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.

  • Review Article
  • Published:

Stem cells and breast cancer: A field in transit

Key Points

  • Stem cells have a long life and a large replicative potential, making them good candidates for the cells of origin of cancer.

  • The adult mammary gland requires stem cells, or a stem-cell-like activity, to fulfil the demands of pregnancy-dependent epithelial expansion and replace cells that are lost through routine cell turnover.

  • Evidence for the existence of specialized adult mammary stem cells comes from transplantation, retroviral tagging and X-chromosome-linked gene-inactivation studies.

  • Current evidence points to an undifferentiated, suprabasal cell, with a slow proliferative rate that has characteristics in common with stem cells from other tissues, as the best candidate for an adult mammary epithelial stem cell.

  • Experimental evidence from the transplantation of tumour-cell subpopulations and from animal models supports the view that breast stem cells are the cells in which mammary cancers are initiated.

  • Therapeutic or prophylactic targeting of breast stem cells provides a novel approach to breast cancer treatment that is aimed directly at the cells of origin of the tumour.

Abstract

Evidence that supports the existence of a tumour stem cell for breast cancer has given fresh impetus to the search for an adult mammary epithelial stem cell in the normal breast. Such a cell might be responsible for routine tissue renewal and the massive expansion in epithelial tissue that the breast undergoes during pregnancy, and might be the cell of origin of most, if not all, breast tumours. So, what evidence is there that an adult mammary epithelial stem cell exists, what is its possible identity and what opportunities might the identification of such a cell present for the development of novel therapeutic and prophylactic strategies for treating breast cancer?

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

Access options

Buy this article

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

Figure 1: The human breast and rodent mammary gland.
Figure 2: The terminal end bud.
Figure 3: The role of stem cells in the adult mammary gland.
Figure 4: Field cancerization.

Similar content being viewed by others

References

  1. Seaberg, R. M. & Van Der Kooy, D. Stem and progenitor cells: the premature desertion of rigorous definitions. Trends Neurosci. 26, 125–131 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Smith, C. Hematopoietic stem cells and hematopoiesis. Cancer Control 10, 9–16 (2003).

    Article  PubMed  Google Scholar 

  3. Galli, R. et al. Neural stem cells: an overview. Circ. Res. 92, 598–608 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Alonso, L. & Fuchs, E. Stem cells in the skin: waste not, Wnt not. Genes Dev. 17, 1189–1200 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Potten, C. S. & Loeffler, M. Stem cells: attributes, cycles, spirals, pitfalls and uncertainties. Lessons for and from the crypt. Development 110, 1001–1020 (1990).

    CAS  PubMed  Google Scholar 

  6. Potten, C. S. et al. The small intestine as a model for evaluating adult tissue stem cell drug targets. Cell Prolif. 36, 115–129 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hedrick, S. M. T-cell development: bottoms-up. Immunity 16, 619–622 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. Ohneda, K. & Yamamoto, M. Roles of hematopoietic transcription factors Gata-1 and Gata-2 in the development of red blood cell lineage. Acta Haematol. 108, 237–245 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Frye, M. et al. Evidence that Myc activation depletes the epidermal stem cell compartment by modulating adhesive interactions with the local microenvironment. Development 130, 2793–2808 (2003). An important demonstration of how perturbation of the expression pattern of a single gene can affect the balance between stem and transit cells.

    Article  CAS  PubMed  Google Scholar 

  10. Owens, D. M. & Watt, F. M. Contribution of stem cells and differentiated cells to epidermal tumours. Nature Rev. Cancer 3, 444–451 (2003).

    Article  CAS  Google Scholar 

  11. Morgan, J. E. & Partridge, T. A. Muscle satellite cells. Int. J. Biochem. Cell Biol. 35, 1151–1156 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Hawke, T. J. & Garry, D. J. Myogenic satellite cells: physiology to molecular biology. J. Appl. Physiol. 91, 534–551 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Seale, P. & Rudnicki, M. A. A new look at the origin, function, and 'stem-cell' status of muscle satellite cells. Dev. Biol. 218, 115–124 (2000).

