Skip to main content

Advertisement

Log in

Eradicating Cancer Stem Cells: Concepts, Issues, and Challenges

  • Hot Topic
  • Published:
Current Treatment Options in Oncology Aims and scope Submit manuscript

Opinion statement:

The cells of malignant cancers result in the evolution of cells with stem-like characteristics, commonly known as cancer stem cells (CSCs). Progress of anticancer therapies is severely hampered because of disease relapse mostly in a more aggressive form due to CSCs. These CSCs are more or less like embryonic or tissue stem cells, known for their capacity of self-renewal, exactly recapitulate of the original tumor. Deregulation of key stem cell pathways like Wnt, Hedgehog (Hh), and Notch is attributed towards the rise of CSCs. Recent breakthroughs offer better insights into CSC signaling. Scientists have developed several combinatorial therapies like targeting one/multiple of these CSC pathways. The article summarized various markers used to identify CSCs and discuss major signaling pathways in them. The futuristic probabilities to use CSC therapeutics in clinical development have been discussed. Our views have been highlighted on the future directions for targeting advances in the clinical development.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Abbreviations

ABC:

ATP-binding cassette

ALDH1:

aldehyde dehydrogenase 1

APC:

adenomatous polyposis coli

BCC:

basal cell carcinoma

Bmi 1:

B cell-specific Moloney murine leukemia virus integration site 1

BrdU:

bromodeoxyuridine

BTSC:

brain tumor stem cell

CBC:

Crypt base columnar

CCL5:

Chemokine C-C motif ligand 5

CCR5:

Chemokine C-C Motif Receptor 5

CKIε:

Casein Kinase Iε

CSCs:

Cancer stem cells

CXCL12:

C-X-C motif chemokine ligand 12

CXCR4:

Chemokine receptor type 4

DKK1:

Dickkopf related protein 1

DLL:

Delta-like ligands

EMT:

Epithelial-to-mesenchymal transition

EpCAM:

Epithelial cell adhesion molecule

ES cells:

Embryonic stem cells

ESA:

Epithelial surface antigen

FGF20:

Fibroblast growth factor 20

GBM:

Glioblastomas

GCG:

Glucagon

GI:

Gastrointestinal

GPI-AP:

Glycosylphosphatidylinositol-anchored protein

GSI:

Gamma secretase inhibitors

Hh:

Hedgehog

HpSCs:

Hepatocellular stem cells

HSPCs:

Hepatic stem/progenitor cells

ICD:

Intracellular domain

IWR compounds:

Inhibitors of Wnt response compounds

JAG1 and JAG2:

Jagged proteins

MB:

Medullo-blastoma

MSCs:

Mesenchymal stem cells

NDRG2:

N myc downregulated gene 2

NICD:

Notch intracellular domain

NLK:

Nemo-like kinase

NOD-SCID:

Non-obese diabetic- severe combined immunodeficient

PPARγ:

Peroxisome proliferator-activated receptor γ

SCFR:

Stem cell factor receptor

SDF-1:

Stromal cell-derived factor

SHh:

Secreted Hedgehog ligands

Shh:

Sonic Hedgehog

SMO:

Small molecule anti-smoothened

SP:

Side population

TACE:

Tumor, necrosis factor alpha-converting enzyme

TCF:

T cell factor

WISP1:

WNT1- inducible signaling pathway

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Pardal R, Clarke MF, Morrison SJ. Applying the principles of stem-cell biology to cancer. Nat Rev Cancer. 2003;3:895–902.

    Article  CAS  PubMed  Google Scholar 

  2. Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 2005;122:947–56.

  3. Camarasa MV. Directed Differentiation of Pluripotent Cells Towards Therapeutic Stem Cells. Recent Pat Regen Med. 2015;5:85–101.

    CAS  Google Scholar 

  4. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–28.

  5. Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol. 2004;36:568–84.

    Article  CAS  PubMed  Google Scholar 

  6. Krampera M, Pizzolo G, Aprili G, Franchini M. Mesenchymal stem cells for bone, cartilage, tendon and skeletal muscle repair. Bone. 2006;39:678–83.

    Article  CAS  PubMed  Google Scholar 

  7. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, et al. Mesenchymal stem cells within tumor stroma promote breast cancer metastasis. Nature. 2007;449:557–63.

  8. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76.

    Article  CAS  PubMed  Google Scholar 

  9. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–20.

  10. Henrique D, Hirsinger E, Adam J, Le Roux I, Pourquié O, Ish-Horowicz D, et al. Maintenance of neuroepithelial progenitor cells by Delta–Notch signaling in the embryonic chick retina. Curr Biol. 1997;7:661–70.

  11. Bandhavkar S. Cancer stem cells: a metastasizing menace! Cancer Med. 2016;5:649–55.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Van Dussen KL, Carulli AJ, Keeley TM, Patel SR, Puthoff BJ, Magness ST, et al. Notch signaling modulates proliferation and differentiation of intestinal crypt base columnar stem cells. Devel. 2012;139:488–97.

