Review Article
J Stem Cells Res, Rev & Rep. 2014;1(2): 1009.
Stem Cells in Gliomas
Assimakis Assimakopoulos1, Konstantinos Polyzoidis1,2 and Athanassios P Kyritsis1,3*
1Neurosurgical Research Institute, University of Ioannina, Ioannina, Greece
2Department of Neurosurgery, Aristotle University of Thessaloniki, Thessaloniki, Greece
3Department of Neurology, University of Ioannina, Ioannina, Greece
*Corresponding author:Athanassios P Kyritsis, Neurosurgical Research Institute, University Campus Ioannina 45500, Greece
Received: Aug 20, 2014; Accepted: September 16, 2014; Published: September 16, 2014
Abstract
Gliomas are central nervous system tumors exhibiting marked cellular heterogeneity, invasiveness and resistance to any therapeutic intervention. Experimental evidence suggests that most of these properties of gliomas are due to the presence of glial stem cells within the gliomas. Markers of glioma stem cells include Nestin, CD133 and CD15. The anatomic location of stem cells within gliomas is predominantly the perivascular areas. Glioma patients with high percentage of glial stem cells have poor survival. Although eradication of the glioma stem cells could extend survival it is difficult to succeed due to their high resistance to therapy. Apart from the glioma stem cells, normal neural stem cells that can be induced from pluripotential stem cells may be used therapeutically for gliomas as carriers for various antitumor agents due to their tropism for neural tissue, if their safety can be attained.
Keywords: Stem cells; Glioma; Brain tumor
Introduction
Gliomas are central nervous system (CNS) tumors of glial origin, exhibiting a profound cellular heterogeneity with tumor cells showing various degrees of differentiation, and genetic heterogeneity with dissimilar gene alterations in neighbourhood cells of the same tumor [1]. This heterogeneity could be due to possible origin of glioma from neural stem cells (NSCs) which after pre-existing or acquired genetic abnormalities drive the NSCs to malignant transformation and formation of glioma stem cells [2]. Glioma stem cells exhibit increased invasiveness, angiogenesis and resistance to therapeutic interventions [3-5]. These glioma stem cells are eventually responsible for tumor malignancy, growth and recurrence [6].
Normal stem cells are capable of infinite proliferation like cancer cells. Apart to the well-known hematopoietic stem cells from bone marrow, stem cell stores exist in other adult tissues. Thus, subcutaneous fat and dermis consist of accessible sources for obtaining stem cells, with minimal discomfort to the patient [7]. Stem cells niche denotes the anatomic location or microenvironment where stem cells are located, and this microenvironment interacts with the stem cells to regulate various cell functions [8]. Recent evidence suggests that human glioma niches are localized in the perivascular areas within gliomas [9].
Study of both normal stem cells and glioma stem cells are important during therapy of gliomas: Normal stem cells may be used during treatment for gliomas, mainly as vehicles to transfer various therapeutic agents to the tumor; study of glioma stem cells is also important to assess the tumor behavior, response to treatment and prognosis.
Stem Cell Markers
Glioma stem cells and NSCs co-express similar markers essential for similar functions in both types of cells (Table 1) [10]. Markers of glioma stem cells include Nestin, CD133 and CD15 [11]. Nestin, is a type-VI intermediate filament that is briefly expressed in glioma tissue during brain development. A study in 70 patients with gliomas that had surgery showed that nestin was expressed in astrocytic gliomas and correlated with the degree of malignancy [12]. Similarly, another immunohistochemichal study in 87 primary CNS tumors showed that nestin was expressed in 95.8% of gliomas with higher expression in malignant tumors and inversely correlated with patient survival. Interestingly, the immunohistochemical staining of nestin in a xenograft model demonstrated its location mainly in the invasive tumor cells at the tumor periphery rather than tumor center [13].
