexchange.org. The mRNA sequencing data are deposited in the Gene Expression Omnibus (GEO) database under accession code GSE124817. Processed transcriptomic data that support the findings of this study are available on reasonable request from the corresponding author.

References

  1. Almendro, V., Marusyk, A. & Polyak, K. Cellular heterogeneity and molecular evolution in cancer. Annu. Rev. Pathol. 8, 277–302 (2013).

    Article  CAS  Google Scholar 

  2. Koren, S. & Bentires-Alj, M. Breast tumor heterogeneity: source of fitness, hurdle for therapy. Mol. Cell 60, 537–546 (2015).

    Article  CAS  Google Scholar 

  3. Polzer, B. & Klein, C. A. Metastasis awakening: the challenges of targeting minimal residual cancer. Nat. Med. 19, 274–275 (2013).

    Article  CAS  Google Scholar 

  4. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  Google Scholar 

  5. Marusyk, A., Almendro, V. & Polyak, K. Intra-tumour heterogeneity: a looking glass for cancer? Nat. Rev. Cancer 12, 323–334 (2012).

    Article  CAS  Google Scholar 

  6. Chaffer, C. L., San Juan, B. P., Lim, E. & Weinberg, R. A. EMT, cell plasticity and metastasis. Cancer Metastasis Rev. 35, 645–654 (2016).

    Article  Google Scholar 

  7. Meacham, C. E. & Morrison, S. J. Tumour heterogeneity and cancer cell plasticity. Nature 501, 328–337 (2013).

    Article  ADS  CAS  Google Scholar 

  8. Vanharanta, S. & Massagué, J. Origins of metastatic traits. Cancer Cell 24, 410–421 (2013).

    Article  CAS  Google Scholar 

  9. Obenauf, A. C. & Massagué, J. Surviving at a distance: organ-specific metastasis. Trends Cancer 1, 76–91 (2015).

    Article  Google Scholar 

  10. Lindström, L. S. et al. Clinically used breast cancer markers such as estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 are unstable throughout tumor progression. J. Clin. Oncol. 30, 2601–2608 (2012).

    Article  Google Scholar 

  11. Alizadeh, A. A. et al. Toward understanding and exploiting tumor heterogeneity. Nat. Med. 21, 846–853 (2015).

    Article  CAS  Google Scholar 

  12. Lawson, D. A. et al. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature 526, 131–135 (2015).

    Article  ADS  CAS  Google Scholar 

  13. Balwierz, P. J. et al. ISMARA: automated modeling of genomic signals as a democracy of regulatory motifs. Genome Res. 24, 869–884 (2014).

    Article  CAS  Google Scholar 

  14. Hosseini, H. et al. Early dissemination seeds metastasis in breast cancer. Nature 540, 552–558 (2016).

    Article  ADS  CAS  Google Scholar 

  15. van der Pompe, G., Antoni, M. H. & Heijnen, C. J. Elevated basal cortisol levels and attenuated ACTH and cortisol responses to a behavioral challenge in women with metastatic breast cancer. Psychoneuroendocrinology 21, 361–374 (1996).

    Article  Google Scholar 

  16. Sephton, S. E., Sapolsky, R. M., Kraemer, H. C. & Spiegel, D. Diurnal cortisol rhythm as a predictor of breast cancer survival. J. Natl. Cancer Inst. 92, 994–1000 (2000).

    Article  CAS  Google Scholar 

  17. Pang, D., Kocherginsky, M., Krausz, T., Kim, S.-Y. & Conzen, S. D. Dexamethasone decreases xenograft response to paclitaxel through inhibition of tumor cell apoptosis. Cancer Biol. Ther. 5, 933–940 (2006).

    Article  CAS  Google Scholar 

  18. Saxena, M. & Christofori, G. Rebuilding cancer metastasis in the mouse. Mol. Oncol. 7, 283–296 (2013).

    Article  CAS  Google Scholar 

  19. Gao, H. et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat. Med. 21, 1318–1325 (2015).

    Article  CAS  Google Scholar 

  20. Marotta, L. L. C. et al. The JAK2/STAT3 signaling pathway is required for growth of CD44+CD24 stem cell-like breast cancer cells in human tumors. J. Clin. Invest. 121, 2723–2735 (2011).