  13. Bray SJ. Notch signaling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006;7:678–89.

    Article  CAS  PubMed  Google Scholar 

  14. Koch U, Lehal R, Radtke F. Stem cells living with a Notch. Devel. 2013;140:689–704.

    CAS  Google Scholar 

  15. Taipale J, Beachy PA. The Hedgehog and Wntsignalling pathways in cancer. Nature. 2001;411:349–54.

    Article  CAS  PubMed  Google Scholar 

  16. Lai K, Kaspar BK, Gage FH, Schaffer DV. Sonic hedgehog regulates adult neural progenitor proliferation in vitro and in vivo. Nature Neurosci. 2003;6:21–7.

    Article  CAS  PubMed  Google Scholar 

  17. Evangelista M, Tian H, de Sauvage FJ. The hedgehog signaling pathway in cancer. Clin Cancer Res. 2006;12:5924–8.

    Article  CAS  PubMed  Google Scholar 

  18. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of human brain tumor initiating cells. Nature. 2004;432:396–401.

  19. Uchida H, Arita K, Yunoue S, Yonezawa H, Shinsato Y, Kawano H, et al. Role of sonic hedgehog signaling in migration of cell lines established from CD133-positive malignant glioma cells. J Neurooncol. 104:697–704.

  20. Irollo E, Pirozzi G. CD133: to be or not to be, is this the real question? Am J Transl Res. 2013;5:563–81.

    PubMed  PubMed Central  Google Scholar 

  21. Giebel B, Corbeil D, Beckmann J, Höhn J, Freund D, Giesen K, et al. Segregation of lipid raft markers including CD133 in polarized human hematopoietic stem and progenitor cells. Blood. 2004;104:2332–8.

  22. Liu G, Yuan X, Zeng Z, Tunici P, Ng H, Abdulkadir IR, Lu L, Irvin D, Black KL, John SY. Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer 2006; 1.

  23. Ma S, Chan KW, Lee TKW, Tang KH, Wo JYH, Zheng BJ, et al. Aldehyde dehydrogenase discriminates the CD133 liver cancer stem cell populations. Mol Cancer Res. 2002;6:1146–53.

  24. Monzani E, Facchetti F, Galmozzi E, Corsini E, Benetti A, Cavazzin C, et al. Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. Eur J Cancer. 2007;43:935–46.

  25. Miki J, Furusato B, Li H, Gu Y, Takahashi H, Egawa S, et al. 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. 2007;67:3153–61.

  26. Ferrandina G, Bonanno G, Pierelli L, Perillo A, Procoli A, Mariotti A, et al. Expression of CD133–1 and CD133–2 in ovarian cancer. Int J Gynecol Cancer. 2008;18:506–14.

  27. Beier D, Hau P, Proescholdt M, Lohmeier A, Wischhusen J, Oefner PJ, et al. CD133+ and CD133− glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res. 2007;67:4010–5.

  28. Lottaz C, Beier D, Meyer K, Kumar P, Hermann A, Schwarz J, et al. Transcriptional profiles of CD133+ and CD133− glioblastoma-derived cancer stem cell lines suggest different cells of origin. Cancer Res. 2010;70:2030–40.

  29. Schmelzer E, Reid LM. EpCAM expression in normal, non-pathological tissues. Front Biosci. 2007;13:3096–100.

    Article  Google Scholar 

  30. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704–15.

  31. Pantel K, Alix-Panabières C. Circulating tumor cells in cancer patients: challenges and perspectives. Trends Mol Med. 2010;16:398–406.

    Article  PubMed  Google Scholar 

  32. Munz M, Baeuerle PA, Gires O. The Emerging Role of EpCAM in Cancer and Stem Cell Signaling. Cancer Res. 2009;69:5627–9.

    Article  CAS  PubMed  Google Scholar 

  33. Maetzel D, Denzel S, Mack B, Canis M, Went P, Benk M, et al. Nuclear signaling by tumor-associated antigen EpCAM. Nat Cell Biol. 2009;11:162–71.

  34. Yamashita T, Budhu A, Forgues M, Wang XW. Activation of hepatic stem cell marker EpCAM by Wnt–β-catenin signaling in hepatocellular carcinoma. Cancer Res. 2007;67:10,831–9.

    Article  CAS  Google Scholar 

  35. Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW, et al. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci U S A. 2007;104:10,158–63.

  36. Jiang F, Qiu Q, Khanna A, Todd NW, Deepak J, Xing L, et al. Aldehyde Dehydrogenase 1 Is a Tumor Stem Cell-Associated Marker in Lung Cancer. Mol Cancer Res. 2009;7:330–8.

  37. Croker AK, Goodale D, Chu J, Postenka C, Hedley BD, Hess DA, et al. High aldehyde dehydrogenase and expression of cancer stem cell markers selects for breast cancer cells with enhanced malignant and metastatic ability. J Cell Mol Med. 2009;13:2236–52.

  38. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007;1:313–23.

  39. Elkord AJA. E, Significance of CD44 and CD24 as Cancer Stem Cell Markers: An Enduring Ambiguity. Clin Dev Immunol. 2012;2012:11.

