The importance of DNA repair as a resistance mechanism in gliomas, the most aggressive form of brain tumor, is a clinically relevant topic. Recent studies show that not all cells are equally malignant in gliomas. Certain subpopulations are particularly prone to drive tumor progression and resist chemo- and radiotherapy. Those cells have been variably named cancer stem cells or cancer-initiating cells or tumor-propagating cells, owing to their possible (but still uncertain) origin from normal stem cells. Although DNA repair reduces the efficacy of chemotherapeutics and ionizing radiation toward bulk gliomas, its contribution to resistance of the rare glioma stem cell subpopulations is less clear. Mechanisms other than DNA repair (in particular low proliferation and activation of the DNA damage checkpoint response) are likely main players of resistance in glioma stem cells and their targeting might yield significant therapeutic gains. (Mol Cancer Res 2009;7(7):989–99)

Primary brain tumors can be classified in terms of what type of cell the tumor began as (reviewed in refs. 1, 2). Gliomas are the most common form of primary brain tumors. Among gliomas, there are astrocytomas [World Health Organization (WHO) grade I and II], which derive from star-shaped astrocyte-like cells. High-grade astrocytomas are called anaplastic astrocytomas (grade III) and multiform glioblastomas [(GBM) grade IV]. Compared with lung and breast cancer, which have rates of about 60 per 100,000 in Western countries, the rate of gliomas is ∼ tenfold lower. Nevertheless these cancers have serious effects on the health-care system in general, and especially on patients and their families. Not only do glioma patients have to cope with the diagnosis of incurable disease, they and their families are usually also confronted with the patient's decrease in cognitive and emotional function as a result of cerebral disease. Despite advances in surgery and radiotherapy, the prognosis for malignant gliomas remains poor, the median survival of GBM patients being 5 to 8 months, depending on the patient's age (2). Alkylating agent-based chemotherapy has been shown to modestly increase response rates and survival times of high-grade glioma patients when used as an adjuvant to surgery and radiation (3, 4). Some more efficacy can be observed in lower-grade gliomas such as oligodendroglioma (5), but serious side effects such as cumulative lymphopenia and consequent high incidence of infections occur after prolonged treatments (6). Although, the most frequently used alkylating agent for glioma treatment, temozolomide (TMZ), is well-tolerated by most patients and has favorable pharmacodynamic and pharmacokinetic properties (3). The inefficacy of currently available chemotherapeutic and radiotherapeutic agents depends on a number of resistance mechanisms among which DNA repair may play a role (7-10).

O6 MethylGua DNA Methyltransferase

Many studies have indicated O6 methylGua DNA methyltransferase (MGMT) as a possible resistance factor to alkylating chemotherapy in brain tumors (3, 11), although negative results have been also reported (Table 1; ref. 12). Two major cell-killing and apoptotic alkylated lesions arising from drugs used in brain cancer chemotherapy are O6-methylguanine (O6-meGua) and O6-chloroethylguanine (O6-chloroethylGua). These lesions are removed by the specific action of MGMT thus counteracting alkylating agents' killing effects. Levels of MGMT expression may in some cases predict TMZ sensitivity in adult human glioma patients (13-16). A stronger correlation with overall survival, regardless of treatment, is observed in pediatric glioma patients (17). MGMT promoter methylation actually represents part of a genetic signature of less-aggressive gliomas, apart from treatment (Fig. 1A; refs. 15, 18, 19). In MGMT-negative glioblastomas TMZ further enhances radiation responses by increasing the degree of radiation-induced double-strand DNA damage (20). However, the role of MGMT may have been overestimated by the relatively simple assay procedures for its promoter methylation status and enzymatic activity, and should not be generalized (21). For instance, no correlation between MGMT promoter methylation and MGMT protein expression was recently observed in serial stereotactic specimens from anaplastic astrocytomas and glioblastomas (22). One advantage of MGMT targeting is that local intracerebral administration of MGMT drug inhibitors that may sensitize tumor cells to TMZ, is feasible and without important side effects (23, 24). Viral inhibitors may further be envisaged, on the basis of the fact that some viral proteins directly target and inactivate some DNA repair genes (25). For instance, a combination of oncolytic adenovirus Delta-24-RGD and TMZ induces a therapeutic synergy in glioma cells and significantly improves the survival of glioma-bearing mice by silencing the MGMT gene (26).

Table 1.

