Submit manuscript...
MOJ
eISSN: 2374-6912

Cell Science & Report

Correspondence:

Received: January 01, 1970 | Published: ,

Citation: DOI:

Download PDF

Abstract

Glioblastoma multiforme (GBM) is the most common and aggressive form of brain cancer. Despite advances in current therapeutic procedures including surgery, chemotherapy and radiation, there have been no significant improvements in patient survival. GBM tumors are highly heterogeneous and it is believed that a small proportion of the tumor mass is comprised of cancer stem-like cells (CSCs). The CSCs behave much like neural stem cells in that they can self-renew and undergo differentiation; however, their high tumor-initiating capacity and therapeutic resistance apparently drive tumorigenesis. Recent evidence depicts the involvement and crosstalk of several different signaling pathways in the regulation and progression of GBM. In this review, we discuss the PI3K-Akt, mTOR, Notch and JAK-STAT signaling pathways that often crosstalk to maintain GBM CSC survival. Furthermore, we discuss the potential role of epigenetic regulation of the CSCs. We believe that a thorough understanding of the signaling pathways that regulate GBM CSCs, and further molecular characterization of the GBM tumors, will lead to the development of more efficient therapies.

Keywords: glioblastoma, cancer stem cells, signaling pathways

Abbreviations

CSCs, cancer stem-like cells; EGFR, epidermal growth factor receptor; GBM, glioblastoma multiforme; JAK, janus tyrosine kinase; MTOR, mechanistic target of rapamycin; MAPK, mitogen-activated protein kinase; NF1, neurofibromin-1; NF-κB, nuclear factor-kappa b; PI3K, phosphoinositide-3-kinase; PDGFRA, platelet derived growth factor receptor-alpha; STAT, signal transducer and activator of transcription

Introduction

Glioblastoma

Glioblastoma multiforme (GBM) is an aggressive and lethal form of brain cancer with an extremely dismal prognosis. Despite advances in the molecular characterization of GBM and new targeted therapeutic approaches, the average patient survival remains only between 12 to 15months.1 GBM tumors are highly heterogeneous and are comprised of multiple differentiated cell types and a small subpopulation of cancer stem-like cells (CSCs). The CSCs are believed to be responsible for tumor heterogeneity,2,3 are relatively resistant to treatment, and are responsible for the aggressive nature of the disease.4,5 Therefore, in order to design new targeted therapeutic approaches it is imperative to identify and characterize the signaling pathways involved in the formation, maintenance and regulation of CSCs. Recent reports on the dedifferentiation of GBM cell lines into more CSC-like cells may have added a new layer of complexity into treating GBM.5,6 Therefore, in GBM it is critical to target not only the CSCs but also the bulk differentiated cells within the tumor.

Multiple signaling pathways, including Notch1, mechanistic target of rapamycin (mTOR), AKT, mitogen-activated protein kinase (MAPK), Hedgehog and the Janus tyrosine kinase (JAK)- signal transducer and activator of transcription (STAT) pathways, have been found to be involved in the maintenance and progression of GBM.7 Additionally, mutations of epidermal growth factor receptor (EGFR), platelet derived growth factor receptor-alpha (PDGFRA), phosphatase and tensin homolog (PTEN), p53, and neurofibromin-1 (NF1) are frequently seen in GBM.8 EGFR is amplified in 40% to 50% of GBM, and gain of function EGFRvIII mutations are found in nearly half of all GBMs.9,10 The activation of EGFR signaling leads to the subsequent activation of the PI3K-Akt signaling cascade,11 which plays a critical role in cell survival. Despite the identification of EGFR mutations in GBM, the EGFR tyrosine kinase inhibitors, Erlotinib and gefitinib, have only limited clinical benefit.12,13 Studies indicate that PTEN loss in GBM, which results in the activation of the PI3K-Akt signaling pathway, promotes resistance to EGFR inhibitors.12

Dysregulating signaling pathways in glioblastoma

Activating mutations of the phosphoinositide-3-kinase (PI3K)-Akt pathway are present in 90% of GBMs.9 Mutations of the PI3K pathway are often accompanied by PTEN loss, and lead to the hyper activation of the mTORC1 pathway.14 The mTOR kinase exists as two distinct complexes, mTORC1 and mTORC2 (Figure 1). mTORC1 lies downstream of the PI3K pathway and promotes cell proliferation, protein translation and maintains the energy status of the cancer cell.14,15 In contrast to mTORC1, the role of mTORC2 is less understood. It was recently found that the combination of the gain of function EGFRvIII mutation and PTEN loss can promote mTORC2 signaling in GBM.16 Despite the significance of mTOR signaling in glioma cell growth, only limited success has been achieved from rapamycin (sirolimus), an allosteric mTORC1 inhibitor, in phase I and phase II clinical trials.17 The limited success of the EGFR and mTORC1 inhibitors indicates that other signaling pathways are involved in GBM and continue to transmit downstream signals thereby promoting cancer cell survival. Furthermore, it is important to define the crosstalk between different pathways that operate in GBM, which can subsequently identify a specific group of targets that can eventually be targeted by therapy.