    Article  CAS  PubMed  Google Scholar 

  14. Williams, J. M. & Daniel, C. W. Mammary ductal elongation: differentiation of myoepithelium and basal lamina during branching morphogenesis. Dev. Biol. 97, 274–290 (1983).

    Article  CAS  PubMed  Google Scholar 

  15. Srinivasan, K. et al. Netrin-1/neogenin interaction stabilizes multipotent progenitor cap cells during mammary gland morphogenesis. Dev. Cell 4, 371–382 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Debnath, J. et al. Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods 30, 256–268 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Smith, G. H. Experimental mammary epithelial morphogenesis in an in vivo model: evidence for distinct cellular progenitors of the ductal and lobular phenotype. Breast Cancer Res. Treat. 39, 21–31 (1996).

    Article  CAS  PubMed  Google Scholar 

  18. Kordon, E. C. & Smith, G. H. An entire functional mammary gland may comprise the progeny from a single cell. Development 125, 1921–1930 (1998). References 17 and 18 provide direct experimental evidence for the existence of dedicated mammary epithelial stem cells.

    CAS  PubMed  Google Scholar 

  19. Wagner, K. U. et al. An adjunct mammary epithelial cell population in parous females: its role in functional adaptation and tissue renewal. Development 129, 1377–1386 (2002).

    CAS  PubMed  Google Scholar 

  20. Welm, B. E. et al. Sca-1pos cells in the mouse mammary gland represent an enriched progenitor cell population. Dev. Biol. 245, 42–56 (2002). This study was the first to demonstrate prospective enrichment of mammary epithelial cells for cells with stem-cell-like properties.

    Article  CAS  PubMed  Google Scholar 

  21. Naylor, S. et al. Retroviral expression of Wnt-1 and Wnt-7b produces different effects in mouse mammary epithelium. J. Cell Sci. 113, 2129–2138 (2000).

    CAS  PubMed  Google Scholar 

  22. Diallo, R. et al. Monoclonality in normal epithelium and in hyperplastic and neoplastic lesions of the breast. J. Pathol. 193, 27–32 (2001).

    Article  CAS  PubMed  Google Scholar 

  23. Chepko, G. & Dickson, R. B. Ultrastructure of the putative stem cell niche in rat mammary epithelium. Tissue Cell 35, 83–93 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Chepko, G. & Smith, G. H. Three division-competent, structurally-distinct cell populations contribute to murine mammary epithelial renewal. Tissue Cell 29, 239–253 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Ellis, S. & Capuco, A. Cell proliferation in bovine mammary epithelium: identification of the primary proliferative cell population. Tissue Cell 34, 155 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Smith, C. A. et al. Basal clear cells of the normal human breast. Virchows Arch. A Pathol. Anat. Histopathol. 402, 319–329 (1984).

    Article  CAS  PubMed  Google Scholar 

  27. Smith, C. A. et al. Epithelial cells of the normal human breast. J. Pathol. 146, 221–226 (1985).

    Article  CAS  PubMed  Google Scholar 

  28. Ferguson, D. J. An ultrastructural study of mitosis and cytokinesis in normal 'resting' human breast. Cell Tissue Res. 252, 581–587 (1988). References 23–28 provide the basis for an understanding of the heterogeneity of the mammary epithelium and identify the basal clear cell as a putative stem cell.

    Article  CAS  PubMed  Google Scholar 

  29. Clarke, R. B. et al. Dissociation between steroid receptor expression and cell proliferation in the human breast. Cancer Res. 57, 4987–4991 (1997).

    CAS  PubMed  Google Scholar 

  30. Clarke, R. B. et al. P27kip1 expression indicates that steroid receptor-positive cells are a non-proliferating, differentiated subpopulation of the normal human breast epithelium. Eur. J. Cancer 36, S28–S29 (2000).

