Skip to main content

Stem Cells in the Normal and Malignant Prostate

  • Chapter
  • First Online:
Prostate Cancer

Part of the book series: Protein Reviews ((PRON,volume 16))

Abstract

Tissues and organs like the prostate are derived from multipotent stem cells, which themselves are the products of differentiation from an original pluripotent embryonic stem cell population. Stem cells that persist into the mature prostate gland are termed tissue stem cells and are required for replenishment of the epithelial and stromal populations after damage, for example, by inflammation or gland involution after castration. While there remains some controversy over the phenotype of these cells, their ability to regenerate tissues and their inherent resistance to mutagenic and cytotoxic insults confer a unique biology on tissue stem cells. When one considers the origins of prostate cancer, the extended life span of tissue stem cells, and their ability to accumulate over time the necessary founder mutations, would imply that this primitive SC population is the cell of origin for prostate cancer. In the cancers, cells with similar primitive phenotypes are rare, but can be identified in varying proportions, depending on the markers used for isolation and the purification techniques. The tumor-initiating capacity of these cancer stem cells is many orders of magnitude higher than the majority cell population in tumors, and they display treatment resistance characteristics, which are sometimes shared with the normal tissue stem cells. The cancer stem cells in prostate cancers may therefore represent a viable target for therapeutic intervention, but there remain real challenges in the design and execution of these stem cell treatments.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

Acute myeloid leukemia (AML):

A rare cancer with the phenotype of an increase in the number of (myeloid) white blood cells in the bone marrow

Aldehyde dehydrogenase (ALDH):

A member of the aldehyde dehydrogenase enzyme family, whose elevated expression levels can be used to enrich for stem-like cells

Androgen receptor (AR):

Protein receptor for the male sex hormone androgen. Present at highest levels in the luminally differentiated cells in prostate

Basement membrane:

A complex proteinaceous boundary to each acinar unit of the prostate: It forms part of an active stem cell niche and signals to both stromal and epithelial components

Cancer cell type of origin (CCTO):

The cell type within prostate from which a tumor develops

Castration-resistant Nkx3.1-expressing cells (CARN cells):

A rare luminal stem cell population, which has been identified in the mouse prostate. CARN cells can give rise to both luminal and basal cells during prostate tissue regeneration induced by androgen depletion

Fluorescent-activated cell sorter (FACS):

Provides a method for sorting a disaggregated heterogeneous mixture of biological cells into two or more fractions, based upon the specific light scattering and fluorescent characteristics of each cell. It is particularly useful for the identification of rare cell populations

Gleason grading:

A morphological classification of the abnormal prostate gland, first established by Donald Gleason (in 1966). The loss of acinar morphology is broadly predictive of patient outcome

Hedgehog, wingless (wnt) and Notch:

Developmental signaling pathways originally defined in Drosophila melanogaster which also influence embryonic prostate development and adult prostate differentiation

Hematopoietic stem cells (HSCs):

Primitive cell type at the top of the hierarchy of cell types which differentiate into multiple cells types in the bloodstream and bone marrow (for example)

Induced pluripotent stem cells (iPS cells):

Biologically engineered stem cells, generated by in vitro treatment of already differentiated cells (e.g., skin fibroblasts) by a cocktail of (normally) four genes, which can differentiate into multiple cell types

Mesenchymal cells (also mesenchymal stem cells, MSCs):

Cells with a broadly stromal elongated morphology, which include an androgen-­receptor expressing population capable of changing the behavior of the epithelial cells by signaling through the basement membrane

Orthotopic xenografts:

Implantation into the tissue of origin (in this case, the murine prostate)

Prostate cancer stem cells (CSCs):

A generic term for the epithelial tumor-­initiating cell in prostate cancer, as like a normal tissue stem cell, CSCs can differentiate into multiple cell types. Also known as tumor-­initiating cells (TICs)

Prostate involution:

Shrinkage of the prostate gland as a consequence of castration, which is accompanied by the loss of structural acinar features

Prostate stem cells:

An epithelial cell, which can reconstitute all of the cells of the epithelial ­component of a prostatic acinus. Its basal/luminal phenotype remains controversial

Prostatic acinus (acinar morphology):

The base subunit of the prostate gland, which consists of an epithelial bilayer, surrounded by an intact basement membrane and bounded by complex fibroblastic (stromal) cells. Progressively disrupted through increasing Gleason grades of cancer

Stem cell quiescence:

A common feature of most reserve and stem cells in tissues, quiescence implies a lack of replicative activity, in the absence of complete cellular degenerative shut down. It can be considered as an inactive slowly metabolizing cell that is primed to respond to various stimuli, including injury

Subcutaneous xenografts:

Describes the implantation site under the skin of the mouse host

Tumor-initiating cells (TICs):

See CSCs

Urogenital sinus mesenchyme (UGM):

A powerful inductive androgen responsive mesenchymal component that defines the earliest stages of prostate gland (and acinar) development in embryos

Xenografts:

Implantation of (in this case) human tissues into an immune-­compromised mouse host

References

  1. Cunha GR et al (2004) Hormonal, cellular, and molecular regulation of normal and neoplastic prostatic development. J Steroid Biochem Mol Biol 92:221

    PubMed  CAS  Google Scholar 

  2. Cunha G et al (1992) Normal and abnormal development of the male urogenital tract. Role of androgens, mesenchymal-epithelial interactions, and growth factors. J Androl 13:465

    PubMed  CAS  Google Scholar 

  3. Thomson A, Marker P (2006) Branching morphogenesis in the prostate gland and seminal vesicles. Differentiation 74:382

    PubMed  CAS  Google Scholar 

  4. Prins GS, Birch L (1995) The developmental pattern of androgen receptor expression in rat prostate lobes is altered after neonatal exposure to estrogen. Endocrinology 136:1303

    PubMed  CAS  Google Scholar 

  5. Hayward SW et al (1996) Epithelial development in the rat ventral prostate, anterior prostate and seminal vesicle. Acta Anat 155:81

    PubMed  CAS  Google Scholar 

  6. Cunha GR (1972) Epithelio-mesenchymal interactions in primordial gland structures which become responsive to androgenic stimulation. Anat Rec 172:179

    PubMed  CAS  Google Scholar 

  7. Cunha GR (1972) Tissue interactions between epithelium and mesenchyme of urogenital and integumental origin. Anat Rec 172:529

    PubMed  CAS  Google Scholar 

  8. Xue Y et al (2001) Proliferative activity and branching morphogenesis in the human prostate: a closer look at pre- and postnatal prostate growth. Prostate 49:132

    PubMed  CAS  Google Scholar 

  9. Cunha GR (1973) The role of androgens in the epithelio-mesenchymal interactions involved in prostatic morphogenesis in embryonic mice. Anat Rec 175:87

