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.

A model of breast cancer heterogeneity reveals vascular mimicry as a driver of metastasis

Subjects

Abstract

Cancer metastasis requires that primary tumour cells evolve the capacity to intravasate into the lymphatic system or vasculature, and extravasate into and colonize secondary sites1. Others have demonstrated that individual cells within complex populations show heterogeneity in their capacity to form secondary lesions2,3,4,5. Here we develop a polyclonal mouse model of breast tumour heterogeneity, and show that distinct clones within a mixed population display specialization, for example, dominating the primary tumour, contributing to metastatic populations, or showing tropism for entering the lymphatic or vasculature systems. We correlate these stable properties to distinct gene expression profiles. Those clones that efficiently enter the vasculature express two secreted proteins, Serpine2 and Slpi, which were necessary and sufficient to program these cells for vascular mimicry. Our data indicate that these proteins not only drive the formation of extravascular networks but also ensure their perfusion by acting as anticoagulants. We propose that vascular mimicry drives the ability of some breast tumour cells to contribute to distant metastases while simultaneously satisfying a critical need of the primary tumour to be fed by the vasculature. Enforced expression of SERPINE2 and SLPI in human breast cancer cell lines also programmed them for vascular mimicry, and SERPINE2 and SLPI were overexpressed preferentially in human patients that had lung-metastatic relapse. Thus, these two secreted proteins, and the phenotype they promote, may be broadly relevant as drivers of metastatic progression in human cancer.

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: Clonal analysis of 4T1 transplantation by molecular barcoding.
Figure 2: Focused analysis of a subset of 4T1 clones throughout metastatic disease progression.
Figure 3: Serpine2 and Slpi are regulators of intravasation into the cardiovascular system.
Figure 4: Vascular mimicry drives metastatic progression.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

All raw and processed data is available through the Gene Expression Omnibus (GEO) under the accession number GSE63180.

References

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

    Article  CAS  Google Scholar 

  2. Miller, F. R., Miller, B. E. & Heppner, G. H. Characterization of metastatic heterogeneity among subpopulations of a single mouse mammary tumor: heterogeneity in phenotypic stability. Invasion Metastasis 3, 22–31 (1983)

    CAS  PubMed  Google Scholar 

  3. Miller, B. E., Miller, F. R., Wilburn, D. & Heppner, G. H. Dominance of a tumor subpopulation line in mixed heterogeneous mouse mammary tumors. Cancer Res. 48, 5747–5753 (1988)

    CAS  PubMed  Google Scholar 

  4. Fidler, I. J. & Kripke, M. L. Metastasis results from preexisting variant cells within a malignant tumor. Science 197, 893–895 (1977)

    Article  ADS  CAS  Google Scholar 

  5. Fidler, I. J. Tumor heterogeneity and the biology of cancer invasion and metastasis. Cancer Res. 38, 2651–2660 (1978)

    CAS  PubMed  Google Scholar 

  6. Ding, L. et al. Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature 464, 999–1005 (2010)

    Article  ADS  CAS  Google Scholar 

  7. Lohr, J. G. et al. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell 25, 91–101 (2014)

    Article  CAS  Google Scholar 

  8. Mullighan, C. G. et al. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science 322, 1377–1380 (2008)

    Article  ADS  CAS  Google Scholar 

  9. Navin, N. et al. Tumour evolution inferred by single-cell sequencing. Nature 472, 90–94 (2011)

    Article  ADS  CAS  Google Scholar 

  10. Braun, S. et al. Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II, or III breast cancer. N. Engl. J. Med. 342, 525–533 (2000)

    Article  CAS  Google Scholar 

  11. Harrell, J. C. et al. Genomic analysis identifies unique signatures predictive of brain, lung, and liver relapse. Breast Cancer Res. Treat. 132, 523–535 (2012)

    Article  CAS  Google Scholar 

  12. Amiano, N. O. et al. Anti-tumor effect of SLPI on mammary but not colon tumor growth. J. Cell. Physiol. 228, 469–475 (2013)

    Article  CAS  Google Scholar 

  13. Fayard, B. et al. The serine protease inhibitor protease nexin-1 controls mammary cancer metastasis through LRP-1-mediated MMP-9 expression. Cancer Res. 69, 5690–5698 (2009)

