DOI QR코드

DOI QR Code

Application of Stem Cells in Targeted Therapy of Breast Cancer: A Systematic Review

  • Madjd, Zahra (Department of Pathology, Iran University of Medical Sciences) ;
  • Gheytanchi, Elmira (Oncopathology Research Centre, Iran University of Medical Sciences) ;
  • Erfani, Elham (Oncopathology Research Centre, Iran University of Medical Sciences) ;
  • Asadi-Lari, Mohsen (Department of Epidemiology, Oncopathology Research Centre, Iran University of Medical Sciences)
  • Published : 2013.05.30

Abstract

Background: The aim of this systematic review was to investigate whether stem cells could be effectively applied in targeted therapy of breast cancer. Material and Method: A systematic literature search was performed for original articles published from January 2007 until May 2012. Results: Nine studies met the inclusion criteria for phase I or II clinical trials, of which three used stem cells as vehicles, two trials used autologous hematopoetic stem cells and in four trials cancer stem cells were targeted. Mesenchymal stem cells (MSCs) were applied as cellular vehicles to transfer therapeutic agents. Cell therapy with MSC can successfully target resistant cancers. Cancer stem cells were selectively targeted via a proteasome-dependent suicide gene leading to tumor regression. $Wnt/{\beta}$-catenin signaling pathway has been also evidenced to be an attractive CSC-target. Conclusions: This systematic review focused on two different concepts of stem cells and breast cancer marking a turning point in the trials that applied stem cells as cellular vehicles for targeted delivery therapy as well as CSC-targeted therapies. Applying stem cells as targeted therapy could be an effective therapeutic approach for treatment of breast cancer in the clinic and in therapeutic marketing; however this needs to be confirmed with further clinical investigations.

Keywords

References

  1. Ablett MP, Singh JK, Clarke RB (2012). Stem cells in breast tumours: are they ready for the clinic? Eur J Cancer, 48, 2104-16. https://doi.org/10.1016/j.ejca.2012.03.019
  2. Aboody KS, Najbauer J, Danks MK (2008). Stem and progenitor cell-mediated tumor selective gene therapy. Gene Ther, 15, 739-52. https://doi.org/10.1038/gt.2008.41
  3. Al-Hajj M, Becker MW, Wicha M, Weissman I, Clarke MF (2004). Therapeutic implications of cancer stem cells. Curr Opin Genet Dev, 14, 43-7. https://doi.org/10.1016/j.gde.2003.11.007
  4. 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, 3983-8. https://doi.org/10.1073/pnas.0530291100
  5. Austin J, Kimble J (1987). glp-1 is required in the germ line for regulation of the decision between mitosis and meiosis in C. elegans. Cell, 51, 589-99. https://doi.org/10.1016/0092-8674(87)90128-0
  6. Ayyanan A, Civenni G, Ciarloni L, et al (2006). Increased Wnt signaling triggers oncogenic conversion of human breast epithelial cells by a Notch-dependent mechanism. Proc Natl Acad Sci USA, 103, 3799-804. https://doi.org/10.1073/pnas.0600065103
  7. Bafico A, Liu G, Goldin L, Harris V, Aaronson SA (2004). An autocrine mechanism for constitutive Wnt pathway activation in human cancer cells. Cancer Cell, 6, 497-506. https://doi.org/10.1016/j.ccr.2004.09.032
  8. Balicki D (2007) Moving forward in human mammary stem cell biology and breast cancer prognostication using ALDH1. Cell Stem Cell, 1, 485-7. https://doi.org/10.1016/j.stem.2007.10.015
  9. Banna GL, Simonelli M, Santoro A (2007) High-dose chemotherapy followed by autologous hematopoietic stemcell transplantation for the treatment of solid tumors in adults: a critical review. Curr Stem Cell Res Ther, 2, 65-82. https://doi.org/10.2174/157488807779316964
  10. Barker N, Clevers H (2006). Mining the Wnt pathway for cancer therapeutics. Nat Rev Drug Discov, 5, 997-1014. https://doi.org/10.1038/nrd2154