    Google Scholar 

  40. Leung ELH, Fiscus RR, Tung JW, Tin VPC, Cheng LC, Sihoe ADL, et al. Non-small cell lung cancer cells expressing CD44 are enriched for stem cell-like properties. PloS one. 2010;5:e14062.

  41. Godar S, Ince TA, Bell GW, Feldser D, Donaher JL, Bergh J, et al. Growth-inhibitory and tumor-suppressive functions of p53 depend on its repression of CD44 expression. Cell. 2008;134:62–73.

  42. Günthert U, Hofmann M, Rudy W, Reber S, Zöller M, Hauβmann I, et al. new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell. 1991;65:13–24.

  43. Weber GF, Bronson RT, Ilagan J, Cantor H, Schmits R, Mak TW. Absence of the CD44 gene prevents sarcoma metastasis. Cancer Res. 2002;62:2281–6.

    CAS  PubMed  Google Scholar 

  44. Nilsson SK, Johnston HM, Whitty GA, Williams B, Webb RJ, Denhardt DT, et al. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive. Blood. 2005;106:1232–9.

  45. Rangaswami H, Bulbule A, Kundu GC. Osteopontin: role in cell signaling and cancer progression. Trends Cell Biol. 2006;16:79–87.

    Article  CAS  PubMed  Google Scholar 

  46. Napier SL, Healy ZR, Schnaar RL, Konstantopoulos K. Selectin Ligand Expression Regulates the Initial Vascular Interactions of Colon Carcinoma Cells: the roles of cd44v and alternative sialofucosylated selectin ligands. J Biol Chem. 2007;282:3433–41.

    Article  CAS  PubMed  Google Scholar 

  47. Bourguignon LY. CD44-mediated oncogenic signaling and cytoskeleton activation during mammary tumor progression. J Mammary Gland Biol Neoplasia. 2001;6:287–97.

    Article  CAS  PubMed  Google Scholar 

  48. Du L, Wang H, He L, Zhang J, Ni B, Wang X, et al. CD44 is of functional importance for colorectal cancer stem cells. Clin Cancer Res. 2008;14:6751–60.

  49. Bapat SA. Human ovarian cancer stem cells. Reproduction. 2010;140:33–41.

    Article  CAS  PubMed  Google Scholar 

  50. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 2005;65:10,946–51.

    Article  CAS  Google Scholar 

  51. Patrawala L, Calhoun T, Schneider-Broussard R, Li H, Bhatia B, Tang S, et al. Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene. 2006;25:1696–708.

  52. Allegra E, Trapasso S, Pisani D, Puzzo L. The role of BMI1 as a biomarker of cancer stem cells in head and neck cancer: a review. Oncology. 2014;86:199–205.

    Article  CAS  PubMed  Google Scholar 

  53. Proctor E, Waghray M, Lee CJ, Heidt DG, Yalamanchili M, Li C, et al. Bmi1 enhances tumorigenicity and cancer stem cell function in pancreatic adenocarcinoma. PloS one. 2013;8:e55820.

  54. Wei XD, He J, Wang JY, Yang XL, Ma BJ. Bmi-1 is essential for the oncogenic potential in CD133(+) human laryngeal cancer cells. Tumor Biol. 2015;36:8931–42.

    Article  CAS  Google Scholar 

  55. Zheng J, Li Y, Yang J, Liu Q, Shi M, Zhang R, et al. NDRG2 inhibits hepatocellular carcinoma adhesion, migration and invasion by regulating CD24 expression. BMC Cancer. 2011;11:1–9.

  56. Aigner S, Ramos CL, Hafezi-moghadam A, Lawrence MB, Friederichs J, Altevogt P, et al. CD24 mediates rolling of breast carcinoma cells on P-selectin. FASEB J. 1998;12:1241–51.

  57. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, et al. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67:1030–7.

  58. Ricardo S, Vieira AF, Gerhard R, Leitão D, Pinto R, Cameselle-Teijeiro JF, Milanezi F, Schmitt F, Paredes J. Breast cancer stem cell markers CD44, CD24 and ALDH1: expression distribution within intrinsic molecular subtype. J Clin Pathol 2011; jcp. 2011.090456.

  59. Rege TA, Hagood JS. Thy-1 as a regulator of cell-cell and cell-matrix interactions in axon regeneration, apoptosis, adhesion, migration, cancer, and fibrosis. FASEB J. 2006;20:1045–54.

    Article  CAS  PubMed  Google Scholar 

  60. Dennis JE, Esterly K, Awadallah A, Parrish CR, Poynter GM, Goltry KL. Clinical-Scale Expansion of a Mixed Population of Bone Marrow-Derived Stem and Progenitor Cells for Potential Use in Bone Tissue Regeneration. Stem Cells. 2007;25:2575–82.

    Article  PubMed  Google Scholar 

  61. Cho RW, Wang X, Diehn M, Shedden K, Chen GY, Sherlock G, et al. Isolation and molecular characterization of cancer stem cells in MMTV-Wnt-1 murine breast tumors. Stem Cells. 2008;26:364–71.