    Article  CAS  Google Scholar 

  14. Martin, K. J., Patrick, D. R., Bissell, M. J. & Fournier, M. V. Prognostic breast cancer signature identified from 3D culture model accurately predicts clinical outcome across independent datasets. PLoS ONE 3, e2994 (2008)

    Article  ADS  Google Scholar 

  15. Rosso, M. et al. Secretory Leukocyte Protease Inhibitor (SLPI) expression downregulates E-cadherin, induces beta-catenin re-localisation and triggers apoptosis-related events in breast cancer cells. Biol. Cell 106, 308–322 (2014)

    Article  CAS  Google Scholar 

  16. Sayers, K. T., Brooks, A. D., Sayers, T. J. & Chertov, O. Increased secretory leukocyte protease inhibitor (SLPI) production by highly metastatic mouse breast cancer cells. PLoS ONE 9, e104223 (2014)

    Article  ADS  Google Scholar 

  17. Valiente, M. et al. Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell 156, 1002–1016 (2014)

    Article  CAS  Google Scholar 

  18. Maniotis, A. J. et al. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am. J. Pathol. 155, 739–752 (1999)

    Article  CAS  Google Scholar 

  19. Hendrix, M. J., Seftor, E. A., Hess, A. R. & Seftor, R. E. Vasculogenic mimicry and tumour-cell plasticity: lessons from melanoma. Nature Rev. Cancer 3, 411–421 (2003)

    Article  CAS  Google Scholar 

  20. Folberg, R., Hendrix, M. J. & Maniotis, A. J. Vasculogenic mimicry and tumor angiogenesis. Am. J. Pathol. 156, 361–381 (2000)

    Article  CAS  Google Scholar 

  21. Ruf, W. et al. Differential role of tissue factor pathway inhibitors 1 and 2 in melanoma vasculogenic mimicry. Cancer Res. 63, 5381–5389 (2003)

    CAS  PubMed  Google Scholar 

  22. Cao, Z. et al. Tumour vasculogenic mimicry is associated with poor prognosis of human cancer patients: a systemic review and meta-analysis. Eur.J. Cancer 49, 3914–3923 (2013)

    Article  Google Scholar 

  23. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009)

    Article  Google Scholar 

  24. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nature Methods 9, 357–359 (2012)

    Article  CAS  Google Scholar 

  25. Anders, S., Pyl, P. T. & Huber, W. HTSeq — A Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015)

    Article  CAS  Google Scholar 

  26. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Howard Hughes Medical Institute as well as grants from the NIH (G.J.H.). This work was performed with assistance from CSHL Shared Resources, which are funded, in part, by the Cancer Center Support Grant 5P30CA045508. We thank M. Mosquera, M. Cahn, J. Coblentz, L. Bianco for support with mouse work; J. Ratcliff and P. Moody for assistance with flow cytometry; D. Hoppe, A. Nourjanova, R. Puzis for histology support and S. Hearn for microscopy assistance. We thank E. Hodges and E. Lee for support with next-generation sequencing; K. Chang for the lentiviral barcode library; E. Mardis and C. Sawyers for comments on the manuscript. E.W. is a Starr Centennial Scholar and is supported by a fellowship from the Boehringer Ingelheim Fonds. J.C.H. and C.M.P. were supported by funds from the NCI Breast SPORE program (P50-CA58223-09A1), the Breast Cancer Research Foundation and the Triple Negative Breast Cancer Foundation. S.R.V.K. is supported by a fellowship from The Hope Funds For Cancer Research.

Author information

Authors and Affiliations

Authors

Contributions

E.W., G.J.H. and S.R.V.K. designed the experiments. E.W., M.S., S.G., C.A.H., A.L.G., A.R.M., N.E., A.M.W., S.Y.K., S.D. and S.R.V.K. performed experiments and analysed data. J.C.H. and C.M.P. assisted with human expression data. A.D.S. helped with analysis. J.E.W. performed histological analysis. E.W., G.J.H. and S.R.V.K. wrote the paper. G.J.H. and S.R.V.K. supervised the research.