  11. Behbod F, Rosen JM (2005) Will cancer stem cells provide new therapeutic targets? Carcinogenesis, 26, 703-11. https://doi.org/10.1093/carcin/bgh293
  12. Berry DA, Ueno NT, Johnson MM, et al (2011) High-dose chemotherapy with autologous stem-cell support as adjuvant therapy in breast cancer: overview of 15 randomized trials. J Clin Oncol, 29, 3214-23. https://doi.org/10.1200/JCO.2010.32.5910
  13. Bohl SR, Pircher A, Hilbe W (2011) Cancer stem cells: characteristics and their potential role for new therapeutic strategies. Onkologie, 34, 269-74. https://doi.org/10.1159/000327815
  14. Chaffer CL, Brueckmann I, Scheel C, et al (2011). Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci USA, 108, 7950-5. https://doi.org/10.1073/pnas.1102454108
  15. Chan EF, Gat U, McNiff JM, Fuchs E (1999). A common human skin tumour is caused by activating mutations in beta-catenin. Nat Genet, 21, 410-3. https://doi.org/10.1038/7747
  16. Clarke MF, Dick JE, Dirks PB, et al (2006). Cancer stem cells--perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res, 66, 9339-44. https://doi.org/10.1158/0008-5472.CAN-06-3126
  17. Coffelt SB, Marini FC, Watson K, et al (2009). The proinflammatory peptide LL-37 promotes ovarian tumor progression through recruitment of multipotent mesenchymal stromal cells. Proc Natl Acad Sci USA, 106, 3806-11. https://doi.org/10.1073/pnas.0900244106
  18. Cosse JP, Michiels C (2008). Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression. Anticancer Agents Med Chem, 8, 790-7. https://doi.org/10.2174/187152008785914798
  19. Curtin JC, Lorenzi MV (2010). Drug discovery approaches to target Wnt signaling in cancer stem Cells. Oncotarget, 1, 552-66.
  20. Del Bufalo D, Biroccio A, Soddu S, et al (1996). Lonidamine induces apoptosis in drug-resistant cells independently of the p53 gene. J Clin Invest, 98, 1165-73. https://doi.org/10.1172/JCI118900
  21. Deonarain MP, Kousparou CA, Epenetos AA (2009) .Antibodies targeting cancer stem cells: a new paradigm in immunotherapy? MAbs, 1, 12-25. https://doi.org/10.4161/mabs.1.1.7347
  22. Dickinson ME, McMahon AP (1992). The role of Wnt genes in vertebrate development. Curr Opin Genet Dev, 2, 562-6. https://doi.org/10.1016/S0959-437X(05)80172-8
  23. Duan X, Guan H, Cao Y, Kleinerman ES (2009). Murine bone marrow-derived mesenchymal stem cells as vehicles for interleukin-12 gene delivery into Ewing sarcoma tumors. Cancer, 115, 13-22. https://doi.org/10.1002/cncr.24013
  24. Dwyer RM, Potter-Beirne SM, Harrington KA, et al (2007). Monocyte chemotactic protein-1 secreted by primary breast tumors stimulates migration of mesenchymal stem cells. Clin Cancer Res 13(17): 5020-7. https://doi.org/10.1158/1078-0432.CCR-07-0731
  25. Dwyer RM, Ryan J, Havelin RJ, et al (2011). Mesenchymal Stem Cell-mediated delivery of the sodium iodide symporter supports radionuclide imaging and treatment of breast cancer. Stem Cells 29(7): 1149-57. https://doi.org/10.1002/stem.665
  26. El-Haibi CP, Karnoub AE (2010). Mesenchymal stem cells in the pathogenesis and therapy of breast cancer. J Mammary Gland Biol Neoplasia, 15, 399-409. https://doi.org/10.1007/s10911-010-9196-7
  27. Ellisen LW, Bird J, West DC, et al (1991). TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms. Cell, 66, 649-61. https://doi.org/10.1016/0092-8674(91)90111-B
  28. Evangelista M, Tian H, de Sauvage FJ (2006). The hedgehog signaling pathway in cancer. Clin Cancer Res, 12, 5924-8. https://doi.org/10.1158/1078-0432.CCR-06-1736
  29. Franovic A, Gunaratnam L, Smith K, et al (2007). Translational up-regulation of the EGFR by tumor hypoxia provides a nonmutational explanation for its overexpression in human cancer. Proc Natl Acad Sci USA, 104, 13092-7. https://doi.org/10.1073/pnas.0702387104
  30. Gailani MR, Bale AE (1999). Acquired and inherited basal cell carcinomas and the patched gene. Adv Dermatol, 14, 261-83; discussion 284.
