Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-2xdlg Total loading time: 0 Render date: 2024-07-06T00:48:51.082Z Has data issue: false hasContentIssue false

4 - Mammospheres and breast carcinoma

from SECTION I - CHARACTERIZATION OF CANCER STEM CELLS

Published online by Cambridge University Press:  15 December 2009

Massimiliano Bonafe
Affiliation:
University of Bologna, Bologna, Italy
Get access

Summary

In this chapter, recent literature on human and mouse normal mammary gland and breast cancer stem/progenitor cells will be reviewed. Part of these data will be gathered from studies performed on stem/progenitor cells in vitro expanded as multicellular spheroids, called mammospheres (MS). It will be highlighted that the available data support the notion that normal and putative cancer stem cell gene expression patterns are close to the basal-like phenotype. Such a phenotype identifies the normal mammary gland cell compartment that is supposed to harbor stem/progenitor cells as well as a subset of highly aggressive/metastatic breast carcinomas lacking estrogen receptor alpha (ERα) expression and overexpressing cytokeratin 5/6 (CK5/6), epidermal growth factor receptor (EGFr), interleukin-6 (IL-6), Notch-3, Jagged-1, carbonic anhydrase isoenzyme-IX (CA-IX), vimentin, and SNAI gene family members. It will be then argued that understanding the regulation of basal-like gene expression profile is expected to provide an insight on normal and cancer stem cell–controlling mechanisms. In particular, it will be pointed out that the pro-inflammatory cytokine IL-6, the stem cell regulatory gene Notch-3, its ligand Jagged-1, and the hypoxia survival gene CA-IX share a molecular machinery that promotes invasive behavior and survival of normal mammary gland and breast cancer stem cells. It will be also reported that, in normal mammary gland and breast cancer cells, the SNAI gene family members govern the onset of an undifferentiated mesenchymal phenotype that parallels the gain of a basal-like/stem cell–like phenotype.

Type
Chapter
Information
Cancer Stem Cells , pp. 49 - 67
Publisher: Cambridge University Press
Print publication year: 2009

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Hoshino, K.Transplantability of mammary gland in brown fat pads of mice. Nature. 1967 Jan 14;213(5072):194–5.CrossRefGoogle Scholar
Kordon, EC and Smith, GH. An entire functional mammary gland may comprise the progeny from a single cell. Development. 1998 May;125(10):1921–30.Google ScholarPubMed
Shackleton, M, et al. Generation of a functional mammary gland from a single stem cell. Nature. 2006 Jan 5;439(7072):84–8.CrossRefGoogle ScholarPubMed
Stingl, J, et al. Purification and unique properties of mammary epithelial stem cells. Nature. 2006 Feb 23;439(7079):993–7.Google ScholarPubMed
Tsai, YC, et al. Contiguous patches of normal human mammary epithelium derived from a single stem cell: implications for breast carcinogenesis. Cancer Res. 1996 Jan 15;56(2):402–4.Google ScholarPubMed
Diallo, R, et al. Monoclonality in normal epithelium and in hyperplastic and neoplastic lesions of the breast. J Pathol. 2001 Jan;193(1):27–32.3.0.CO;2-H>CrossRefGoogle ScholarPubMed
Boecker, W, et al. Evidence of progenitor cells of glandular and myoepithelial cell lineages in the human adult female breast epithelium: a new progenitor (adult stem) cell concept. Cell Prolif. 2003 Oct;36(Suppl 1):73–84.CrossRefGoogle ScholarPubMed
Korsching, E, et al. Cytogenetic alterations and cytokeratin expression patterns in breast cancer: integrating a new model of breast differentiation into cytogenetic pathways of breast carcinogenesis. Lab Invest. 2002 Nov;82(11):1525–33.CrossRefGoogle ScholarPubMed
Villadsen, R, et al. Evidence for a stem cell hierarchy in the adult human breast. J Cell Biol. 2007 Apr 9;177(1):87–101.CrossRefGoogle ScholarPubMed
Gudjonsson, T, et al. Isolation, immortalization, and characterization of a human breast epithelial cell line with stem cell properties. Genes Dev. 2002 Mar 15;16(6):693–706.CrossRefGoogle ScholarPubMed
Stingl, J and Caldas, C. Molecular heterogeneity of breast carcinomas and the cancer stem cell hypothesis. Nat Rev Cancer. 2007 Oct;7(10):791–9.CrossRefGoogle ScholarPubMed