  62. Herrera MB, Bruno S, Buttiglieri S, Tetta C, Gatti S, Deregibus MC, et al. Isolation and characterization of a stem cell population from adult human liver. Stem Cells. 2006;24:2840–50.

  63. Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P, et al. Significance of CD90+ Cancer Stem Cells in Human Liver Cancer. Cancer Cell. 2008;13:153–66.

  64. Dallas NA, Samuel S, Xia L, Fan F, Gray MJ, Lim SJ, et al. Endoglin (CD105): A Marker of Tumor Vasculature and Potential Target for Therapy. Clin Cancer Res. 2008;14:1931–7.

  65. Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, et al. Microvesicles Released from Human Renal Cancer Stem Cells Stimulate Angiogenesis and Formation of Lung Premetastatic Niche. Cancer Res. 2011;71:5346–56.

  66. Bussolati B, Bruno S, Grange C, Ferrando U, Camussi G. Identification of a tumor-initiating stem cell population in human renal carcinomas. FASEB J. 2008;22:3696–705.

    Article  CAS  PubMed  Google Scholar 

  67. Chiou SH, Yu CC, Huang CY, Lin SC, Liu CJ, Tsai TH, et al. Positive Correlations of Oct-4 and Nanog in Oral Cancer Stem-Like Cells and High-Grade Oral Squamous Cell Carcinoma. Clin Cancer Res. 2008;14:4085–95.

  68. Margaritescu C, Pirici D, Simionescu C, Stepan A. The utility of CD44, CD117 and CD133 in identification of cancer stem cells (CSC) in oral squamous cell carcinomas (OSCC). Rom J Morphol Embryol. 2011;52:985–93.

    PubMed  Google Scholar 

  69. Zhou S, Schuetz JD, Bunting KD, Colapietro AM, Sampath J, Morris JJ, et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med. 2001;7:1028–34.

  70. Wu C, Alman BA. Side population cells in human cancers. Cancer Lett. 2008;268:1–9.

    Article  CAS  PubMed  Google Scholar 

  71. Burkert J, Otto W, Wright N. Side populations of gastrointestinal cancers are not enriched in stem cells. J Pathol. 2008;214:564–73.

    Article  CAS  PubMed  Google Scholar 

  72. Shi GM, Xu Y, Fan J, Zhou J, Yang XR, Qiu SJ, et al. Identification of side population cells in human hepatocellular carcinoma cell lines with stepwise metastatic potentials. J Cancer Res Clin Oncol. 2008;134:1155–63.

  73. Zhang SN, Huang FT, Huang YJ, Zhong W, Characterization YZ. of a cancer stem cell-like side population derived from human pancreatic adenocarcinoma cells. Tumori. 2010;96:985–92.

    PubMed  Google Scholar 

  74. Patrawala L, Calhoun T, Schneider-Broussard R, Zhou J, Claypool K, Tang DG. Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and ABCG2− cancer cells are similarly tumorigenic. Cancer Res. 2005;65:6207–19.

  75. Kondo T, Setoguchi T, Taga T. Persistence of a small subpopulation of cancer stem-like cells in the C6 glioma cell line. Proc Natl Acad Sci U S A. 2004;101:781–6.

  76. Haraguchi N, Utsunomiya T, Inoue H, Tanaka F, Mimori K, Barnard GF, et al. Characterization of a side population of cancer cells from human gastrointestinal system. Stem Cells. 2006;24:506–13.

  77. Hirschmann-Jax C, Foster A, Wulf G, Nuchtern J, Jax T, Gobel U, et al. A distinct “side population” of cells with high drug efflux capacity in human tumor cells. Proc Natl Acad Sci U S A. 2004;101:14,228–33.

  78. Deonarain MP, Kousparou CA, Epenetos AA. Antibodies targeting cancer stem cells: A new paradigm in immunotherapy? MAbs. 2009;1:12–25.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med. 2006;12:1167–74.

    Article  PubMed  Google Scholar 

  80. Siddique HR, Saleem M. Role of BMI1, a stem cell factor, in cancer recurrence and chemoresistance: preclinical and clinical evidences. Stem Cells. 2012;30:372–8.

    Article  CAS  PubMed  Google Scholar 

  81. Chen J, Wang J, Chen D, Yang J, Yang C, Zhang Y, et al. Evaluation of characteristics of CD44 + CD117+ ovarian cancer stem cells in three dimensional basement membrane extract scaffold versus two dimensional monocultures. BMC Cell Biol. 2013;14:1–11.

  82. Syed IS, Pedram A, Farhat WA. Role of Sonic Hedgehog (Shh) Signaling in Bladder Cancer Stemness and Tumorigenesis. Curr Urol Rep. 2016;17:1–7.

    Article  Google Scholar 

  83. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444:756–60.

  84. Wang WJ, Wu MY, Shen M, Zhi Q, Liu ZY, Gong FR, et al. Cantharidin and norcantharidin impair stemness of pancreatic cancer cells by repressing the beta-catenin pathway and strengthen the cytotoxicity of gemcitabine and erlotinib. Int J Oncol. 2015;47:1912–22.