  31. Giles RH, van Es JH, Clevers H (2003). Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta, 1653, 1-24.
  32. Ginestier C, Hur MH, Charafe-Jauffret E, 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, 555-67. https://doi.org/10.1016/j.stem.2007.08.014
  33. Ginestier C, Wicha MS (2007). Mammary stem cell number as a determinate of breast cancer risk. Breast Cancer Res, 9, 109. https://doi.org/10.1186/bcr1741
  34. Grisendi G, Bussolari R, Cafarelli L, et al (2010). Adiposederived mesenchymal stem cells as stable source of tumor necrosis factor-related apoptosis-inducing ligand delivery for cancer therapy. Cancer Res, 70, 3718-29. https://doi.org/10.1158/0008-5472.CAN-09-1865
  35. Gupta PB, Fillmore CM, Jiang G, et al (2011). Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell, 146, 633-44. https://doi.org/10.1016/j.cell.2011.07.026
  36. Han ZB, Ren H, Zhao H, et al (2008). Hypoxia-inducible factor (HIF)-1 alpha directly enhances the transcriptional activity of stem cell factor (SCF) in response to hypoxia and epidermal growth factor (EGF). Carcinogenesis, 29, 1853-61. https://doi.org/10.1093/carcin/bgn066
  37. Harris AL (2002). Hypoxia - a key regulatory factor in tumour growth. Nat Rev Cancer, 2, 38-47. https://doi.org/10.1038/nrc704
  38. Harris AL, Hochhauser D (1992). Mechanisms of multidrug resistance in cancer treatment. Acta Oncol, 31, 205-13. https://doi.org/10.3109/02841869209088904
  39. Henrique D, Hirsinger E, Adam J, et al (1997). Maintenance of neuroepithelial progenitor cells by Delta-Notch signalling in the embryonic chick retina. Curr Biol, 7, 661-70. https://doi.org/10.1016/S0960-9822(06)00293-4
  40. Heppner GH (1984). Tumor heterogeneity. Cancer Res, 44, 2259-65.
  41. Hoey T, Yen WC, Axelrod F, et al (2009). DLL4 blockade inhibits tumor growth and reduces tumor-initiating cell frequency. Cell Stem Cell, 5, 168-77. https://doi.org/10.1016/j.stem.2009.05.019
  42. Jain M, Arvanitis C, Chu K, et al (2002). Sustained loss of a neoplastic phenotype by brief inactivation of MYC. Science, 297, 102-4. https://doi.org/10.1126/science.1071489
  43. Jaiswal S, Traver D, Miyamoto T, et al (2003). Expression of BCR/ABL and BCL-2 in myeloid progenitors leads to myeloid leukemias. Proc Natl Acad Sci USA, 100, 10002-7. https://doi.org/10.1073/pnas.1633833100
  44. Jo M, Kim TH, Seol DW, et al (2000). Apoptosis induced in normal human hepatocytes by tumor necrosis factor-related apoptosis-inducing ligand. Nat Med, 6, 564-7. https://doi.org/10.1038/75045
  45. Karnoub AE, Dash AB, Vo AP, et al (2007). Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature, 449, 557-63. https://doi.org/10.1038/nature06188
  46. Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser A (2007). Tumor growth need not be driven by rare cancer stem cells. Science, 317, 337. https://doi.org/10.1126/science.1142596
  47. Khakoo AY, Pati S, Anderson SA, et al (2006). Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi's sarcoma. J Exp Med, 203, 1235-47 https://doi.org/10.1084/jem.20051921
  48. Kim K, Lu Z, Hay ED (2002). Direct evidence for a role of betacatenin/LEF-1 signaling pathway in induction of EMT. Cell Biol Int, 26, 463-76. https://doi.org/10.1006/cbir.2002.0901
  49. Kim SM, Oh JH, Park SA, et al (2010). Irradiation enhances the tumor tropism and therapeutic potential of tumor necrosis factor-related apoptosis-inducing ligand-secreting human umbilical cord blood-derived mesenchymal stem cells in glioma therapy. Stem Cells, 28, 2217-28. https://doi.org/10.1002/stem.543
  50. Kintner C (1992). Molecular bases of early neural development in Xenopus embryos. Annu Rev Neurosci, 15, 251-84. https://doi.org/10.1146/annurev.ne.15.030192.001343
  51. Klopocki E, Kristiansen G, Wild PJ, et al (2004). Loss of SFRP1 is associated with breast cancer progression and poor prognosis in early stage tumors. Int J Oncol, 25, 641-9.