Sleeman, KE, et al. Dissociation of estrogen receptor expression and in vivo stem cell activity in the mammary gland. J Cell Biol. 2007 Jan 1;176(1):19–26.CrossRefGoogle ScholarPubMed
Vaillant, F, et al. The emerging picture of the mouse mammary stem. Stem Cell Rev. 2007 Jun;3(2):114–23.CrossRefGoogle ScholarPubMed
Asselin-Labat, ML, et al. Steroid hormone receptor status of mouse mammary stem cells. J Natl Cancer Inst. 2006 Jul 19;98(14):1011–4.CrossRefGoogle ScholarPubMed
Regan, J and Smalley, M. Prospective isolation and functional analysis of stem and differentiated cells from the mouse mammary gland. Stem Cell Rev. 2007 Jun;3(2):124–36.CrossRefGoogle ScholarPubMed
Nielsen, TO, et al. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res. 2004 Aug 15;10(16):5367–74.CrossRefGoogle ScholarPubMed
Fulford, LG, et al. Specific morphological features predictive for the basal phenotype in grade 3 invasive ductal carcinoma of breast. Histopathology. 2006 Jul;49(1):22–34.CrossRefGoogle ScholarPubMed
Rakha, EA, et al. Impact of basal-like breast carcinoma determination for a more specific therapy. Pathobiology. 2008 Jun;75(2):95–103.CrossRefGoogle ScholarPubMed
Livasy, CA, et al. Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol. 2006 Feb;19(2):264–71.CrossRefGoogle ScholarPubMed
Reis-Filho, JS, et al. Triple negative tumours: a critical reviewHistopathology. 2008 Jan;52(1):108–18.CrossRefGoogle ScholarPubMed
Dontu, G, et al. In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev. 2003 May 15;17(10):1253–70.CrossRefGoogle ScholarPubMed
Dontu, G, et al. Role of Notch signaling in cell-fate determination of human mammary stem/progenitor cells. Breast Cancer Res. 2004 Aug;6(6):R605–15.CrossRefGoogle ScholarPubMed
Dontu, G, and Wicha, MS. Survival of mammary stem cells in suspension culture: implications for stem cell biology and neoplasia. J Mammary Gland Biol Neoplasia. 2005 Jan;10(1):75–86.CrossRefGoogle ScholarPubMed
Dontu, G, et al. Stem cells in mammary development and carcinogenesis: implications for prevention and treatment. Stem Cell Rev. 2005;1(3):207–13.CrossRefGoogle ScholarPubMed
Liu, S, et al. BRCA1 regulates human mammary stem/progenitor cell fate. Proc Natl Acad Sci U S A. 2008 Feb 5;105(5):1680–5.CrossRefGoogle ScholarPubMed
Sansone, P, et al. IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. J Clin Invest. 2007 Dec;117(12):3988–4002.CrossRefGoogle ScholarPubMed
Sansone, P, et al. p66Shc/Notch-3 interplay controls self-renewal and hypoxia survival in human stem/progenitor cells of the mammary gland expanded in vitro as mammospheres. Stem Cells. 2007 Mar;25(3):807–15.CrossRefGoogle ScholarPubMed
Storci, G, et al. The basal-like breast carcinoma phenotype is regulated by SLUG gene expression. J Pathol. 2008 Jan;214(1):25–37.CrossRefGoogle ScholarPubMed
Liu, R, et al. The prognostic role of a gene signature from tumorigenic breast-cancer cells. N Engl J Med. 2007 Jan 18;356(3):217–26.CrossRefGoogle ScholarPubMed
Bertucci, F, et al. A gene signature in breast cancer. N Engl J Med. 2007 May 3;356(18):1887–8.Google ScholarPubMed
Ben-Porath, I, et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 2008 May;40(5):499–507.CrossRefGoogle ScholarPubMed
Nakshatri, H, et al. Breast cancer stem cells and intrinsic subtypes: controversies rage on. Curr Stem Cell Res Ther. 2009 Jan;4(1):50–60.CrossRefGoogle Scholar
James, CR, et al. BRCA1, a potential predictive biomarker in the treatment of breast cancer. Oncologist. 2007 Feb;(12):142–50.CrossRefGoogle ScholarPubMed
Herschkowitz, JI, et al. Identification of conserved gene expression features between murine mammari carcinoma models and human breast tumors. Genome Biol. 2007 May;8(5):R76.CrossRefGoogle Scholar
Al-Hajj, M, et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003 Apr 1;100(7):3983–88.CrossRefGoogle ScholarPubMed
Cobaleda, C, et al. The emerging picture of human breast cancer as a stem cell-based disease. Stem Cell Rev. 2008 Summer;4(2):67–79.CrossRefGoogle ScholarPubMed
Wicha, MS, et al. Cancer stem cells: an old idea – a paradigm shift. Cancer Res. 2006 Feb 15;66(4):1883–90.CrossRefGoogle ScholarPubMed
Honeth, G, et al. The CD44+/CD24- phenotype is enriched in basal-like breast tumors. Breast Cancer Res. 2008 Jun;10(3):R53.CrossRefGoogle ScholarPubMed