    PubMed  CAS  Google Scholar 

  10. Cunha GR, Chung LW (1981) Stromal-epithelial interactions–I. Induction of prostatic phenotype in urothelium of testicular feminized (Tfm/y) mice. J Steroid Biochem 14:1317

    PubMed  CAS  Google Scholar 

  11. Cunha GR et al (1987) The endocrinology and developmental biology of the prostate. Endocr Rev 8:338

    PubMed  CAS  Google Scholar 

  12. Prins GS et al (2001) Influence of neonatal estrogens on rat prostate development. Reprod Fertil Dev 13:241

    PubMed  CAS  Google Scholar 

  13. Huang L, Pu Y, Alam S, Birch L, Prins GS (2004) Estrogenic regulation of signaling pathways and homeobox genes during rat prostate development. J Androl 25:330

    PubMed  CAS  Google Scholar 

  14. Seo R, McGuire M, Chung M, Bushman W (1997) Inhibition of prostate ductal morphogenesis by retinoic acid. J Urol 158:931

    PubMed  CAS  Google Scholar 

  15. Peehl DM, Wong ST, Stamey TA (1993) Vitamin A regulates proliferation and differentiation of human prostatic epithelial cells. Prostate 23:69

    PubMed  CAS  Google Scholar 

  16. Lohnes D et al (1995) Developmental roles of the retinoic acid receptors. J Steroid Biochem Mol Biol 53:475

    PubMed  CAS  Google Scholar 

  17. Donjacour A, Cunha G (1993) Assessment of prostatic protein secretion in tissue recombinants made of urogenital sinus mesenchyme and urothelium from normal or androgen-­insensitive mice. Endocrinology 132:2342

    PubMed  CAS  Google Scholar 

  18. Notini A, Davey R, McManus J, Bate K, Zajac J (2005) Genomic actions of the androgen receptor are required for normal male sexual differentiation in a mouse model. J Mol Endocrinol 35:547

    PubMed  CAS  Google Scholar 

  19. Bhatia-Gaur R et al (1999) Roles for Nkx3.1 in prostate development and cancer. Genes Dev 13:966

    PubMed  CAS  Google Scholar 

  20. Freestone S et al (2003) Sonic hedgehog regulates prostatic growth and epithelial differentiation. Dev Biol 264:352

    PubMed  CAS  Google Scholar 

  21. Schneider A, Brand T, Zweigerdt R, Arnold H (2000) Targeted disruption of the Nkx3.1 gene in mice results in morphogenetic defects of minor salivary glands: parallels to glandular duct morphogenesis in prostate. Mech Dev 95:163

    PubMed  CAS  Google Scholar 

  22. Tanaka M et al (2000) Nkx3.1, a murine homolog of Ddrosophila bagpipe, regulates epithelial ductal branching and proliferation of the prostate and palatine glands. Dev Dyn 219:248

    PubMed  CAS  Google Scholar 

  23. Podlasek C et al (1999) Hoxa-10 deficient male mice exhibit abnormal development of the accessory sex organs. Dev Dyn 214:1

    PubMed  CAS  Google Scholar 

  24. Warot X, Fromental-Ramain C, Fraulob V, Chambon P, Dolle P (1997) Gene dosage-­dependent effects of the Hoxa-13 and Hoxd-13 mutations on morphogenesis of the terminal parts of the digestive and urogenital tracts. Development 124:4781

    PubMed  CAS  Google Scholar 

  25. Podlasek C, Duboule D, Bushman W (1997) Male accessory sex organ morphogenesis is altered by loss of function of Hoxd-13. Dev Dyn 208:454

    PubMed  CAS  Google Scholar 

  26. Donjacour A, Thomson A, Cunha G (2003) FGF-10 plays an essential role in the growth of the fetal prostate. Dev Biol 261:39

    PubMed  CAS  Google Scholar 

  27. Guo L, Degenstein L, Fuchs E (1996) Keratinocyte growth factor is required for hair development but not for wound healing. Genes Dev 10:165

    PubMed  CAS  Google Scholar 

  28. Huang L, Pu Y, Alam S, Birch L, Prins G (2005) The role of Fgf10 signaling in branching morphogenesis and gene expression of the rat prostate gland: lobe-specific suppression by neonatal estrogens. Dev Biol 278:396

    PubMed  CAS  Google Scholar 

  29. Sugimura Y et al (1996) Keratinocyte growth factor (KGF) can replace testosterone in the ductal branching morphogenesis of the rat ventral prostate. Int J Dev Biol 40:941

    PubMed  CAS  Google Scholar 

  30. Wilhelm D, Koopman P (2006) The makings of maleness: towards an integrated view of male sexual development. Nat Rev Genet 7:620

    PubMed  CAS  Google Scholar 

  31. Cancilla B et al (2001) Regulation of prostate branching morphogenesis by activin A and follistatin. Dev Biol 237:145

    PubMed  CAS  Google Scholar 

  32. Gakunga P et al (1997) Hyaluronan is a prerequisite for ductal branching morphogenesis. Development 124:3987

    PubMed  CAS  Google Scholar 

  33. Marker P, Donjacour A, Dahiya R, Cunha G (2003) Hormonal, cellular, and molecular control of prostatic development. Dev Biol 253:165

    PubMed  CAS  Google Scholar 

  34. Kurita T, Medina R, Mills A, Cunha G (2004) Role of p63 and basal cells in the prostate. Development 131:4955

    PubMed  CAS  Google Scholar 

  35. Signoretti S et al (2000) p63 is a prostate basal cell marker and is required for prostate development. Am J Pathol 157:1769

    PubMed  CAS  Google Scholar 

  36. Signoretti S et al (2005) p63 regulates commitment to the prostate cell lineage. Proc Natl Acad Sci USA 102:11355

    PubMed  CAS  Google Scholar 

  37. Shou J, Ross S, Koeppen H, de Sauvage F, Gao W (2001) Dynamics of notch expression during murine prostate development and tumorigenesis. Cancer Res 61:7291

    PubMed  CAS  Google Scholar 

  38. Wang X, Shou J, Wong P, French D, Gao W (2004) Notch1-expressing cells are indispensable for prostatic branching morphogenesis during development and re-growth following castration and androgen replacement. J Biol Chem 279:24733

    PubMed  CAS  Google Scholar 

  39. Grishina I, Kim S, Ferrara C, Makarenkova H, Walden P (2005) BMP7 inhibits branching morphogenesis in the prostate gland and interferes with Notch signaling. Dev Biol 288:334