    Article  CAS  Google Scholar 

  21. Liu, J. et al. Synaptopodin-2 suppresses metastasis of triple-negative breast cancer via inhibition of YAP/TAZ activity. J. Pathol. 244, 71–83 (2018).

    Article  CAS  Google Scholar 

  22. Li, C. et al. A ROR1–HER3–lncRNA signalling axis modulates the Hippo–YAP pathway to regulate bone metastasis. Nat. Cell Biol. 19, 106–119 (2017).

    Article  CAS  Google Scholar 

  23. Cui, B. et al. Targeting ROR1 inhibits epithelial–mesenchymal transition and metastasis. Cancer Res. 73, 3649–3660 (2013).

    Article  CAS  Google Scholar 

  24. Chien, H.-P. et al. Expression of ROR1 has prognostic significance in triple negative breast cancer. Virchows Arch. 468, 589–595 (2016).

    Article  CAS  Google Scholar 

  25. Pongor, L. et al. A genome-wide approach to link genotype to clinical outcome by utilizing next generation sequencing and gene chip data of 6,697 breast cancer patients. Genome Med. 7, 104 (2015).

    Article  Google Scholar 

  26. Robinson, D. R. et al. Integrative clinical genomics of metastatic cancer. Nature 548, 297–303 (2017).

    Article  ADS  CAS  Google Scholar 

  27. Curtis, C. et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 486, 346–352 (2012).

    Article  CAS  Google Scholar 

  28. Pereira, B. et al. The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes. Nat. Commun. 7, 11479 (2016).

    Article  ADS  CAS  Google Scholar 

  29. West, D. C. et al. Discovery of a glucocorticoid receptor (GR) Activity signature using selective GR antagonism in ER-negative breast cancer. Clin. Cancer Res. 24, 3433–3446 (2018).

    Article  CAS  Google Scholar 

  30. Braun, S. et al. A pooled analysis of bone marrow micrometastasis in breast cancer. N. Engl. J. Med. 353, 793–802 (2005).

    Article  CAS  Google Scholar 

  31. Arora, V. K. et al. Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade. Cell 155, 1309–1322 (2013).

    Article  CAS  Google Scholar 

  32. Skor, M. N. et al. Glucocorticoid receptor antagonism as a novel therapy for triple-negative breast cancer. Clin. Cancer Res. 19, 6163–6172 (2013).

    Article  CAS  Google Scholar 

  33. DeRose, Y. S. et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat. Med. 17, 1514–1520 (2011).

    Article  CAS  Google Scholar 

  34. Britschgi, A. et al. The Hippo kinases LATS1 and 2 control human breast cell fate via crosstalk with ERα. Nature 541, 541–545 (2017).

    Article  ADS  CAS  Google Scholar 

  35. Wyckoff, J., Gligorijevic, B., Entenberg, D., Segall, J. & Condeelis, J. High-resolution multiphoton imaging of tumors in vivo. Cold Spring Harb. Protoc. 2011, 1167–1184 (2011).

    PubMed  PubMed Central  Google Scholar 

  36. Bonapace, L. et al. If you don’t look, you won’t see: intravital multiphoton imaging of primary and metastatic breast cancer. J. Mammary Gland Biol. Neoplasia 17, 125–129 (2012).

    Article  Google Scholar 

  37. Bonapace, L. et al. Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature 515, 130–133 (2014).

    Article  ADS  CAS  Google Scholar 

  38. Ahrné, E. et al. Evaluation and improvement of quantification accuracy in isobaric mass tag-based protein quantification experiments. J. Proteome Res. 15, 2537–2547 (2016).

    Article  Google Scholar 

  39. Post, H. et al. Robust, sensitive, and automated phosphopeptide enrichment optimized for low sample amounts applied to primary hippocampal neurons. J. Proteome Res. 16, 728–737 (2017).

    Article  CAS  Google Scholar 

  40. Wang, Y. et al. Reversed-phase chromatography with multiple fraction concatenation strategy for proteome profiling of human MCF10A cells. Proteomics 11, 2019–2026 (2011).

    Article  CAS  Google Scholar 

  41. Gaidatzis, D., Lerch, A., Hahne, F. & Stadler, M. B. QuasR: quantification and annotation of short reads in R. Bioinformatics 31, 1130–1132 (2015).