Charafe-Jauffret, E, et al. Gene expression profiling of breast cell lines identifies potential new basal markers. Oncogene. 2006 Apr 6;25(15):2273–84.CrossRefGoogle ScholarPubMed
Shipitsin, M, et al. Molecular definition of breast tumor heterogeneityCancer Cell. 2007 Mar;11(3):259–73.CrossRefGoogle ScholarPubMed
Bertucci, F, et al. How different are luminal A and basal breast cancers?Int J Cancer. 2009 Mar 15;124(6):1338–48.CrossRefGoogle ScholarPubMed
Reedijk, M, et al. High-level coexpression of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res. 2005 Sep 15; 65(18):8530–7.CrossRefGoogle ScholarPubMed
Reedijk, M, et al. JAG1 expression is associated with a basal phenotype and recurrence in lymph node-negative breast cancer. Breast Cancer Res Treat. 2008 Oct;111(3):439–48.CrossRefGoogle ScholarPubMed
Dickson, BC, et al. High-level JAG1 mRNA and protein predict poor outcome in breast cancer. Mod Pathol. 2007 Jun;20(6):685–93.CrossRefGoogle ScholarPubMed
Brennan, DJ, et al. CA IX is an independent prognostic marker in premenopausal breast cancer patients with one to three positive lymph nodes and a putative marker of radiation resistance. Clin Cancer Res. 2006 Nov;12(21):439–48.CrossRefGoogle Scholar
Kuroda, N, et al. Basal-like carcinoma of the breast: further evidence of the possibility that most metaplastic carcinomas may be actually basal-like carcinomas. Med Mol Morphol. 2008 Jun;41(2):117–20.CrossRefGoogle ScholarPubMed
Sarrió, D, et al. Epithelial-mesenchymal transition in breast cancer relates to the basal-like phenotype. Cancer Res. 2008 Feb 15;68(4):989–97.CrossRefGoogle ScholarPubMed
Rakha, EA, et al. Breast carcinoma with basal differentiation: a proposal for pathology definition based on basal cytokeratin expression. Histopathology. 2007 Mar;50(4): 434–8.CrossRefGoogle ScholarPubMed
Sleeman, KE, et al. Dissociation of estrogen receptor expression and in vivo stem cell activity in the mammary gland. J Cell Biol. 2007 Jan 1;176(1):19–26.CrossRefGoogle ScholarPubMed
Liao, MJ, et al. Hypoxia-inducible factor-1alpha is a key regulator of metastasis in a transgenic model of cancer initiation and progression. Cancer Res. 2007 Sep 1;67(17):8131–8.CrossRefGoogle Scholar
Mani, SA, et al. The epithelial-mesenchymal transition generates cells with properties of stem Cells. Cell. 2008 May 16;133(4):704–15.CrossRefGoogle ScholarPubMed
Phillips, TM, et al. The response of CD24(−/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst. 2006 Dec 20;98(24):1777–85.CrossRefGoogle Scholar
Ponti, D, et al. Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res. 2005 Jul 1;65(13):5506–11.CrossRefGoogle ScholarPubMed
Horwitz, KB, et al. Rare steroid receptor-negative basal-like tumorigenic cells in luminal subtype human breast cancer xenografts. Proc Natl Acad Sci U S A. 2008 Apr 15;105(15):5774–9.CrossRefGoogle ScholarPubMed
Sheridan, C, et al. CD44+/CD24− breast cancer cells exhibit enhanced invasive properties: an early step necessary for metastasis. Breast Cancer Res. 2006;8(5):R59.CrossRefGoogle ScholarPubMed
Fillmore, CM, et al. Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res. 2008 Mar;10(2):R25.CrossRefGoogle ScholarPubMed
Bolós, V, et al. The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: a comparison with Snail and E47 repressors. J Cell Sci. 2003 Feb 1;116(Pt 3):499–511.CrossRefGoogle ScholarPubMed
Morel, AP, et al. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS ONE. 2008 Aug 6;3(8):e2888.CrossRefGoogle ScholarPubMed
Gordon, , et al. Breast cell invasive potential relates to the myoepithelial phenotypeInt J Cancer. 2003 Aug 10;106(1):8–16.CrossRefGoogle ScholarPubMed
Neve, RM, et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006 Dec;10(6):515–27.CrossRefGoogle Scholar
Hugo, H, et al. Epithelial – mesenchymal and mesenchymal – epithelial transitions in carcinoma progression. J Cell Physiol. 2007 Nov;213(2):374–83.CrossRefGoogle ScholarPubMed
Korsching, E, et al. The origin of vimentin expression in invasive breast cancer: epithelial-mesenchymal transition, myoepithelial histogenesis or histogenesis from progenitor cells with bilinear differentiation potential. J Pathol. 2005 Aug;206(4):451–7.CrossRefGoogle ScholarPubMed