    PubMed  CAS  Google Scholar 

  40. Lamm M et al (2001) Mesenchymal factor bone morphogenetic protein 4 restricts ductal budding and branching morphogenesis in the developing prostate. Dev Biol 232:301

    PubMed  CAS  Google Scholar 

  41. Itoh N, Patel U, Cupp A, Skinner M (1998) Developmental and hormonal regulation of transforming growth factor-beta1 (TGFbeta1), -2, and -3 gene expression in isolated prostatic epithelial and stromal cells: epidermal growth factor and TGFbeta interactions. Endocrinology 139:1378

    PubMed  CAS  Google Scholar 

  42. Tomlinson D, Freestone S, Grace O, Thomson A (2004) Differential effects of transforming growth factor-beta1 on cellular proliferation in the developing prostate. Endocrinology 145:4292

    PubMed  CAS  Google Scholar 

  43. Thomson A, Foster B, Cunha G (1997) Analysis of growth factor and receptor mRNA levels during development of the rat seminal vesicle and prostate. Development 124:2431

    PubMed  CAS  Google Scholar 

  44. Shen MM, Abate-Shen C (2010) Molecular genetics of prostate cancer: new prospects for old challenges. Genes Dev 24:1967

    PubMed  CAS  Google Scholar 

  45. Maitland N (2008) Pathobiology of the Human prostate. Trends Urol Gynaecol Sex Health 13:12–19

    Google Scholar 

  46. Taylor R et al (2006) Formation of human prostate tissue from embryonic stem cells. Nat Methods 3:179

    PubMed  CAS  Google Scholar 

  47. Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663

    PubMed  CAS  Google Scholar 

  48. Cheng T et al (2000) Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 287:1804

    PubMed  CAS  Google Scholar 

  49. Li L, Clevers H (2010) Coexistence of quiescent and active adult stem cells in mammals. Science 327:542

    PubMed  CAS  Google Scholar 

  50. Buczacki S, Davies RJ, Winton DJ (2011) Stem cells, quiescence and rectal carcinoma: an unexplored relationship and potential therapeutic target. Br J Cancer 105:1253

    PubMed  CAS  Google Scholar 

  51. Moore N, Houghton J, Lyle S (2012) Slow-cycling therapy-resistant cancer cells. Stem Cells Dev 21:1822

    PubMed  CAS  Google Scholar 

  52. Miller SJ, Lavker RM, Sun T-T (2005) Interpreting epithelial cancer biology in the context of stem cells: tumor properties and therapeutic implications. Biochim Biophys Acta 1756:25

    PubMed  CAS  Google Scholar 

  53. Kyprianou N, Isaacs JT (1988) Activation of programmed cell death in the rat ventral prostate after castration. Endocrinology 122:552

    PubMed  CAS  Google Scholar 

  54. English HF, Kyprianou N, Isaacs JT (1989) Relationship between DNA fragmentation and apoptosis in the programmed cell death in the rat prostate following castration. Prostate 15:233

    PubMed  CAS  Google Scholar 

  55. Walensky LD, Coffey DS, Chen TH, Wu TC, Pasternack GR (1993) A novel M(r) 32,000 nuclear phosphoprotein is selectively expressed in cells competent for self-renewal. Cancer Res 53:4720

    PubMed  CAS  Google Scholar 

  56. Isaacs JT (1987) Development and characteristics of the available animal model systems for the study of prostatic cancer. Prog Clin Biol Res 239:513

    PubMed  CAS  Google Scholar 

  57. Isaacs JT, Coffey DS (1989) Etiology and disease process of benign prostatic hyperplasia. Prostate Suppl 2:33

    PubMed  CAS  Google Scholar 

  58. Evans GS, Chandler JA (1987) Cell proliferation studies in the rat prostate: II. The effects of castration and androgen-induced regeneration upon basal and secretory cell proliferation. Prostate 11:339

    PubMed  CAS  Google Scholar 

  59. English HF, Santen RJ, Isaacs JT (1987) Response of glandular versus basal rat ventral prostatic epithelial cells to androgen withdrawal and replacement. Prostate 11:229

    PubMed  CAS  Google Scholar 

  60. Scholzen T, Gerdes J (2000) The Ki-67 protein: from the known and the unknown. J Cell Physiol 182:311

    PubMed  CAS  Google Scholar 

  61. De Marzo AM, Meeker AK, Epstein JI, Coffey DS (1998) Prostate stem cell compartments: expression of the cell cycle inhibitor p27Kip1 in normal, hyperplastic, and neoplastic cells. Am J Pathol 153:911

    PubMed  Google Scholar 

  62. van der Kwast TH et al (1991) Androgen receptors in endocrine-therapy-resistant human prostate cancer. Int J Cancer 48:189

    PubMed  Google Scholar 

  63. Storms RW et al (2005) Distinct hematopoietic progenitor compartments are delineated by the expression of aldehyde dehydrogenase and CD34. Blood 106:95

    PubMed  CAS  Google Scholar 

  64. Sugimura Y (1986) Morphogenesis of ductal networks in the mouse prostate. Stem Cells 30:344

    Google Scholar 

  65. Kinbara H, Cunha GR, Boutin E, Hayashi N, Kawamura J (1996) Evidence of stem cells in the adult prostatic epithelium based upon responsiveness to mesenchymal inductors. Prostate 29:107

    PubMed  CAS  Google Scholar 

  66. Tsujimura A et al (2002) Proximal location of mouse prostate epithelial stem cells: a model of prostatic homeostasis. J Cell Biol 157:1257

    PubMed  CAS  Google Scholar 

  67. Spangrude GJ, Heimfeld S, Weissman IL (1988) Purification and characterization of mouse hematopoietic stem cells. Science 241:58

    PubMed  CAS  Google Scholar 

  68. Xin L, Lawson DA, Witte ON (2005) The Sca-1 cell surface marker enriches for a prostate-­regenerating cell subpopulation that can initiate prostate tumorigenesis. Proc Natl Acad Sci USA 102:6942

    PubMed  CAS  Google Scholar 

  69. Burger PE et al (2005) Sca-1 expression identifies stem cells in the proximal region of prostatic ducts with high capacity to reconstitute prostatic tissue. Proc Natl Acad Sci USA 102:7180

    PubMed  CAS  Google Scholar 

  70. Lawson DA, Xin L, Lukacs RU, Cheng D, Witte ON (2007) Isolation and functional characterization of murine prostate stem cells. Proc Natl Acad Sci USA 104:181

    PubMed  CAS  Google Scholar 

  71. Leong K, Wang B-E, Johnson L, Gao W-Q (2008) Generation of a prostate from a single adult stem cell. Nature 456:804

    PubMed  CAS  Google Scholar 

  72. Jiao J et al (2012) Identification of CD166 as a surface marker for enriching prostate stem/progenitor and cancer initiating cells. PLoS One 7:e42564