DNA Repair Capacity and Therapeutic Efficacy in Bulk Gliomas

DNA Repair Mechanism-EnzymeHigherSimilarLowerTherapeutic AgentTherapeutic EfficacyReference
IncreasedUnchangedDecreased
MGMT    CENU    12 
MGMT    TMZ    11 
MGMT    TMZ+IR    20 
MGMT    TMZ    13 
MGMT    TMZ    
MGMT    TMZ    26 
MGMT        17 
MGMT    TMZ    17 
MGMT        18 
MGMT    TMZ    14 
MGMT    TMZ ACNU BCNU Procarbazine    15, 129 
MGMT    TMZ    16 
MGMT    TMZ+IR    19 
MGMT    TMZ    140 
MLH1        32 
MSH2,MSH6,PMS2    TMZ    13 
MSH6    TMZ    30 
MSH6    TMZ    31 
MSH6    Alkylating agents    29 
APE1/Ref-1        37 
APE1/Ref-1    MMS TMZ BCNU    38 
APE1/Ref-1    IR BCNU CCNU Procarbazine    39 
APE1/Ref-1    Me-Lex    42 
AAG    Me-Lex    42 
PARP    TMZ    43 
PARP    TMZ    44 
DSB repair    TMZ    158 
Rad51    IR    45 
Rad51    IR    46 
BRCA1    IR Cisplatin    48 
DNA-PKcs    IR    49 
KUB3    IR    49 
DNA-PK    Cisplatin    50 
Ku86    Cisplatin    50 
Ku70    Cisplatin    50 
Resolution of γ-H2AX foci    IR    131 
Phosphorylation of histone H2AX    IR    47 
FA pathway    TMZ BCNU    51 
Various DNA repair activities (MGMT DNA-PK Ku70 Ku80 ERCC-1)    Cisplatin Etoposide Camptothec.Doxorubicin IR    54 
Expression of various DNA repair genes    TMZ    60 
DNA Repair Mechanism-EnzymeHigherSimilarLowerTherapeutic AgentTherapeutic EfficacyReference
IncreasedUnchangedDecreased
MGMT    CENU    12 
MGMT    TMZ    11 
MGMT    TMZ+IR    20 
MGMT    TMZ    13 
MGMT    TMZ    
MGMT    TMZ    26 
MGMT        17 
MGMT    TMZ    17 
MGMT        18 
MGMT    TMZ    14 
MGMT    TMZ ACNU BCNU Procarbazine    15, 129 
MGMT    TMZ    16 
MGMT    TMZ+IR    19 
MGMT    TMZ    140 
MLH1        32 
MSH2,MSH6,PMS2    TMZ    13 
MSH6    TMZ    30 
MSH6    TMZ    31 
MSH6    Alkylating agents    29 
APE1/Ref-1        37 
APE1/Ref-1    MMS TMZ BCNU    38 
APE1/Ref-1    IR BCNU CCNU Procarbazine    39 
APE1/Ref-1    Me-Lex    42 
AAG    Me-Lex    42 
PARP    TMZ    43 
PARP    TMZ    44 
DSB repair    TMZ    158 
Rad51    IR    45 
Rad51    IR    46 
BRCA1    IR Cisplatin    48 
DNA-PKcs    IR    49 
KUB3    IR    49 
DNA-PK    Cisplatin    50 
Ku86    Cisplatin    50 
Ku70    Cisplatin    50 
Resolution of γ-H2AX foci    IR    131 
Phosphorylation of histone H2AX    IR    47 
FA pathway    TMZ BCNU    51 
Various DNA repair activities (MGMT DNA-PK Ku70 Ku80 ERCC-1)    Cisplatin Etoposide Camptothec.Doxorubicin IR    54 
Expression of various DNA repair genes    TMZ    60 
FIGURE 1.

DNA repair-dependent tumor progression in glioma patients. A. Overall survival of glioblastoma patients treated with IR + TMZ by presence of MGMT promoter methylation status. Dark gray line, patients with methylated MGMT promoter; light gray line, patients with unmethylated MGMT promoter status (Reprinted with permission from 2008 American Society of Clinical Oncology. All rights reserved. Brandes AA, et al.: J Clin Oncol Vol. 26, 2008:2192–7). B. Progression-free survival for grade III gliomas treated with alkylating agent-based chemotherapy according to AP endo activity categorized as quartiles. All patients were treated with surgery and radiation therapy before receiving alkylator treatment. Activity was measured in tumors obtained before alkylator therapy. Time to tumor progression is the interval between the initiation of alkylating agent therapy and radiological evidence of progression. Curves were calculated by the method of Kaplan-Meier (Reprinted by permission from the American Assoiciation for Cancer Research: Clin Cancer Res 10:7875–83, 2004).

FIGURE 1.

DNA repair-dependent tumor progression in glioma patients. A. Overall survival of glioblastoma patients treated with IR + TMZ by presence of MGMT promoter methylation status. Dark gray line, patients with methylated MGMT promoter; light gray line, patients with unmethylated MGMT promoter status (Reprinted with permission from 2008 American Society of Clinical Oncology. All rights reserved. Brandes AA, et al.: J Clin Oncol Vol. 26, 2008:2192–7). B. Progression-free survival for grade III gliomas treated with alkylating agent-based chemotherapy according to AP endo activity categorized as quartiles. All patients were treated with surgery and radiation therapy before receiving alkylator treatment. Activity was measured in tumors obtained before alkylator therapy. Time to tumor progression is the interval between the initiation of alkylating agent therapy and radiological evidence of progression. Curves were calculated by the method of Kaplan-Meier (Reprinted by permission from the American Assoiciation for Cancer Research: Clin Cancer Res 10:7875–83, 2004).

Close modal

Mismatch Repair

The relatively low incidence of microsatellite instability in pediatric and adult high-grade gliomas would suggest that the involvement of mismatch repair (MMR) defects in alkylating agent resistance is limited (27, 28). However, inactivating somatic mutations of the MMR gene MSH6 confer resistance to alkylating agents in glioma in vivo and concurrently unleash accelerated mutagenesis in resistant clones as a consequence of continued exposure to alkylating agents. Hence, when MSH6 is inactivated in gliomas, alkylating agents may convert from inducers of tumor cell death to promoters of neoplastic progression (Table 1; ref. 29). Loss of MSH6 has been recently found in a subset of ionizing radiation (IR) + TMZ-treated glioblastoma recurrences (30). The association with tumor progression during TMZ treatment was significant, mirroring the alkylator resistance conferred by MSH6 inactivation in vitro. MSH6 deficiency may therefore contribute to the emergence of recurrent glioblastomas during TMZ treatment (30). These conclusions have been yet recently questioned by Maxwell and coworkers (31) who could not find any effect of MMR deficiency on resistance of adult malignant glioma to TMZ. Promoter hypermethylations but not structural alterations in the MLH1 gene have been observed in low grade gliomas, associated with their development and progression (32). MLH1 promoter hypermethylation may be an early event in the development and progression or the clonal evolution of gliomas, this gene inactivation proving stable even on tumor recurrence (32). Conversely, proficient MMR is required for repair of interstrand crosslinks via stabilization of RAD51-containing repair intermediates and may thus contribute to resistance to crosslinking agents (33).