  52. Klopp AH, Spaeth EL, Dembinski JL, et al (2007). Tumor irradiation increases the recruitment of circulating mesenchymal stem cells into the tumor microenvironment. Cancer Res, 67, 11687-95. https://doi.org/10.1158/0008-5472.CAN-07-1406
  53. Knudson AG, Strong LC, Anderson DE (1973). Heredity and cancer in man. Prog Med Genet, 9, 113-58.
  54. Komarova S, Kawakami Y, Stoff-Khalili MA, Curiel DT, Pereboeva L (2006). Mesenchymal progenitor cells as cellular vehicles for delivery of oncolytic adenoviruses. Mol Cancer Ther, 5, 755-66. https://doi.org/10.1158/1535-7163.MCT-05-0334
  55. Lehmann BD, Bauer JA, Chen X, et al (2011). Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest, 121, 2750-67. https://doi.org/10.1172/JCI45014
  56. Leong KG, Karsan A (2006). Recent insights into the role of Notch signaling in tumorigenesis. Blood, 107, 2223-33. https://doi.org/10.1182/blood-2005-08-3329
  57. Li C, Heidt DG, Dalerba P, et al (2007) Identification of pancreatic cancer stem cells. Cancer Res, 67, 1030-7. https://doi.org/10.1158/0008-5472.CAN-06-2030
  58. Li L, Clevers H (2010) Coexistence of quiescent and active adult stem cells in mammals. Science, 327, 542-5 https://doi.org/10.1126/science.1180794
  59. Li X, Lewis MT, Huang J, et al (2008). Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst, 100, 672-9. https://doi.org/10.1093/jnci/djn123
  60. Li YC, Fung KP, Kwok TT, et al (2002). Mitochondrial targeting drug lonidamine triggered apoptosis in doxorubicin-resistant HepG2 cells. Life Sci, 71, 2729-40. https://doi.org/10.1016/S0024-3205(02)02103-3
  61. Liebner S, Cattelino A, Gallini R, et al (2004). Beta-catenin is required for endothelial-mesenchymal transformation during heart cushion development in the mouse. J Cell Biol, 166, 359-67. https://doi.org/10.1083/jcb.200403050
  62. Lin SY, Xia W, Wang JC, et al (2000). Beta-catenin, a novel prognostic marker for breast cancer: its roles in cyclin D1 expression and cancer progression. Proc Natl Acad Sci USA, 97, 4262-6. https://doi.org/10.1073/pnas.060025397
  63. Lindeman GJ, Visvader JE (2010). Insights into the cell of origin in breast cancer and breast cancer stem cells. Asia Pac J Clin Oncol, 6, 89-97. https://doi.org/10.1111/j.1743-7563.2010.01279.x
  64. Liu S, Dontu G, Mantle ID, et al (2006). Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res, 66, 6063-71. https://doi.org/10.1158/0008-5472.CAN-06-0054
  65. Luistro L, He W, Smith M, et al (2009). Preclinical profile of a potent gamma-secretase inhibitor targeting notch signaling with in vivo efficacy and pharmacodynamic properties. Cancer Res, 69, 7672-80. https://doi.org/10.1158/0008-5472.CAN-09-1843
  66. Madjd Z, Ramezani B, Molanae S, Asadi-Lari M (2012). High expression of stem cell marker ALDH1 is associated with reduced BRCA1 in invasive breast carcinomas. Asian Pac J Cancer Prev, 13, 2973-8. https://doi.org/10.7314/APJCP.2012.13.6.2973