Normal neural stem cell
Normal mesenchymal stem cell
Glioma stem cell
Glioma cell
Nestin
+
+
+
-
CD-133
+
+
+
-
Capable to infinite proliferation
+
+
+
+
Malignant
-
-
+
+
Invasive
-
-
+
+
Affect patient prognosis
-
-
+
+
Resistance to chemotherapy
Unknown
Unknown
++
+
Resistance to radiotherapy
+/-
Unknown
++
+
Resistance to apoptotic agents
-
-
++
+
Localization
Subventricular zone of brain
Umbilical cord blood, adipose tissue, muscle, cornea
Perivascular area of glioma
N/A
Tropism to glioma
+++
+
+
-
Carrier of anti-glioma agents*
+++
+++
-
-
* Chemotherapy, therapeutic genes, viruses, or tumor-toxic molecules
Table 1: Characteristics of normal stem cells and glioma stem cells.
Prognostic significance of glioma stem cells
Immunohistochemical analysis in 125 patients with gliomas of various grades revealed that the presence of glioma stem cells in the tumor, as manifested by Nestin and/or CD133 expression, especially the co-expression of both, was an independent predictor of poor survival [14]. Another study in 95 gliomas of various grade revealed that both the proportion of CD133-positive cells and their ability to organize in clusters were significant independent negative prognostic factors. In addition, the presence of CD133-positive cells was an independent risk factor for tumor recurrence [15].
There is evidence for a crucial role of the expression of glioma SC genes and tumor recurrence as well as response to therapy [16]. Bmi1, an oncogene that is expressed in stem cells and associated with increased cell proliferation and invasion potential of gliomas was able to be downregulated by miR-218, a microRNA involved in its function. These findings suggested that miR-218 may be functioning as tumor suppressor, inhibiting invasion and proliferation of glioma cells [17].
Glioma stem cells mediating resistance to therapy
Gliomas, are tumors highly resistant to chemotherapy [18] or any other therapy [19]. Evidence suggests that GSCs may play a significant role mediating such a resistance [20]. Established human glioblastoma cell lines, such as U-87 MG possess subpopulation of glioma stem cells expressing CD133 and resistant to Fas-activated apoptosis in contrast to the non stem glioma cells that exhibit sensitivity to Fasmediated apoptosis [21]. The apparent heterogeneity of glial tumors [1,2] appears to be a crucial element for in vitro studies in glioma cell lines, since most of them consist of only a small cell subpopulation of the original tumor, making tissue culture results of preclinical test of new therapeutic agents difficult to interpret. Thus, establishment of glial stem cell lines would be more appropriate to test potential therapeutic agents for further in vivo testing.
GSCs have been also resistant to TRAIL even at high concentration of 100-2,000 ng/ml, in contrast to glioma cells with non stem cell characteristics. Their resistance to TRAIL has been attributed to hypermethylation of caspase-8 promoter and low caspase-8 levels for TRAIL-mediated apoptosis. However, reversion of expression of caspase-8 by 5-Aza-2’-deoxycytidine was not enough to reinstate TRAIL effectiveness suggesting interplay of additional mechanisms to TRAIL resistance [22].
The serine/threonine kinase maternal embryonic leucine-zipper kinase (MELK) is an enzyme encoded by the MELK gene, highly expressed in gliomas and significantly associated with the malignant phenotype. MELK expression is reduced by p53 expression leading to increased GSCs apoptosis. MELK is able to form a complex with the oncoprotein c-JUN in GSCs but not normal stem cells and mediate the JNK-driven MELK/c-JUN signaling to maintain tumor survival and resistance to therapeutic interventions [20]. GSCs but not normal neural stem cells synthesize nitric oxide through nitric oxide synthase-2 (NOS2) that is associated with tumor growth and reduced patient survival. Thus, NOS2 inhibition may be a possible target for glioma treatment [23].
Although GSCs exhibit prolonged cell cycle and checkpoint, there appears to be no enhanced DNA repair capability during the checkpoints to explain cell resistance due to DNA repair [24]. After radiotherapy of GSCs in vitro, early postradiation resistance was noted in cells under the presence of EGF and FGF-2, but late postradiation apoptosis was encountered in cells with non-functional p53 [25].
Survivin, a protein encoded by the BIRC5 gene, is expressed during G2-M phase of the cell cycle and abundant in malignant glioma cells and GSCs, mediating inhibition of apoptosis through caspase inactivation. Comparison of Survivin immunohistochemical expression in glioblastomas of 44 untreated and 31 recurrent postchemoradiation and resistant to chemotherapy patients, demonstrated higher expression in the tumors of the recurrent patients especially in the perivascular areas [26].