    Article  CAS  Google Scholar 

  42. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  Google Scholar 

  43. Szklarczyk, D. et al. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 45 (D1), D362–D368 (2017).

    Article  CAS  Google Scholar 

  44. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    Article  Google Scholar 

  45. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  Google Scholar 

  46. Györffy, B. et al. An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res. Treat. 123, 725–731 (2010).

    Article  Google Scholar 

Download references

Acknowledgements

We thank members of the Bentires-Alj laboratory for advice and discussion. Tissue samples that correspond to PDX1, PDX2 and PDX4–PDX11 were provided by the Cooperative Human Tissue Network, which is funded by the National Cancer Institute. Other investigators may have received specimens from the same subjects. We thank A. L. Welm (University of Utah) for the PDX3 and PDX12–PDX16 models; H.-R. Hotz for offering the QuasR and edgeR tools in the FMI Galaxy server; and S. Bichet and P. Hirschmann for help with immunohistochemistry. We are grateful for the support of the FMI, DBM and Biozentrum core facilities. Research in the Bentires-Alj laboratory is supported by the Swiss Initiative for Systems Biology- SystemsX, the European Research Council, the Swiss National Science Foundation, Novartis, the Krebsliga Beider Basel, the Swiss Cancer League, the Swiss Personalized Health Network (Swiss Personalized Oncology driver project) and the Department of Surgery of the University Hospital Basel.

Reviewer information

Nature thanks Melanie Flint and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

M.M.S.O. conceived the study, designed and performed the experiments, analysed the data, interpreted the results and wrote the manuscript. B.H. performed experiments on gene expression and helped with ROR1 shRNA and mouse experiments, analysed the data and interpreted the results. N.M. established methods and measured stress hormones levels in plasma, analysed the data and interpreted the results. J.P.C. designed experiments, analysed the data and interpreted the results. S.M. performed histopathological analysis of the PDX models, analysed the data and interpreted the results. R.O. characterized the metastatic potential of PDX models, analysed the data and interpreted the results. A. Sethi performed computational analysis of metastatic breast cancer samples, analysed the data and interpreted the results. H.K. performed fluorescence-activated cell sorting experiments, analysed the data and interpreted the results. M.-M.C. performed intravital imaging, analysed the data and interpreted the results. A. Schmidt performed proteomics and phosphoproteomics experiments, analysed the data and interpreted the results. M.B.-A. conceived the study, designed the experiments and interpreted the results. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Mohamed Bentires-Alj.

Ethics declarations

Competing interests

B.H., R.O., S.M., M.-M.C., A. Sethi, A. Schmidt and M.B.-A. declare no competing interests. N.M. is an employee of UCB Pharma. H.K. and J.P.C. are employees of Novartis. M.M.S.O. is an employee of Wellmera AG.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Increase in GR activation in breast cancer metastases.

a, Tumours and matched lung, liver, ovary and spleen metastases in the MDA-MB 231 model (haematoxylin and eosin staining). Scale bar, 200 μm. Right, frequency of metastases detected in distant organs upon tumour resection. n = 10 from 5 independent experiments. b, FACS analysis of organs affected with distant metastases in the MDA-MB 231 model. n = 10. c, Tumour growth kinetics after orthotopic transplantation in the MDA-MB 231 model (n = 9) and PDX1, PDX2 and PDX3 models (n = 5, 4 and 9 respectively). Mean ± s.e.m. Right, tumour and matched lung metastases in the PDX1 and PDX2 models (haematoxylin and eosin staining). Scale bar, 200 μm. d, Tumour and matched lung, liver or ovary metastases in the PDX3 model (haematoxylin and eosin staining). Scale bar, 200 μm. Right, frequency of metastases detected in distant organs upon tumour resection. n = 10. e, FACS analysis of PDX3 tumour and organs with matched metastases. n = 5. f, Principal component analysis of PDX3 tumours (n = 4) and matched liver (n = 3) and lung (n = 3) metastases. gi, Heat maps of genes that are differentially expressed and upstream regulator analysis, for the MDA-MB 231 model. n = 3, Fisher’s exact test. g, Tumours and liver metastases. h, Tumours and circulating tumour cells. i, Tumour and spleen. jm, Heat maps of genes that are differentially expressed for PDX models. j, PDX1 tumour (n = 4) and lung metastases (n = 3). k, PDX2 tumour and lung metastases (n = 4). l, PDX3 tumour and lung metastases (n = 4). m, PDX3 tumour and liver metastases (n = 4). n represents biological replicates (mice) in all panels. Threshold criteria for all differential-expression heat map analyses are a fold-change ≥ 2 and FDR < 0.05. The statistical approach for differential-expression analysis is provided in Methods.

