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:

Microenvironmental control of breast cancer subtype elicited through paracrine platelet-derived growth factor-CC signaling

Abstract

Breast tumors of the basal-like, hormone receptor–negative subtype remain an unmet clinical challenge, as there is high rate of recurrence and poor survival in patients following treatment. Coevolution of the malignant mammary epithelium and its underlying stroma instigates cancer-associated fibroblasts (CAFs) to support most, if not all, hallmarks of cancer progression. Here we delineate a previously unappreciated role for CAFs as determinants of the molecular subtype of breast cancer. We identified paracrine crosstalk between cancer cells expressing platelet-derived growth factor (PDGF)-CC and CAFs expressing the cognate receptors in human basal-like mammary carcinomas. Genetic or pharmacological intervention of PDGF-CC activity in mouse models of cancer resulted in conversion of basal-like breast cancers into a hormone receptor-positive state that enhanced sensitivity to endocrine therapy in previously resistant tumors. We conclude that specification of breast cancer to the basal-like subtype is under microenvironmental control and is therapeutically actionable.

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: Epithelial expression of PDGF-CC is associated with a poor outcome in patients with breast carcinoma.
Figure 2: Expression of PDGF-CC in breast carcinomas is associated with the hormone receptor–negative, basal-like molecular subtype.
Figure 3: CAF-derived factors whose expression is induced by PDGF-CC reduce the sensitivity of breast tumor cells to endocrine therapy.
Figure 4: Genetic or pharmacological targeting of PDGF-CC induces expression of ERα and sensitizes tumors to endocrine therapy.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Perou, C.M. et al. Molecular portraits of human breast tumours. Nature 406, 747–752 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Goldhirsch, A. et al. Thresholds for therapies: highlights of the St Gallen International Expert Consensus on the primary therapy of early breast cancer 2009. Ann. Oncol. 20, 1319–1329 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sørlie, T. et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl. Acad. Sci. USA 98, 10869–10874 (2001).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Goldhirsch, A. et al. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 24, 2206–2223 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Goldhirsch, A. et al. Strategies for subtypes—dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann. Oncol. 22, 1736–1747 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Prat, A. et al. Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast 24 (Suppl. 2), S26–S35 (2015).

    Article  PubMed  Google Scholar 

  7. Ignatiadis, M. & Sotiriou, C. Luminal breast cancer: from biology to treatment. Nat. Rev. Clin. Oncol. 10, 494–506 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Voduc, K.D. et al. Breast cancer subtypes and the risk of local and regional relapse. J. Clin. Oncol. 28, 1684–1691 (2010).

    Article  PubMed  Google Scholar 

  9. Prat, A. & Perou, C.M. Deconstructing the molecular portraits of breast cancer. Mol. Oncol. 5, 5–23 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Hanahan, D. & Coussens, L.M. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21, 309–322 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Pietras, K. & Ostman, A. Hallmarks of cancer: interactions with the tumor stroma. Exp. Cell Res. 316, 1324–1331 (2010).

    Article  CAS  PubMed  Google Scholar 

  12. Kalluri, R. & Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 6, 392–401 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Cortez, E., Roswall, P. & Pietras, K. Functional subsets of mesenchymal cell types in the tumor microenvironment. Semin. Cancer Biol. 25, 3–9 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Augsten, M. Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment. Front. Oncol. 4, 62 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Kim, H.M., Jung, W.H. & Koo, J.S. Expression of cancer-associated fibroblast related proteins in metastatic breast cancer: an immunohistochemical analysis. J. Transl. Med. 13, 222 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Anderberg, C. et al. Paracrine signaling by platelet-derived growth factor-CC promotes tumor growth by recruitment of cancer-associated fibroblasts. Cancer Res. 69, 369–378 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Pietras, K., Pahler, J., Bergers, G. & Hanahan, D. Functions of paracrine PDGF signaling in the proangiogenic tumor stroma revealed by pharmacological targeting. PLoS Med. 5, e19 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Li, X. et al. PDGF-C is a new protease-activated ligand for the PDGF α-receptor. Nat. Cell Biol. 2, 302–309 (2000).

    Article  CAS  PubMed  Google Scholar 

  19. Cao, R. et al. Angiogenesis stimulated by PDGF-CC, a novel member in the PDGF family, involves activation of PDGFR-αα and -αβ receptors. FASEB J. 16, 1575–1583 (2002).