Blick, T, et al. Epithelial mesenchymal transition traits in human breast cancer cell lines. Clin Exp Metastasis. 2008 May;25(6):629–42.CrossRefGoogle ScholarPubMed
Knüpfer, H and Preiss, R. Significance of interleukin-6 (IL-6) in breast cancer. Breast Cancer Res Treat. 2007 Apr;102(2):129–35.CrossRefGoogle Scholar
Lien, HC, et al. Molecular signatures of metaplastic carcinoma of the breast by large-scale transcriptional profiling: identification of genes potentially related to epithelial–mesenchymal transition. Oncogene. 2007 Dec 13;26(57):7859–71.CrossRefGoogle ScholarPubMed
Sasser, AK, et al. Interleukin-6 is a potent growth factor for ER-alpha-positive human breast cancer. FASEB J. 2007 Nov;21(13):3763–70.CrossRefGoogle ScholarPubMed
Ancrile, B, et al. Oncogenic Ras-induced secretion of IL6 is required for tumorigenesis. Genes Dev. 2007 Jul 15;21(14):1714–19.CrossRefGoogle ScholarPubMed
Bolós, V, et al. Notch signaling in development and cancer. Endocr Rev. 2007 May;28(3):339–63.CrossRefGoogle ScholarPubMed
Tan, EY, et al. The key hypoxia regulated gene CAIX is upregulated in basal-like breast tumours and is associated with resistance to chemotherapy. Br J Cancer. 2009 Jan;100(2):405–11.CrossRefGoogle ScholarPubMed
Gao, SP, et al. Mutations in the EGFR kinase domain mediate STAT3 activation via IL-6 production in human lung adenocarcinomas. J Clin Invest. 2007 Dec;117(12):3846–56.CrossRefGoogle ScholarPubMed
Studebaker, AW, et al. Fibroblasts isolated from common sites of breast cancer metastasis enhance cancer cell growth rates and invasiveness in an interleukin-6-dependent manner. Cancer Res. 2008 Nov;68(21):9087–95.CrossRefGoogle Scholar
Hoadley, KA, et al. EGFR associated expression profiles vary with breast tumor subtype. BMC Genomics. 2007 Jul 31;8:258.CrossRefGoogle ScholarPubMed
Zhao, L, et al. Mammary gland remodeling depends on gp130 signaling through Stat3 and MAPK. J Biol Chem. 2004 Oct 15;279(42):44093–100.CrossRefGoogle ScholarPubMed
Viti, J, et al. Epidermal growth factor receptors control competence to interpret leucemia inhibitory factor as an astrocyte inducer in developing cortex. J Neurosci. 2003 Apr 15;23(8):3385–93.CrossRefGoogle Scholar
Shimazaki, T, et al. The ciliary neurotrophic factor/leukemia inhibitory factor/gp130 receptor complex operates in the maintenance of mammalian forebrain neural stem cells. J Neurosci. 2001 Oct 1;21(19):7642–53.CrossRefGoogle ScholarPubMed
Reynolds, BA and Rietze, RL. Neural stem cells and neurospheres – re-evaluating the relationship. Nat Methods. 2005 May;2(5):333–6.CrossRefGoogle ScholarPubMed
Farnie, GJ, et al. Novel cell culture technique for primary ductal carcinoma in situ: role of Notch and epidermal growth factor receptor signaling pathways. J Natl Cancer Inst. 2007 Apr 18;99(8):616–27.CrossRefGoogle ScholarPubMed
Chojnacki, A, et al. Glycoprotein 130 signaling regulates Notch1 expression and activation in the self-renewal of mammalian forebrain neural stem cells. J Neurosci. 2003 Mar 1;23(5):1730–41.CrossRefGoogle ScholarPubMed
Haruki, N, et al. Dominant-negative Notch3 receptor inhibits mitogen-activated protein kinase pathway and the growth of human lung cancers. Cancer Res. 2005 May 1;65(9):3555–61.CrossRefGoogle ScholarPubMed
Talora, C, et al. Cross talk among Notch3, pre-TCR, and Tal1 in T-cell development and leukemogenesis. Blood. 2006 Apr 15;107(8):3313–20.CrossRefGoogle ScholarPubMed
Fitzgerald, K, et al. Ras pathway signals are required for notch-mediated oncogenesis. Oncogene. 2000 Aug 31;19(37):4191–8.CrossRefGoogle ScholarPubMed
Raouf, A, et al. Transcriptome analysis of the normal human mammary cell commitment and differentiation process. Cell Stem Cell. 2008 Jul 3;3(1):109–18.CrossRefGoogle ScholarPubMed
Yamaguchi, N, et al. NOTCH3 signaling pathway plays crucial roles in the proliferation of ErbB2-negative human breast cancer cells. Cancer Res. 2008 Mar 15;68(6): 1881–8.CrossRefGoogle ScholarPubMed
Grivennikov, S and Karin, M. Autocrine IL-6 signaling: a key event in tumorigenesis?Cancer Cell. 2008 Jan;13(1):7–9.CrossRefGoogle ScholarPubMed
Schafer, ZT, and Brugge, JS, IL-6 involvement in epithelial cancers. J Clin Invest. 2007 Dec;117(12):3660–3.CrossRefGoogle ScholarPubMed