Corresponding author

Correspondence to Gregory J. Hannon.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Overview of vector plasmids and clonal abundances in the primary tumour and secondary sites after 4T1 transplantation.

a, Schematic of the retroviral barcode vector (used for Figs 1, 2d, 4e and Extended Data Fig. 8f). b, Schematic of the lentiviral barcode vector (used for Fig. 2a–c). c, Schematic of the retroviral shRNA vector used for single gene knockdown. d, Schematic of the tandem retroviral shRNA vector used for double gene knockdown. e, Schematic of the retroviral cDNA vector used for gene overexpression. f, Distribution of clone abundances in the primary tumours and secondary sites for the 4T1 clones that engrafted and contributed to tumour formation in all five animals after orthotopic injection. g, Overlap of abundant clones in the primary tumour, lymph nodes, blood and in all blood-borne metastases (BBM: lung, liver and brain) (P < 0.001, hypergeometric test). h, Overlap of the abundant clones in the lymph nodes, liver, lung and brain.

Extended Data Figure 2 Morphology and proliferation rates of all clonal lines grown.

Microscopic phase-contrast images of the 23 clonal lines discussed in Figs 2, 3, 4. Doubling times (DT), as calculated over a 3-day period, are colour-coded in the image borders. Original magnification, ×10.

Extended Data Figure 3 Tumour growth and metastases rates for individually injected clones and clonal composition of intracardiacally injected pool.

a, Primary tumour volumes resulting from orthotopically injected individual clonal cell lines. Measurements were taken 14 and 24 days after injection (n = 4 mice). For comparison, the clonal composition of the primary tumour when the pool of clones is injected orthotopically is shown below. b, The percentage of lung metastatic burden for the animals discussed in a 24 days after injection (n = 4 mice). For comparison, the clonal composition of the lung when the pool of clones is injected orthotopically is shown below. c, A comparison of the clonal composition of CTCs and lung metastases when the pool of 23 clonal lines was orthotopically injected (data from Fig. 2c) versus injected into the left cardiac ventricle of mice (n = 10 mice for orthotopic injections and n = 8 mice for intracardiac injections). d, Clonal compositions of the primary tumours, CTCs and lung metastases analysed in Fig. 2d. e, Clonal composition of CTCs and lung metastases after orthotopic injection when the individual clones were frozen down 3–5 times and cultured for a week each time (n = 5 mice). For all bar graphs, error bars extend to the values q3 + w(q3 − q1), and q1 − w(q3 − q1), in which w is 1.5 and q1 and q3 are the twenty-fifth and seventy-fifth percentiles.

Extended Data Figure 4 SERPINE2 and SLPI expression in human patients and Serpine2 and Slpi shRNAs abundances in lung and CTCs after orthotopic and intracardiac injection.

a, Analysis of Serpine2 and Slpi human orthologues SERPINE2 and SLPI, respectively in basal, Her2 or claudin-low breast cancer patients with no relapse as compared to patients with relapse in the lung (P < 0.005, Wilcoxon rank-sum, n indicated in figure). b, A non-targeting shRNA and two targeting each of Serpine2 and Slpi were infected separately into 4T1-T cells. After selection, the separately infected cells were pooled in equal amounts with the remaining 22 clonal lines and orthotopically injected into mice. The proportions of shRNAs shown are that of the CTCs and lung metastases in comparison to those in the primary tumour (P < 0.01, Wilcoxon rank-sum, n = 10 mice). c, 4T1-T cells were infected with a non-targeting shRNA or shRNAs targeting Serpine2 or Slpi. The shRNAs were cotained in a vector that constitutively expressed mCherry. The infected cells were then pooled with the other 22 clones and injected orthotopically (n = 10 mice). Shown are representative images of the resultant mCherry positive lung metastatic nodules. d, Quantification of all mCherry positive metastatic lung nodules in all mice (P < 0.01, Wilcoxon rank-sum, n = 10 mice). e, 4T1-parental cells were infected with a non-targeting shRNA or shRNAs targeting Serpine2 or Slpi. In each case the cells were then orthotopically injected into mice. Shown are representative images of haematoxylin-and-eosin-stained lung sections (n = 10 mice). f, Quantification of lung metastatic nodules in all haematoxylin-and-eosin-stained sections described in e (P < 0.01 and P < 0.005 for single- and double-knockdowns, respectively, Wilcoxon rank-sum, n = 10 mice). For all box plots, the edges of the box are the twenty-fifth and seventy-fifth percentiles. The error bars extend to the values q3 + w(q3 − q1), and q1 − w(q3 − q1), in which w is 1.5 and q1 and q3 are the twenty-fifth and seventy-fifth percentiles.