  85. • McAuliffe SM, Morgan SL, Wyant GA, Tran LT, Muto KW, Chen YS, et al. Targeting Notch, a key pathway for ovarian cancer stem cells, sensitizes tumors to platinum therapy. Proc Natl Acad Sci U S A. 2012;109:E2939–48. Described the cisplatin/GSI combination as an efficient treatment to eradicate both CSCs and the bulk of tumor cells in Notch-dependent tumor cells.

  86. •• Yahyanejad S, King H, Iglesias VS, Granton PV, Barbeau LM, van Hoof SJ, Groot AJ, Habets R, Prickaerts J, Chalmers AJ, Eekers DB, Theys J, Short SC, Verhaegen F, Vooijs M. NOTCH blockade combined with radiation therapy and temozolomide prolongs survival of orthotopic glioblastoma. Oncotarget 2016. NOTCH/γ-secretase inhibitor (GSI) RO4929097 combined with temozolomide and radiotherapy reduced tumor growth.

  87. •• Zhao ZL, Zhang L, Huang CF, Ma SR, Bu LL, Liu JF, et al. NOTCH1 inhibition enhances the efficacy of conventional chemotherapeutic agents by targeting head neck cancer stem cell. Sci Rep. 2016;6:24704. DAPT (GSI-IX) reduces CSC frequency either alone or in combination with chemotherapeutic agents.

  88. •• Yokogi S, Tsubota T, Kanki K, Azumi J, Itaba N, Oka H, et al. Wnt/Beta-Catenin Signal Inhibitor HC-1 Sensitizes Oral Squamous Cell Carcinoma Cells to 5-Fluorouracil through Reduction of CD44-Positive Population. Yonago Acta Med. 2016;59:93–9. It is a translational and clinical study to improve cancer remedy using Wnt/beta-catenin signal inhibitor HC-1.

  89. Fevr T, Robine S, Louvard D, Huelsken J. Wnt/β-Catenin Is Essential for Intestinal Homeostasis and Maintenance of Intestinal Stem Cells. Mol Cell Biol. 2007;27:7551–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Chen B, Dodge ME, Tang W, Lu J, Ma Z, Fan CW, et al. Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer. Nat Chem Biol. 2009;5:100–7.

  91. Liu L, Zhi Q, Shen M, Gong FR, Zhou BP, Lian L, Shen B, Chen K, Duan W, Wu MY, Tao M, Li W. FH535, a beta-catenin pathway inhibitor, represses pancreatic cancer xenograft growth and angiogenesis. Oncotarget 2016.

  92. Jang GB, Hong IS, Kim RJ, Lee SY, Park SJ, Lee ES, et al. Wnt/β-catenin small-molecule inhibitor CWP232228 preferentially inhibits the growth of breast cancer stem-like cells. Cancer Res. 2015;75:1691–702.

  93. •• Kim JY, Lee HY, Park KK, Choi YK, Nam JS, Hong IS. CWP232228 targets liver cancer stem cells through Wnt/β-catenin signaling: a novel therapeutic approach for liver cancer treatment. Oncotarget. 2016;7:20,395–409. Liver CSCs are responsible for tumor relapse, but CWP232228 targets liver cancer stem cells through Wnt/β-catenin signaling.

  94. • Li X, Bai B, Liu L, Ma P, Kong L, Yan J, et al. Novel β-carbolines against colorectal cancer cell growth via inhibition of Wnt/β-catenin signaling. Cell Death Discov. 2015;1:15,033. isopropyl 9-ethyl-1- (naphthalen-1-yl)-9H-pyrido[3,4-b]indole-3-carboxylate (novel Wnt signaling inhibitor) inhibited the growth of colorectal cancer cells selectively and caused obvious G1-phase arrest of the cell cycle via Wnt signaling pathway.

  95. Yakisich JS. Challenges and limitations of targeting cancer stem cells and/or the tumor microenvironment. Drugs Ther Stud. 2012;2:10.

    Article  Google Scholar 

  96. Katoh M, Katoh M. WNT signaling pathway and stem cell signaling network. Clin Cancer Res. 2007;13:4042–5.

    Article  CAS  PubMed  Google Scholar 

  97. Naka K, Hoshii T, Hirao A. Novel therapeutic approach to eradicate tyrosine kinase inhibitor resistant chronic myeloid leukemia stem cells. Cancer Sci. 2010;101:1577–81.

    Article  CAS  PubMed  Google Scholar 

  98. Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12:445–64.

  99. • Qu Y, Gharbi N, Yuan X, Olsen JR, Blicher P, Dalhus B, et al. Axitinib blocks Wnt/beta-catenin signaling and directs asymmetric cell division in cancer. Proc Natl Acad Sci U S A. 2016;113:9339–44. Therapeutic benefits to cancer patients with aberrant nuclear β-catenin activation.

  100. • Huang SMA, Mishina YM, Liu S, Cheung A, Stegmeier F, Michaud GA, et al. Tankyrase inhibition stabilizes axin and antagonizes Wntsignalling. Nature. 2009;461:614–20. Wnt pathway inhibitor (XAV939) stimulates degradation of β-catenin as a good strategy to treat cancer.