Base Excision Repair

Albeit resistance to TMZ has been ascribed to elevated levels of MGMT and or reduced MMR, >80% of the DNA lesions induced by TMZ are N-methylated bases that are recognized by DNA glycosylases and not by MGMT (34, 35). Studies with isogenic cells deficient or overexpressing the DNA base excision repair (BER) enzymes pol beta and alkyladenine DNA glycosylase (AAG) have shown that the BER pathway is a major contributor to cellular resistance to TMZ and its efficacy depends on specific BER gene expression and activity (35, 36). APE1/Ref-1, the main AP endonuclease of mammalian cells, significantly increases during gliomagenesis (37) and may contribute to alkylating agent resistance by incising at abasic (AP) sites left after removal of methylated bases (Table 1; refs. 38, 39). Silber and coworkers found that antisense oligonucleotides directed against APE1/Ref-1 in a human glioma cell line lacking MGMT, mediated both reduction in APE1/Ref-1 protein and AP endo activity and concurrent reduction in resistance to methylmethane sulfonate (MMS), TMZ, and 1,3-(2-chloroethyl)-1-nitrosourea (BCNU; ref. 38). The same laboratory also found that AP endo activity promoted resistance to IR plus chemotherapy with BCNU and TMZ in medulloblastoma and primitive neuroectodermal tumors (40). APE1/Ref-1 may thus represent a potential marker of treatment outcome and use of AP endo inhibitors may help in overcoming resistance in bulk gliomas (Fig. 1B; refs. 39-41). Methyl-lexitropsin (Me-lex) is a sequence-specific alkylator that produces 3-methyl adenine (3-meAde) as the predominant (>90%) DNA lesion (42). Me-lex is cytotoxic in human glioma cells and AAG promotes resistance, indicating that 3-meAde is a lethal lesion in these cells. Further, AP sites resulting from 3-meAde repair are cytotoxic and Ape1/Ref-1 promotes resistance to these derivative lesions. MGMT-proficient lines are more resistant than MGMT-deficient lines, an unexpected finding because Me-Lex produces very little O6 methylGua. Me-lex may have clinical utility in the adjuvant therapy of gliomas, and AAG and Ape1/Ref-1 inhibitors may be useful in targeting alkylating agent resistance in bulk gliomas (42). Finally, increased sensitivity to TMZ can be observed in combination with inhibitors of poly(ADP-ribose) polymerase, a molecular sensor of single strand breaks that interacts with many BER components (43, 44).

Strand Break Repair

Rad51-mediated homology-directed repair (HDR) may represent an important determinant of glioma radiosensitivity that can be modulated. Gleevec is a relatively specific inhibitor of c-Abl, a tyrosine kinase that participates in the regulation of Rad51. When glioma cells are pretreated with Gleevec, radiation-induced Rad51 expression and nuclear foci formation are reduced and result in radiosensitivity enhancement (Table 1) (ref. 45). Hence, agents that disrupt Rad51-dependent repair or prevent G2 checkpoint activation may selectively sensitize glioma cells to radiation (46). One of those agent could be TMZ itself (47). A number of additional DSB repair factors including BRCA1 (48), DNA-protein kinase catalytic subunit (DNA-PKcs), and the novel potential end-joining factor KUB3 also influence the cellular radiation response in rodent glioma models (49). DNA-PK influences the response to cisplatin as well (50).

Fanconi Anemia Pathway

The Fanconi anemia (FA) pathway may contribute to resistance of glioma tumors to alkylating agents (51). A glioma cell line deficient in the FA repair pathway (HT16) exhibited increased sensitivity to TMZ and BCNU relative to FA-proficient glioma cells. Moreover, inhibition of FA pathway by curcumin, a polyphenol present in the curry spice turmeric, or by small interfering RNA (siRNA) suppression caused increased sensitivity to TMZ/BCNU in the wild-type U87 glioma cell line. The BCNU sensitizing effect of FA inhibition seemed additive to that of MGMT inhibition. Yet, curcumin has many effects beyond targeting the FA pathway, being e.g., a nuclear factor kappa-B (NF-KB) inhibitor and a reactive oxygen species (ROS) scavenger (52, 53), and curcumin sensitization of glioma cells to several clinically used chemotherapeutic agents (cisplatin, etoposide, camptothecin, and doxorubicin) and IR also correlates with reduced expression of bcl-2 family members as well as DNA repair enzymes that do not take part in the FA pathway (MGMT, DNA-PK, Ku70, Ku80, and ERCC-1; ref. 54). Leaving aside the underlying mechanism, a role for curcumin as an adjunct to traditional chemotherapy and radiation in the treatment of brain cancer could be worth exploring (54).