Figure 1 Dysregulated signaling pathways in GBM.

The Notch signaling cascade is an evolutionarily conserved pathway that regulates cell fate decisions during early embryonic development, and plays important roles in cell differentiation, proliferation, survival, angiogenesis and migration,18–20 Notch is involved with the maintenance of neural progenitors and the generation of glia during normal brain development,21 and plays critical roles in the maintenance of glioma CSCs (Figure 1).22 Recent reports have indicated that Notch inhibitors can induce differentiation of glioma CSCs.23 Our recent studies indicate that the STAT3 and nuclear factor-kappa B (NF-κB) transcription factors are involved in regulating Notch1 expression and downstream signaling in glioma CSCs (Figure 1).24 Furthermore, glioma CSCs were relatively sensitive to STAT3 and NF-κB inhibitors, and the STAT3 inhibitor WP1066 led to marked tumor regression and loss of tumorigenicity in vivo.24,25

Epigenetic modifications in glioblastoma

While the identification of the multiple pathways operating in GBM provides targets for therapy, recent evidence suggests that epigenetic changes in collaboration with genetic alterations in GBM provide a new layer of complexity into approaching therapeutic intervention.26,27 The reactivation of the Oct4 and Sox2 transcription factors by DNA methyltransferase promoter transactivation enables GBM cells to develop more stem-like characteristics.28 Due to the reversibility of epigenetic marks, treatment of GBM CSCs with HDAC inhibitors and demethylating agents, which modulate the conformation of chromatin and regulate gene expression,29 can reactivate silenced tumor suppressor genes.27 Studies indicate that DNA methyltransferase 5-azacytidine reduced glioma cell proliferation, induced differentiation and reduced tumor growth.30 Identification of several GBM CSC targets enables the use of combination therapies that may provide maximum benefit. Recent studies indicate that the HDAC inhibitors trichostatin A and valproic acid reduce GBM CSC growth, reduce the expression of stem cell markers and promote cell differentiation.31 HDAC inhibitors used in combination with the EGFR inhibitor erlotinib has provided promising results in patients with GBM that over express EGFR.32

Cancer stem-like cells in glioblastoma

Another important point to consider is that the heterogeneity in GBM tumors may reflect that there is more than one population of GBM CSCs, which would provide an additional complication for therapeutic intervention to selectively target the CSC compartment. Previous studies using genomic profiling identified four molecular subtypes of GBM: Proneural, Neural, Classical and Mesenchymal.33 However, single cell analysis of GBM tumor xenografts has demonstrated that multiple molecular subtypes exist within a single tumor and that individual cells varied markedly in their gene expression patterns.34 Using traditional tumorsphere culture conditions to enrich for GBM CSCs, as well as growth as adherent CSCs on laminin-coated plates, we have recently shown that both of these CSC populations display stem cell properties and initiate histologically indistinguishable GBM tumors following xenotransplantation.25 However, these CSC populations differ in their gene expression patterns when grown in vitro, and induce molecularly distinct tumors in vivo. Taken together, these results suggest that GBM CSCs are not monoclonal but rather are a mosaic of different CSC populations, which are capable of initiating tumors with different gene signatures.

Conclusion

Molecular characterization of GBM over the recent years has enabled the identification of new therapeutic targets. However, the heterogeneous nature of the disease makes it critical to identify multiple pathways that promote cell survival. An important future direction of research is to understand the role of the signaling pathways that are dysregulated in GBM CSCs, and are responsible for the therapeutic resistance and tumor invasiveness of GBM. Moreover, much needs to be learned about the mechanism that underlie the interconversion of bulk GBM cells into GBM CSCs, as well as the different molecular properties of the multiple GBM CSC populations.

Acknowledgements

This work was supported in part by grants from the National Institutes of Health CA133322, the Department of Defense W81XWH-11-1-0533 (L.M.P.), and by the Muirhead Chair Endowment of the UTHSC. The authors would like to thank the members of our laboratory at the University of Tennessee Health Science Center for reading and assistance in editing the manuscript.

Conflict of interest

The author declares no conflict of interest.