Glioma SCs exhibit a deregulated balance between cell proliferation and differentiation, specifically increased cellular proliferation and decreased cellular differentiation, partially mediated by the Notch signaling pathway. Inhibition of this pathway by Notch-1 small interfering RNA (siRNA), it was able to inhibit growth of glioma SCs in vitro and in vivo in nude mice, suggestive that Notch-1 gene may represent a possible target for glioma therapy [27]. Stem cells isolated from glioma specimens could induce tumors in immunocompromised mice that secreted vascular endothelial growth factor (VEGF) leading to endothelial cell migration and excessive angiogenesis. These angiogenic effects could be suppressed by bevacizumab a potent inhibitor of angiogenesis and currently used therapy for gliomas [28].
Experimental evidence in nude mice bearing C6 gliomas suggests that disruption of GSC niche by antiangiogenic therapy could sensitize the GSCs to chemotherapy [25]. Furthermore, clinical evidence indicates that radiotherapy of the stem cell niches in patients with gliomas could extend survival. Thus, a study in 55 patients with malignant gliomas treated with radiotherapy showed that patients subjected to bilateral subventricular zone that harbors the GSCs niches had a significant improvement in progression-free survival (15.0 vs 7.2 months) than patients whose radiotherapy field did not include these areas. In addition, higher radiation fractions may be more efficient that lower fractions [29].
Normal stem cells as carriers of therapeutic agents to gliomas
Normal NSCs and mesenchymal stem cells (MSCs) may be used as cellular vehicles for targeted delivery of various agents to glioma cells (Table 1). However, the normal stem cells that carry anti-tumor substances may be used therapeutically only if their malignant transformation potential can be eliminated [30]. MSCs isolated from the umbilical cord blood could migrate towards glioma cells via a partially dependent on the PDGF/PGGFR system glioma tropism, and provoke a Fas-mediated apoptosis in glioma cells [31]. NSCs secrete factors that inhibited the proliferation of glioma cells, both in vitro and in vivo in gliomas growing in the cisterna magna of mice [32]. In addition to secretion of growth inhibitory factors, NSCs could be used as cellular vehicles to deliver chemotherapy, therapeutic genes, viruses, or tumor-toxic molecules to malignant gliomas due to their tropism for glioma cells [33,34]. Preclinical comparison of the two cell lines revealed that although both NSCs and MSCs allowed adenoviral replication intracellularly, the efficiency of the NSCs was much higher to MSCs [35].
Difficult tumor areas to attain enough concentration of a therapeutic agent consist of hypoxic areas with necrosis and poor blood circulation, such as the tumor necrotic core and the adjacent to tumor areas that contain infiltrating tumor cells with still poor new blood vessel formation. Interestingly, NSCs tropism is predominantly directed towards the hypoxic areas of the malignant gliomas located both in tumor core and the periphery of the tumor. This function appears to be mediated via stromal cell-derived factor-1 (SDF-1) SDF-1/CXCR4, uPA/uPAR, VEGF/VEGFR2, and hepatocyte growth factor/c-Met signaling pathways [36].
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) demonstrated glioma-directed killing activity suggestive of a promising antitumor treatment strategy in clinic [37,38]. Thus, employment of TRAIL-secreting human MSCs could represent a tumor specific targeted therapy for gliomas alone or in combination of a chemotherapeutic agent such as temozolomide [39]. Another example is the use of normal NSCs carrying secretable TRAIL in an orthotopic mouse model of gliomas in combination with systemically administered other specific therapies to enhance the anti-tumor effect [40].
Normal stem cells can be used to deliver enzymes that convert a chemotherapeutic prodrug to an active chemotherapy drug. An in vivo example of using normal NSCs to this effect consists of usage of a cytosine deaminase (CD)-expressing clonal human NSC line, HB1.F3.CD, to deliver the enzyme to gliomas growing in brains of mice in order to convert the systemically administered prodrug 5-fluorocytosine to the active chemotherapeutic 5-fluorouracil locally [41].