Source Data

Extended Data Fig. 2 GR activation in distant metastases and circulating tumour cells.

a, ISMARA transcription-factor-activity plot of the tumour, lung, liver metastases and circulating tumour cells in the MDA-MB 231 model. n = 3 biological replicates. b, ISMARA transcription-factor activity of PDX models. n = 4 (apart from PDX1 lung metastases, n = 3) biological replicates (mice). c, GR transcription-factor binding sites in lung metastases of PDX1, PDX2, PDX3, MDA-MB 231 and BALB/c–NeuT models14. n = 4 (apart from PDX1 lung metastases, n = 3) biological replicates (mice). d, GR expression in tumours and matched lung metastases in MDA-MB 231, PDX1, PDX2 and PDX3 models. n = 3 biological replicates (mice). Scale bar,100 μm. eh, Expression of genes involved in glucocorticoid synthesis, with HPRT1 as an internal control. e, MDA-MB 231, n = 3. f, PDX1, n = 4 tumours and n = 3 lung metastases. g, PDX2, n = 4. h, PDX3 models, n = 4. Mean ± s.d., n indicates biological replicates (mice). ik, Subgroup analysis of plasma hormone levels in mice of the MDA-MB 231 model before tumour resection (M0 mice). The M0 group has been split at the median into two groups, one with smaller tumours (mean volume, 446 mm3) and the other with larger tumours (mean volume, 692 mm3). i, Cortisol levels. j, Corticosterone levels. k, Adrenocorticotropic hormone levels. Means and single data points are represented. n = 5 biological replicates (mice), two-tailed Student’s t-test.

Source Data

Extended Data Fig. 3 Glucocorticoids promote colonization via GR.

a, Expression of GR targets (qPCR). Mean ± s.d., n = 6 biological replicates, in technical duplicates; two-tailed Student’s t-test. b, Expression of GR targets three weeks after discontinuation of GR activation by dexamethasone. Mean ± s.d., n = 3 biological replicates in technical duplicates, two-tailed Student’s t-test. c, Number of metastatic foci in lungs of mice injected with 4T1 dexamethasone or vehicle-treated cells. n = 9 mice, two-tailed Student’s t-test. d, GR downregulation in MDA-MB 231 cells (left, qPCR; right, immunoblotting). Mean ± s.d., n = 3 biological replicates, two-tailed Student’s t-test. e, MDA-MB 231 cells were propagated in the presence of dexamethasone or vehicle for seven days. n = 7 biological replicates. f, GR-downregulated MDA-MB 231 cells did not express the GR-activation marker gene set upon dexamethasone treatment. Mean ± s.d., n = 3 biological replicates in technical duplicates, two-tailed Student’s t-test. g, Bioluminescence imaging 12 h, 24 h and 48 h after intravenous injection of control and dexamethasone-treated MDA-MB 231 cells transduced with control or GR shRNA. n represents biological replicates (mice), mean ± s.d., two-tailed Student’s t-test. h, Bioluminescence imaging of mice two weeks after intravenous injection. n = 15 for vehicle + control shRNA, dexamethasone + control shRNA, and dexamethasone + GR shRNA 2 (shGR2); n = 14 for dexamethasone + GR shRNA 1 (shGR1); n = 13 for vehicle + GR shRNA 2; n = 12 for vehicle + GR shRNA 1. Three independent experiments; n represents biological replicates (mice), two-tailed Student’s t-test. i, Bioluminescence imaging two weeks after intravenous injection of GR-activated, mifepristone- or vehicle-treated MDA-MB 231 cells. n = 10 mice, 2 independent experiments, two-tailed Student’s t-test. j, Kaplan–Meier survival analysis of mice upon intravenous injection of GR-activated, mifepristone- or vehicle-treated MDA-MB 231 cells. n = 10 mice for vehicle and dexamethasone groups; n = 9 for mifepristone group. Two independent experiments, two-tailed log-rank test, vehicle-treated versus mifepristone-treated groups, P = 0.054. In box plots, the centre line indicates the median, the box extends from the 25th to 75th percentiles and whiskers extend to the most extreme data points.