Other Resistance Mechanisms in Bulk Gliomas

Abrogation of p53 wild-type function strongly attenuates TMZ cytotoxicity. Conversely, agents designed to stabilize the wild-type conformation of p53 sensitize glioma cells to TMZ cytotoxicity. Thus, determination of p53 status may help in identification of glioma patients who will or will not respond to TMZ (13). Contrary to what can be observed for TMZ, chloroethylating drugs are more toxic for p53-mutated glioma cells in which both apoptosis and necrosis are induced (55). Inactivation of p53 by pifithrin-alpha or siRNA downregulation sensitize p53 wild-type but not p53-mutated glioma cells to 3-[(4-amino-2-methyl-5-pyrimidinyl) methyl]-1-(2-chloroethyl)-1-nitrosourea (ACNU) and BCNU. ACNU and BCNU provoke the formation of DNA double strand breaks (DSBs) in glioma cells that precede the onset of apoptosis and necrosis. These DSBs are repaired in p53 wild-type cells but accumulate in p53-mutated cells. Therefore, functional p53 seems to stimulate the repair of chloroethylnitrosourea (CENU)-induced cross-links and or DSBs generated from CENU-induced lesions. In conclusion, p53 may have opposing effects in gliomas treated with methylating or chloroethylating agents and, therefore, the p53 status should be taken into account when deciding which therapeutic drug to use (55). After IR, expression of dominant-negative p53 selectively sensitizes glioma cells (56) and p53-mutated glioma cell lines repair DSBs more slowly and less effectively than p53-wild-type glioma cell lines or normal human astrocytes (46).

Expression of apoptotic proteins correlates in part with resistance of gliomas to chemotherapeutic agents (57, 58). Astrocytes per se have been found to express relatively high levels of antiapoptotic Bcl-2 and Bcl-XL proteins and low levels of proapoptotic Bad and Bid proteins suggesting that astrocytes may be less vulnerable to proapoptotic stimuli compared with other cell types. Hypermethylation of the proapoptotic gene caspase-8 is also a common hallmark of relapsing gliomas (59).

Microarray analyses showed that 78 out of 17,000 genes were differentially expressed between parental cells and TMZ-resistant variants isolated in the human glioma cell line SNB-19 (60). None were implicated in DNA repair whereas several genes involved in differentiation were downregulated. Thus, in this model, the acquisition of resistance to TMZ may result from selection of less differentiated preexistent resistant cells in the parental tumor (60).

Glioma Stem Cells

More than 25 years ago, Rosenblum and coworkers seminally proposed that chemo- and radio sensitivity of human malignant brain tumors could be dependent on inherent differences in the sensitivity of clonogenic stem cells to radiation and or chemotherapeutic agents (61). As for other cancers, the existence of glioma cells endowed with features of primitive progenitor cells and tumor-initiating function has been shown (62-65). Most gliomas contain considerable portions of cells expressing stem cell markers such as CD133, Nestin, Sox-2, and Musashi-1 (66, 67), and expression of some of them has been shown to be prognostic of decreased overall survival (68-70). In particular, expression of the CD133 stem cell antigen negatively correlates with glioma patient survival in several studies (71-75). CD133+ is often expressed on the surface of human glioma stem cells and importantly, CD133+ cells are preserved during orthotopic transplantations of human gliomas in the mouse (76). However, significant subpopulations of CD133− stem cells may also exist in gliomas (77). For instance, Beier and coworkers (78) have observed that 11 out of 15 primary glioblastomas contained a significant CD133+ subpopulation that displayed asymmetrical cell divisions yielding cells expressing markers characteristic for all three neural lineages. However, the remaining four cell lines were driven by CD133− tumor cells that also fulfilled stem cell criteria (e.g., pluripotency and tumorigenicity in nude mice in vivo). Consistently, analysis of the expression profiles of nine glioma cell lines established under neural stem cell conditions yielded two distinct clusters (79). Four cell lines were characterized by the expression of neurodevelopmental genes, a multipotent differentiation profile, expression of CD133, and formation of highly invasive tumors in vivo. The other five cell lines shared expression signatures enriched for extracellular matrix-related genes, had a more restricted differentiation capacity, contained no or fewer CD133+ cells, and displayed reduced tumorigenicity and invasion in vivo. Hence, CD133+ stem cells may maintain only a subset of primary glioblastomas, the remainder deriving from CD133− tumor stem cells with distinct phenotypical features (78-82). Additional cell surface markers have been investigated to hallmark glioma stem cells including the aforementioned Nestin, Sox-2, and Musashi-1 as well as CD117, CD71, CD45, and A2B5 (83-85), but at the moment, CD133 remains prominent. Whether the currently identified glioma stem cells may be the true cell-of-origin for tumor initiation and progression or the results of such processes is still an open question (86, 87). Further, gliomas produce factors [e.g., urokinase plasminogen activator (uPA) and its receptor (uPAR)] that selectively attract neural stem cells (88-93) and this has originated the question about whether stem cells may be attracted by, rather than originate, the tumor (66, 94-96). Notwithstanding, gliomas do not clash with the general cancer-stem-cell-related concept that if only a rare subset of tumor stem cells, whatever their origin, drives tumor formation, then it is important to identify this population and develop therapies that target it (97-103).

Some established glioma cell lines that have been maintained over years in culture contain a subpopulation of stem cells (104, 105). For instance, in the absence of serum a combination of basic fibroblast growth factor (FGF) and platelet-derived growth factor (PDGF) maintains in the C6 glioma cell line a population enriched in stem cells (referred to as “side population”) with the ability to efflux the fluorescent dye Hoechst 33342. Moreover, C6 side population cells, but not nonside population cells, can generate both side population and nonside population cells in culture and are largely responsible for the in vivo malignancy of that cell line. Importantly, C6 side population cells can produce both neurons and glial cells in vitro and in vivo. Thus, some cancer cell lines may contain a minor subpopulation of stem cells that can be maintained indefinitely in culture, and which is crucial for their malignancy (104-108). This cannot be generalized yet: controversial data exist about the presence and properties of cancer stem cells in the established and tumorigenic glioma cell lines D54, TJ905, U87, and U251 (109-111).