Extended Data Figure 5 Leakiness index of clonal cell lines.

a, DAPI and dextran Alexa 647 (+ dextran inverted)-stained tumour sections from orthotopic tumours derived from parental-4T1, 4T1-L and 4T1-T cells. b, Leakiness index of primary tumours resulting from orthotopic injection of parental-4T1, 4T1-L and 4T1-T cells (P < 0.05, Wilcoxon rank-sum, n = 15 mice). c, The leakiness index of primary tumours derived from 4T1-T cells that have been infected with either a non-targeting shRNA or an shRNA targeting Serpine2 or Slpi (P < 0.03, Wilcoxon rank-sum, n = 18 mice). For all box plots, the edges of the box are the twenty-fifth and seventy-fifth percentiles. The error bars extend to the values q3 + w(q3 − q1), and q1 − w(q3 − q1), in which w is 1.5 and q1 and q3 are the twenty-fifth and seventy-fifth percentiles.

Extended Data Figure 6 Vascular mimicry in 4T1-T and pooled primary tumours.

a, PAS/CD31 (left) and PAS/mCherry (right) staining of serially sectioned 4T1-T derived primary tumours, where the tumour cells constitutively express an empty vector or mCherry. Orange arrows indicate CD31+ endothelial blood vessels and blue arrows show PAS+/CD31 channels. The purple arrows indicate mCherry-positive tumour cells adjacent to PAS+ channels (green scale bars = 100 μm, grey scale bars = 50 μm). b, PAS/CD31 (left) and PAS/mCherry (right) staining of serially sectioned primary tumours resulting from orthotopic injection of a pool of the 23 clones, in which the 4T1-T clonal line constitutively expresses mCherry. Orange arrows indicate CD31+ endothelial blood vessels and blue arrows show PAS+/CD31 channels. The purple arrows indicate mCherry-positive tumour cells adjacent to PAS+ channels (green scale bars = 100 μm, grey scale bars = 50 μm).

Extended Data Figure 7 Overexpression of Serpine2 and Slpi in parental 4T1 and clonal cell lines.

a, Images of HUVECs, parental 4T1, 4T1-L, 4T1-I, 4T1-E and 4T1-T cells grown on Matrigel to assess tube formation ability. b, Number of tubular structures identified per ×5 microscopic field when 4T1-T cells had been infected with a non-targeting shRNA or shRNAs targeting Serpine2 or Slpi and grown on Matrigel (P < 0.0002, Wilcoxon rank-sum, n = 8 fields). c, Number of tubular structures identified per ×5 microscopic image when non-intravasating 4T1 clonal lines had been infected with an empty retroviral vector or vectors for overexpression of Serpine2 or Slpi (P < 0.03, Wilcoxon rank-sum, with the exception of 4T1-B overexpressing Slpi, n = 4 fields). d, Relative CTC proportions of the cells described in c after they had been pooled with the remaining clonal lines and orthotopically injected (P < 0.05, Wilcoxon rank-sum, with the exception of 4T1-F and 4T1-B Slpi, n = 10 mice). e, PAS+/CD31 channels in primary tumours resulting from orthotopic injection of parental 4T1 cells infected with an empty retroviral vector or vectors for overexpression of Serpine2 or Slpi (P < 0.01, Wilcoxon rank-sum, n = 10 fields). f, Quantification of the number of lung metastatic nodules resulting from the tumours described in e after 18 days (n = 4 mice). g, Quantification of the number of lung metastatic nodules resulting from the tumours described in e after 24 days (P < 0.05, Friedman, n = 10 mice). For all box plots, the edges of the box are the twenty-fifth and seventy-fifth percentiles. The error bars extend to the values q3 + w(q3 − q1), and q1 − w(q3 − q1), in which w is 1.5 and q1 and q3 are the twenty-fifth and seventy-fifth percentiles.