Hilvo, M, et al. Biochemical characterization of CA IX, one of the most active carbonic anhydrase isozymes. J Biol Chem. 2008 Oct, 283(41): 27799–809.CrossRefGoogle ScholarPubMed
Supuran, CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov. 2008 Feb;7(2):168–81.CrossRefGoogle ScholarPubMed
Dirix, LY, et al. Inflammatory breast cancer: current understanding. Curr Opin Oncol. 2006 Nov;18(6):563–71.CrossRefGoogle ScholarPubMed
Ben Hamida, A, et al. Markers of subtypes in inflammatory breast cancer studied by immunohistochemistry: prominent expression of P-cadherin. BMC Cancer. 2008 Jan 29;8:28.CrossRefGoogle ScholarPubMed
Xiao, Y, et al. The lymphovascular embolus of inflammatory breast cancer expresses a stem cell-like phenotype. Am J Pathol. 2008 Aug;173(2):561–74.CrossRefGoogle ScholarPubMed
Colpaert, CG, et al. Inflammatory breast cancer shows angiogenesis with high endothelial proliferation rate and strong E-cadherin expression. Br J Cancer. 2003 Mar 10;88(5):718–25.CrossRefGoogle ScholarPubMed
Moreno-Bueno, G, et al. Genetic profiling of epithelial cells expressing E-cadherin repressors reveals a distinct role for Snail, Slug, and E47 factors in epithelial–mesenchymal transition. Cancer Res. 2006 Oct 1;66(19):9543–56.CrossRefGoogle ScholarPubMed
Hajra, KM, et al. The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Res. 2002 Mar 15;62(6):1613–8.Google ScholarPubMed
Thiery, JP and Sleeman, JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 2006 Feb;7(2):131–42.CrossRefGoogle ScholarPubMed
Zajchowski, DA, et al. Identification of gene expression profiles that predict the aggressive behaviour of breast cancer cells. Cancer Res. 2001 Jul 1;61(13):5168–78.Google Scholar
Dhasarathy, A, et al. The transcription factor snail mediates epithelial to mesenchymal transitions by repression of estrogen receptor-alpha. Mol Endocrinol. 2007 Dec;21(12):2907–18.CrossRefGoogle ScholarPubMed
Nelson, WJ, et al. Convergence of Wnt, beta-catenin, and cadherin pathways. Science. 2004 Mar 5;303(5663):1483–7.CrossRefGoogle ScholarPubMed
Leong, KG, et al. Jagged1-mediated Notch activation induces epithelial-to-mesenchymal transition through Slug-induced repression of E-cadherin. J Exp Med. 2007 Nov 26;204(12):2935–48.CrossRefGoogle ScholarPubMed
Prasad, CP, et al. Epigenetic alterations of CDH1 and APC genes: relationship with activation of Wnt/beta-catenin pathway in invasive ductal carcinoma of breast. Life Sci. 2008 Aug;83(9–10):318–25.CrossRefGoogle ScholarPubMed
Woodward, WA, et al. WNT/beta-catenin mediates radiation resistance of mouse mammary progenitor cells. Proc Natl Acad Sci U S A. 2007 Jan 9;104(2):618–23.CrossRefGoogle ScholarPubMed
Lindvall, C, et al. Wnt signaling, stem cells, and the cellular origin of breast cancer. Stem Cell Rev. 2007 Jun;3(2):157–68.CrossRefGoogle ScholarPubMed
Schwartz, DR, et al. Novel candidate targets of beta-catenin/T-cell factor signaling identified by gene expression profiling of ovarian endometrioid adenocarcinomas. Cancer Res. 2003 Jun 1;63(11):2913–22.Google ScholarPubMed
Estrach, S, et al. Jagged 1 is a beta-catenin target gene required for ectopic hair follicle formation in adult epidermis. Development. 2006 Nov;133(22):4427–38.CrossRefGoogle ScholarPubMed
Kaidi, A, et al. Interaction between beta-catenin and HIF-1 promotes cellular adaptation to hypoxia. Nat Cell Biol. 2007 Feb;9(2):210–17.CrossRefGoogle ScholarPubMed
Lo, HW, et al. Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res. 2007 Oct 1;67(19):9066–76.CrossRefGoogle ScholarPubMed
Sahlgren, C, et al. Notch signaling mediates hypoxia-induced tumor cell migration and invasion. Proc Natl Acad Sci U S A. 2008 Apr 29;105(17):6392–7.CrossRefGoogle ScholarPubMed
McCarthy, A, et al. A mouse model of basal-like breast carcinoma with metaplastic elements. J Pathol. 2007 Mar;211(4):389–98.CrossRefGoogle ScholarPubMed
Liu, BY, et al. The transforming activity of Wnt effectors correlates with their ability to induce the accumulation of mammary progenitor cells. Proc Natl Acad Sci U S A. 2004 Mar 23;101(12):4158–63.CrossRefGoogle ScholarPubMed
Li, Y, et al. Evidence that transgenes encoding components of the Wnt signaling pathway preferentially induce mammary cancers from progenitor cells. Proc Natl Acad Sci U S A. 2003 Dec 23;100(26):15853–8.CrossRefGoogle ScholarPubMed