Altered Pathways in Glioma Stem Cells

It is difficult to find normal pathways in glioma stem cells. p53 regulates the proliferation of stem and progenitor cells in the brain (112). In p53-mutant neural stem cells, transcriptome analyses identified the dysregulation of several cell cycle regulators, in particular a pronounced downregulation of p21 expression, implicating p53 as a growth suppressor of glioma-initiating stem cells (113). Deregulation of additional processes that control cell cycle progression, such as the p16-CDK4-RB pathway and other cyclin-dependent kinase pathways may also promote the generation of brain cancer stem-like cells (114, 115). Subventricular zone stem cells often express the PDGF receptor, perhaps contributing to their peculiar ability to respond to PDGF (116). Activation of signaling pathways like this, which is often accompanied by Ras inactivation (117), has been implicated in neural stem cell transformation (116). Within the vertebrate central nervous system (CNS), the Sonic Hedgehog (Shh) pathway acts as a morphogen or mitogen that regulates the patterning, proliferation, and survival of neural stem cells (118, 119). Gli1 is a primary mediator of Shh signaling (120) that likely serves as a protective mechanism against premature mitosis in nonmalignant stem cells. Deregulation of Gli1 has been observed in brain tumor-derived stem cells (120). Aberrant expression of additional developmental pathways, such as WNT, Notch, and transforming growth factor-beta/BMP has been also reported (121, 122). In comparison with matched nonstem glioma cell populations, glioma stem cells consistently secrete markedly elevated levels of the angiogenic factor vascular endothelial growth factor (VEGF), which are further induced by hypoxia (123-126). The proangiogenic effects of stem cells on endothelial cells are specifically abolished by the anti-VEGF neutralizing antibody bevacizumab that also suppresses growth of xenografts derived from those cells (127, 128). A number of epigenetic injuries such as CpG island aberrant hypermethylation, histone modification, and altered chromatin states have been also reported in malignant glioma stem cells (129).

Resistance Mechanisms in Glioma Stem Cells

Gliomas frequently display hyperactivation of the phosphatidylinositol-3-kinase (PI3K)-Akt pathway, a protumorigenic signaling cascade that contributes to the pathogenesis of several human cancers (130-134). The PI3K-Akt pathway can be activated in tumors through different mechanisms, including inactivation of the inhibitory effects of the phosphatase and tensin homolog (PTEN) tumor suppressor (130-134). Hyperactive Akt signaling promotes tumorigenic cell behaviors by increasing cell survival, proliferation, invasion, and angiogenesis. Treatment with an Akt inhibitor potently reduces the numbers of viable glioma stem cells relative to matched nonstem cells (135). Further, delayed resolution of γ-H2AX foci after IR and radiosensitization have been observed in glioma cells after induction of PTEN and concomitant decrease of activated Akt (131). These results together hint at a link between Akt activation and radioresistance in glioma stem cells. Bao and coworkers (136) have shown that glioma stem cells may resist radiation through preferential activation of the DNA damage checkpoint response. The fraction of tumor cells expressing CD133 was enriched after radiation in gliomas. In both cell culture and the brains of immunocompromised mice, CD133+ glioma cells survived IR in increased proportions relative to most tumor cells, which lack CD133. CD133+ tumor cells isolated from both human glioma xenografts and primary patient glioblastoma specimens preferentially activated the DNA damage checkpoint in response to radiation and repaired IR-induced-DNA damage more effectively than CD133− tumor cells (136, 137). In addition, the radioresistance of CD133+ glioma stem cells could be reversed with a specific inhibitor of the Chk1 and Chk2 checkpoint kinases. In the mouse, G2 and G1-arrested neural stem cells rapidly increase after IR and this correlates with phosphorylation of cdc2 and p53, respectively, and inactivation of the Notch signal (138). Resistance of glioma stem cells to chemotherapeutic drugs has been further reported (139, 140). An expression signature dominated by HOX genes, which comprises CD133, emerged as a predictor for poor survival in glioma patients treated with concomitant chemo- and radiotherapy (141). Taken together, these results suggest that CD133+ tumor cells represent the cellular population that confers glioma resistance to chemo- and radiotherapy and that activation of the DNA damage checkpoint response may be a major mechanism in this regard. One further important resistance mechanism could be multidrug resistance. As aforementioned (see “Glioma Stem Cells”), a peculiarity of many stem cell populations (side populations) is their relatively high expression of ATP-binding cassette (ABC) drug transporters, which confers them the ability to efflux the fluorescent dye Hoechst 33342 and protection from cytotoxic agents (142). Side-population cells have been isolated in the CNS, and similar to hematological malignancies, expression of multidrug resistance-associated protein genes has been found elevated in cancer stem-like cells isolated from glioblastoma and astrocytoma (143-145). Finally, elevated expression of antiapoptic proteins [e.g., the myeloid cell leukemia-1 (Mcl-1) protein] can be found in glioma stem cells (144, 146) and reduced expression of major histocompatibility complex genes may also contribute to their resistance to therapies (147, 148).

The relative importance of DNA repair as a resistance mechanism in glioma stem cells still is an open question. We have recently examined DNA repair in five stem and nonstem glioma cell lines (Fig. 2; ref. 111). The stem lines fulfilled the defining criteria proposed by Vescovi and coworkers (142). The population doubling time was significantly increased in stem as compared with nonstem cells and enhanced activation of Chk1 and Chk2 kinases was observed in untreated CD133+ as compared with CD133− cells (Fig. 3). In this regard, cell-cycle restriction limits DNA damage and maintains self-renewal of leukemia stem cells as well (149). In our laboratory, neither DNA base excision, nor single strand break repair, nor resolution of pH2AX nuclear foci were increased in CD133+ as compared with CD133− glioma cells (Fig. 4). Hence, reduced proliferation may be a major mechanism in glioma stem cells to resist chemo- and radiotherapeutic-induced killing, whereas if enhanced DNA repair (per time unit) is a common resistance mechanism in glioma stem cells still requires further studies.