  101. Varnat F, Duquet A, Malerba M, Zbinden M, Mas C, Gervaz P, et al. Human colon cancer epithelial cells harbor active HEDGEHOG-GLI signaling that is essential for tumor growth, recurrence, metastasis and stem cell survival and expansion. EMBO Mol Med. 2009;1:338–51.

  102. Von Hoff DD, LoRusso PM, Rudin CM, Reddy JC, Yauch RL, Tibes R, et al. Inhibition of the hedgehogpathway in advancedbasal-cell carcinoma. N Engl J Med. 2009;361:1164–72.

  103. Yauch RL, Dijkgraaf GJP, Alicke B, Januario T, Ahn CP, Holcomb T, et al. Smoothened Mutation Confers Resistance to a Hedgehog Pathway Inhibitor in Medulloblastoma. Science. 2009;326:572–4.

  104. Justilien V, Fields AP. βMolecular Pathways: Novel Approaches for Improved Therapeutic Targeting of Hedgehog Signaling in Cancer Stem Cells. Clin Cancer Res. 2015;21:505–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Mamaeva V, Niemi R, Beck M, Ozliseli E, Desai D, Landor S, et al. Inhibiting Notch Activity in Breast Cancer Stem Cells by Glucose Functionalized Nanoparticles Carrying [gamma]-secretase Inhibitors. Mol Ther. 2016;24:926–36.

  106. Yabuuchi S, Pai SG, Campbell NR, de Wilde RF, De Oliveira E, Korangath P, et al. Notch signaling pathway targeted therapy suppresses tumor progression and metastatic spread in pancreatic cancer. Cancer Lett. 2013;335:41–51.

  107. Garg M. Emerging role of microRNAs in cancer stem cells: Implications in cancer therapy. World J Stem Cells. 2015;7:1078.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Hasegawa S, Eguchi H, Nagano H, Konno M, Tomimaru Y, Wada H, et al. MicroRNA-1246 expression associated with CCNG2-mediated chemoresistance and stemness in pancreatic cancer. Br J Cancer. 2014;111:1572–80.

  109. Hwang-Verslues WW, Chang PH, Wei PC, Yang CY, Huang CK, Kuo WH, et al. miR-495 is upregulated by E12/E47 in breast cancer stem cells, and promotes oncogenesis and hypoxia resistance via downregulation of E-cadherin and REDD1. Oncogene. 2011;30:2463–74.

  110. Zhou AD, Diao LT, Xu H, Xiao ZD, Li JH, Zhou H, et al. β-Catenin/LEF1 transactivates the microRNA-371-373 cluster that modulates the Wnt/β-catenin-signaling pathway. Oncogene. 2012;31:2968–78.

  111. Cairo S, Wang Y, de Reyniès A, Duroure K, Dahan J, Redon MJ, et al. Stem cell-like micro-RNA signature driven by Myc in aggressive liver cancer. Proc Natl Acad Sci USA. 2010;107:20,471–6.

  112. Xia H, Ooi LL, Hui KM. MicroRNA-216a/217-induced epithelial-mesenchymal transition targets PTEN and SMAD7 to promote drug resistance and recurrence of liver cancer. Hepatology. 2013;58:629–41.

    Article  CAS  PubMed  Google Scholar 

  113. Bao B, Ali S, Ahmad A, Azmi AS, Li Y, Banerjee S, et al. Hypoxia-induced aggressiveness of pancreatic cancer cells is due to increased expression of VEGF, IL-6 and miR-21, which can be attenuated by CDF treatment. PLoS One. 2012;7:e50165.

  114. Xu W, Ji J, Xu Y, Liu Y, Shi L, Liu Y, Lu X, Zhao Y, Luo F, Wang B, Ziang R. MicroRNA-191, by promoting the EMT and increasing CSC-like properties, is involved in neoplastic and metastatic properties of transformed human bronchial epithelial cells. Mol Carcinog 2015; (S1), E148–161.

  115. Ma S, Tang KH, Chan YP, Lee TK, Kwan PS, Castilho A, et al. miR-130b Promotes CD133(+) liver tumor-initiating cell growth and self-renewal via tumor protein 53-induced nuclear protein 1. Cell Stem Cell. 2010;7:694–707.

  116. Han YC, Park CY, Bhagat G, Zhang J, Wang Y, Fan JB, et al. microRNA-29a induces aberrant self-renewal capacity in hematopoietic progenitors, biased myeloid development, and acute myeloid leukemia. J Exp Med. 2010;207:475–89.

  117. Sureban SM, May R, Qu D, Weygant N, Chandrakesan P, Ali N, et al. DCLK1 regulates pluripotency and angiogenic factors via microRNA-dependent mechanisms in pancreatic cancer. PLoS One. 2013;8:e73940.

  118. King CE, Cuatrecasas M, Castells A, Sepulveda AR, Lee JS, Rustgi AK. LIN28B promotes colon cancer progression and metastasis. Cancer Res. 2011;71:4260–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Xi S, Xu H, Shan J, Tao Y, Hong JA, Inchauste S, et al. Cigarette smoke mediates epigenetic repression of miR-487b during pulmonary carcinogenesis. J Clin Invest. 2013;123:1241–61.