Brabletz, T, et al. Opinion: migrating cancer stem cells – an integrated concept of malignant tumour progression. Nat Rev Cancer. 2005 Sep;5(9):744–9.CrossRefGoogle ScholarPubMed
Grimshaw, MJ, et al. Mammosphere culture of metastatic breast cancer cells enriches for tumorigenic breast cancer cells. Breast Cancer Res. 2008;10(3):R52.CrossRefGoogle ScholarPubMed
Olmeda, D, et al. SNAI1 is required for tumor growth and lymph node metastasis of human breast carcinoma MDA-MB-231 cells. Cancer Res. 2007 Dec 15;67(24):11721–31.CrossRefGoogle ScholarPubMed
Kurrey, NK and Bapat, SA. Snail and Slug are major determinants of ovarian cancer invasiveness at the transcription level. Gynecol Oncol. 2005;97:155–65.CrossRefGoogle ScholarPubMed
Dong, R, et al. Role of nuclear factor kappa B and reactive oxygen species in the tumor necrosis factor-alpha-induced epithelial-mesenchymal transition of MCF-7 cells. Braz J Med Biol Res. 2007 Aug;40(8):1071–8.CrossRefGoogle ScholarPubMed
Huber, MA, et al. NF-kappaB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J Clin Invest. 2004 Aug;114(4):569–81.CrossRefGoogle Scholar
Grund, EM, et al. Tumor necrosis factor-alpha regulates inflammatory and mesenchymal responses via mitogen-activated protein kinase kinase, p38, and nuclear factor kappaB in human endometriotic epithelial cells. Mol Pharmacol. 2008 May;73(5):1394–404.CrossRefGoogle ScholarPubMed
Lester, RD, et al. uPAR induces epithelial-mesenchymal transition in hypoxic breast cancer cells. J Cell Biol. 2007 Jul 30;178(3):425–36.CrossRefGoogle ScholarPubMed
Julien, S, et al. Activation of NF-kappaB by Akt upregulates Snail expression and induces epithelium mesenchyme transition. Oncogene. 2007 Nov 22;26(53):7445–56.CrossRefGoogle ScholarPubMed
Chua, HL, et al. NF-kappaB represses E-cadherin expression and enhances epithelial to mesenchymal transition of mammary epithelial cells: potential involvement of ZEB-1 and ZEB-2. Oncogene. 2007 Feb 1;26(5):711–24.CrossRefGoogle ScholarPubMed
Brantley, DM, et al. Nuclear factor-kappaB (NF-kappaB) regulates proliferation and branching in mouse mammary epithelium. Mol Biol Cell. 2001 May;12(5):1445–55.CrossRefGoogle ScholarPubMed
Demicco, EG, et al. RelB/p52 NF-kappaB complexes rescue an early delay in mammary gland development in transgenic mice with targeted superrepressor IkappaB-alpha expression and promote carcinogenesis of the mammary gland. Mol Cell Biol. 2005 Nov;25(22):10136–47.CrossRefGoogle ScholarPubMed
Cao, Y, et al. IkappaB kinase alpha kinase activity is required for self-renewal of ErbB2/Her2-transformed mammary tumor-initiating cells. Proc Natl Acad Sci U S A. 2007 Oct 2;104(40):15852–7.CrossRefGoogle ScholarPubMed
Guzman, ML, et al. The sesquiterpene lactone parthenolide induces apoptosis of human acute myelogenous leukemia stem and progenitor cells. Blood. 2005 Jun 1; 105(11):4163–9.CrossRefGoogle ScholarPubMed
Finetti, P, et al. Sixteen-kinase gene expression identifies luminal breast cancers with poor prognosis. Cancer Res. 2008 Feb 1;68(3):767–76.CrossRefGoogle ScholarPubMed
Sun, C, et al. Aurora kinase inhibition downregulates NF-kappaB and sensitises tumour cells to chemotherapeutic agents. Biochem Biophys Res Commun. 2007 Jan 5;352(1):220–5.CrossRefGoogle ScholarPubMed
Laere, SJ, et al. NF-kappaB activation in inflammatory breast cancer is associated with oestrogen receptor downregulation, secondary to EGFR and/or ErbB2 overexpression and MAPK hyperactivation. Br J Cancer. 2007 Sep 3;97(5):659–69.CrossRefGoogle ScholarPubMed
Laffin, B, et al. Loss of singleminded-2s in the mouse mammary gland induces an epithelial-mesenchymal transition associated with up-regulation of slug and matrix metalloprotease 2. Mol Cell Biol. 2008 Mar;28(6):1936–46.CrossRefGoogle ScholarPubMed
Belguise, K, et al. Green tea polyphenols reverse cooperation between c-Rel and CK2 that induces the aryl hydrocarbon receptor, slug, and an invasive phenotype. Cancer Res. 2007 Dec 15;67(24):11742–50.CrossRefGoogle ScholarPubMed
Ikuta, T, and Kawajiri, K, Zinc finger transcription factor Slug is a novel target gene of aryl hydrocarbon receptor. Exp Cell Res. 2006 Nov 1;312(18):3585–94.CrossRefGoogle ScholarPubMed