FIGURE 2.

Differentiation of glioma stem cells. Morphology of stem (COMI; DEMI; top) and nonstem (D54; U373; bottom) glioma cell lines. Cells were cultured in the presence of EGF-FGF (stem conditions) or fetal calf serum [(FCS) - differentiating conditions] for 21 days and their morphology photographed with an inverted microscope (Reprinted by permission from the American Association for Cancer Research: Mol Cancer Res 7:383–92, 2009).

FIGURE 2.

Differentiation of glioma stem cells. Morphology of stem (COMI; DEMI; top) and nonstem (D54; U373; bottom) glioma cell lines. Cells were cultured in the presence of EGF-FGF (stem conditions) or fetal calf serum [(FCS) - differentiating conditions] for 21 days and their morphology photographed with an inverted microscope (Reprinted by permission from the American Association for Cancer Research: Mol Cancer Res 7:383–92, 2009).

Close modal
FIGURE 3.

A. Population doubling times of stem (solid bars) and nonstem (open bars) cells. Appropriate numbers of cells were seeded and grown for 7 to 21 days. The total number of cells was then counted and after determination of the plating efficiency, the population doublings were calculated according to Glaab and Tyndall (157). The mean number ± standard error of the mean (SEM) of hours required to double the cell population is shown. Average values ± SEM on five stem and nonstem cell lines are indicated with gray bars at far right. The P < 0.01 statistical significance of the difference between nonstem and stem cells is indicated with two stars. B. Chk1 and Chk2 activation in CD133+ and CD133− cells. The phosphorylated state of the Chk1 (Ser345) and Chk2 (Ser19) checkpoint kinases was assessed in BORRU CD133+ and CD133− cells without (−) and 1 hour after IR treatment (+). Whole cell lysates were prepared, resolved by SDS-PAGE, and immunoblotted with primary rabbit polyclonal antibodies raised against phospho-Chk1 (Ser345) and phospho-Chk2 (Ser19). Immunoblotting for actin served as a loading control (Reprinted by permission from the American Association for Cancer Research: Mol Cancer Res 7:383–92, 2009).

FIGURE 3.

A. Population doubling times of stem (solid bars) and nonstem (open bars) cells. Appropriate numbers of cells were seeded and grown for 7 to 21 days. The total number of cells was then counted and after determination of the plating efficiency, the population doublings were calculated according to Glaab and Tyndall (157). The mean number ± standard error of the mean (SEM) of hours required to double the cell population is shown. Average values ± SEM on five stem and nonstem cell lines are indicated with gray bars at far right. The P < 0.01 statistical significance of the difference between nonstem and stem cells is indicated with two stars. B. Chk1 and Chk2 activation in CD133+ and CD133− cells. The phosphorylated state of the Chk1 (Ser345) and Chk2 (Ser19) checkpoint kinases was assessed in BORRU CD133+ and CD133− cells without (−) and 1 hour after IR treatment (+). Whole cell lysates were prepared, resolved by SDS-PAGE, and immunoblotted with primary rabbit polyclonal antibodies raised against phospho-Chk1 (Ser345) and phospho-Chk2 (Ser19). Immunoblotting for actin served as a loading control (Reprinted by permission from the American Association for Cancer Research: Mol Cancer Res 7:383–92, 2009).

Close modal
FIGURE 4.

Contrasting results on enhanced DNA strand break repair in CD133+ cells. A. CD133+ and CD133− cells derived from primary glioblastoma T3539 were irradiated with 3 Gy of IR. The presence of DNA damage at sequential time points after damage was assessed by single-cell gel electrophoresis assay under alkaline conditions (alkaline comet assay). The histogram quantitates the percentages of cells with comet tails at different time points after IR in CD133+ (open bars) and CD133− (solid bars) populations. Data are the means ± standard deviation (SD; *, P = 0.001; **, P = 0.002; reprinted by permission from Macmillan Publishers Ltd: Nature 444:756–60, 2006). B. CD133+ and CD133− derived from primary glioblastoma COMI were separated and plated. Sixteen hours later adherent cells were irradiated with 3 Gy IR and DNA strand break repair assessed by measuring the percentages of DNA in comets' tails under alkaline conditions. The histogram quantitates the percentages of DNA in comet tails at different time points after IR in CD133+ (open bars) and CD133− (solid bars) populations. Data are the means ± SEM (Reprinted by permission from the American Association for Cancer Research: Mol Cancer Res 7:383–92, 2009).

FIGURE 4.

Contrasting results on enhanced DNA strand break repair in CD133+ cells. A. CD133+ and CD133− cells derived from primary glioblastoma T3539 were irradiated with 3 Gy of IR. The presence of DNA damage at sequential time points after damage was assessed by single-cell gel electrophoresis assay under alkaline conditions (alkaline comet assay). The histogram quantitates the percentages of cells with comet tails at different time points after IR in CD133+ (open bars) and CD133− (solid bars) populations. Data are the means ± standard deviation (SD; *, P = 0.001; **, P = 0.002; reprinted by permission from Macmillan Publishers Ltd: Nature 444:756–60, 2006). B. CD133+ and CD133− derived from primary glioblastoma COMI were separated and plated. Sixteen hours later adherent cells were irradiated with 3 Gy IR and DNA strand break repair assessed by measuring the percentages of DNA in comets' tails under alkaline conditions. The histogram quantitates the percentages of DNA in comet tails at different time points after IR in CD133+ (open bars) and CD133− (solid bars) populations. Data are the means ± SEM (Reprinted by permission from the American Association for Cancer Research: Mol Cancer Res 7:383–92, 2009).