    PubMed  CAS  Google Scholar 

  73. Storms RW et al (1999) Isolation of primitive human hematopoietic progenitors on the basis of aldehyde dehydrogenase activity. Proc Natl Acad Sci USA 96:9118

    PubMed  CAS  Google Scholar 

  74. Eirew P et al (2012) Aldehyde dehydrogenase activity is a biomarker of primitive normal human mammary luminal cells. Stem Cells 30:344

    PubMed  CAS  Google Scholar 

  75. Blum R et al (2009) Molecular signatures of prostate stem cells reveal novel signaling pathways and provide insights into prostate cancer. PLoS One 4:e5722

    PubMed  Google Scholar 

  76. Wang X et al (2009) A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature 461:495

    PubMed  CAS  Google Scholar 

  77. Banerjee P (1995) Lobe-specific apoptotic cell death in rat prostate after androgen ablation by castration. Endocrinology 136:4368

    PubMed  CAS  Google Scholar 

  78. Barker N, van de Wetering M, Clevers H (2008) The intestinal stem cell. Genes Dev 22:1856

    PubMed  CAS  Google Scholar 

  79. Choi N, Zhang B, Zhang L, Ittmann M, Xin L (2012) Adult murine prostate basal and luminal cells are self-sustained lineages that can both serve as targets for prostate cancer initiation. Cancer Cell 21:253

    PubMed  CAS  Google Scholar 

  80. Ousset M et al (2012) Multipotent and unipotent progenitors contribute to prostate postnatal development. Nat Cell Biol 14:1131–1138

    PubMed  CAS  Google Scholar 

  81. Collins AT, Habib FK, Maitland NJ, Neal DE (2001) Identification and isolation of human prostate epithelial stem cells based on alpha(2)beta(1)-integrin expression. J Cell Sci 114:3865

    PubMed  CAS  Google Scholar 

  82. Freund D et al (2006) Comparative analysis of proliferative potential and clonogenicity of MACS-immunomagnetic isolated CD34+ and CD133+ blood stem cells derived from a single donor. Cell Prolif 39:325

    PubMed  CAS  Google Scholar 

  83. Richardson GD et al (2004) CD133, a novel marker for human prostatic epithelial stem cells. J Cell Sci 117:3539

    PubMed  CAS  Google Scholar 

  84. Huss WJ, Gray DR, Werdin ES, Funkhouser WK Jr, Smith GJ (2004) Evidence of pluripotent human prostate stem cells in a human prostate primary xenograft model. Prostate 60:77

    PubMed  Google Scholar 

  85. Birnie R et al (2008) Gene expression profiling of human prostate cancer stem cells reveals a pro-inflammatory phenotype and the importance of extracellular matrix interactions. Genome Biol 9:R83

    PubMed  Google Scholar 

  86. Goldstein A, Huang J, Guo C, Garraway I, Witte O (2010) Identification of a cell of origin for human prostate cancer. Science 329:568

    PubMed  CAS  Google Scholar 

  87. Garraway IP et al (2010) Human prostate sphere-forming cells represent a subset of basal epithelial cells capable of glandular regeneration in vivo. Prostate 70:491

    PubMed  Google Scholar 

  88. Blackwood JK et al (2011) In situ lineage tracking of human prostatic epithelial stem cell fate reveals a common clonal origin for basal and luminal cells. J Pathol 225:181

    PubMed  CAS  Google Scholar 

  89. Frame FM et al (2010) Development and limitations of lentivirus vectors as tools for tracking differentiation in prostate epithelial cells. Exp Cell Res 316:3161

    PubMed  CAS  Google Scholar 

  90. Bruce WR, Van Der Gaag H (1963) A quantitative assay for the number of murine lymphoma cells capable of proliferation in vivo. Nature 199:79

    PubMed  CAS  Google Scholar 

  91. Hamburger AW, Salmon SE (1977) Primary bioassay of human tumor stem cells. Science 197:461

    PubMed  CAS  Google Scholar 

  92. Sabbath KD, Ball ED, Larcom P, Davis RB, Griffin JD (1985) Heterogeneity of clonogenic cells in acute myeloblastic leukemia. J Clin Invest 75:746

    PubMed  CAS  Google Scholar 

  93. Bonnet D, Dick JE (1997) Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 3:730

    PubMed  CAS  Google Scholar 

  94. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 100:3983–3988

    PubMed  CAS  Google Scholar 

  95. Singh SK et al (2003) Identification of a cancer stem cell in human brain tumors. Cancer Res 63:5821

    PubMed  CAS  Google Scholar 

  96. Singh SK et al (2004) Identification of human brain tumour initiating cells. Nature 432:396

    PubMed  CAS  Google Scholar 

  97. Li C et al (2007) Identification of pancreatic cancer stem cells. Cancer Res 67:1030

    PubMed  CAS  Google Scholar 

  98. Ma S et al (2007) Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology 132:2542

    PubMed  CAS  Google Scholar 

  99. Ricci-Vitiani L et al (2007) Identification and expansion of human colon-cancer-initiating cells. Nature 445:111

    PubMed  CAS  Google Scholar 

  100. Eramo A et al (2008) Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ 15:504

    PubMed  CAS  Google Scholar 

  101. Hubbard SA et al (2009) Evidence for cancer stem cells in human endometrial carcinoma. Cancer Res 69:8241

    PubMed  CAS  Google Scholar 

  102. Collins A, Berry P, Hyde C, Stower M, Maitland N (2005) Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 65:10946

    PubMed  CAS  Google Scholar 

  103. Schepers AG et al (2012) Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science 337:730

    PubMed  CAS  Google Scholar 

  104. Wang S et al (2003) Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell 4:209

    PubMed  CAS  Google Scholar 

  105. Wang S et al (2006) Pten deletion leads to the expansion of a prostatic stem/progenitor cell subpopulation and tumor initiation. Proc Natl Acad Sci USA 103:1480

    PubMed  CAS  Google Scholar 

  106. Greenberg NM et al (1995) Prostate cancer in a transgenic mouse. Proc Natl Acad Sci USA 92:3439

    PubMed  CAS  Google Scholar 

  107. Asamoto M et al (2001) Prostate carcinomas developing in transgenic rats with SV40 T antigen expression under probasin promoter control are strictly androgen dependent. Cancer Res 61:4693

    PubMed  CAS  Google Scholar 

  108. Ma X et al (2005) Targeted biallelic inactivation of Pten in the mouse prostate leads to prostate cancer accompanied by increased epithelial cell proliferation but not by reduced apoptosis. Cancer Res 65:5730