    Article  CAS  PubMed  Google Scholar 

  31. Shoker, B. S. et al. Estrogen receptor-positive proliferating cells in the normal and precancerous breast. Am. J. Pathol. 155, 1811–1815 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zeps, N. et al. Estrogen receptor-negative epithelial cells in mouse mammary gland development and growth. Differentiation 62, 221–226 (1998).

    Article  CAS  PubMed  Google Scholar 

  33. Zeps, N. et al. Detection of a population of long-lived cells in mammary epithelium of the mouse. Cell Tissue Res. 286, 525–536 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. O'Hare, M. J. et al. Characterization in vitro of luminal and myoepithelial cells isolated from the human mammary gland by cell sorting. Differentiation 46, 209–221 (1991).

    Article  CAS  PubMed  Google Scholar 

  35. Pechoux, C. et al. Human mammary luminal epithelial cells contain progenitors to myoepithelial cells. Dev. Biol. 206, 88–99 (1999).

    Article  CAS  PubMed  Google Scholar 

  36. Stingl, J. et al. Phenotypic and functional characterization in vitro of a multipotent epithelial cell present in the normal adult human breast. Differentiation 63, 201–213 (1998).

    Article  CAS  PubMed  Google Scholar 

  37. Stingl, J. et al. Characterization of bipotent mammary epithelial progenitor cells in normal adult human breast tissue. Breast Cancer Res. Treat. 67, 93–109 (2001). References 35–37, together with reference 50, relate in situ heterogeneity of the breast epithelium to in vitro differentiative capacity.

    Article  CAS  PubMed  Google Scholar 

  38. Dontu, G. et al. In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev. 17, 1253–1270 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dundas, S. R. et al. Characterization of luminal and basal cells flow-sorted from the adult rat mammary parenchyma. J. Cell Sci. 100, 459–471 (1991).

    PubMed  Google Scholar 

  40. Smalley, M. J. Cell Sorting and Clonal Analysis of Mouse Mammary Luminal Epithelial and Myoepithelial Cells. Thesis, Univ. London (1995).

    Google Scholar 

  41. Smalley, M. J. et al. Clonal characterization of mouse mammary luminal epithelial and myoepithelial cells separated by fluorescence-activated cell sorting. In Vitro Cell. Dev. Biol. Anim. 34, 711–721 (1998).

    Article  CAS  PubMed  Google Scholar 

  42. Smalley, M. J. et al. Differentiation of separated mouse mammary luminal epithelial and myoepithelial cells cultured on ehs matrix analyzed by indirect immunofluorescence of cytoskeletal antigens. J. Histochem. Cytochem. 47, 1513–1524 (1999).

    Article  CAS  PubMed  Google Scholar 

  43. Goodell, M. A. et al. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J. Exp. Med. 183, 1797–1806 (1996). The first description of the 'side population' phenomenon.

    Article  CAS  PubMed  Google Scholar 

  44. Zhou, S. et al. The ABC transporter BCRP1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nature Med. 7, 1028–1034 (2001).

    Article  CAS  PubMed  Google Scholar 

  45. Bunting, K. D. ABC transporters as phenotypic markers and functional regulators of stem cells. Stem Cells 20, 11–20 (2002).

    Article  CAS  PubMed  Google Scholar 

  46. Bhattacharya, S. et al. Direct identification and enrichment of retinal stem cells/progenitors by Hoechst dye efflux assay. Invest. Ophthalmol. Vis. Sci. 44, 2764–2773 (2003).