Facilitated delivery of gene therapy agents is another potential application of normal stem cell technology for treatment of gliomas. Animal experiments have demonstrated that transfer of the interleukin-4 gene into C57BL6J mouse NSCs and injection into syngeneic brain gliomas significantly increased the survival of most tumor-bearing mice [42]. Similarly, NSCs transduced with herpes simplex virus-thymidine kinase gene (NSCtk) were injected in distant sites of rat brains harboring C6 glioma cells, following by the systemic administration of ganciclovir (GCV), a drug against herpes virus. The reason for the injection in distant areas was to study the migratory potential of neural stem cells and their effectiveness to reach the glioma cells. The result was active migration of neural stem cells towards the tumors, even when implanted at the controlateral hemisphere, and marked inhibition of tumor growth and prolonged survival of the animals [43]. Induced pluripotent stem cells derived from primary mouse embryonic fibroblasts were used to generate NSCs. Subsequently, the NSCs were transduced with a baculoviral vector having the HSV TK gene and were injected into the controlateral to the tumor hemisphere in mice. Systemic administration of ganciclovir, resulted in inhibition of glioma growth suggesting that NSCs may be used as vehicles for gene therapy [44,45]. Apart from intratumoral delivery of normal stem cells, intravascular delivery of NSCs appears to be an effective strategy to target tumors of neural origin, inside the brain [46].
Conclusion
The profound cellular heterogeneity of gliomas in association with their invasiveness and resistance to therapeutic interventions is at least partially due to the presence of glioma stem cells within the malignant types of these tumors. The anatomic location or microenvironment where these cells are located is denoted as stem cell niches in the perivascular areas within gliomas. Study of glioma stem cells is important to assess the tumor behavior, delivery of chemotherapeutic agents, response to treatment and prognosis. In addition to glioma stem cells, normal neural stem cells exist in the brain of human beings in addition to other locations. These normal stem cells have high tropism for glioma tumors and can migrate even from distant areas towards gliomas. This property renders them good candidates as transfer vehicles to carry toxic agents to gliomas. Further research is needed to assess their safety prior to their routine utilization as carriers of therapeutic agents to humans.
References
- Kyritsis AP, Zhang B, Zhang W, Xiao M, Takeshima H, Bondy ML, Cunningham JE. Mutations of the p16 gene in gliomas. Oncogene. 1996; 12: 63-67.
- Kyritsis AP, Bondy ML, Rao JS, Sioka C. Inherited predisposition to glioma. Neuro Oncol. 2010; 12: 104-113.
- Kyritsis AP, Sioka C, Rao JS. Viruses, gene therapy and stem cells for the treatment of human glioma. Cancer Gene Ther. 2009; 16: 741-752.
- Heddleston JM, Hitomi M, Venere M, Flavahan WA, Yang K, Kim Y, et al. Glioma stem cell maintenance: the role of the microenvironment. Curr Pharm Des. 2011; 17: 2386-2401.
- Baronchelli S, Bentivegna A, Redaelli S, Riva G, Butta V, Paoletta L, et al. Delineating the cytogenomic and epigenomic landscapes of glioma stem cell lines. PLoS One. 2013; 8: e57462.
- 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: 781-786.
- Freitas CS, Dalmau SR. Multiple sources of non-embryonic multipotent stem cells: processed lipoaspirates and dermis as promising alternatives to bone-marrow-derived cell therapies. Cell Tissue Res. 2006; 325: 403-411.
- Mannino M, Chalmers AJ. Radioresistance of glioma stem cells: intrinsic characteristic or property of the 'microenvironment-stem cell unit'? Mol Oncol. 2011; 5: 374-386.
- He H, Li MW, Niu CS. The pathological characteristics of glioma stem cell niches. J Clin Neurosci. 2012; 19: 121-127.
- Holmberg J, He X, Peredo I, Orrego A, Hesselager G, Ericsson C, et al. Activation of neural and pluripotent stem cell signatures correlates with increased malignancy in human glioma. PLoS One. 2011; 6: e18454.
- Jin X, Jin X, Jung JE, Beck S, Kim H. Cell surface Nestin is a biomarker for glioma stem cells. Biochem Biophys Res Commun. 2013; 433: 496-501.
