Cancer Letters

Cancer Letters

Volume 322, Issue 2, 28 September 2012, Pages 139-147
Cancer Letters

Mini-review
Targeting a cornerstone of radiation resistance: Cancer stem cell

https://doi.org/10.1016/j.canlet.2012.03.024Get rights and content

Abstract

In radiation oncology, cancer stem cells (CSCs) have become an important research field. In fact, it appears that most cancer types contain populations of cells that exhibit stem-cell properties. CSCs have the ability to renew indefinitely, which can drive tumor development and metastatic invasion. As those cells are classically resistant to conventional chemotherapy and to radiation therapy, they may contribute to treatment failure and relapse. Over past decades, preclinical research has highlighted that variations in the CSCs content within tumor could affect their radiocurability by interfering with mechanisms of DNA repair, redistribution in the cell cycle, tumor cells repopulation, and hypoxia. It is now possible to isolate particular cells expressing specific surface markers and thus better investigating CSCs pathways. Numerous inhibitory agents targeting these specific signaling pathways, such as Notch and Wnt/B-catenin, are currently evaluated in early clinical trials. By targeting CSCs, tumor radioresistance could be potentially overcome to improve outcome for patients with solid malignancies. Radiation therapy using ion particles (proton and carbon) may be also more effective than classic photon on CSCs. This review presents the major pathophysiological mechanisms involved in CSCs radioresistance and recent developments for targeted strategies.

Introduction

For over a half a century, cancer stem cells (CSCs) have been an important area of radiation research. It appears that most cancers contain populations of cells that exhibit stem-cell properties. Those are characterized by their ability to self-renew and may produce a much larger set of cells with very limited proliferative potential that are named the bulk. The definition of a CSC requires three characteristics: (i) a selective capacity to initiate tumor and drive neoplastic proliferation, (ii) an ability to generate endless copies of themselves through self-renewal, and (iii) the potential to give rise to more differentiated non-stem cell cancer progeny [1].

Since CSCs have been described for first time in leukaemia by the Dick laboratory in 1994, other groups have demonstrated the presence of CSCs in numerous solid tumors (glioblastoma, colorectal cancer, prostate, head and neck, liver, melanoma, etc.), suggesting that the majority of malignancies are dependent on such a cell compartment. Anyhow, properties of CSCs in different tumors were not similar, and the proportion of stem cells in tumors was different [2]. Moreover, the origin of CSCs remains unclear and many works tried to identify this. Barker et al. [3], Zhu et al. [4], and Alcantara Llaguno et al. [5] demonstrated that CSCs were derived from normal stem cells if oncogenes were activated. But, there is evidence that more differentiated cancer cell populations may acquire CSC properties through epithelial mesenchymal transition (EMT) [6], [7]. Chaffer et al. [8] recently observed in a subpopulation of basal-like human mammary epithelial cells that spontaneously dedifferentiated into stem-like cells. Then, some non-stem cancer cells gave rise to CSCs. Classically, the definition of adult stem cell implicates that those are capable to generate full cellular components of the relevant organ from a single stem cell. This could be revealed by in vivo functional assays, which consists in implanting a progressively smaller number of tumor cells in immunodeficient animals. The number of cells could be an appropriate surrogate when analyzing the frequency of proper cancer stem cells. Now, through the development of immunofluorescence tools, it is possible to more easily isolate CSCs using their surface proteins (e.g. CD133, CD44, CD29).