  67. Massard C, Deutsch E, Soria JC (2006) Tumour stem celltargeted treatment: elimination or differentiation. Ann Oncol, 17, 1620-4. https://doi.org/10.1093/annonc/mdl074
  68. Menon LG, Picinich S, Koneru R, et al (2007). Differential gene expression associated with migration of mesenchymal stem cells to conditioned medium from tumor cells or bone marrow cells. Stem Cells, 25, 520-8. https://doi.org/10.1634/stemcells.2006-0257
  69. Milane L, Duan Z, Amiji M (2011). Therapeutic efficacy and safety of paclitaxel/lonidamine loaded EGFR-targeted nanoparticles for the treatment of multi-drug resistant cancer. PLoS One, 6, 24075. https://doi.org/10.1371/journal.pone.0024075
  70. Morrison SJ, Qian D, Jerabek L, et al (2002). A genetic determinant that specifically regulates the frequency of hematopoietic stem cells. J Immunol, 168, 635-42. https://doi.org/10.4049/jimmunol.168.2.635
  71. Muller T, Bain G, Wang X, Papkoff J (2002). Regulation of epithelial cell migration and tumor formation by beta-catenin signaling. Exp Cell Res, 280, 119-33. https://doi.org/10.1006/excr.2002.5630
  72. Nakamizo A, Marini F, Amano T, et al (2005). Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res, 65, 3307-18.
  73. Nakshatri H (2010). Radiation resistance in breast cancer: are CD44+/CD24-/proteosome low/PKH26+ cells to blame? Breast Cancer Res, 12, 105. https://doi.org/10.1186/bcr2559
  74. O'Brien CA, Pollett A, Gallinger S, Dick JE (2007) A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature, 445, 106-10. https://doi.org/10.1038/nature05372
  75. Orian-Rousseau V (2010) CD44, a therapeutic target for metastasising tumours. Eur J Cancer, 46, 1271-7. https://doi.org/10.1016/j.ejca.2010.02.024
  76. Ozawa K, Sato K, Oh I, et al (2008). Cell and gene therapy using mesenchymal stem cells (MSCs). J Autoimmun, 30, 121-7. https://doi.org/10.1016/j.jaut.2007.12.008
  77. Park SY, Lee HE, Li H, et al (2010). Heterogeneity for stem cellrelated markers according to tumor subtype and histologic stage in breast cancer. Clin Cancer Res, 16, 876-87 https://doi.org/10.1158/1078-0432.CCR-09-1532
  78. Parkin DM, Bray F, Ferlay J, Pisani P (2001). Estimating the world cancer burden: Globocan 2000. Int J Cancer, 94, 153-6. https://doi.org/10.1002/ijc.1440
  79. Polakis P (2000). Wnt signaling and cancer. Genes Dev, 14, 1837-51.
  80. Prince ME, Sivanandan R, Kaczorowski A, et al (2007). Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA, 104, 973-8 https://doi.org/10.1073/pnas.0610117104
  81. Quintana E, Shackleton M, Sabel MS, et al (2008). Efficient tumour formation by single human melanoma cells. Nature, 456, 593-8. https://doi.org/10.1038/nature07567
  82. Ratajczak MZ (2005). Cancer stem cells--normal stem cells "Jedi" that went over to the "dark side". Folia Histochem Cytobiol, 43, 175-81.