    PubMed  CAS  Google Scholar 

  109. Korsten H, Ziel-van der Made A, Ma X, van der Kwast T, Trapman J (2009) Accumulating progenitor cells in the luminal epithelial cell layer are candidate tumor initiating cells in a Pten knockout mouse prostate cancer model. PLoS One 4:e5662

    PubMed  Google Scholar 

  110. Liao CP et al (2007) Mouse models of prostate adenocarcinoma with the capacity to monitor spontaneous carcinogenesis by bioluminescence or fluorescence. Cancer Res 67:7525

    PubMed  CAS  Google Scholar 

  111. Lawson DA et al (2010) Basal epithelial stem cells are efficient targets for prostate cancer initiation. Proc Natl Acad Sci USA 107:2610

    PubMed  CAS  Google Scholar 

  112. Liao C-P, Adisetiyo H, Liang M, Roy-Burman P (2010) Cancer-associated fibroblasts enhance the gland-forming capability of prostate cancer stem cells. Cancer Res 70:7294

    PubMed  CAS  Google Scholar 

  113. Driessens G, Beck B, Caauwe A, Simons BD, Blanpain C (2012) Defining the mode of tumour growth by clonal analysis. Nature 488:527

    PubMed  CAS  Google Scholar 

  114. Mulholland DJ et al (2009) Lin-Sca-1 + CD49fhigh stem/progenitors are tumor-initiating cells in the Pten-null prostate cancer model. Cancer Res 69:8555

    PubMed  CAS  Google Scholar 

  115. Wu CT et al (2007) Increased prostate cell proliferation and loss of cell differentiation in mice lacking prostate epithelial androgen receptor. Proc Natl Acad Sci USA 104:12679

    PubMed  CAS  Google Scholar 

  116. Liu AY et al (1997) Cell-cell interaction in prostate gene regulation and cytodifferentiation. Proc Natl Acad Sci USA 94:10705

    PubMed  CAS  Google Scholar 

  117. Dilworth S (1990) Cell alterations induced by the large T-antigens of SV40 and polyoma virus. Semin Cancer Biol 1:407

    PubMed  CAS  Google Scholar 

  118. Sontag E et al (1993) The interaction of SV40 small tumor antigen with protein phosphatase 2A stimulates the map kinase pathway and induces cell proliferation. Cell 75:887

    PubMed  CAS  Google Scholar 

  119. Cussenot O et al (1991) Immortalization of human adult normal prostatic epithelial cells by liposomes containing large T-SV40 gene. J Urol 146:881

    PubMed  CAS  Google Scholar 

  120. Hayward SW et al (1995) Establishment and characterization of an immortalized but non-­transformed human prostate epithelial cell line: BPH-1. In Vitro Cell Dev Biol Anim 31:14

    PubMed  CAS  Google Scholar 

  121. Hayward SW et al (2001) Malignant transformation in a nontumorigenic human prostatic epithelial cell line. Cancer Res 61:8135

    PubMed  CAS  Google Scholar 

  122. Lang SH, Maitland NJ (2004) Coculture of prostate cancer cells. Methods Mol Med 88:295

    PubMed  Google Scholar 

  123. Goldstein AS, Huang J, Guo C, Garraway IP, Witte ON (2010) Identification of a cell of origin for human prostate cancer. Science 329:568

    PubMed  CAS  Google Scholar 

  124. Lilja H et al (2011) Prediction of significant prostate cancer diagnosed 20 to 30 years later with a single measure of prostate-specific antigen at or before age 50. Cancer 117:1210

    PubMed  Google Scholar 

  125. Calabrese P, Tavare S, Shibata D (2004) Pretumor progression: clonal evolution of human stem cell populations. Am J Pathol 164:1337

    PubMed  Google Scholar 

  126. Reid R (1983) Genital warts and cervical cancer. II. Is human papillomavirus infection the trigger to cervical carcinogenesis? Gynecol Oncol 15:239

    PubMed  CAS  Google Scholar 

  127. Zhuang TN, Ly LP, Cumming RG, Handelsman DJ (2005) Growth and development during early manhood as determinants of prostate size in later life. J Clin Endocrinol Metab 90:6055

    PubMed  CAS  Google Scholar 

  128. Key TJ (2011) Diet, insulin-like growth factor-1 and cancer risk. Proc Nutr Soc 3:1–4

    Google Scholar 

  129. Sfanos KS, De Marzo AM (2012) Prostate cancer and inflammation: the evidence. Histopathology 60:199

    PubMed  Google Scholar 

  130. Maitland NJ, Collins AT (2008) Inflammation as the primary aetiological agent of human prostate cancer: a stem cell connection? J Cell Biochem 105:931

    PubMed  CAS  Google Scholar 

  131. Rajasekhar VK, Studer L, Gerald W, Socci ND, Scher HI (2011) Tumour-initiating stem-like cells in human prostate cancer exhibit increased NF-kappaB signalling. Nat Commun 2:162

    PubMed  Google Scholar 

  132. Shepherd C et al (2008) Expression profiling of CD133+ and CD133- epithelial cells from human prostate. Prostate 68:1007

    PubMed  CAS  Google Scholar 

  133. Luo JL et al (2007) Nuclear cytokine-activated IKKalpha controls prostate cancer metastasis by repressing Maspin. Nature 446:690

    PubMed  CAS  Google Scholar 

  134. Brown MD et al (2007) Characterization of benign and malignant prostate epithelial Hoechst 33342 side populations. Prostate 67:1384

    PubMed  Google Scholar 

  135. Vander Griend D et al (2008) The role of CD133 in normal human prostate stem cells and malignant cancer-initiating cells. Cancer Res 68:9703

    PubMed  CAS  Google Scholar 

  136. Vander Griend D et al (2010) Cell-autonomous intracellular androgen receptor signaling drives the growth of human prostate cancer initiating cells. Prostate 70:90

    PubMed  CAS  Google Scholar 

  137. Pfeiffer M, Schalken J (2010) Stem cell characteristics in prostate cancer cell lines. Eur Urol 57:246

    PubMed  CAS  Google Scholar 

  138. Pellacani D et al (2011) Regulation of the stem cell marker CD133 is independent of promoter hypermethylation in human epithelial differentiation and cancer. Mol Cancer 10:94

    PubMed  CAS  Google Scholar 

  139. Gerber JM et al (2011) Characterization of chronic myeloid leukemia stem cells. Am J Hematol 86:31

    PubMed  Google Scholar 

  140. Ginestier C et al (2007) ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1:555

    PubMed  CAS  Google Scholar 

  141. Yu C et al (2011) ALDH activity indicates increased tumorigenic cells, but not cancer stem cells, in prostate cancer cell lines. In Vivo 25:69