Cancer stem cells account for a minor fraction of a tumor population, but those might be particularly involved in tumor initiation, proliferation, or in metastatic process. CSCs are believed to share many properties with normal stem cells that render them relatively insensitive to conventional cytotoxic agents, such as chemotherapy and to radiation therapy. This is an advocated factor of treatment failure and relapse. The mechanisms through which solid tumors become resistant to conventional therapy are only partially understood. Those involve numerous factors implicated in the resistance of CSC to ionizing radiation: quiescence propensity, enhanced DNA repair, upregulated cell cycle control mechanisms, mechanisms of free-radical scavenging, and specific interaction with stromal microenvironnement [9], [10]. From a radiobiological point of view, four factors are classically involved in the efficiency of ionizing radiation: repair of DNA damage systems, redistribution of the cell cycle, tumor cells repopulation, and level of intratumor hypoxia. Although the identity of cancer cell clonogens has not been definitively resolved, the understanding of CSCs pathways continues to grow and new potential molecular therapeutic targets have recently emerged. Numerous inhibitory agents targeting these specific signaling pathways, such as Notch and Wnt/B-catenin, are currently evaluated in early clinical trials (http://clinicaltrials.gov: NCT01122901, NCT01351103). Overcoming radioresistance by concurrently targeting cancer stem cells pathways may potentially improve outcome for patients with localized solid tumors. Radiation therapy using heavy particles (proton and carbon) may be also more effective than classic photon on CSCs [11], [12]. The aim of this paper is to report the various intracellular pathways involved in CSCs radioresistance for which potential intracellular target and highlight strategies for enhancing antitumor effectiveness.

Section snippets

Mechanism of radiation resistance

Although debated, CSCs’ radioresistance could give explanation for their capacity to cause tumor recurrence. Few clinical data have corroborated this hypothesis. In 2006, Bao et al. [13] determined a subpopulation of glioblastoma, which contributes to radioresistance. This fraction of tumor cells expressed CD133 and had a lower sensitivity to radiation-induced apoptosis. Authors also reported that their frequency was 2- to 4- fold higher after radiation in both primary tumors and xenografts.

Therapeutical strategies to target CSCs

Several studies have investigated the possibility of specifically target CSC’s molecular pathways for overcoming radiation resistance.

Conclusion

Despite substantial research and important progress in the anti-cancer therapeutic arsenal, about 50% of cancer patients will experience local or distant tumor relapse despite treatment. The fact that most current strategies did not target CSCs but rather more differentiated tumor cells may have a putative role in the failure of numerous intent-to-cure strategies. Since local tumor seems to be potentially related to CSCs control, radiobiological research should focus in radiation effects on

References (94)

  • C. Calabrese et al.

    A perivascular niche for brain tumor stem cells

    Cancer Cell

    (2007)
  • M. Tsatmali et al.

    Newborn neurons acquire high levels of reactive oxygen species and increased mitochondrial proteins upon differentiation from progenitors

    Brain Res.

    (2005)
  • J. Boonstra et al.

    Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells

    Gene

    (2004)
  • B.J. Moeller et al.

    Radiation activates HIF1 regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules

    Cancer Cell

    (2004)
  • Z. Li et al.

    Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells

    Cancer Cell

    (2009)
  • M.J. Bissell et al.

    Context, tissue plasticity, and cancer: are tumor stem cells also regulated by the microenvironment?

    Cancer Cell

    (2005)
  • N.A. Theodosiou et al.

    Wnt signaling during development of the gastrointestinal tract

    Dev. Biol.

    (2003)
  • V. Clement et al.

    HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity

    Curr. Biol.

    (2007)
  • J. Lee et al.

    Epigenetic-mediated dysfunction of the bone morphogenetic protein pathway inhibits differentiation of glioblastoma-initiating cells

    Cancer Cell

    (2008)
  • A. Soeda et al.

    Epidermal growth factor plays a crucial role in mitogenic regulation of human brain tumor stem cells

    J. Biol. Chem.

    (2008)
  • H.W. Lo et al.

    Nuclear interaction of EGFR and STAT3 in the activation of the iNOS/NO pathway

    Cancer Cell

    (2005)
  • N.P. Nguyen et al.

    Molecular biology of breast cancer stem cells: potential clinical applications

    Cancer Treat. Rev.

    (2010)
  • R. Ellsworth et al.

    The radiosensitizing effects of a novel tyrosine kinase inhibitor, Mp470 in glioblastoma multiforme stem cells

    Int. J. Radiat. Oncol. Biol. Phys.

    (2008)
  • Y. Sun et al.

    Gene profiling of cancer stem cells-like cells reveals IGF-1R and IGFBP3 pathway as a promising therapeutic target for lung cancer

    Int. J. Radiat. Oncol. Biol. Phys.

    (2010)
  • S. Kahana et al.

    Proteasome inhibitors sensitize glioma cells and glioma stem cells to TRAIL-induced apoptosis by PKCε-dependent downregulation of AKT and XIAP expressions

    Cell. Signal.