  120. Gal H, Pandi G, Kanner AA, Ram Z, Lithwick-Yanai G, Amariglio N, et al. MIR-451 and Imatinib mesylate inhibit tumor growth of Glioblastoma stem cells. Biochem Biophys Res Commun. 2008;376:86–90.

  121. Babashah S, Sadeghizadeh M, Hajifathali A, Tavirani MR, Zomorod MS, Ghadiani M, et al. Targeting of the signal transducer Smo links microRNA-326 to the oncogenic Hedgehog pathway in CD34+ CML stem/progenitor cells. Int J Cancer. 2013;133:579–89.

    Article  CAS  PubMed  Google Scholar 

  122. Ying Z, Li Y, Wu J, Zhu X, Yang Y, Tian H, et al. Loss of miR-204 expression enhances glioma migration and stem cell-like phenotype. Cancer Res. 2013;73:990–9.

    Article  CAS  PubMed  Google Scholar 

  123. Lu Y, Lu J, Li X, Zhu H, Fan X, Zhu S, et al. MiR-200a inhibits epithelial-mesenchymal transition of pancreatic cancer stem cell. BMC Cancer. 2014;14:85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Wang Y, Yu Y, Tsuyada A, Ren X, Wu X, Stubblefield K, et al. Transforming growth factor-β regulates the sphere-initiating stem cell-like feature in breast cancer through miRNA-181 and ATM. Oncogene. 2011;30:1470–80.

    Article  CAS  PubMed  Google Scholar 

  125. Morris VA, Zhang A, Yang T, Stirewalt DL, Ramamurthy R, Meshinchi S, et al. MicroRNA-150 expression induces myeloid differentiation of human acute leukemia cells and normal hematopoietic progenitors. PLoS One. 2013;8:e75815.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Pramanik D, Campbell NR, Karikari C, Chivukula R, Kent OA, Mendell JT, et al. Restitution of tumor suppressor microRNAs using a systemic nanovector inhibits pancreatic cancer growth in mice. Mol Cancer Ther. 2011;10:1470–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Godlewski J, Nowicki MO, Bronisz A, Williams S, Otsuki A, Nuovo G, et al. Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal. Cancer Res. 2008;68:9125–30.

    Article  CAS  PubMed  Google Scholar 

  128. Collet G, Skrzypek K, Grillon C, Matejuk A, El Hafni-Rahbi B, Lamerant-Fayel N, et al. Hypoxia control to normalize pathologic angiogenesis: potential role for endothelial precursor cells and miRNAs regulation. Vascul Pharmacol. 2012;56:252–61.

    Article  CAS  PubMed  Google Scholar 

  129. Ji Q, Hao X, Zhang M, Tang W, Yang M, Li L, et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS One. 2009;4:e681.

    Google Scholar 

  130. Di Fiore R, Drago-Ferrante R, Pentimalli F, Di Marzo D, Forte IM, D’Anneo A, et al. MicroRNA-29b-1 impairs in vitro cell proliferation, self-renewal and chemoresistance of human osteosarcoma 3AB-OS cancer stem cells. Int J Oncol. 2014;45:2013–23.

    Article  PubMed  Google Scholar 

  131. Scheibner KA, Teaboldt B, Hauer MC, Chen X, Cherukuri S, Guo Y, et al. MiR-27a functions as a tumor suppressor in acute leukemia by regulating 14–3-3theta. PLoS One. 2012;7:e50895.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Geng J, Luo H, Pu Y, Zhou Z, Wu X, Xu W, et al. Methylation mediated silencing of miR-23b expression and its role in glioma stem cells. Neurosci Lett. 2012;528:185–9.

    Article  CAS  PubMed  Google Scholar 

  133. Sun X, Jiao X, Pestel TG, Fan C, Qin S, Mirabelli E, et al. MicroRNAs and cancer stem cells: the sword and the shield. Oncogene. 2014;33:4967–77.

    Article  CAS  PubMed  Google Scholar 

  134. Bimonte S, Barbieri A, Leongito M, Palma G, Del Vecchio V, Falco M, et al. The Role of miRNAs in the Regulation of Pancreatic Cancer Stem Cells. Stem Cells Int. 2016;2016:8352684.

    PubMed  PubMed Central  Google Scholar 

  135. Li Y, Guessous F, Zhang Y, DiPierro C, Kefas B, Johnson E, et al. MicroRNA-34a inhibits glioblastoma growth by targeting multiple oncogenes. Cancer Res. 2009;69:7569–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Corney DC, Flesken-Nikitin A, Godwin AK, Wang W, Nikitin AY. MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in control of cell proliferation and adhesion-independent growth. Cancer Res. 2007;67:8433–8.

    Article  CAS  PubMed  Google Scholar 

  137. •• Ji J, Yamashita T, Budhu A, Forgues M, Jia HL, Li C, et al. Identification of microRNA-181 by genome-wide screening as a critical player in EpCAM-positive hepatic cancer stem cells. Hepatology. 2009;50:472–80. The highly expressed miR-181 promotes differentiation and directly targets CDX2, GATA6, and NLK in hepatocellular stem cells (HpSCs).