    PubMed  Google Scholar 

  142. van den Hoogen C et al (2010) High aldehyde dehydrogenase activity identifies tumor-­initiating and metastasis-initiating cells in human prostate cancer. Cancer Res 70:5163

    PubMed  Google Scholar 

  143. Hurt EM, Kawasaki BT, Klarmann GJ, Thomas SB, Farrar WL (2008) CD44+ CD24(-) prostate cells are early cancer progenitor/stem cells that provide a model for patients with poor prognosis. Br J Cancer 98:756

    PubMed  CAS  Google Scholar 

  144. Patrawala L et al (2006) Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene 25:1696

    PubMed  CAS  Google Scholar 

  145. Maitland N, Frame F, Polson E, Lewis J, Collins A (2011) Prostate Cancer Stem Cells: Do They Have a Basal or Luminal Phenotype? Horm Cancer 2:47

    PubMed  Google Scholar 

  146. Ishizawa K et al (2010) Tumor-initiating cells are rare in many human tumors. Cell Stem Cell 7:279

    PubMed  CAS  Google Scholar 

  147. Quintana E et al (2008) Efficient tumour formation by single human melanoma cells. Nature 456:593

    PubMed  CAS  Google Scholar 

  148. Boiko A et al (2010) Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271. Nature 466:133

    PubMed  CAS  Google Scholar 

  149. Knouf EC et al (2009) Multiple integrated copies and high-level production of the human retrovirus XMRV (xenotropic murine leukemia virus-related virus) from 22Rv1 prostate carcinoma cells. J Virol 83:7353

    PubMed  CAS  Google Scholar 

  150. Duhagon MA, Hurt EM, Sotelo-Silveira JR, Zhang X, Farrar WL (2010) Genomic profiling of tumor initiating prostatospheres. BMC Genomics 11:324

    PubMed  Google Scholar 

  151. Chen S, Principessa L, Isaacs JT (2012) Human prostate cancer initiating cells isolated directly from localized cancer do not form prostaspheres in primary culture. Prostate 72:1478

    PubMed  CAS  Google Scholar 

  152. Adams MD et al (1995) Initial assessment of human gene diversity and expression patterns based upon 83 million nucleotides of cDNA sequence. Nature 377:3

    PubMed  CAS  Google Scholar 

  153. Blum R et al (2009) Molecular signatures of prostate stem cells reveal novel signaling pathways and provide insights into prostate cancer. PLoS ONE 4(5):e5722

    PubMed  Google Scholar 

  154. Glinsky GV, Berezovska O, Glinskii AB (2005) Microarray analysis identifies a death-from-cancer signature predicting therapy failure in patients with multiple types of cancer. J Clin Invest 115:1503

    PubMed  CAS  Google Scholar 

  155. De Marzo AM et al (2007) Inflammation in prostate carcinogenesis. Nat Rev Cancer 7:256

    PubMed  Google Scholar 

  156. Pascal LE et al (2011) Lineage relationship of prostate cancer cell types based on gene expression. BMC Med Genomics 4:46

    PubMed  Google Scholar 

  157. Liu C et al (2012) Distinct microRNA expression profiles in prostate cancer stem/progenitor cells and tumor-suppressive functions of let-7. Cancer Res 72:3393

    PubMed  CAS  Google Scholar 

  158. Salvatori L et al (2012) Cell-to-cell signaling influences the fate of prostate cancer stem cells and their potential to generate more aggressive tumors. PLoS One 7:e31467

    PubMed  CAS  Google Scholar 

  159. van Leenders GJ et al (2011) Activation of c-MET induces a stem-like phenotype in human prostate cancer. PLoS One 6:e26753

    PubMed  Google Scholar 

  160. Chen W, Wang GM (2012) Gene expression profiling of cancer stem cells in the Du145 prostate cancer cell line. Oncol lett 3:791

    PubMed  CAS  Google Scholar 

  161. Nishida S et al (2012) Gene expression profiles of prostate cancer stem cells isolated by aldehyde dehydrogenase activity assay. J Urol 188:294–299

    PubMed  CAS  Google Scholar 

  162. Rowehl RA, Crawford H, Dufour A, Ju J, Botchkina GI (2008) Genomic analysis of prostate cancer stem cells isolated from a highly metastatic cell line. Cancer Genomics Proteomics 5:301

    PubMed  CAS  Google Scholar 

  163. Singh S et al (2012) Chemoresistance in prostate cancer cells is regulated by miRNAs and Hedgehog pathway. PLoS One 7:e40021

    PubMed  CAS  Google Scholar 

  164. Ma Y et al (2011) Prostate cancer cell lines under hypoxia exhibit greater stem-like ­properties. PLoS One 6:e29170

    PubMed  CAS  Google Scholar 

  165. Tomlins SA et al (2005) Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310:644

    PubMed  CAS  Google Scholar 

  166. Demichelis F, Rubin MA (2007) TMPRSS2-ETS fusion prostate cancer: biological and ­clinical implications. J Clin Pathol 60:1185

    PubMed  Google Scholar 

  167. Cerveira N et al (2006) TMPRSS2-ERG gene fusion causing ERG overexpression precedes chromosome copy number changes in prostate carcinomas and paired HGPIN lesions. Neoplasia 8:826

    PubMed  CAS  Google Scholar 

  168. Polson ES et al (2013) Mono-allelic expression of TMPRSS2/ERG in prostate cancer stem cells. Nature Commun (in press)

    Google Scholar 

  169. Setlur SR et al (2008) Estrogen-dependent signaling in a molecularly distinct subclass of aggressive prostate cancer. J Natl Cancer Inst 100:815

    PubMed  CAS  Google Scholar 

  170. Qin J et al (2012) The PSA(-/lo) prostate cancer cell population harbors self-renewing long-­term tumor-propagating cells that resist castration. Cell Stem Cell 10:556

    PubMed  CAS  Google Scholar 

  171. Feinberg AP, Ohlsson R, Henikoff S (2006) The epigenetic progenitor origin of human ­cancer. Nat Rev Genet 7:21

    PubMed  CAS  Google Scholar 

  172. Liu C, Tang DG (2011) MicroRNA regulation of cancer stem cells. Cancer Res 71:5950

    PubMed  CAS  Google Scholar 

  173. Huggins C, Stevens R, Hodges CV (1941) Studies on prostatic cancer ii. The effects of castration on advanced carcinoma of the prostate gland. Arch Surg 43(209)