  83. Ravagnan L, Marzo I, Costantini P, et al (1999). Lonidamine triggers apoptosis via a direct, Bcl-2-inhibited effect on the mitochondrial permeability transition pore. Oncogene, 18, 2537-46. https://doi.org/10.1038/sj.onc.1202625
  84. Resetkova E, Reis-Filho JS, Jain RK, et al (2010). Prognostic impact of ALDH1 in breast cancer: a story of stem cells and tumor microenvironment. Breast Cancer Res Treat, 123, 97-108. https://doi.org/10.1007/s10549-009-0619-3
  85. Reya T, Duncan AW, Ailles L, et al (2003). A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature, 423, 409-14. https://doi.org/10.1038/nature01593
  86. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001). Stem cells, cancer, and cancer stem cells. Nature, 414, 105-11. https://doi.org/10.1038/35102167
  87. Robarge KD, Brunton SA, Castanedo GM, et al (2009). GDC-0449-a potent inhibitor of the hedgehog pathway. Bioorg Med Chem Lett, 19, 5576-81. https://doi.org/10.1016/j.bmcl.2009.08.049
  88. Roukos DH, Murray S, Briasoulis E (2007). Molecular genetic tools shape a roadmap towards a more accurate prognostic prediction and personalized management of cancer. Cancer Biol Ther, 6, 308-12. https://doi.org/10.4161/cbt.6.3.3994
  89. Ruiz I, Altaba A (1999). Gli proteins and Hedgehog signaling: development and cancer. Trends Genet, 15, 418-25. https://doi.org/10.1016/S0168-9525(99)01840-5
  90. Schultz LB, Weber BL (1999). Recent advances in breast cancer biology. Curr Opin Oncol, 11, 429-34. https://doi.org/10.1097/00001622-199911000-00002
  91. Sell S, Pierce GB (1994). Maturation arrest of stem cell differentiation is a common pathway for the cellular origin of teratocarcinomas and epithelial cancers. Lab Invest, 70, 6-22.
  92. Semenza GL (2003). Targeting HIF-1 for cancer therapy. Nat Rev Cancer, 3, 721-32. https://doi.org/10.1038/nrc1187
  93. Semenza GL (2008). Hypoxia-inducible factor 1 and cancer pathogenesis. IUBMB Life, 60, 591-7. https://doi.org/10.1002/iub.93
  94. Shackleton M, Quintana E, Fearon ER, Morrison SJ (2009) Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell, 138, 822-9. https://doi.org/10.1016/j.cell.2009.08.017
  95. Singh SK, Clarke ID, Terasaki M, et al (2003). Identification of a cancer stem cell in human brain tumors. Cancer Res, 63, 5821-8.
  96. Snippert HJ, Haegebarth A, Kasper M, et al (2010). Lgr6 marks stem cells in the hair follicle that generate all cell lineages of the skin. Science, 327, 1385-9. https://doi.org/10.1126/science.1184733
  97. Soltanian S, Matin MM (2011). Cancer stem cells and cancer therapy. Tumour Biol, 32, 425-40. https://doi.org/10.1007/s13277-011-0155-8
  98. Spaeth E, Klopp A, Dembinski J, Andreeff M, Marini F (2008). Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells. Gene Ther, 15, 730-8. https://doi.org/10.1038/gt.2008.39
  99. Stockler M, Wilcken NR, Ghersi D, Simes RJ (2000). Systematic reviews of chemotherapy and endocrine therapy in metastatic breast cancer. Cancer Treat Rev, 26, 151-68. https://doi.org/10.1053/ctrv.1999.0161
  100. Studeny M, Marini FC, Dembinski JL, et al (2004). Mesenchymal stem cells: potential precursors for tumor stroma and targeted-delivery vehicles for anticancer agents. J Natl Cancer Inst, 96, 1593-603. https://doi.org/10.1093/jnci/djh299
  101. Thiery JP, Acloque H, Huang RY, Nieto MA (2009). Epithelialmesenchymal transitions in development and disease. Cell, 139, 871-90. https://doi.org/10.1016/j.cell.2009.11.007
  102. Ueno NT, de Souza JA, Booser D, et al (2009). Pilot study of targeted skeletal radiation therapy for bone-only metastatic breast cancer. Clin Breast Cancer, 9, 173-7. https://doi.org/10.3816/CBC.2009.n.028