    Article  PubMed  Google Scholar 

  47. Kubota, H. et al. Spermatogonial stem cells share some, but not all, phenotypic and functional characteristics with other stem cells. Proc. Natl Acad. Sci. USA 100, 6487–6492 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhou, S. et al. Bcrp1 gene expression is required for normal numbers of side population stem cells in mice, and confers relative protection to mitoxantrone in hematopoietic cells in vivo. Proc. Natl Acad. Sci. USA 99, 12339–12344 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Alvi, A. J. et al. Functional and molecular characterisation of mammary side population cells. Breast Cancer Res. 5, R1–R8 (2002). The first demonstration of side population in human breast epithelium.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Gudjonsson, T. et al. Isolation, immortalization, and characterization of a human breast epithelial cell line with stem cell properties. Genes Dev. 16, 693–706 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chepko, G. & Smith, G. H. Mammary epithelial stem cells: our current understanding. J. Mammary Gland Biol. Neoplasia 4, 35–52 (1999).

    Article  CAS  PubMed  Google Scholar 

  52. Reya, T. et al. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Al-Hajj, M., Wicha, M. S., Benito-Hernandez, A., Morrison, S. J. & Clarke, M. F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl Acad. Sci. USA 100, 6890 (2003). Describes the prospective isolation of candidate breast-tumour stem cells.

    Article  CAS  Google Scholar 

  54. Perez-Losada, J. & Balmain, A. Stem-cell hierarchy in skin cancer. Nature Rev. Cancer 3, 434–443 (2003).

    Article  CAS  Google Scholar 

  55. Boulanger, C. A. & Smith, G. H. Reducing mammary cancer risk through premature stem cell senescence. Oncogene 20, 2264–2272 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. Russo, J. & Russo, I. H. Differentiation and breast cancer. Medicina 57, S81–S91 (1997).

    Google Scholar 

  57. Russo, J. et al. Influence of age and parity on the development of the human breast. Breast Cancer Res. Treat. 23, 211–218 (1992).

    Article  CAS  PubMed  Google Scholar 

  58. Layde, P. M. et al. The independent associations of parity, age at first full term pregnancy, and duration of breastfeeding with the risk of breast cancer. Cancer and steroid hormone study group. J. Clin. Epidemiol. 42, 963–973 (1989).

    Article  CAS  PubMed  Google Scholar 

  59. Chie, W. C. et al. Age at any full-term pregnancy and breast cancer risk. Am. J. Epidemiol. 151, 715–722 (2000).

    Article  CAS  PubMed  Google Scholar 

  60. Musey, V. C. et al. Long term effects of a first pregnancy on the hormonal environment: estrogens and androgens. J. Clin. Endocrinol. Metab. 64, 111–118 (1987).

    Article  CAS  PubMed  Google Scholar 

  61. Musey, V. C. et al. Long-term effect of a first pregnancy on the secretion of prolactin. N. Engl. J. Med. 316, 229–234 (1987).

    Article  CAS  PubMed  Google Scholar 

  62. Slaughter, D. P. et al. 'Field cancerization' in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer 6, 963–968 (1953). The original proposal of the field-cancerization concept.

    Article  CAS  PubMed  Google Scholar 

  63. Braakhuis, B. J. et al. A genetic explanation of Slaughter's concept of field cancerization: evidence and clinical implications. Cancer Res. 63, 1727–1730 (2003).

    CAS  PubMed  Google Scholar 

  64. Lakhani, S. R. et al. Genetic alterations in 'normal' luminal and myoepithelial cells of the breast. J. Pathol. 189, 496–503 (1999).

    Article  CAS  PubMed  Google Scholar 

  65. Stappenbeck, T. S. et al. Molecular features of adult mouse small intestinal epithelial progenitors. Proc. Natl Acad. Sci. USA 100, 1004–1009 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Ramalho-Santos, M. et al. 'Stemness': transcriptional profiling of embryonic and adult stem cells. Science 298, 597–600 (2002).

    Article  CAS  PubMed  Google Scholar 

  67. Ivanova, N. B. et al. A stem cell molecular signature. Science 298, 601–604 (2002). References 65–67 describe comparative gene-profile expression studies of different stem-cell types.