    Google Scholar 

  174. Denmeade SR, Isaacs JT (2002) A history of prostate cancer treatment. Nat Rev Cancer 2:389

    PubMed  CAS  Google Scholar 

  175. Gingrich JR et al (1996) Metastatic prostate cancer in a transgenic mouse. Cancer Res 56:4096

    PubMed  CAS  Google Scholar 

  176. Craft N et al (1999) Evidence for clonal outgrowth of androgen-independent prostate cancer cells from androgen-dependent tumors through a two-step process. Cancer Res 59:5030

    PubMed  CAS  Google Scholar 

  177. Chen CD et al (2004) Molecular determinants of resistance to antiandrogen therapy. Nat Med 10:33

    PubMed  Google Scholar 

  178. Taplin M, Rajeshkumar B (2011) Integrin alphav expression is required for the acquistion of a metastatic stem/progenitor cell phenotype in human prostate cancer. Am J Pathol 179(2559)

    Google Scholar 

  179. Li P et al (2002) Heterogeneous expression and functions of androgen receptor co-factors in primary prostate cancer. Am J Pathol 161:1467

    PubMed  CAS  Google Scholar 

  180. Bubendorf L et al (1999) Survey of gene amplifications during prostate cancer progression by high-throughout fluorescence in situ hybridization on tissue microarrays. Cancer Res 59:803

    PubMed  CAS  Google Scholar 

  181. Oldridge EE, Pellacani D, Collins AT, Maitland NJ (2011) Prostate cancer stem cells: Are they androgen-responsive? Mol Cell Endocrinol 5:14–24

    Google Scholar 

  182. Diehn M et al (2009) Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 458:780

    PubMed  CAS  Google Scholar 

  183. Frame FM, Maitland NJ (2011) Cancer stem cells, models of study and implications of therapy resistance mechanisms. Adv Exp Med Biol 720:105

    Google Scholar 

  184. Bristow RG, Hill RP (2008) Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability. Nat Rev Cancer 8:180

    PubMed  CAS  Google Scholar 

  185. Hennessey D et al. (2011) Exposure to hypoxia following irradiation increases radioresistance in prostate cancer cells. Urol Oncol. Epub ahead of print

    Google Scholar 

  186. Goda N et al (2003) Hypoxia-inducible factor 1alpha is essential for cell cycle arrest during hypoxia. Mol Cell Biol 23:359

    PubMed  CAS  Google Scholar 

  187. Patrawala L et al (2005) Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and ABCG2- cancer cells are similarly tumorigenic. Cancer Res 65:6207

    PubMed  CAS  Google Scholar 

  188. Yan H et al (2011) Drug-tolerant cancer cells show reduced tumor-initiating capacity: depletion of CD44 cells and evidence for epigenetic mechanisms. PLoS One 6:e24397

    PubMed  CAS  Google Scholar 

  189. Hudson DL et al (2001) Epithelial cell differentiation pathways in the human prostate: identification of intermediate phenotypes by keratin expression. J Histochem Cytochem 49:271

    PubMed  CAS  Google Scholar 

  190. Morimoto K et al (2009) Stem cell marker aldehyde dehydrogenase 1-positive breast cancers are characterized by negative estrogen receptor, positive human epidermal growth factor receptor type 2, and high Ki67 expression. Cancer Sci 100:1062

    PubMed  CAS  Google Scholar 

  191. Pietras EM, Warr MR, Passegue E (2011) Cell cycle regulation in hematopoietic stem cells. J Cell Biol 195:709

    PubMed  CAS  Google Scholar 

  192. Andriole GL et al (2010) Effect of dutasteride on the risk of prostate cancer. N Engl J Med 362:1192

    PubMed  CAS  Google Scholar 

  193. Kucway R et al (2002) Prostate volume reduction with androgen deprivation therapy before interstitial brachytherapy. J Urol 167:2443

    PubMed  CAS  Google Scholar 

  194. Kallioniemi OP, Visakorpi T (1996) Genetic basis and clonal evolution of human prostate cancer. Adv Cancer Res 68:225

    PubMed  CAS  Google Scholar 

  195. Liu W et al (2009) Copy number analysis indicates monoclonal origin of lethal metastatic prostate cancer. Nat Med 15:559

    PubMed  CAS  Google Scholar 

  196. Wu C et al (2012) Integrated genome and transcriptome sequencing identifies a novel form of hybrid and aggressive prostate cancer. J Pathol 227:53

    PubMed  CAS  Google Scholar 

  197. Rane JK, Pellacani D, Maitland NJ (2012) Advanced prostate cancer-a case for adjuvant differentiation therapy. Nat Rev Urol 9:595–602

    PubMed  CAS  Google Scholar 

  198. Pierce GB, Speers WC (1988) Tumors as caricatures of the process of tissue renewal: prospects for therapy by directing differentiation. Cancer Res 48:1996

    PubMed  CAS  Google Scholar 

  199. Sanz MA et al (2009) Management of acute promyelocytic leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood 113:1875

    PubMed  CAS  Google Scholar 

  200. Hacein-Bey-Abina S et al (2003) A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N Engl J Med 348:255

    PubMed  Google Scholar 

  201. Zhang K, Waxman DJ (2010) PC3 prostate tumor-initiating cells with molecular profile FAM65B(high)/MFI2(low)/LEF1(low) increase tumor angiogenesis. Mol Cancer 9:319

    PubMed  CAS  Google Scholar 

  202. Fan X, Liu S, Su F, Pan Q, Lin T (2012) Effective enrichment of prostate cancer stem cells from spheres in a suspension culture system. Urol Oncol 30:314

    PubMed  CAS  Google Scholar 

  203. van den Hoogen C et al (2011) Integrin aplha v expression is required for the acquisition of a metastatic stem/progenitor cell phenotype in human prostate cancer. Am J Pathol 179:2559

    PubMed  Google Scholar 

  204. Wei C, Guomin W, Yujun L, Ruizhe Q (2007) Cancer stem-like cells in human prostate carcinoma cells DU145: the seeds of the cell line? Cancer Biol Ther 6:763

    PubMed  CAS  Google Scholar 

  205. Miki J et al (2007) Identification of putative stem cell markers, CD133 and CXCR4, in hTERT-immortalized primary nonmalignant and malignant tumor-derived human prostate epithelial cell lines and in prostate cancer specimens. Cancer Res 67:3153

    PubMed  CAS  Google Scholar 

  206. Germann M et al (2012) Stem-like cells with luminal progenitor phenotype survive castration in human prostate cancer. Stem Cells 30:1076

    PubMed  CAS  Google Scholar 

  207. Patrawala L, Calhoun-Davis T, Schneider-Broussard R, Tang DG (2007) Hierarchical organization of prostate cancer cells in xenograft tumors: the CD44 + alpha2beta1+ cell population is enriched in tumor-initiating cells. Cancer Res 67:6796