Belaiba, RS, et al. Hypoxia up-regulates hypoxia-inducible factor-1alpha transcription by involving phosphatidylinositol 3-kinase and nuclear factor kappaB in pulmonary artery smooth muscle cells. Mol Biol Cell. 2007 Dec;18(12):4691–7.CrossRefGoogle ScholarPubMed
Rius, J, et al. NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature. 2008 Jun 5;453(7196):807–11.CrossRefGoogle ScholarPubMed
Uden, P, et al. Regulation of hypoxia-inducible factor-1alpha by NF-kappaB. Biochem J. 2008 Jun 15;412(3):477–84.Google ScholarPubMed
Xu, Q, et al. Targeting Stat3 blocks both HIF-1 and VEGF expression induced by multiple oncogenic growth signaling pathways. Oncogene. 2005 Aug 25;24(36):5552–60.CrossRefGoogle ScholarPubMed
Bertucci, F, et al. Gene expression profiling for molecular characterization of inflammatory breast cancer and prediction of response to chemotherapy. Cancer Res. 2004 Dec;64(23):8558–65.CrossRefGoogle ScholarPubMed
Eynden, GG, et al. Gene expression profiles associated with the presence of a fibrotic focus and the growth pattern in lymph node-negative breast cancer. Clin Cancer Res. 2008 May 15;14(10):2944–52.CrossRefGoogle ScholarPubMed
Simon, MC and Keith, B.The role of oxygen availability in embryonic development and stem cell function. Nat Rev Mol Cell Biol. 2008 Apr;9(4):285–96.CrossRefGoogle ScholarPubMed
Cipolleschi, MG, et al. The role of hypoxia in the maintenance of hematopoietic stem cells. Blood. 1993 Oct 1;82(7):2031–7.Google ScholarPubMed
Holmquist, L, et al. Effect of hypoxia on the tumor phenotype: the neuroblastoma and breast cancer models. Adv Exp Med Biol. 2006;587:179–93.CrossRefGoogle ScholarPubMed
Axelson, H, et al. Hypoxia-induced dedifferentiation of tumor cells – a mechanism behind heterogeneity and aggressiveness of solid tumors. Semin Cell Dev Biol. 2005 Aug–Oct;16(4–5):554–63.CrossRefGoogle Scholar
Jögi, A, et al. Hypoxia alters gene expression in human neuroblastoma cells toward an immature and neural crest-like phenotype. Proc Natl Acad Sci U S A. 2002 May 14;99(10):7021–6.CrossRefGoogle ScholarPubMed
Parmar, K, et al. Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc Natl Acad Sci U S A. 2007 Mar 27;104(13):5431–6.CrossRefGoogle ScholarPubMed
Gustafsson, MV, et al. Hypoxia requires notch signaling to maintain the undifferentiated cell state. Dev Cell. 2005 Nov;9(5):617–28.CrossRefGoogle ScholarPubMed
Das, B, et al. Hypoxia enhances tumor stemness by increasing the invasive and tumorigenic side population fraction. Stem Cells. 2008 Jul;26(7):1818–30.CrossRefGoogle ScholarPubMed
Vorbach, C, et al. Evolution of the mammary gland from the innate immune system?Bioessays. 2006 Jun;28(6):606–16.CrossRefGoogle ScholarPubMed
Romieu-Mourez, R, et al. Mouse mammary tumor virus c-rel transgenic mice develop mammary tumors. Mol Cell Biol. 2003 Aug;23(16):5738–54.CrossRefGoogle ScholarPubMed
Mantovani, A, et al. Inflammation and cancer: breast cancer as a prototype. Breast. 2007 Dec;16(Suppl 2):27–33.CrossRefGoogle ScholarPubMed
Rao, VP, et al. Breast cancer: should gastrointestinal bacteria be on our radar screen?Cancer Res. 2007 Feb 1;67(3):847–50.CrossRefGoogle ScholarPubMed
Rao, VP, et al. Innate immune inflammatory response against enteric bacteria Helicobacter hepaticus induces mammary adenocarcinoma in mice. Cancer Res. 2006 Aug 1;66(15):7395–400.CrossRefGoogle ScholarPubMed
Mantovani, A, et al. Cancer-related inflammation. Nature. 2008 Jul 24;454(7203):436–44.CrossRefGoogle ScholarPubMed
Shin, SR, et al. 7,12-dimethylbenz(a)anthracene treatment of a c-rel mouse mammary tumor cell line induces epithelial to mesenchymal transition via activation of nuclear factor-kappaB. Cancer Res. 2006 Mar 1;66(5):2570–5.CrossRefGoogle ScholarPubMed
Benz, CC. Impact of aging on the biology of breast cancer. Crit Rev Oncol Hematol. 2008 Apr;66(1):65–74.CrossRefGoogle ScholarPubMed
Balducci, L and Ershler, WB. Cancer and ageing: a nexus at several levels. Nat Rev Cancer. 2005 Aug;5(8):655–62.CrossRefGoogle ScholarPubMed
Franceschi, C, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000 Jun;908:244–54.CrossRefGoogle ScholarPubMed