Close modal

Targeting Cell Cycle Restriction in Glioma Stem Cells

The ATM (ataxia-telangiectasia mutated) and ATR (ATM and Rad3-related) kinases are transducers that recognize DNA damage and coordinate the initiation, amplification, and activation of the checkpoint through phosphorylation of many different targets (Fig. 5) (reviewed in refs. 150, 151). Checkpoint kinase-1 (Chk1) and checkpoint kinase-2 (Chk2) are the checkpoint transducer kinases that function downstream of ATR and ATM in the DNA-damage checkpoint signaling pathway. Although structurally dissimilar, Chk1 and Chk2 are serine- threonine kinases that serve as functional analogs. ATM phosphorylates Chk2 at various sites including serine 19 and threonine 68, whereas ATR phosphorylates Chk1 at serines 280, 296, 317, and 345. Significant crosstalk exists between the ATM/Chk2 and ATR/Chk1 pathways (152). Together, the proximal transducers ATM and ATR and the distal transducers Chk1 and Chk2 phosphorylate a variety of effector molecules, such as p53 and CDC25 phosphatases, culminating in cell cycle arrest. In particular, the three CDC25 isoforms, A, B, and C, are active in different phases of the cell cycle. CDC25 phosphatases remove inhibitory phosphate groups from cyclin-cyclin-dependent kinase (CDK) complexes, promoting cell cycle progression. In response to DNA damage, the checkpoint kinases phosphorylate CDC25 phosphatases, resulting in CDC25 inactivation through either ubiquitin-mediated degradation or cytoplasmic sequestration. In this manner, the checkpoint kinases serve as negative regulators of the CDC25 phosphatases.

FIGURE 5.

Cell cycle checkpoint pathways, possible targets in glioma stem cells. Once DNA damage is identified with the aid of sensors, the checkpoint transducers ATM and ATR undergo conformational change and or localization, resulting in their activation. ATM and ATR activate a series of downstream molecules, including the checkpoint kinases 1 (A, Chk1) and 2 (B, Chk2). Chk1 and Chk2 inactivate CDC25 phosphatases, culminating in cell cycle arrest. AZD7762 (AstraZeneca) is a specific inhibitor of Chk1 and Chk2 kinases. CP466722 (Pfizer) is a specific inhibitor of ATM.

FIGURE 5.

Cell cycle checkpoint pathways, possible targets in glioma stem cells. Once DNA damage is identified with the aid of sensors, the checkpoint transducers ATM and ATR undergo conformational change and or localization, resulting in their activation. ATM and ATR activate a series of downstream molecules, including the checkpoint kinases 1 (A, Chk1) and 2 (B, Chk2). Chk1 and Chk2 inactivate CDC25 phosphatases, culminating in cell cycle arrest. AZD7762 (AstraZeneca) is a specific inhibitor of Chk1 and Chk2 kinases. CP466722 (Pfizer) is a specific inhibitor of ATM.

Close modal

Treatment with ATM and Chk1 and Chk2 inhibitors used alone or in combination may selectively sensitize glioma stem cells to IR and temozolomide (reviewed in refs. 153, 154). In this regard, two interesting novel drugs have been recently developed. The preclinical profile of AZD7762, a potent ATP-competitive checkpoint kinase inhibitor in clinical trials was described by Zabludoff and coworkers (155). AZD7762 was profiled extensively in vitro and in vivo in combination with DNA-damaging agents and was shown to potentiate response in several different settings in which inhibition of checkpoint kinase resulted in the abrogation of DNA damage-induced cell cycle arrest. Dose-dependent potentiation of antitumor activity, when AZD7762 was administered in combination with DNA-damaging agents, was observed in multiple xenograft models with several DNA-damaging agents, further supporting the potential of checkpoint kinase inhibitors to enhance the efficacy of both conventional chemotherapy and radiotherapy and increase patient response rates.

Rainey and coworkers (156) screened a targeted compound library for inhibitors of the ATM kinase, and CP466722 was identified. The compound was nontoxic and did not inhibit PI3K or PI3K-like protein kinase family members in cells. CP466722 inhibited cellular ATM-dependent phosphorylation events and disruption of ATM function resulted in characteristic cell cycle checkpoint defects. Inhibition of cellular ATM kinase activity was rapidly and completely reversed by removing CP466722. Clonogenic survival assays done on HeLa or AT GM02052 cells showed that transient inhibition of ATM was sufficient to sensitize cells to IR and suggested that therapeutic radiosensitization may only require ATM inhibition for short periods of time. The ability of AZD7762 and CP466722 to rapidly and reversibly regulate Chk1, Chk2, and ATM activities thus provides new tools to inhibit constitutive activation of cell cycle checkpoints in CD133+ glioma stem cells, reverse their cell cycle block, and sensitize them to IR and chemotherapy.

In 31 out of 40 (77%) studies on bulk high-grade gliomas, an inverse correlation between DNA repair capacity and therapeutic efficacy has been observed, indicating that nonstem cells in these tumors may frequently adopt DNA repair as a resistance mechanism (Table 1). However, currently available therapies for gliomas seldom are curative, possibly for their inability to eradicate a small subpopulation of highly resistant cells that almost invariably cause tumor relapse (called with some imprecision “glioma stem cells”). Low proliferation rate and constitutive activation of the DNA damage checkpoint response may be major mechanisms of resistance in these cells, conferring increased time for lesion removal or bypass. Those features may be common to other tumors as well (149). Drugs targeting cell cycle restriction in glioma stem cells could be of help for complete eradication of the tumor and several novel agents of this kind are under development. In particular ATM and Chk1 and Chk2 kinase inhibitors may effectively sensitize glioma stem cells to IR and alkylating agents by reversing their cell cycle delay. Treating checkpoint activation, rather than DNA repair, might be a fruitful research direction in improving glioma therapies.