    (2011)
  • S. Supiot et al.

    “Dose-painting”: myth or reality?

    Cancer Radiother.

    (2010)
  • C.C. Ling et al.

    Towards multidimensional radiotherapy (MD-CRT): biological imaging and biological conformality

    Int. J. Radiat. Oncol. Biol. Phys.

    (2000)
  • L.S. Piao et al.

    CD133+ liver cancer stem cells modulate radioresistance in human hepatocellular carcinoma

    Cancer Lett.

    (2012)
  • Q. Zhang et al.

    A subpopulation of CD133(+) cancer stem-like cells characterized in human oral squamous cell carcinoma confer resistance to chemotherapy

    Cancer Lett.

    (2010)
  • T. Schatton et al.

    Identification and targeting of cancer stem cells

    BioEssays

    (2009)
  • N. Barker et al.

    Crypts stem cells as the cells-of-origin of intestinal cancer

    Nature

    (2009)
  • L. Zhu et al.

    Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation

    Nature

    (2009)
  • K. Polyak et al.

    Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits

    Nat. Rev. Cancer

    (2009)
  • C.L. Chaffer et al.

    Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state

    Proc. Natl. Acad. Sci. USA

    (2011)
  • F. Pajonk et al.

    Radiation resistance of cancer stem cells: the 4 R’s of radiobiology revisited

    Stem Cells

    (2010)
  • L. Mils et al.

    Cancer stem cells and tumour response to therapy: current problems and future prospects

    Semin. Radiat. Oncol.

    (2009)
  • X. Cui et al.

    Effects of carbon ion beam on putative colon cancer stem cells and its comparison with X-rays

    Cancer Res.

    (2011)
  • S. Bao et al.

    Glioma stem cells promote radioresistance by preferential activation of the DNA damage response

    Nature

    (2006)
  • T.M. Phillips et al.

    The response of CD24(-/low)/CD44+ breast cancer-initiating cells to radiation

    J. Natl Cancer Inst.

    (2006)
  • W.A. Woodward et al.

    WNT/b-catenin mediates radiation resistance of mouse mammary progenitor cells

    Proc. Natl. Acad. Sci. USA

    (2007)
  • B. Campos et al.

    Insight into the complex regulation of CD133 in glioma

    Int. J. Cancer

    (2011)
  • T. Miyabayashi et al.

    Wnt/beta-catenin/CBP signaling maintains long-term murine embryonic stem cell pluripotency

    Proc. Natl. Acad. Sci. USA

    (2007)
  • E. Kendziorra et al.

    Silencing of the Wnt transcription factor TCF4 sensitizes colorectal cancer cells to (chemo-) radiotherapy

    Carcinogenesis

    (2011)
  • J.N. Rich

    Cancer stem cells in radiation resistance

    Cancer Res.

    (2007)
  • H. Yin et al.

    The phenotypic radiation resistance of CD44+/CD24(-or low) breast cancer cells is mediated through the enhanced activation of ATM signaling

    PLoS ONE

    (2011)
  • S. Facchino et al.

    BMI confers radioresistance to normal and cancerous neural stem cells through recruitment of the DNA damage response machinery

    J. Neurosci.

    (2010)
  • R.S. Gieni et al.

    Polycomb group proteins in the DNA damage response: a link between radiation resistance and “stemness”

    Cell Cycle

    (2011)
  • Cited by (118)

    • Pan-cancer genomic amplifications underlie a WNT hyperactivation phenotype associated with stem cell-like features leading to poor prognosis

      2019, Translational Research
      Citation Excerpt :

      Moreover, EZH2 inhibits E-cadherin expression via lncRNA H19 to promote bladder cancer metastasis.53 To further confirm that the 16 Wnt drivers represent potential markers of cancer stem cells, we performed correlation analyses between 16-Wnt-gene scores and 9 well-studied cancer stem cell markers: CD133,54,55 NESTIN,56,57 CD200,58,59 CD44,60,61 CD105,62,63 CD24,64,65 CD90,66,67 CD73,68,69 and CD29.70,71 Cancer stem cell markers are often cancer type specific.

    View all citing articles on Scopus
    View full text