- Tomita T, Akimoto J, Haraoka J, Kudo M. Clinicopathological significance of expression of nestin, a neural stem/progenitor cell marker, in human glioma tissue. Brain Tumor Pathol. 2014; 31: 162-171.
- Strojnik T, Røsland GV, Sakariassen PO, Kavalar R, Lah T. Neural stem cell markers, nestin and musashi proteins, in the progression of human glioma: correlation of nestin with prognosis of patient survival. Surg Neurol. 2007; 68: 133-143.
- Zhang M, Song T, Yang L, Chen R, Wu L, Yang Z, et al. Nestin and CD133: valuable stem cell-specific markers for determining clinical outcome of glioma patients. J Exp Clin Cancer Res. 2008; 27: 85.
- Zeppernick F, Ahmadi R, Campos B, Dictus C, Helmke BM, Becker N, et al. Stem cell marker CD133 affects clinical outcome in glioma patients. Clin Cancer Res. 2008; 14: 123-129.
- Balbous A, Cortes U, Guilloteau K, Villalva C, Flamant S, Gaillard A, Milin S. A mesenchymal glioma stem cell profile is related to clinical outcome. Oncogenesis. 2014; 3: e91.
- Tu Y, Gao X, Li G, Fu H, Cui D, Liu H, et al. MicroRNA-218 inhibits glioma invasion, migration, proliferation, and cancer stem-like cell self-renewal by targeting the polycomb group gene Bmi1. Cancer Res. 2013; 73: 6046-6055.
- Kyritsis AP, Levin VA. An algorithm for chemotherapy treatment of recurrent glioma patients after temozolomide failure in the general oncology setting. Cancer Chemother Pharmacol. 2011; 67: 971-983.
- Kyritsis AP, Bondy ML, Levin VA. Modulation of glioma risk and progression by dietary nutrients and antiinflammatory agents. Nutr Cancer. 2011; 63: 174-184.
- Gu C, Banasavadi-Siddegowda YK, Joshi K, Nakamura Y, Kurt H, Gupta S, et al. Tumor-specific activation of the C-JUN/MELK pathway regulates glioma stem cell growth in a p53-dependent manner. Stem Cells. 2013; 31: 870-881.
- Bertrand J, Begaud-Grimaud G, Bessette B, Verdier M, Battu S, Jauberteau MO. Cancer stem cells from human glioma cell line are resistant to Fas-induced apoptosis. Int J Oncol. 2009; 34: 717-727.
- Capper D, Gaiser T, Hartmann C, Habel A, Mueller W, Herold-Mende C, et al. Stem-cell-like glioma cells are resistant to TRAIL/Apo2L and exhibit down-regulation of caspase-8 by promoter methylation. Acta Neuropathol. 2009; 117: 445-456.
- Eyler CE, Wu Q, Yan K, MacSwords JM, Chandler-Militello D, Misuraca KL, et al. Glioma stem cell proliferation and tumor growth are promoted by nitric oxide synthase-2. Cell. 2011; 146: 53-66.
- Ropolo M, Daga A, Griffero F, Foresta M, Casartelli G, Zunino A, et al. Comparative analysis of DNA repair in stem and nonstem glioma cell cultures. Mol Cancer Res. 2009; 7: 383-392.
- Firat E, Gaedicke S, Tsurumi C, Esser N, Weyerbrock A, Niedermann G. Delayed cell death associated with mitotic catastrophe in γ-irradiated stem-like glioma cells. Radiat Oncol. 2011; 6: 71.
- Guvenc H, Pavlyukov MS, Joshi K, Kurt H, Banasavadi-Siddegowda YK, Mao P, et al. Impairment of glioma stem cell survival and growth by a novel inhibitor for Survivin-Ran protein complex. Clin Cancer Res. 2013; 19: 631-642.
- Wang J, Wang C, Meng Q, Li S, Sun X, Bo Y, et al. siRNA targeting Notch-1 decreases glioma stem cell proliferation and tumor growth. Mol Biol Rep. 2012; 39: 2497-2503.
- Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, et al. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006; 66: 7843-7848.
- Evers P, Lee PP, DeMarco J, Agazaryan N, Sayre JW, Selch M, et al. Irradiation of the potential cancer stem cell niches in the adult brain improves progression-free survival of patients with malignant glioma. BMC Cancer. 2010; 10: 384.