    Article  CAS  PubMed  Google Scholar 

  20. Theurillat, J.P. et al. NY-ESO-1 protein expression in primary breast carcinoma and metastases: correlation with CD8+ T-cell and CD79a+ plasmacytic/B-cell infiltration. Int. J. Cancer 120, 2411–2417 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Falck, A.K. et al. Biomarker expression and St Gallen molecular subtype classification in primary tumours, synchronous lymph node metastases and asynchronous relapses in primary breast cancer patients with 10 years' follow-up. Breast Cancer Res. Treat. 140, 93–104 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Falck, A.K. et al. Analysis of and prognostic information from disseminated tumour cells in bone marrow in primary breast cancer: a prospective observational study. BMC Cancer 12, 403 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Guy, C.T., Cardiff, R.D. & Muller, W.J. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol. Cell. Biol. 12, 954–961 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lin, E.Y. et al. Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am. J. Pathol. 163, 2113–2126 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Ding, H. et al. A specific requirement for PDGF-C in palate formation and PDGFR-alpha signaling. Nat. Genet. 36, 1111–1116 (2004).

    Article  CAS  PubMed  Google Scholar 

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

  27. Badve, S. et al. FOXA1 expression in breast cancer—correlation with luminal subtype A and survival. Clin. Cancer Res. 13, 4415–4421 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Thorat, M.A. et al. Forkhead box A1 expression in breast cancer is associated with luminal subtype and good prognosis. J. Clin. Pathol. 61, 327–332 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. 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 

  30. Kong, S.L., Li, G., Loh, S.L., Sung, W.K. & Liu, E.T. Cellular reprogramming by the conjoint action of ERα, FOXA1, and GATA3 to a ligand-inducible growth state. Mol. Syst. Biol. 7, 526 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kojima, Y. et al. Autocrine TGF-β and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc. Natl. Acad. Sci. USA 107, 20009–20014 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Ray, P.S. et al. FOXC1 is a potential prognostic biomarker with functional significance in basal-like breast cancer. Cancer Res. 70, 3870–3876 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Chebil, G., Bendahl, P.O., Idvall, I. & Fernö, M. Comparison of immunohistochemical and biochemical assay of steroid receptors in primary breast cancer—clinical associations and reasons for discrepancies. Acta Oncol. 42, 719–725 (2003).

    Article  CAS  PubMed  Google Scholar 

  34. Hammond, M.E. et al. American Society of Clinical Oncology/College Of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J. Clin. Oncol. 28, 2784–2795 (2010).

    PubMed  PubMed Central  Google Scholar 

  35. Polyak, K. Breast cancer: origins and evolution. J. Clin. Invest. 117, 3155–3163 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gupta, P.B. et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 146, 633–644 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Lim, E. et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat. Med. 15, 907–913 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Molyneux, G. & Smalley, M.J. The cell of origin of BRCA1 mutation-associated breast cancer: a cautionary tale of gene expression profiling. J. Mammary Gland Biol. Neoplasia 16, 51–55 (2011).

    Article  PubMed  Google Scholar 

  39. Liu, S. et al. BRCA1 regulates human mammary stem/progenitor cell fate. Proc. Natl. Acad. Sci. USA 105, 1680–1685 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Molyneux, G. et al. BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells. Cell Stem Cell 7, 403–417 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Su, Y. et al. Somatic cell fusions reveal extensive heterogeneity in basal-like breast cancer. Cell Rep. 11, 1549–1563 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Yamamoto, S. et al. JARID1B is a luminal lineage-driving oncogene in breast cancer. Cancer Cell 25, 762–777 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bernardo, G.M. et al. FOXA1 represses the molecular phenotype of basal breast cancer cells. Oncogene 32, 554–563 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Sflomos, G. et al. A preclinical model for ERα-positive breast cancer points to the epithelial microenvironment as determinant of luminal phenotype and hormone response. Cancer Cell 29, 407–422 (2016).