    Article  CAS  PubMed  Google Scholar 

  68. Alexander, C. M. et al. Syndecan-1 is required for Wnt-1-induced mammary tumorigenesis in mice. Nature Genet. 25, 329–332 (2000).

    Article  CAS  PubMed  Google Scholar 

  69. Watt, F. M. The stem cell compartment in human interfollicular epidermis. J. Dermatol. Sci. 28, 173–180 (2002).

    Article  CAS  PubMed  Google Scholar 

  70. Rosner, A. et al. Pathway pathology: histological differences between ErbB/Ras and wnt pathway transgenic mammary tumors. Am. J. Pathol. 161, 1087–1097 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lewis, J. Notch signalling and the control of cell fate choices in vertebrates. Semin. Cell Dev. Biol. 9, 583–589 (1998).

    Article  CAS  PubMed  Google Scholar 

  72. Brunner, H. G. et al. P63 gene mutations and human developmental syndromes. Am. J. Med. Genet. 112, 284–290 (2002).

    Article  PubMed  Google Scholar 

  73. Lin, H. The stem-cell niche theory: lessons from flies. Nature Rev. Genet. 3, 931–940 (2002).

    Article  CAS  PubMed  Google Scholar 

  74. Nishimura, E. K. et al. Dominant role of the niche in melanocyte stem-cell fate determination. Nature 416, 854–860 (2002).

    Article  CAS  PubMed  Google Scholar 

  75. Kai, T. & Spradling, A. An empty Drosophila stem cell niche reactivates the proliferation of ectopic cells. Proc. Natl Acad. Sci. USA 100, 4633–4638 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Barcellos-Hoff, M. H. & Ravani, S. A. Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 60, 1254–1260 (2000).

    CAS  PubMed  Google Scholar 

  77. De Ome, K. B. et al. Development of mammary tumours from hyperplastic alveolar nodules transplanted into gland-free mammary fat pads of female C3H mice. Cancer Res. 78, 515–520 (1959).

    Google Scholar 

  78. Cairns, J. Mutation selection and the natural history of cancer. Nature 255, 197–200 (1975).

    Article  CAS  PubMed  Google Scholar 

  79. Potten, C. S. et al. Intestinal stem cells protect their genome by selective segregation of template DNA strands. J. Cell Sci. 115, 2381–2388 (2002).

    CAS  PubMed  Google Scholar 

  80. Merok, J. R. et al. Cosegregation of chromosomes containing immortal DNA strands in cells that cycle with asymmetric stem cell kinetics. Cancer Res. 62, 6791–6795 (2002).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge all those who have contributed to this field but whose work could not be included due to space constraints. The authors would also like to thank G. Chepko for access to unpublished material and all those who have critically read this manuscript and offered suggestions for its improvement. Work in the authors' laboratory is funded by Breakthrough Breast Cancer and Cancer Research UK.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alan Ashworth.

Related links

Related links

DATABASES

Cancer.gov

breast cancer

LocusLink

KIP1

ESA

MUC1

CD10

integrin-α6

cytokeratin 19

SCA1

ABCG2

CD44

CD24

BRCA2

TP53

syndecan 1

FURTHER INFORMATION

The Biology of the Mammary Gland home page

Breakthough Breast Cancer

The Breakthrough Centre

The Stem Cell Database

Glossary

TERMINAL DIFFERENTIATION

The process by which stem-cell or transit-amplifying-cell progeny commit to becoming the mature, fully functioning cell type of a tissue. Probably irreversible.

MICROENVIRONMENT

The local growth conditions of the cell, formed by interactions between the extracellular matrix, blood supply, local cell types, and circulating hormones and growth factors.

LUMINAL EPITHELIAL CELLS

The cells that line the lumen — also used to refer to the non-myoepithelial component of the mammary epithelium in general.