Extended Data Figure 8 Overexpression of SERPINE2 and SLPI in human breast cancer cell lines.

a, MDA-MB-231 cells infected with an empty overexpression vector plated on Matrigel (top) and MDA-MB-231 cells overexpressing SERPINE2 (NM_0016216) plated on Matrigel (bottom). b, Full quantification of tubular structures in basal cell lines MDA-MB-231 and MDA-MB-436 when infected with an empty overexpression vector or when infected with vectors for overexpressing SERPINE2 or SLPI (P < 0.05, Friedman test, for SERPINE2 isoform NM_0016216 and SLPI, n = 4 fields). c, Quantification of CD31/PAS+ channels in MDA-MB-436-derived primary tumours cells that have been infected with an empty retroviral vector or vectors for overexpression of SERPINE2 or SLPI (P < 0.002, Wilcoxon rank-sum, for SERPINE2 isoform NM_001136528 and SLPI, n = 20 fields for empty, SERPINE2 NM_001136528 and NM_0016216 and n = 25 fields for SLPI). d, The percentage of lung metastatic burden after orthotopic injection of MDA-MB-231 cells that have been infected with an empty retroviral vector or vectors for overexpression of SERPINE2 or SLPI (P < 0.05, Wilcoxon rank-sum, n = 9 mice). e, Quantification of the number of lung metastatic nodules after orthotopic injection of MDA-MB-436 cells that have been infected with an empty retroviral vector or vectors for overexpression of SERPINE2 or SLPI (Wilcoxon rank-sum P < 0.05 for SERPINE2 isoform NM_001136528 and SLPI, n = 9 mice). f, Sub-clonal analysis of MDA-MB-436 cells that have been infected with an empty retroviral vector or vectors for overexpression of SERPINE2 or SLPI. Each cell line was infected with a barcode library and then the number of clones were quantified in the primary tumour, cardiovascular CTCs and lung metastatic cells (P < 0.02, Wilcoxon rank-sum, for SERPINE2 isoform NM_001136528 and SLPI in CTCs and lung metastasis, n = 5 mice). For all box plots, the edges of the box are the twenty-fifth and seventy-fifth percentiles. The error bars extend to the values q3 + w(q3 − q1) and q1 − w(q3 − q1), where w is 1.5 and q1 and q3 are the twenty-fifth and seventy-fifth percentiles.

Extended Data Figure 9 Effect of warfarin on leakiness, metastasis and clonal abundance.

a, Levels of prothrombin fragments F1 and F2 (active component of blood coagulation), as quantified by ELISA, after administration of warfarin in the drinking water of mice (P < 0.01, Wilcoxon rank-sum, n = 4 mice). b, DAPI and dextran Alexa 647 staining to visualize vascular leakage in primary tumours derived from the pool of 23 clonal lines. Animals were administered drinking water either with or without 10 mg ml−1warfarin. c, Leakiness index of primary tumours resulting from orthotopic injection of the pool of 23 clonal lines. After injection mice were administered drinking water with no warfarin or water containing 10 mg ml−1 warfarin (P < 0.000005, Wilcoxon rank-sum, n = 10 mice). d, Counts of lung metastatic nodules in animals that were injected intravenously with the pool of 23 clonal lines administered drinking water with no warfarin or water containing 10 mg ml−1 of warfarin (P < 0.05, Wilcoxon rank-sum, n = 10 mice). e, The pool of 23 clonal lines was injected orthotopically and the mice administered drinking water with no warfarin or 10 mg ml−1 warfarin. The percentage abundance of each clone in the cardiovascular CTCs of animals (either pre- or post-injection, P < 0.002, n = 7 mice for no warfarin, n = 7 mice for pre-injection and n = 5 mice for post-injection). f, The clone proportions in the resultant primary tumours and lung metastases described in (e) (P < 0.003, Wilcoxon rank-sum, for lung metastases, n = 7 mice for no warfarin, n = 7 mice for pre-injection and n = 5 mice for post-injection). For all box plots, the edges of the box are the twenty-fifth and seventy-fifth percentiles. The error bars extend to the values q3 + w(q3 − q1), and q1 − w(q3 − q1), in which w is 1.5 and q1 and q3 are the twenty-fifth and seventy-fifth percentiles.

Supplementary information

Supplementary Table 1

This file shows the hazard analysis of SLPI and SERPINE2 in human patients. (XLSX 10 kb)

Supplementary Table 2

This file shows the knock-down efficiency of shRNAs and gene expression levels after cDNA overexpression. (XLSX 43 kb)

Supplementary Table 3

This file shows the viral integration site of each barcode. (XLSX 29 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wagenblast, E., Soto, M., Gutiérrez-Ángel, S. et al. A model of breast cancer heterogeneity reveals vascular mimicry as a driver of metastasis. Nature 520, 358–362 (2015). https://doi.org/10.1038/nature14403

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14403

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