    Article  CAS  PubMed  Google Scholar 

  45. Tam, W.L. et al. Protein kinase C α is a central signaling node and therapeutic target for breast cancer stem cells. Cancer Cell 24, 347–364 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Meng, F. et al. PDGFRα and β play critical roles in mediating Foxq1-driven breast cancer stemness and chemoresistance. Cancer Res. 75, 584–593 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Jansson, S. et al. The three receptor tyrosine kinases c-KIT, VEGFR2 and PDGFRα, closely spaced at 4q12, show increased protein expression in triple-negative breast cancer. PLoS One 9, e102176 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Horikawa, S. et al. PDGFRα plays a crucial role in connective tissue remodeling. Sci. Rep. 5, 17948 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kim, Y.J. et al. MET is a potential target for use in combination therapy with EGFR inhibition in triple-negative/basal-like breast cancer. Int. J. Cancer 134, 2424–2436 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Ho-Yen, C.M. et al. C-Met in invasive breast cancer: is there a relationship with the basal-like subtype? Cancer 120, 163–171 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Ponzo, M.G. et al. Met induces mammary tumors with diverse histologies and is associated with poor outcome and human basal breast cancer. Proc. Natl. Acad. Sci. USA 106, 12903–12908 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Graveel, C.R. et al. Met induces diverse mammary carcinomas in mice and is associated with human basal breast cancer. Proc. Natl. Acad. Sci. USA 106, 12909–12914 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Gastaldi, S. et al. Met signaling regulates growth, repopulating potential and basal cell-fate commitment of mammary luminal progenitors: implications for basal-like breast cancer. Oncogene 32, 1428–1440 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Marzec, K.A., Baxter, R.C. & Martin, J.L. Targeting insulin-like growth factor binding protein-3 signaling in triple-negative breast cancer. BioMed Res. Int. 2015, 638526 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bissell, M.J. & Hines, W.C. Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat. Med. 17, 320–329 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Marsh, T., Pietras, K. & McAllister, S.S. Fibroblasts as architects of cancer pathogenesis. Biochim. Biophys. Acta 1832, 1070–1078 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Gascard, P. & Tlsty, T.D. Carcinoma-associated fibroblasts: orchestrating the composition of malignancy. Genes Dev. 30, 1002–1019 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Brechbuhl, H.M. et al. Fibroblast subtypes regulate responsiveness of luminal breast cancer to estrogen. Clin. Cancer Res. 23, 1710–1721 (2017).

    Article  CAS  PubMed  Google Scholar 

  59. Özdemir, B.C. et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25, 719–734 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Rhim, A.D. et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 25, 735–747 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  62. Bray, N.L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    Article  CAS  PubMed  Google Scholar 

  63. Pimentel, H. et al. Differential analysis of RNA-seq incorporating quantification uncertainty. Nat. Methods 14, 687–690 (2017).

    Article  CAS  PubMed  Google Scholar 

  64. Mei, S. et al. Cistrome Data Browser: a data portal for ChIP–seq and chromatin accessibility data in human and mouse. Nucleic Acids Res. 45 D1, D658–D662 (2017).

    Article  CAS  PubMed  Google Scholar 

  65. Sikora-Wohlfeld, W., Ackermann, M., Christodoulou, E.G., Singaravelu, K. & Beyer, A. Assessing computational methods for transcription factor target gene identification based on ChIP–seq data. PLOS Comput. Biol. 9, e1003342 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. 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 

  67. Mootha, V.K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge expert help with pathology assessments from the late D. Grabau and provision of the Tam2Y cohort by M. Fernö. Further, we would like to thank D. Cao, M. O'Brien and C. Murone for their technical support and P.-O. Bendahl for statistical assistance. K.P. is the Göran & Birgitta Grosskopf Professor at Lund University. The research presented herein was supported by grants from the following agencies to K.P.: a Consolidator Grant from the European Research Council (the TUMORGAN project, grant 309322), the Swedish Research Council, the Swedish Cancer Society, the STARGET consortium (a Swedish Research Council Linnaeus network), BioCARE and Lund University. U.E. acknowledges funding support from the Swedish Research Council, the Swedish Cancer Society, Karolinska Institutet and Ludwig Institute for Cancer Research. A.M.S. acknowledges funding support from National Health and Medical Research Council (NHMRC) Fellowship 1084178 and Grant 10927888 and the Operational Infrastructure Support Program provided by the Victorian Government, Australia.

Author information

Authors and Affiliations

Authors

Contributions

P.R. generated and analyzed data and conceived the study. M. Bocci, M. Bartoschek, H.L., G.K., S.J., S.L., J.S., C.L., P.E., S.R., C.A., E. Cortez, L.H.S., C.O.-P., B.K.H. and J.H. generated and analyzed data. E. Cordero, I.J.G.B. and E.L. generated data. A.O. provided exclusive reagents. M.H. analyzed data. L.R., H.M. and A.M.S. analyzed data and provided exclusive reagents. U.E. analyzed data, provided exclusive reagents and conceived the study. K.P. generated and analyzed data, conceived the study, managed the study and wrote the manuscript.

Corresponding author

Correspondence to Kristian Pietras.

Ethics declarations

Competing interests

K.P., U.E. and P.R. are named inventors on Patent Cooperation Treaty (PCT) application no. PCT/EP2016/077295, which is related to the findings of the current study. K.P., U.E. and A.M.S. are shareholders of Paracrine Therapeutics, which develops inhibitory agents to PDGF-CC.

Supplementary information

Supplementary Figures & Tables

Supplementary Figures 1–12 & Supplementary Tables 1–8 (PDF 4066 kb)

Life Sciences Reporting Summary (PDF 133 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roswall, P., Bocci, M., Bartoschek, M. et al. Microenvironmental control of breast cancer subtype elicited through paracrine platelet-derived growth factor-CC signaling. Nat Med 24, 463–473 (2018). https://doi.org/10.1038/nm.4494

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4494

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