Source Data

Extended Data Fig. 4 Dexamethasone offsets the response to paclitaxel.

a, Kaplan–Meier survival analysis of mice intravenously injected with 4T1 GR-activated or control cells. Dexamethasone decreases 4T1 response to paclitaxel in vivo. Paclitaxel was administrated five days after 4T1 cell inoculation. n = 9 control; n = 10 dexamethasone-treated biological replicates (mice) per group; 2 mice were censored; two-tailed log-rank test. b, Dexamethasone offsets paclitaxel effect in the MDA-MB 231 model. Analysis of colonization potential under paclitaxel treatment of MDA-MB 231 cells transduced with one of the two GR shRNAs or control shRNA, and treated with dexamethasone or vehicle, intravenously injected into NSG mice. Two paclitaxel injections (15 and 22 days after cell injection). Mean ± s.d., n = 5 mice per group, two-tailed Student’s t-test. c, Bioluminescence imaging corresponding to day 21 of b. d, Kaplan–Meier survival analysis of mice shown in b. n = 5 biological replicates (mice) per group, two-tailed log-rank test.

Source Data

Extended Data Fig. 5 Dexamethasone reduces overall survival and GR downregulation increases cancer cell dissemination from the primary site.

a, b, Upon tumour removal from the 4th mammary gland, randomized mice were treated with dexamethasone or vehicle on 5 consecutive days (intraperitoneal injection of 0.1 mg kg−1 dexamethasone once daily). Kaplan–Meier survival analysis in PDX1 model (n = 8 control; n = 7 dexamethasone-treated biological replicates (mice)) (a) or 4T1 model (n = 8 control; n = 9 dexamethasone-treated biological replicates (mice)) (b). Two 4T1 mice were censored. Two-tailed log-rank test (a, b). c, GR downregulation in MDA-MB 231 cells does not affect tumour volume, relative to tumour cells transduced with control shRNA, at resection. Mean ± s.d., n = 14 biological replicates (mice), pooled data from 3 independent experiments, two-tailed Student’s t-test. d, Circulating tumour cell count measured by the number of in vitro propagated colonies upon circulating tumour cell isolation from peripheral blood of tumour-bearing mice at tumour resection time point. Mean ± s.d., n = 5 biological replicates (mice), two-tailed Student’s t-test. e, In vivo bioluminescence imaging upon tumour removal. In the box plot, the centre line is the median, the box extends from the 25th to 75th percentiles, and whiskers extend to the most extreme data points. n = 17 control shRNA; n = 10 GR shRNA 1 and 2 biological replicates (mice), pooled data from 3 independent experiments, two-tailed Student’s t-test. f, Kaplan–Meier survival analysis of mice upon of removal of MDA-MB 231 tumours transduced with control shRNA or one of the two GR shRNAs, and treatment with dexamethasone or vehicle. n = 13 biological replicates (mice) per group, pooled data from 3 independent experiments, two-tailed log-rank test. g, Tumour volumes at resection. In vitro dexamethasone- or vehicle-treated MDA-MB 231 cells transduced with control shRNA, GR shRNA 1 or GR shRNA 2 inoculated into the mammary fat pad of NSG mice. n = 13 control shRNA; n = 8 GR shRNA 1 and 2 biological replicates (mice), pooled data from 2 independent experiments, two-tailed Student’s t-test. All tumours in all experiments were resected at the same time point.

Source Data

Extended Data Fig. 6 Differential expression of protein kinases in tumours and matched metastases.

ad, Expression of protein kinases in MDA-MB 231 model, n = 3 biological replicates (mice) (a); PDX1 model, n = 4 tumour and n = 3 matched lung metastases; biological replicates (mice) (b); PDX2 model, n = 4 biological replicates (mice) (c); and PDX3 model, n = 4 biological replicates (mice) (d). The threshold criteria used for the analysis are fold-change ≥ 2 and P < 0.05. Further details of the statistical analysis are provided in Methods.