  138. •• Yao S. MicroRNA biogenesis and their functions in regulating stem cell potency and differentiation. Biol Proced Online. 2016;18:8. Described the role of miR-128, miR-181, miR-16, miR-103, and miR-107 in cancer cell proliferation.

  139. Hong M, Tan HY, Li S, Cheung F, Wang N, Nagamatsu T, et al. Cancer Stem Cells: The Potential Targets of Chinese Medicines and Their Active Compounds. Int J Mol Sci. 2016;17:893.

    Article  PubMed Central  Google Scholar 

  140. Zahnow C, Topper M, Stone M, Murray-Stewart T, Li H, Baylin SB, et al. Chapter Two-Inhibitors of DNA Methylation, Histone Deacetylation, and Histone Demethylation: A Perfect Combination for Cancer Therapy. Adv Cancer Res. 2016;130:55–111.

    Article  CAS  PubMed  Google Scholar 

  141. Taniura H, Sng JC, Yoneda Y. Histone modifications in the brain. Neurochem Int. 2007;51:85–91.

    Article  CAS  PubMed  Google Scholar 

  142. Chikamatsu K, Ishii H, Murata T, Sakakura K, Shino M, Toyoda M, et al. Alteration of cancer stem cell-like phenotype by histone deacetylase inhibitors in squamous cell carcinoma of the head and neck. Cancer Sci. 2013;104:1468–75.

    Article  CAS  PubMed  Google Scholar 

  143. Loriot A, Parvizi GK, Reister S, De Smet C. Silencing of cancer-germline genes in human preimplantation embryos: evidence for active de novo DNA methylation in stem cells. Biochem Biophys Res Commun. 2012;417:187–91.

    Article  CAS  PubMed  Google Scholar 

  144. Brehm A, Miska EA, McCance DJ, Reid JL, Bannister AJ, Kouzarides T. Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature. 1998;391:597–601.

    Article  CAS  PubMed  Google Scholar 

  145. Giudice FS, Pinto DS Jr, Nör JE, Squarize CH, Castilho RM. Inhibition of Histone Deacetylase Impacts Cancer Stem Cells and Induces Epithelial-Mesenchyme Transition of Head and Neck Cancer. PLoS ONE. 2013;8:e58672.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Haffner MC, Chaux A, Meeker AK, Esopi DM, Gerber J, Pellakuru LG, et al. Global 5-hydroxymethylcytosine content is significantly reduced in tissue stem/progenitor cell compartments and in human cancers. Oncotarget. 2011;2:627–37.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Matsubara N. Epigenetic regulation and colorectal cancer. Dis Colon Rectum. 2012;55:96–104.

    Article  PubMed  Google Scholar 

  148. Tsujii M. Cyclooxygenase, Cancer Stem Cells and DNA Methylation Play Important Roles in Colorectal Carcinogenesis. Digestion. 2013;87:12–6.

    Article  CAS  PubMed  Google Scholar 

  149. Liu CC, Lin JH, Hsu TW, Su K, Li AF, Hsu HS, et al. IL-6 enriched lung cancer stem-like cell population by inhibition of cell cycle regulators via DNMT1 upregulation. Int J Cancer. 2015;136:547–59.

    CAS  PubMed  Google Scholar 

  150. Wongtrakoongate P. Epigenetic therapy of cancer stem and progenitor cells by targeting DNA methylation machineries. World J Stem Cells. 2015;7:137–48.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Sakajiri S, Kumagai T, Kawamata N, Saitoh T, Said JW, Koeffler HP. Histone deacetylase inhibitors profoundly decrease proliferation of human lymphoid cancer cell lines. Exp Hematol. 2005;33:53–61.

    Article  CAS  PubMed  Google Scholar 

  152. Kikuchi J, Takashina T, Kinoshita I, Kikuchi E, Shimizu Y, Sakakibara-Konishi J, et al. Epigenetic therapy with 3-deazaneplanocin A, an inhibitor of the histone methyltransferase EZH2, inhibits growth of non-small cell lung cancer cells. Lung Cancer. 2012;78:138–43.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Kondo Y. Targeting histone methyltransferase EZH2 as cancer treatment. J Biochem. 2014;156:249–57.

    Article  CAS  PubMed  Google Scholar 

  154. Momparler RL, Cote S. Targeting of cancer stem cells by inhibitors of DNA and histone methylation. Expert Opin Investi Drug. 2015;24:1031–43.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

All the authors thank the Vice-Chancellor of CUPB for the support.

Funding

The work was supported by CUPB-RSM and UGC startup grants.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sandeep Singh PhD.

Ethics declarations

Conflict of Interest

Gurpreet Kaur, Praveen Sharma, Nilambra Dogra, and Sandeep Singh declare they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kaur, G., Sharma, P., Dogra, N. et al. Eradicating Cancer Stem Cells: Concepts, Issues, and Challenges. Curr. Treat. Options in Oncol. 19, 20 (2018). https://doi.org/10.1007/s11864-018-0533-1

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11864-018-0533-1

Keywords

Navigation