Bonafè, M, et al. A gender-dependent genetic predisposition to produce high levels of IL-6 is detrimental for longevity. Eur J Immunol. 2001 Aug;31(8):2357–61.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Ferrucci, L, et al. The origins of age-related proinflammatory state. Blood. 2005 Mar 15;105(6):2294–9.CrossRefGoogle ScholarPubMed
Krtolica, A, et al. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc Natl Acad Sci U S A. 2001 Oct 9;98(21):12072–7.CrossRefGoogle ScholarPubMed
Yang, G, et al. The chemokine growth-regulated oncogene 1 (Gro-1) links RAS signaling to the senescence of stromal fibroblasts and ovarian tumorigenesis. Proc Natl Acad Sci U S A. 2006 Oct 31;103(44):16472–7.CrossRefGoogle ScholarPubMed
Schedin, P, et al. Microenvironment of the involuting mammary gland mediates mammary cancer progression. J Mammary Gland Biol Neoplasia. 2007 Mar;12(1):71–82.CrossRefGoogle ScholarPubMed
Balic, M, et al. Most early disseminated cancer cells detected in bone marrow of breast cancer patients have a putative breast cancer stem cell phenotype. Clin Cancer Res. 2006 Oct 1;12(19):5615–21.CrossRefGoogle ScholarPubMed
Graeber, TG, et al. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature. 1996;379:88–91.CrossRefGoogle ScholarPubMed
Conze, D, et al. Autocrine production of interleukin 6 causes multidrug resistance in breast cancer cells. Cancer Res. 2001 Dec 15;61(24):8851–8.Google ScholarPubMed
Vannini, I, et al. Short interfering RNA directed against the SLUG gene increases cell death induction in human melanoma cell lines exposed to cisplatin and fotemustine. Cell Oncol. 2007;29(4):279–87.Google ScholarPubMed
Toole, BP, et al. Hyaluronan: a constitutive regulator of chemoresistance and malignancy in cancer cells. Semin Cancer Biol. 2008 Aug;18(4):244–50.CrossRefGoogle ScholarPubMed
Biswas, DK, et al. NF-kappa B activation in human breast cancer specimens and its role in cell proliferation and apoptosis. Proc Natl Acad Sci U S A. 2004 Jul 6;101(27):10137–42.CrossRefGoogle ScholarPubMed
Cariati, M, et al. Alpha-6 integrin is necessary for the tumourigenicity of a stem cell-like subpopulation within the MCF7 breast cancer cell line. Int J Cancer. 2008 Jan 15; 122(2):298–304.CrossRefGoogle ScholarPubMed
Chen, MS, et al. Wnt/beta-catenin mediates radiation resistance of Sca1+ progenitors in an immortalized mammary gland cell line. J Cell Sci. 2007 Feb 1;120(Pt 3):468–77.CrossRefGoogle Scholar
Shafee, N, et al. Cancer stem cells contribute to cisplatin resistance in Brca1/p53-mediated mouse mammary tumors. Cancer Res. 2008 May 1;68(9):3243–50.CrossRefGoogle ScholarPubMed
Li, X, et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst. 2008 May 7;100(9):672–9.CrossRefGoogle Scholar
Li, HZ, et al. Suspension culture combined with chemotherapeutic agents for sorting of breast cancer stem cells. BMC Cancer. 2008 May 14;8:135.CrossRefGoogle ScholarPubMed
Krishnamurthy, P, et al. The stem cell marker Bcrp/ABCG2 enhances hypoxic cell survival through interactions with heme. J Biol Chem. 2004 Jun 4;279(23):24218–25.CrossRefGoogle ScholarPubMed
Patrawala, L, et al. Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and ABCG2− cancer cells are similarly tumorigenic. Cancer Res. 2005 Jul 15;65(14):6207–19.CrossRefGoogle ScholarPubMed
Eyler, CE and Rich, JN. Survival of the fittest: cancer stem cells in therapeutic resistance and angiogenesis. J Clin Oncol. 2008 Jun 10;26(17):2839–45.CrossRefGoogle ScholarPubMed
Zhou, J, et al. Cancer stem/progenitor cell active compound 8-quinolinol in combination with paclitaxel achieves an improved cure of breast cancer in the mouse model. Breast Cancer Res Treat. 2008 May 28.
Zhou, J, et al. NF-kappaB pathway inhibitors preferentially inhibit breast cancer stem-like cells. Breast Cancer Res Treat. 2008 Oct;111(3):419–27.CrossRefGoogle ScholarPubMed
Jordan, CT, et al. Cancer stem cells. N Engl J Med. 2006 Sep 21;355(12):1253–61.CrossRefGoogle ScholarPubMed
Schlotter, CM, et al. Molecular targeted therapies for breast cancer treatment. Breast Cancer Res. 2008 Jul 24;10(4):211.CrossRefGoogle ScholarPubMed
Singh, S, et al. Nuclear factor-kappaB activation: a molecular therapeutic target for estrogen receptor-negative and epidermal growth factor receptor family receptor-positive human breast cancerMol Cancer Ther. 2007 Jul;6(7):1973–82.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×