  103. Viens P, Tarpin C, Roche H, Bertucci F (2010) Systemic therapy of inflammatory breast cancer from high-dose chemotherapy to targeted therapies: the French experience. Cancer, 116, 2829-36. https://doi.org/10.1002/cncr.25168
  104. Visvader JE, Lindeman GJ (2008). Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer, 8, 755-68. https://doi.org/10.1038/nrc2499
  105. Vlashi E, Kim K, Lagadec C, et al (2009). In vivo imaging, tracking, and targeting of cancer stem cells. J Natl Cancer Inst, 101, 350-9. https://doi.org/10.1093/jnci/djn509
  106. Wechsler-Reya RJ, Scott MP (1999). Control of neuronal precursor proliferation in the cerebellum by Sonic Hedgehog. Neuron, 22, 103-14. https://doi.org/10.1016/S0896-6273(00)80682-0
  107. Weissman IL (2000). Stem cells: units of development, units of regeneration, and units in evolution. Cell, 100, 157-68. https://doi.org/10.1016/S0092-8674(00)81692-X
  108. Wicha MS, Liu S, Dontu G (2006). Cancer stem cells: an old idea--a paradigm shift. Cancer Res, 66, 1883-90. https://doi.org/10.1158/0008-5472.CAN-05-3153
  109. Wong SC, Lo SF, Lee KC, et al (2002). Expression of frizzledrelated protein and Wnt-signalling molecules in invasive human breast tumours. J Pathol, 196, 145-53. https://doi.org/10.1002/path.1035
  110. Woodward WA, Chen MS, Behbod F, et al (2007). WNT/betacatenin mediates radiation resistance of mouse mammary progenitor cells. Proc Natl Acad Sci USA, 104, 618-23. https://doi.org/10.1073/pnas.0606599104
  111. Yague E, Arance A, Kubitza L, et al (2007). Ability to acquire drug resistance arises early during the tumorigenesis process. Cancer Res, 67, 1130-7. https://doi.org/10.1158/0008-5472.CAN-06-2574
  112. Yu F, Yao H, Zhu P, et al (2007). let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell, 131, 1109-23. https://doi.org/10.1016/j.cell.2007.10.054
  113. Yuan Z, Goetz JA, Singh S, et al (2007). Frequent requirement of hedgehog signaling in non-small cell lung carcinoma. Oncogene, 26, 1046-55. https://doi.org/10.1038/sj.onc.1209860
  114. Zhang Y, Kalderon D (2001). Hedgehog acts as a somatic stem cell factor in the Drosophila ovary. Nature, 410, 599-604. https://doi.org/10.1038/35069099
  115. Zhu AJ, Watt FM (1999). beta-catenin signalling modulates proliferative potential of human epidermal keratinocytes independently of intercellular adhesion. Development, 126, 2285-98.
  116. Zielske SP, Livant DL, Lawrence TS (2009). Radiation increases invasion of gene-modified mesenchymal stem cells into tumors. Int J Radiat Oncol Biol Phys, 75, 843-53. https://doi.org/10.1016/j.ijrobp.2008.06.1953

Cited by

  1. ALDH1 in Combination with CD44 as Putative Cancer Stem Cell Markers are Correlated with Poor Prognosis in Urothelial Carcinoma of the Urinary Bladder vol.15, pp.5, 2014, https://doi.org/10.7314/APJCP.2014.15.5.2013
  2. ALDH1A1 Maintains Ovarian Cancer Stem Cell-Like Properties by Altered Regulation of Cell Cycle Checkpoint and DNA Repair Network Signaling vol.9, pp.9, 2014, https://doi.org/10.1371/journal.pone.0107142
  3. Combined expression of aldehyde dehydrogenase 1A1 and β-catenin is associated with lymph node metastasis and poor survival in breast cancer patients following cyclophosphamide treatment vol.34, pp.6, 2015, https://doi.org/10.3892/or.2015.4273
  4. Role of ALDH1a and CD44 in transitional and squamous cell carcinomas of urinary bladder vol.37, pp.1, 2017, https://doi.org/10.1097/01.XEJ.0000516729.34502.81
  5. Co-culture with lung cancer A549 cells promotes the proliferation and migration of mesenchymal stem cells derived from bone marrow vol.14, pp.4, 2017, https://doi.org/10.3892/etm.2017.4909