References

  1. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996.
  2. Sottoriva A, Spiteri I, Piccirillo SG, et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci U S A. 2013;110(10):4009–4014.
  3. Singh SK, Hawkins C, Clarke ID, et al. Identification of human brain tumour initiating cells. Nature. 2004;432(7015):396–401.
  4. Jackson M, Hassiotou F, Nowak A. Glioblastoma stem–like cells: at the root of tumor recurrence and a therapeutic target. Carcinogenesis. 2015;36(2):177–185.
  5. Safa AR, Saadatzadeh MR, Cohen–Gadol AA, et al. Glioblastoma stem cells (GSCs) epigenetic plasticity and interconversion between differentiated non–GSCs and GSCs. Genes Dis. 2015;2(2):152–163.
  6. Gao X, McDonald JT, Naidu M, et al. A proposed quantitative index for assessing the potential contribution of reprogramming to cancer stem cell kinetics. Stem Cells Int. 2014;2014:249309.
  7. Cheng L, Bao S, Rich JN. Potential therapeutic implications of cancer stem cells in glioblastoma. Biochem Pharmacol. 2010;80(5):654–665.
  8. Kondo Y, Katsushima K, Ohka F, et al. Epigenetic dysregulation in glioma. Cancer Sic. 2014;105(4):363–369.
  9. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–1068.
  10. Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321(5897):1807–1812.
  11. Mellinghoff IK, Wang MY, Vivanco I, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med. 2005;353(19):2012–2024.
  12. Fenton TR, Nathanson D, Ponte de Albuquerque C, et al. Resistance to EGF receptor inhibitors in glioblastoma mediated by phosphorylation of the PTEN tumor suppressor at tyrosine 240. Proc Natl Acad Sci U S A. 2012;109(35):14164–14169.
  13. Raizer JJ, Abrey LE, Lassman AB, et al. A phase II trial of erlotinib in patients with recurrent malignant gliomas and nonprogressive glioblastoma multiforme postradiation therapy. Neuro Oncol. 2010;12(1):95–103.
  14. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149(2):274–293.
  15. Sonenberg N, Pause A. Signal transduction. Protein synthesis and oncogenesis meet again. Science. 2006;314(5798):428–429.
  16. Tanaka K, Babic I, Nathanson D, et al. Oncogenic EGFR signaling activates an mTORC2–NF–kappaB pathway that promotes chemotherapy resistance. Cancer Discov. 2011;1(6):524–538.
  17. Cloughesy TF, Yoshimoto K, Nghiemphu P, et al. Antitumor activity of rapamycin in a Phase I trial for patients with recurrent PTEN–deficient glioblastoma. PLoS Med. 2008;5(1):e8.
  18. Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137(2):216–233.
  19. Fortini ME. Notch signaling: the core pathway and its posttranslational regulation. Dev Cell. 2009;16(5):633–647.
  20. Kageyama R, Ohtsuka T. The Notch–Hes pathway in mammalian neural development. Cell Res. 1999;9(3):179–188.
  21. Gaiano N, Fishell G. The role of notch in promoting glial and neural stem cell fates. Annu Rev Neurosci. 2002;25:471–490.
  22. Zhu TS, Costello MA, Talsma CE, et al. Endothelial cells create a stem cell niche in glioblastoma by providing NOTCH ligands that nurture self–renewal of cancer stem–like cells. Cancer Res. 2011;71(18):6061–6072.
  23. Saito N, Fu J, Zheng S, et al. A high Notch pathway activation predicts response to gamma secretase inhibitors in proneural subtype of glioma tumor–initiating cells. Stem Cells. 2014;32(1):301–312.
  24. Garner JM, Fan M, Yang CH, et al. Constitutive Activation of Signal Transducer and Activator of Transcription 3 (STAT3) and Nuclear Factor kappaB Signaling in Glioblastoma Cancer Stem Cells Regulates the Notch Pathway. J Biol Chem. 2013;288(36):26167–26176.
  25. Garner JM, Ellison DW, Finkelstein D, et al. Molecular heterogeneity in a patient–derived glioblastoma xenoline is regulated by different cancer stem cell populations. PLoS One. 2015;10(12):e0145052.
  26. Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128(4):683–692.
  27. Baylin SB, Esteller M, Rountree MR, et al. Aberrant patterns of DNA methylation, chromatin formation and gene expression in cancer. Hum Mol Genet. 2001;10(7):687–692.
  28. Lopez–Bertoni H, Lal B, Li A, et al. DNMT–dependent suppression of microRNA regulates the induction of GBM tumor–propagating phenotype by Oct4 and Sox2. Oncogene. 2015;34(30):3994–4004.
  29. Wongtrakoongate P. Epigenetic therapy of cancer stem and progenitor cells by targeting DNA methylation machineries. World J Stem Cells. 2015;7(1):137–148.
  30. Christman JK. 5–Azacytidine and 5–aza–2'–deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene. 2002;21(35):5483–5495.
  31. Alvarez AA, Field M, Bushnev S, et al. The effects of histone deacetylase inhibitors on glioblastoma–derived stem cells. J Mol Neurosci. 2015;55(1):7–20.
  32. Liffers K, Kolbe K, Westphal M, et al. Histone Deacetylase Inhibitors Resensitize EGFR/EGFRvIII–Overexpressing, Erlotinib–Resistant Glioblastoma Cells to Tyrosine Kinase Inhibition. Target Oncol. 2016;11(1):29–40.
  33. Verhaak RG, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17(1):98–110.
  34. Patel AP, Tirosh I, Trombetta JJ, et al. Single–cell RNA–seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344(6190):1396–1401.
Creative Commons Attribution License

© . This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.