- Bexell D, Svensson A, Bengzon J. Stem cell-based therapy for malignant glioma. Cancer Treat Rev. 2013; 39: 358-365.
- Gondi CS, Veeravalli KK, Gorantla B, Dinh DH, Fassett D, Klopfenstein JD, et al. Human umbilical cord blood stem cells show PDGF-D-dependent glioma cell tropism in vitro and in vivo. Neuro Oncol. 2010; 12: 453-465.
- Suzuki T, Izumoto S, Wada K, Fujimoto Y, Maruno M, Yamasaki M, et al. Inhibition of glioma cell proliferation by neural stem cell factor. J Neurooncol. 2005; 74: 233-239.
- Ehtesham M, Stevenson CB, Thompson RC. Stem cell therapies for malignant glioma. Neurosurg Focus. 2005; 19: E5.
- Xu Q, Yuan X, Yu JS. Glioma stem cell research for the development of immunotherapy. Adv Exp Med Biol. 2012; 746: 216-225.
- Ahmed AU, Tyler MA, Thaci B, Alexiades NG, Han Y, Ulasov IV, et al. A comparative study of neural and mesenchymal stem cell-based carriers for oncolytic adenovirus in a model of malignant glioma. Mol Pharm. 2011; 8: 1559-1572.
- Zhao D, Najbauer J, Garcia E, Metz MZ, Gutova M, Glackin CA, et al. Neural stem cell tropism to glioma: critical role of tumor hypoxia. Mol Cancer Res. 2008; 6: 1819-1829.
- Puduvalli VK, Sampath D, Bruner JM, Nangia J, Xu R, Kyritsis AP. TRAIL-induced apoptosis in gliomas is enhanced by Akt-inhibition and is independent of JNK activation. Apoptosis. 2005; 10: 233-243.
- Tsamis KI, Alexiou GA, Vartholomatos E, Kyritsis AP. Combination treatment for glioblastoma cells with tumor necrosis factor-related apoptosis-inducing ligand and oncolytic adenovirus delta-24. Cancer Invest. 2014; 31: 630-638.
- Kim SM, Woo JS, Jeong CH, Ryu CH, Jang JD, Jeun SS. Potential application of temozolomide in mesenchymal stem cell-based TRAIL gene therapy against malignant glioma. Stem Cells Transl Med. 2014; 3: 172-182.
- Bagci-Onder T, Wakimoto H, Anderegg M, Cameron C, Shah K. A dual PI3K/mTOR inhibitor, PI-103, cooperates with stem cell-delivered TRAIL in experimental glioma models. Cancer Res. 2011; 71: 154-163.
- Aboody KS, Najbauer J, Metz MZ, D'Apuzzo M, Gutova M, Annala AJ, et al. Neural stem cell-mediated enzyme/prodrug therapy for glioma: preclinical studies. Sci Transl Med. 2013; 5: 184ra59.
- Benedetti S, Pirola B, Pollo B, Magrassi L, Bruzzone MG, Rigamonti D, et al. Gene therapy of experimental brain tumors using neural progenitor cells. Nat Med. 2000; 6: 447-450.
- Li S, Gao Y, Tokuyama T, Yamamoto J, Yokota N, Yamamoto S, et al. Genetically engineered neural stem cells migrate and suppress glioma cell growth at distant intracranial sites. Cancer Lett. 2007; 251: 220-227.
- Lee EX, Lam DH, Wu C, Yang J, Tham CK, Ng WH, et al. Glioma gene therapy using induced pluripotent stem cell derived neural stem cells. Mol Pharm. 2011; 8: 1515-1524.
- Zhao Y, Lam DH, Yang J, Lin J, Tham CK, Ng WH, et al. Targeted suicide gene therapy for glioma using human embryonic stem cell-derived neural stem cells genetically modified by baculoviral vectors. Gene Ther. 2012; 19: 189-200.
- Brown AB, Yang W, Schmidt NO, Carroll R, Leishear KK, Rainov NG, et al. Intravascular delivery of neural stem cell lines to target intracranial and extracranial tumors of neural and non-neural origin. Hum Gene Ther. 2003; 14: 1777-1785.