    PubMed  CAS  Google Scholar 

  208. Liu T et al (2010) Establishment and characterization of multi-drug resistant, prostate carcinoma-­initiating stem-like cells from human prostate cancer cell lines 22RV1. Mol Cell Biochem 340:265

    PubMed  CAS  Google Scholar 

  209. Borovski T, Melo FDE, Vermeulen L, Medema JP (2011) Cancer stem cell niche: the place to be. Cancer Res 71:634

    PubMed  CAS  Google Scholar 

  210. Donnenberg VS, Donnenberg AD (2005) Multiple drug resistance in cancer revisited: the cancer stem cell hypothesis. J Clin Pharmacol 45:872

    PubMed  CAS  Google Scholar 

  211. Szotek PP et al (2006) Ovarian cancer side population defines cells with stem cell-like characteristics and Mullerian Inhibiting Substance responsiveness. Proc Natl Acad Sci USA 103:11154

    PubMed  CAS  Google Scholar 

  212. Hong SP, Wen J, Bang S, Park S, Song SY (2009) CD44-positive cells are responsible for gemcitabine resistance in pancreatic cancer cells. Int J Cancer 125:2323

    PubMed  CAS  Google Scholar 

  213. Ma J, Dong C, Ji C (2010) MicroRNA and drug resistance. Cancer Gene Ther 17:523

    PubMed  CAS  Google Scholar 

  214. Shi L et al (2012) MicroRNA-125b-2 confers human glioblastoma stem cells resistance to temozolomide through the mitochondrial pathway of apoptosis. Int J Oncol 40:119

    PubMed  Google Scholar 

  215. Pajonk F, Vlashi E, McBride WH (2010) Radiation resistance of cancer stem cells – the 4R’s of radiobiology revisited. Stem Cells 28:639

    PubMed  CAS  Google Scholar 

  216. Bao S et al (2006) Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444:756

    PubMed  CAS  Google Scholar 

  217. Harper LJ et al (2010) Normal and malignant epithelial cells with stem-like properties have an extended G2 cell cycle phase that is associated with apoptotic resistance. BMC Cancer 10:166

    PubMed  Google Scholar 

  218. Hoey T et al (2009) DLL4 blockade inhibits tumor growth and reduces tumor-initiating cell frequency. Cell Stem Cell 5:168

    PubMed  CAS  Google Scholar 

  219. Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K (2009) Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res 69:7507

    PubMed  CAS  Google Scholar 

  220. Gupta PB et al (2009) Identification of selective inhibitors of cancer stem cells by high-­throughput screening. Cell 138:645

    PubMed  CAS  Google Scholar 

  221. Ji Q et al (2009) MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS One 4:e6816

    PubMed  Google Scholar 

  222. Li Y et al (2009) MicroRNA-34a inhibits glioblastoma growth by targeting multiple oncogenes. Cancer Res 69:7569

    PubMed  CAS  Google Scholar 

  223. Liu C et al (2011) The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med 17:211

    PubMed  CAS  Google Scholar 

  224. Herman-Antosiewicz A, Johnson DE, Singh SV (2006) Sulforaphane causes autophagy to inhibit release of cytochrome C and apoptosis in human prostate cancer cells. Cancer Res 66:5828

    PubMed  CAS  Google Scholar 

  225. Kallifatidis G et al (2009) Sulforaphane targets pancreatic tumour-initiating cells by NF-kappaB-induced antiapoptotic signalling. Gut 58:949

    PubMed  CAS  Google Scholar 

  226. Li Y et al (2010) Sulforaphane, a dietary component of broccoli/broccoli sprouts, inhibits breast cancer stem cells. Clin Cancer Res 16:2580

    PubMed  CAS  Google Scholar 

  227. Rausch V et al (2010) Synergistic activity of sorafenib and sulforaphane abolishes pancreatic cancer stem cell characteristics. Cancer Res 70:5004

    PubMed  CAS  Google Scholar 

  228. Kakarala M et al (2010) Targeting breast stem cells with the cancer preventive compounds curcumin and piperine. Breast Cancer Res Treat 122:777

    PubMed  CAS  Google Scholar 

  229. Lin L et al (2011) Targeting colon cancer stem cells using a new curcumin analogue, GO-Y030. Br J Cancer 105:212

    PubMed  CAS  Google Scholar 

  230. Doggrell SA (2010) The hedgehog pathway inhibitor GDC-0449 shows potential in skin and other cancers. Expert Opin Investig Drugs 19:451

    PubMed  CAS  Google Scholar 

  231. Visbal AP, Lewis MT (2010) Hedgehog signaling in the normal and neoplastic mammary gland. Curr Drug Targets 11:1103

    PubMed  CAS  Google Scholar 

  232. Al-Hussaini H, Subramanyam D, Reedijk M, Sridhar SS (2011) Notch signaling pathway as a therapeutic target in breast cancer. Mol Cancer Ther 10:9

    PubMed  CAS  Google Scholar 

  233. Harrison H, Farnie G, Brennan KR, Clarke RB (2010) Breast cancer stem cells: something out of notching? Cancer Res 70:8973

    PubMed  CAS  Google Scholar 

  234. Pandey PR et al (2011) Resveratrol suppresses growth of cancer stem-like cells by inhibiting fatty acid synthase. Breast Cancer Res Treat 130:387

    PubMed  CAS  Google Scholar 

  235. Puissant A et al (2010) Resveratrol promotes autophagic cell death in chronic myelogenous leukemia cells via JNK-mediated p62/SQSTM1 expression and AMPK activation. Cancer Res 70:1042

    PubMed  CAS  Google Scholar 

  236. Shankar S et al (2011) Resveratrol inhibits pancreatic cancer stem cell characteristics in human and KrasG12D transgenic mice by inhibiting pluripotency maintaining factors and epithelial-mesenchymal transition. PLoS One 6:e16530

    PubMed  CAS  Google Scholar 

Download references

Acknowledgments

I wish to thank all the members of the YCR Cancer Research Unit who have contributed in so many ways to the ideas and data I have described. In particular I would like to acknowledge the seminal contributions of Anne Collins and willingness of Fiona Frame and Jayant Rane to let me rework their artwork. Michelle Scaife heroically collated the reference list and transcribed the text. Finally, my sincere thanks are due to Yorkshire Cancer Research, which has supported my research on prostate cancer and stem cells since its inception.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Norman J. Maitland B.Sc., Ph.D. .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2013 Mayo Clinic

About this chapter

Cite this chapter

Maitland, N.J. (2013). Stem Cells in the Normal and Malignant Prostate. In: Tindall, D. (eds) Prostate Cancer. Protein Reviews, vol 16. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-6828-8_1

Download citation

Publish with us

Policies and ethics