MYOEPITHELIAL CELLS

Contractile cells that form a basket-like network around the secretory alveoli and a sheath around the ducts. These squeeze the milk down the ducts and out of the nipple in response to the hormone oxytocin.

APOPTOSIS

Programmed cell death that is characterized by regulated DNA digestion and phagocytosis of cell debris with minimal tissue inflammation.

LUMEN

The space in the centre of the mammary ducts and alveoli into which milk is secreted, and along which it passes to the nipple.

SECRETORY ALVEOLI

Structures that resemble bunches of grapes in the mammary epithelium that produce milk products. They are few in number in the virgin or non-parous animal, but appear in huge numbers during pregnancy and fill the gland during lactation.

INVOLUTION

The name given to the process of apoptosis and tissue remodelling by which the mammary gland changes from the epithelial-cell-rich lactational state to the epithelial-cell-sparse non-parous state following weaning of offspring.

ADIPOSE MATRIX

The fatty connective tissue of the mammary gland that supports the epithelium.

RETROVIRAL TAGGING

Uses retroviral infection of cells as a lineage marker to enable the progeny of marked cells to be followed over many generations. Retroviral sequences incorporated into host DNA are detected by Southern blotting, and common insertion patterns are used to infer lineage relationships.

X-CHROMOSOME INACTIVATION

The process by which one X chromosome is stably transcriptionally inactivated. This occurs in almost all mammalian female (XX) cells to achieve comparable gene dosage to XY males.

MAMMARY TREE

The complete epithelial structure of the mammary gland that, with its branching ducts and associated alveoli, resembles the branching of a tree.

TERMINAL DUCTAL LOBULO-ALVEOLAR UNITS

(TDLUs). The structures in the human breast that are equivalent to the secretory alveoli of the rodent. They have a higher order of organization than rodent alveoli, and consist of alveoli that are clustered around a distinct duct and ductal side branches. They also have a distinctive stromal component — the intralobular fibroblasts — as opposed to the interlobular fibroblasts of the breast connective tissue.

HISTOLOGY

The analysis of tissue samples by routine pathological methods and light microscopy.

ULTRASTRUCTURE

Cell and tissue morphology at the electron-microscope level.

CYTOKERATIN

A structural cellular protein that is typical of epithelia.

XENOGRAFTS

The growth of primary human tissue, cancer cells or cancer cell lines in animal hosts in an attempt to recapitulate aspects of normal, or cancerous, cell growth and morphology.

MAMMOSPHERES

Balls of mammary epithelial cells that form under specialized culture conditions in vitro and that are capable of functional differentiation in the correct hormonal environment. Mammospheres that grow in suspension culture are analogous to neurospheres grown under similar conditions.

ABC TRANSPORTER PROTEINS

Transmembrane protein pumps that can eliminate various small molecules from the cell.

POST-TRANSPLANT BONE-MARROW RECONSTITUTING ACTIVITY

The ability of bone-marrow cells, transplanted into an animal that has received lethal whole-body irradiation, to regenerate the haematopoietic stem-cell compartment and keep the animal alive.

CHIMERIC

Composed of cells that originate from more than one source.

LOSS OF HETEROZYGOSITY

The elimination of the remaining normal copy of a gene from a cell that already carries one mutant copy of that gene. This results in the complete loss of function of the gene from that cell.

PROPHYLACTIC

Preventative treatment.

HEPARAN-SULPHATE PROTEOGLYCANS

Cell-surface molecules that might have a role in regulating cell–matrix or receptor–ligand interactions.

WNT1

A potent mammary oncoprotein that is activated by the mouse mammary tumour virus. Its normal role is as a developmental regulator.

RNA INTERFERENCE

A technique that triggers a natural defence mechanism against certain viruses and tricks the cells into suppressing expression of endogenous genes.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Smalley, M., Ashworth, A. Stem cells and breast cancer: A field in transit. Nat Rev Cancer 3, 832–844 (2003). https://doi.org/10.1038/nrc1212

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc1212

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing