Skip to main content

Notch Signaling in Cancer Stem Cells

  • Chapter

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 727))

Abstract

Subpopulations of cancer cells with stem cell-like characteristics, termed cancer stem cells, have been identified in a wide range of human cancers. Cancer stem cells are defined by their ability to self-renew as well as recapitulate the original heterogeneity of cancer cells in culture and in serial xenotransplants. Not only are cancer stem cells highly tumorigenic, but these cells are implicated in tumor resistance to conventional chemotherapy and radiotherapy, thus highlighting their significance as therapeutic targets. Considerable similarities have been found between cancer stem cells and normal stem cells on their dependence on certain signaling pathways. More specifically, the core stem cell signaling pathways, such as the Wnt, Notch and Hedgehog pathways, also critically regulate the self-renewal and survival of cancer stem cells. While the oncogenic functions of Notch pathway have been well documented, its role in cancer stem cells is just emerging. In this chapter, we will discuss recent advances in cancer stem cell research and highlight the therapeutic potential of targeting Notch in cancer stem cells.

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

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   149.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever

Tax calculation will be finalised at checkout

Purchases are for personal use only

Learn about institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Park CY, Tseng D, Weissman IL. Cancer stem cell-directed therapies: recent data from the laboratory and clinic. Mol Ther 2009; 17:219–230.

    PubMed  CAS  Google Scholar 

  2. Eyler CE, Rich JN. Survival of the fittest: cancer stem cells in therapeutic resistance and angiogenesis. J Clin Oncol 2008; 26:2839–2845.

    PubMed  CAS  Google Scholar 

  3. Huang EH, Heidt DG, Li CW et al. Cancer stem cells: a new paradigm for understanding tumor progression and therapeutic resistance. Surgery 2007; 141:415–419.

    PubMed  Google Scholar 

  4. Pajonk F, Vlashi E, McBride WH. Radiation resistance of cancer stem cells: the 4 R’s of radiobiology revisited. Stem Cells 2010; 28:639–648.

    PubMed  CAS  Google Scholar 

  5. Stylianou S, Clarke RB, Brennan K. Aberrant activation of notch signaling in human breast cancer. Cancer Res 2006; 66:1517–1525.

    PubMed  CAS  Google Scholar 

  6. Demarest RM, Ratti F, Capobianco AJ. It’s T-ALL about Notch. Oncogene 2008; 27:5082–5091.

    PubMed  CAS  Google Scholar 

  7. van Es JH, van Gijn ME, Riccio O et al. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature 2005; 435:959–963.

    PubMed  Google Scholar 

  8. Weng AP, Ferrando AA, Lee W et al. Activating mutations of NOTCH1 in human T-cell acute lymphoblastic leukemia. Science 2004; 306:269–271.

    PubMed  CAS  Google Scholar 

  9. Farnie G, Clarke RB. Mammary stem cells and breast cancer—role of Notch signalling. Stem Cell Rev 2007; 3:169–175.

    PubMed  CAS  Google Scholar 

  10. Pierfelice TJ, Schreck KC, Eberhart CG et al. Notch, neural stem cells and brain tumors. Cold Spring Harb Symp Quant Biol 2008; 73:367–375.

    PubMed  CAS  Google Scholar 

  11. Dick JE. Looking ahead in cancer stem cell research. Nat Biotechnol 2009; 27:44–46.

    PubMed  CAS  Google Scholar 

  12. Lapidot T, Sirard C, Vormoor J et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994; 367:645–648.

    PubMed  CAS  Google Scholar 

  13. Al-Hajj M, Wicha MS, Benito-Hernandez A et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 2003; 100:3983–3988.

    PubMed  CAS  Google Scholar 

  14. Singh SK, Clarke ID, Terasaki M et al. Identification of a cancer stem cell in human brain tumors. Cancer Res 2003; 63:5821–5828.

    PubMed  CAS  Google Scholar 

  15. Singh SK, Hawkins C, Clarke ID et al. Identification of human brain tumour initiating cells. Nature 2004; 432:396–401.

    PubMed  CAS  Google Scholar 

  16. O’Brien CA, Pollett A, Gallinger S et al. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007; 445:106–110.

    PubMed  Google Scholar 

  17. Ricci-Vitiani L, Lombardi DG, Pilozzi E et al. Identification and expansion of human colon-cancer-initiating cells. Nature 2007; 445:111–115.

    PubMed  CAS  Google Scholar 

  18. Li C, Heidt DG, Dalerba P et al. Identification of pancreatic cancer stem cells. Cancer Res 2007; 67:1030–1037.

    PubMed  CAS  Google Scholar 

  19. Collins AT, Berry PA, Hyde C et al. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 2005; 65:10946–10951.

    PubMed  CAS  Google Scholar 

  20. Eramo A, Lotti F, Sette G et al. Cell Death Differ 2008; 15:504–514.

    PubMed  CAS  Google Scholar 

  21. Yang ZF, Ho DW, Ng MN et al. Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell 2008; 13:153–166.

    PubMed  CAS  Google Scholar 

  22. Prince ME, Sivanandan R, Kaczorowski A et al. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA 2007; 104:973–978.

    PubMed  CAS  Google Scholar 

  23. Moserle L, Ghisi M, Amadori A et al. Side population and cancer stem cells: therapeutic implications. Cancer Lett. 288:1–9.

    Google Scholar 

  24. Schatton T, Murphy GF, Frank NY et al. Identification of cells initiating human melanomas. Nature 2008; 451:345–349.

    PubMed  CAS  Google Scholar 

  25. Armstrong L, Stojkovic M, Dimmick I et al. Phenotypic characterization of murine primitive hematopoietic progenitor cells isolated on basis of aldehyde dehydrogenase activity. Stem Cells 2004; 22:1142–1151.

    PubMed  Google Scholar 

  26. Hess DA, Meyerrose TE, Wirthlin L et al. Functional characterization of highly purified human hematopoietic repopulating cells isolated according to aldehyde dehydrogenase activity. Blood 2004; 104:1648–1655.

    PubMed  CAS  Google Scholar 

  27. Ginestier C, Hur MH, Charafe-Jauffret E et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 2007; 1:555–567.

    PubMed  CAS  Google Scholar 

  28. Liu R, Wang X, Chen GY et al. The prognostic role of a gene signature from tumorigenic breast-cancer cells. N Engl J Med 2007; 356:217–226.

    PubMed  CAS  Google Scholar 

  29. Abraham BK, Fritz P, McClellan M et al. Prevalence of CD44+/CD24-/low cells in breast cancer may not be associated with clinical outcome but may favor distant metastasis. Clin Cancer Res 2005; 11:1154–1159.

    PubMed  CAS  Google Scholar 

  30. van Rhenen A, Feller N, Kelder A et al. High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival. Clin Cancer Res 2005; 11:6520–6527.

    PubMed  Google Scholar 

  31. Zeppernick F, Ahmadi R, Campos B et al. Stem cell marker CD133 affects clinical outcome in glioma patients. Clin Cancer Res 2008; 14:123–129.

    PubMed  CAS  Google Scholar 

  32. Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer 2005; 5:275–284.

    PubMed  CAS  Google Scholar 

  33. Diehn M, Cho RW, Clarke MF. Therapeutic implications of the cancer stem cell hypothesis. Semin Radiat Oncol 2009; 19:78–86.

    PubMed  Google Scholar 

  34. Li X, Lewis MT, Huang J et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 2008; 100:672–679.

    PubMed  CAS  Google Scholar 

  35. Eramo A, Ricci-Vitiani L, Zeuner A et al. Chemotherapy resistance of glioblastoma stem cells. Cell Death Differ 2006; 13:1238–1241.

    PubMed  CAS  Google Scholar 

  36. Hermann PC, Huber SL, Herrler T et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 2007; 1:313–323.

    PubMed  CAS  Google Scholar 

  37. Todaro M, Alea MP, Di Stefano AB et al. Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4. Cell Stem Cell 2007; 1:389–402.

    PubMed  CAS  Google Scholar 

  38. L evina V, Marrangoni AM, DeMarco R et al. Drug-selected human lung cancer stem cells: cytokine network, tumorigenic and metastatic properties. PLoS ONE 2008; 3:e3077.

    Google Scholar 

  39. Guan Y, Gerhard B, Hogge DE. Detection, isolation and stimulation of quiescent primitive leukemic progenitor cells from patients with acute myeloid leukemia (AML). Blood 2003; 101:3142–3149.

    PubMed  CAS  Google Scholar 

  40. Holyoake T, Jiang X, Eaves C et al. Isolation of a highly quiescent subpopulation of primitive leukemic cells in chronic myeloid leukemia. Blood 1999; 94:2056–2064.

    PubMed  CAS  Google Scholar 

  41. Zheng H, Ying H, Yan H et al. p53 and Pten control neural and glioma stem/progenitor cell renewal and differentiation. Nature 2008; 455:1129–1133.

    PubMed  CAS  Google Scholar 

  42. Wang J, Wang H, Li Z et al. c-Myc is required for maintenance of glioma cancer stem cells. PLoS ONE 2008; 3:e3769.

    Google Scholar 

  43. Liu G, Yuan X, Zeng Z et al. Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer 2006; 5:67.

    PubMed  Google Scholar 

  44. Bao S, Wu Q, McLendon RE et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006; 444:756–760.

    PubMed  CAS  Google Scholar 

  45. Blazek ER, Foutch JL, Maki G. Daoy medulloblastoma cells that express CD133 are radioresistant relative to CD133-cells and the CD133+ sector is enlarged by hypoxia. Int J Radiat Oncol Biol Phys 2007; 67:1–5.

    PubMed  CAS  Google Scholar 

  46. Phillips TM, McBride WH, Pajonk F. The response of CD24(-/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst 2006; 98:1777–1785.

    PubMed  Google Scholar 

  47. Diehn M, Cho RW, Lobo NA et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009; 458:780–783.

    PubMed  CAS  Google Scholar 

  48. Leach JK, Van Tuyle G, Lin PS et al. Ionizing radiation-induced, mitochondria-dependent generation of reactive oxygen/nitrogen. Cancer Res 2001; 61:3894–3901.

    PubMed  CAS  Google Scholar 

  49. Woodward WA, Chen MS, Behbod F et al. WNT/beta-catenin mediates radiation resistance of mouse mammary progenitor cells. Proc Natl Acad Sci USA 2007; 104:618–623.

    PubMed  CAS  Google Scholar 

  50. Muller JM, Chevrier L, Cochaud S et al. Notch and Wnt developmental pathways as targets for anti-cancer drugs. Drug Discovery Today: Disease Mechanisms 2007; 4:285–291.

    Google Scholar 

  51. Wang Z, Li Y, Banerjee S et al. Emerging role of Notch in stem cells and cancer. Cancer Lett 2009; 279:8–12.

    PubMed  CAS  Google Scholar 

  52. Bolos V, Grego-Bessa J, de la Pompa JL. Notch signaling in development and cancer. Endocr Rev 2007; 28:339–363.

    PubMed  CAS  Google Scholar 

  53. Pece S, Serresi M, Santolini E et al. Loss of negative regulation by Numb over Notch is relevant to human breast carcinogenesis. J Cell Biol 2004; 167:215–221.

    PubMed  CAS  Google Scholar 

  54. Politi K, Feirt N, Kitajewski J. Notch in mammary gland development and breast cancer. Semin Cancer Biol 2004; 14:341–347.

    PubMed  CAS  Google Scholar 

  55. Kanamori M, Kawaguchi T, Nigro JM et al. Contribution of Notch signaling activation to human glioblastoma multiforme. J Neurosurg 2007; 106:417–427.

    PubMed  Google Scholar 

  56. Purow BW, Haque RM, Noel MW et al. Expression of Notch-1 and its ligands, Delta-like-1 and Jagged-1, is critical for glioma cell survival and proliferation. Cancer Res 2005; 65:2353–2363.

    PubMed  CAS  Google Scholar 

  57. Nicolas M, Wolfer A, Raj K et al. Notch1 functions as a tumor suppressor in mouse skin. Nat Genet 2003; 33:416–421.

    PubMed  CAS  Google Scholar 

  58. Proweller A, Tu L, Lepore JJ et al. Impaired notch signaling promotes de novo squamous cell carcinoma formation. Cancer Res 2006; 66:7438–7444.

    PubMed  CAS  Google Scholar 

  59. Okuyama R, Nguyen BC, Talora C et al. High commitment of embryonic keratinocytes to terminal differentiation through a Notch1-caspase 3 regulatory mechanism. Dev Cell 2004; 6:551–562.

    PubMed  CAS  Google Scholar 

  60. Rangarajan A, Talora C, Okuyama R et al. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. EMBO J 2001; 20:3427–3436.

    PubMed  CAS  Google Scholar 

  61. Yoon K Gaiano N. Notch signaling in the mammalian central nervous system: insights from mouse mutants. Nat Neurosci 2005; 8:709–715.

    PubMed  CAS  Google Scholar 

  62. Androutsellis-Theotokis A, Leker RR, Soldner F et al. Notch signalling regulates stem cell numbers in vitro and in vivo. Nature 2006; 442:823–826.

    PubMed  CAS  Google Scholar 

  63. Fan X, Matsui W, Khaki L et al. Notch pathway inhibition depletes stem-like cells and blocks engraftment in embryonal brain tumors. Cancer Res 2006; 66:7445–7452.

    PubMed  CAS  Google Scholar 

  64. Fan X, Khaki L, Zhu TS et al. NOTCH pathway blockade depletes CD133 positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts. Stem Cells 2010; 28:5–16.

    PubMed  CAS  Google Scholar 

  65. Liu S, Dontu G, Wicha MS. Mammary stem cells, self-renewal pathways and carcinogenesis. Breast Cancer Res 2005; 7:86–95.

    PubMed  CAS  Google Scholar 

  66. Dontu G, Jackson KW, McNicholas E et al. Role of Notch signaling in cell-fate determination of human mammary stem/progenitor cells. Breast Cancer Res 2004; 6:R605–R615.

    PubMed  CAS  Google Scholar 

  67. Bouras T, Pal B, Vaillant F et al. Notch signaling regulates mammary stem cell function and luminal cell-fate commitment. Cell Stem Cell 2008; 3:429–441.

    PubMed  CAS  Google Scholar 

  68. Buono KD, Robinson GW, Martin C et al. The canonical Notch/RBP-J signaling pathway controls the balance of cell lineages in mammary epithelium during pregnancy. Dev Biol 2006; 293:565–580.

    PubMed  CAS  Google Scholar 

  69. Dickson BC, Mulligan AM, Zhang H et al. High-level JAG1 mRNA and protein predict poor outcome in breast cancer. Mod Pathol 2007; 20:685–693.

    PubMed  CAS  Google Scholar 

  70. Reedijk M, Odorcic S, Chang L 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; 65:8530–8537.

    PubMed  CAS  Google Scholar 

  71. Magnifico A, Albano L, Campaner S et al. Tumor-initiating cells of HER2-positive carcinoma cell lines express the highest oncoprotein levels and are sensitive to trastuzumab. Clin Cancer Res 2009; 15:2010–2021.

    PubMed  CAS  Google Scholar 

  72. Phillips TM, Kim K, Vlashi E et al. Effects of recombinant erythropoietin on breast cancer-initiating cells. Neoplasia 2007; 9:1122–1129.

    PubMed  CAS  Google Scholar 

  73. Sansone P, Storci G, Giovannini C 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; 25:807–815.

    PubMed  CAS  Google Scholar 

  74. Sansone P, Storci G, Tavolari S et al. IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. J Clin Invest 2007; 117:3988–4002.

    PubMed  CAS  Google Scholar 

  75. Gal H, Amariglio N, Trakhtenbrot L et al. Gene expression profiles of AML derived stem cells; similarity to hematopoietic stem cells. Leukemia 2006; 20:2147–2154.

    PubMed  CAS  Google Scholar 

  76. DeAngelis LM, Brain tumors. N Engl J Med 2001; 344:114–123.

    CAS  Google Scholar 

  77. Grossman SA, Batara JF. Current management of glioblastoma multiforme. Semin Oncol 2004; 31:635–644.

    PubMed  CAS  Google Scholar 

  78. Stupp R, Mason WP, van den Bent MJ et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005; 352:987–996.

    PubMed  CAS  Google Scholar 

  79. Scharpfenecker M, Kruse JJ, Sprong D et al. Ionizing radiation shifts the PAI-1/ID-1 balance and activates notch signaling in endothelial cells. Int J Radiat Oncol Biol Phys 2009; 73:506–513.

    PubMed  CAS  Google Scholar 

  80. Wang J, Wakeman TP, Lathia JD et al. Notch Promotes Radioresistance of Glioma Stem Cells. Stem Cells 2010; 28:17–28.

    PubMed  CAS  Google Scholar 

  81. Sade H, Krishna S, Sarin A. The anti-apoptotic effect of Notch-1 requires p56lck-dependent, Akt/PKB-mediated signaling in T-cells. J Biol Chem 2004; 279:2937–2944.

    PubMed  CAS  Google Scholar 

  82. Perumalsamy LR, Nagala M, Banerjee P et al. A hierarchical cascade activated by noncanonical Notch signaling and the mTOR-Rictor complex regulates neglect-induced death in mammalian cells. Cell Death Differ 2009; 16:879–889.

    PubMed  CAS  Google Scholar 

  83. Palomero T, Sulis ML, Cortina M et al. Mutational loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia. Nat Med 2007; 13:1203–1210.

    PubMed  CAS  Google Scholar 

  84. Gutierrez A, Look AT. NOTCH and PI3K-AKT pathways intertwined. Cancer Cell 2007; 12:411–413.

    PubMed  CAS  Google Scholar 

  85. Calzavara E, Chiaramonte R, Cesana D et al. Reciprocal regulation of Notch and PI3K/Akt signalling in T-AL cells in vitro. J Cell Biochem 2008; 103:1405–1412.

    PubMed  CAS  Google Scholar 

  86. Mungamuri SK, Yang X, Thor AD et al. Survival signaling by Notch1: mammalian target of rapamycin (mTOR)-dependent inhibition of p53. Cancer Res 2006; 66:4715–4724.

    PubMed  CAS  Google Scholar 

  87. Eyler CE, Foo WC, LaFiura KM et al. Brain cancer stem cells display preferential sensitivity to Akt inhibition. Stem Cells 2008; 26:3027–3036.

    PubMed  CAS  Google Scholar 

  88. Phillips HS, Kharbanda S, Chen R et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression and resemble stages in neurogenesis. Cancer Cell 2006; 9:157–173.

    PubMed  CAS  Google Scholar 

  89. Pannuti A, Foreman K, Rizzo P et al. Targeting Notch to target cancer stem cells. Clin Cancer Res 2010; 16:3141–3152.

    PubMed  CAS  Google Scholar 

  90. Takebe N Ivy, SP. Controversies in cancer stem cells: targeting embryonic signaling pathways. Clin Cancer Res 2010; 16:3106–3112.

    PubMed  CAS  Google Scholar 

  91. Shih Ie M, Wang TL. Notch signaling, gamma-secretase inhibitors and cancer therapy. Cancer Res 2007; 67:1879–1882.

    Google Scholar 

  92. Armogida M, Petit A, Vincent B et al. Endogenous beta-amyloid production in presenilin-deficient embryonic mouse fibroblasts. Nat Cell Biol 2001; 3:1030–1033.

    PubMed  CAS  Google Scholar 

  93. Huppert SS, Le A, Schroeter EH et al. Embryonic lethality in mice homozygous for a processing-deficient allele of Notch1. Nature 2000; 405:966–970.

    PubMed  CAS  Google Scholar 

  94. Imbimbo BP. Alzheimer’s disease: [gamma]-secretase inhibitors. Drug Discovery Today: Therapeutic Strategies 2008; 5:169–175.

    Google Scholar 

  95. O’Neil J, Calvo J, McKenna K et al. Activating Notch1 mutations in mouse models of T-ALL. Blood 2006; 107:781–785.

    PubMed  Google Scholar 

  96. Curry CL, Reed LL, Golde TE et al. Gamma secretase inhibitor blocks Notch activation and induces apoptosis in Kaposi’s sarcoma tumor cells. Oncogene 2005; 24:6333–6344.

    PubMed  CAS  Google Scholar 

  97. Hallahan AR, Pritchard JI, Hansen S et al. The SmoA1 mouse model reveals that notch signaling is critical for the growth and survival of sonic hedgehog-induced medulloblastomas. Cancer Res 2004; 64:7794–7800.

    PubMed  CAS  Google Scholar 

  98. Wang L, Rahn JJ, Lun X et al. Gamma-secretase represents a therapeutic target for the treatment of invasive glioma mediated by the p75 neurotrophin receptor. PLoS Biol 2008; 6:e289.

    Google Scholar 

  99. Loane DJ, Pocivavsek A, Moussa CE et al. Amyloid precursor protein secretases as therapeutic targets for traumatic brain injury. Nat Med 2009; 15:377–379.

    PubMed  CAS  Google Scholar 

  100. Imbimbo BP. Therapeutic potential of gamma-secretase inhibitors and modulators. Curr Top Med Chem 2008; 8:54–61.

    PubMed  CAS  Google Scholar 

  101. Dovey HF, John V, Anderson JP et al. Functional gamma-secretase inhibitors reduce beta-amyloid peptide levels in brain. J Neurochem 2001; 76:173–181.

    PubMed  CAS  Google Scholar 

  102. Arumugam TV, Chan SL, Jo DG et al. Gamma secretase-mediated Notch signaling worsens brain damage and functional outcome in ischemic stroke. Nat Med 2006; 12:621–623.

    PubMed  CAS  Google Scholar 

  103. Osipo C, Golde TE, Osborne BA et al. Off the beaten pathway: the complex cross talk between Notch and NF-kappaB. Lab Invest 2008; 88:11–17.

    PubMed  CAS  Google Scholar 

  104. Dotto GP. Notch tumor suppressor function. Oncogene 2008; 27:5115–5123.

    PubMed  CAS  Google Scholar 

  105. Fan X, Mikolaenko I, Elhassan I et al. Notch1 and notch2 have opposite effects on embryonal brain tumor growth. Cancer Res 2004; 64:7787–7793.

    PubMed  CAS  Google Scholar 

  106. Graziani I, Eliasz S, De Marco MA et al. Opposite effects of Notch-1 and Notch-2 on mesothelioma cell survival under hypoxia are exerted through the Akt pathway. Cancer Res 2008; 68:9678–9685.

    PubMed  CAS  Google Scholar 

  107. Aste-Amézaga M, Zhang N, Lineberger JE et al. Characterization of Notch1 antibodies that inhibit signaling of both normal and mutated Notch1 receptors. PLoS ONE5:e9094.

    Google Scholar 

  108. Hoey T, Yen WC, Axelrod F et al. DLL4 blockade inhibits tumor growth and reduces tumor-initiating cell frequency. Cell Stem Cell 2009; 5:168–177.

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jialiang Wang .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2012 Landes Bioscience and Springer Science+Business Media

About this chapter

Cite this chapter

Wang, J., Sullenger, B.A., Rich, J.N. (2012). Notch Signaling in Cancer Stem Cells. In: Reichrath, J., Reichrath, S. (eds) Notch Signaling in Embryology and Cancer. Advances in Experimental Medicine and Biology, vol 727. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-0899-4_13

Download citation

Publish with us

Policies and ethics