Extended Data Fig. 7 Differential protein abundance upon GR activation.

a, Volcano plot of protein abundance after GR activation in MDA-MB 231 cells. n = 3 control; n = 4 dexamethasone-treated biological replicates, Bayes-moderated t-statistics, P values corrected for multiple testing using the Benjamini–Hochberg method, calculations performed in R using the LIMMA package, Bioconductor. b, Heat map of differentially abundant proteins in dexamethasone-treated and vehicle-treated (control) cells. n = 3 control; n = 4 dexamethasone-treated biological replicates; FDR < 0.05, Bayes-moderated t-statistics; P values were corrected for multiple testing using the Benjamini–Hochberg method, calculations performed in R using the LIMMA package, Bioconductor. c, Abundance of proteins used for generation of the GR activation signature. n = 3 control; n = 4 dexamethasone-treated biological replicates, mean ± s.d., two-tailed Student’s t-test. d, Pathway enrichment analysis of all phospho-proteins with significant abundance changes against all phospho-proteins quantified as a background using MetaCore (Clarivate Analytics). Enrichment P values and FDRs were determined by the software-specific algorithms using default parameters.

Source Data

Extended Data Fig. 8 GR activation increases the expression of kinases that are predictive of survival in breast cancer.

Survival based on the expression of the protein kinase signature that is upregulated in the metastases. a, Relapse-free survival, two-tailed log-rank test. b, Distant-metastasis-free survival, two-tailed log-rank test. c, Postprogression survival, two-tailed log-rank test. Number of patients (n) and P values are presented in the panels. d, Individual protein kinases, relapse-free survival, n = 1,764, two-tailed log-rank test. e, Co-occurrence of GR and protein kinases in publically available breast cancer datasets, Fisher’s exact test, n = 2,509 (refs. 27,28).

Extended Data Fig. 9 ROR1 expression in breast cancer and metastases.

a, Relapse-free survival analysis of patients with the ROR1 signature (G-2-0, Kaplan–Meier), n = 4,029, two-tailed log-rank test. b, GR-activation signature correlates with increased levels of ROR1 in breast cancer metastases. n = 21 lymph node; n = 34 liver metastases. Pearson correlation. c, Co-occurrence of GR-activation gene signature with GR and ROR1, n = 2,509 (refs. 27,28), Fisher’s exact test. d, Breast cancers that express high levels of GR mRNA were enriched in the claudinlow profile, n = 299 (refs. 27,28).

Extended Data Fig. 10 Dexamethasone increases metastases and precipitates death, via ROR1.

a, ROR1 expression in in vitro-propagated control and GR-downregulated cells. Mean ± s.d., n = 6 biological replicates, two-tailed Student’s t-test. b, c, ROR1 qPCR. RNA from tissues of mice injected with control or GR-downregulated cells in tumours (n = 4 biological replicates (mice)) (b) and lung metastases (n = 4, vehicle + control shRNA, dexamethasone + control shRNA and dexamethasone +GR shRNA 1 or 2; n = 3 vehicle + GR shRNA 1 or 2, biological replicates (mice) in technical duplicates or triplicates) (c). Mean ± s.d., two-tailed Student’s t-test. d, Levels of WNT5A protein in supernatant of dexamethasone-treated or vehicle-treated MDA-MB 231 cells transduced with control or one of the two GR shRNAs. Mean ± s.d., n = 3 biological replicates, two-tailed Student’s t-test. e, WNT5A qPCR in dexamethasone-treated or vehicle-treated MDA-MB 231 cells transduced with control or one of the two GR shRNAs. Mean ± s.d., n = 4 biological replicates in technical triplicates, two-tailed Student’s t-test. f, Levels of WNT5A protein in tumours transduced with control or one of the two GR shRNAs, and their matched metastases. Mean ± s.d., n = 3 biological replicates (mice) in technical duplicates, two-tailed Student’s t-test. g, Pearson correlation of GR activation, ROR1, WNT5A and Wnt signalling pathway members in breast cancer metastases. n = 88 breast cancer metastases; n = 21 lymph node; n = 34 liver; n = 7 bone metastases. h, ROR1 downregulation in MDA-MB 231 cells. Mean ± s.d., n = 3 biological replicates in technical duplicates, two-tailed Student’s t-test, qPCR (top) and flow cytometry (bottom). ik, Kaplan–Meier survival analysis of mice intravenously inoculated with vehicle-treated or dexamethasone-treated MDA-MB 231cells transduced with control shRNA (i), ROR1 shRNA 1 (shROR1-1) (j) or ROR1 shRNA 2 (shROR1-2) (k). n = 5, two-tailed log-rank test. l, Kaplan–Meier survival analysis of mice injected in the mammary fat pad with MDA-MB 231 cells transduced with control or one of the two ROR1 shRNAs. n = 21 control shRNA; n = 23 ROR1 shRNA 1 or 2, two-tailed log-rank test. mo, Kaplan–Meier survival analysis of mice inoculated in the mammary fat pad with MDA-MB 231 cells transduced with control shRNA (m), ROR1 shRNA 1 (n) or ROR1 shRNA 2 (o), propagated in the presence of dexamethasone or vehicle. n = 8 control shRNA; n = 9 ROR1 shRNA 1 or 2, two-tailed log-rank test.

Source Data

Supplementary information

Supplementary Figure 1

This file contains the uncropped western blots.

Reporting Summary

Supplementary Table 1

Characterization of the PDX models. Characterization of 17 primary-derived xenografts (PDX) or cell lines implanted into the 4th mammary gland of NOD-scid IL2rγnull (NSG) immunodeficient mice. Upon primary tumour resection, metastatic potential (metastatic onset and pattern) was analysed.

Supplementary Table 2

Gene Set Enrichment Analysis (GSEA). a, MDA-MB 231, n=3; b, PDX3 n=4; c, PDX2 n=4; d, PDX1 n=4 tumours and n=3 lung metastases; n represents biological replicates (mice). GSEA was performed using the JAVA application from the Broad Institute v2.0. The GSEA use the Kolmogorov-Smirnov statistic.

Supplementary Table 3

Proteomic analysis. Protein abundance analysis of DEX GR-activated and vehicle-treated MDA-MB 231, n=3 “vehicle” biological replicates, n=4 “DEX” biological replicates, Bayes moderated t-statistics, P-values were corrected for multiple testing using the Benjamini−Hochberg method.

Supplementary Table 4

Phosphoproteomic analysis. Phosphoproteomic analysis of DEX GR-activated and vehicle-treated MDA-MB 231, n=3 “vehicle” biological replicates, n=4 “DEX” biological replicates. Used statistical analysis described in the methods section.

Supplementary Table 5

Proteomic and Phosphoproteomic analysis. a, YAP targets protein abundance- DEX GR-activated and vehicle-treated MDA-MB 231. b-c, Phosphoproteomic analysis of DEX GR-activated and vehicle-treated MDA-MB 231: b, HIPPO pathway and c, MAPK-ERK pathway. d, Proteomic and Phosphoproteomic analysis of Wnt signalling members. Bayes moderated t-statistics, P-values were corrected for multiple testing using the Benjamini−Hochberg method. n=3 “vehicle” biological replicates, n=4 “DEX” biological replicates for all provided analysis. Further details on statistical analysis are provided in the methods section.

Video 1: Intravital tumour imaging.

Time-lapse sequence (30-min; 2D) of GFP-labelled shCTRL MDA-MB-231 tumour cells (green) in the 4th mammary gland of a living mouse, n=4. Scale bar 100 μm.

Video 2: Intravital tumour imaging.

Time-lapse sequence (30-min; 2D) of GFP-labelled shGR1 MDA-MB-231 tumour cells (green) in the 4th mammary gland of a living mouse, n=3. Scale bar 100 μm.

Video 3: Intravital tumour imaging.

Time-lapse sequence (30-min; 2D) of GFP-labelled shGR2 MDA-MB-231 tumour cells (green) in the 4th mammary gland of a living mouse, n=4. Scale bar 100 μm.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Obradović, M.M.S., Hamelin, B., Manevski, N. et al. Glucocorticoids promote breast cancer metastasis. Nature 567, 540–544 (2019). https://doi.org/10.1038/s41586-019-1019-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1019-4

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: 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