No potential conflicts of interest were disclosed.

1
Furnari
FB
,
Fenton
T
,
Bachoo
RM
, et al
. 
Malignant astrocytic glioma: genetics, biology, and paths to treatment
.
Genes Dev
2007
;
21
:
2683
710
.
2
Ohgaki
H
,
Kleihues
P
. 
Genetic pathways to primary and secondary glioblastoma
.
Am J Pathol
2007
;
170
:
1445
53
.
3
Stupp
R
,
Hegi
ME
,
van den Bent
MJ
, et al
. 
Changing paradigms - an update of the multidisciplinary management of malignant glioma
.
Oncologist
2006
;
11
:
165
80
.
4
Mutter
N
,
Stupp
R
. 
Temozolomide: a milestone in neuro-oncology and beyond?
Expert Rev Anticancer Ther
2006
;
6
:
1187
204
.
5
Levin
N
,
Lavon
I
,
Zelikovitsh
B
, et al
. 
Progressive low-grade oligodendrogliomas: response to temozolomide and correlation between genetic profile and O6-methylguanine DNA methyltransferase protein expression
.
Cancer
2006
;
106
:
1759
65
.
6
Tosoni
A
,
Cavallo
G
,
Ermani
M
, et al
. 
Is protracted low-dose temozolomide feasible in glioma patients?
Neurology
2006
;
66
:
427
9
.
7
Helleday
T
,
Petermann
E
,
Lundin
C
,
Hodgson
B
,
Sharma
RA
. 
DNA repair pathways as targets for cancer therapy
.
Nat Rev Cancer
2008
;
8
:
193
204
.
8
Kelley
MR
,
Fishel
ML
. 
DNA repair proteins as molecular targets for cancer therapeutics
.
Anticancer Agents Med Chem
2008
;
8
:
417
25
.
9
Parkinson
JF
,
Wheeler
HT
,
McDonald
KL
. 
Contribution of DNA repair mechanisms to determining chemotherapy response in high-grade glioma
.
J Clin Neurosci
2008
;
15
:
1
8
.
10
Sarkaria
JN
,
Kitange
GJ
,
James
CD
, et al
. 
Mechanisms of chemoresistance to alkylating agents in malignant glioma
.
Clin Cancer Res
2008
;
14
:
2900
8
.
11
Hegi
ME
,
Diserens
AC
,
Gorlia
T
, et al
. 
MGMT gene silencing and benefit from temozolomide in glioblastoma
.
N Engl J Med
2005
;
352
:
997
1003
.
12
Blanc
JL
,
Wager
M
,
Guilhot
J
, et al
. 
Correlation of clinical features and methylation status of MGMT gene promoter in glioblastomas
.
J Neurooncol
2004
;
68
:
275
83
.
13
Hermisson
M
,
Klumpp
A
,
Wick
W
, et al
. 
O6-methylguanine DNA methyltransferase and p53 status predict temozolomide sensitivity in human malignant glioma cells
.
J Neurochem
2006
;
96
:
766
76
.
14
Hau
P
,
Stupp
R
,
Hegi
ME
. 
MGMT methylation status: the advent of stratified therapy in glioblastoma?
Dis Markers
2007
;
23
:
97
104
.
15
Martinez
R
,
Schackert
G
,
Yaya-Tur
R
,
Rojas-Marcos
I
,
Herman
JG
,
Esteller
M
. 
Frequent hypermethylation of the DNA repair gene MGMT in long-term survivors of glioblastoma multiforme
.
J Neurooncol
2007
;
83
:
91
3
.
16
Stupp
R
,
Hegi
ME
,
Gilbert
MR
,
Chakravarti
A
. 
Chemoradiotherapy in malignant glioma: standard of care and future directions
.
J Clin Oncol
2007
;
25
:
4127
36
.
17
Donson
AM
,
Addo-Yobo
SO
,
Handler
MH
,
Gore
L
,
Foreman
NK
. 
MGMT promoter methylation correlates with survival benefit and sensitivity to temozolomide in pediatric glioblastoma
.
Pediatr Blood Cancer
2007
;
48
:
403
7
.
18
Eoli
M
,
Menghi
F
,
Bruzzone
MG
, et al
. 
Methylation of O6-methylguanine DNA methyltransferase and loss of heterozygosity on 19q and/or 17p are overlapping features of secondary glioblastomas with prolonged survival
.
Clin Cancer Res
2007
;
13
:
2606
13
.
19
Brandes
AA
,
Franceschi
E
,
Tosoni
A
, et al
. 
MGMT promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant radiochemotherapy in newly diagnosed glioblastoma patients
.
J Clin Oncol
2008
;
26
:
2192
7
.
20
Chakravarti
A
,
Erkinnen
MG
,
Nestler
U
, et al
. 
Temozolomide-mediated radiation enhancement in glioblastoma: a report on underlying mechanisms
.
Clin Cancer Res
2006
;
12
:
4738
46
.
21
Vlassenbroeck
I
,
Califice
S
,
Diserens
AC
, et al
. 
Validation of real-time methylation-specific PCR to determine O6-methylguanine-DNA methyltransferase gene promoter methylation in glioma
.
J Mol Diagn
2008
;
10
:
332
7
.
22
Grasbon-Frodl
EM
,
Kreth
FW
,
Ruiter
M
, et al
. 
Intratumoral homogeneity of MGMT promoter hypermethylation as demonstrated in serial stereotactic specimens from anaplastic astrocytomas and glioblastomas
.
Int J Cancer
2007
;
121
:
2458
64
.
23
Koch
D
,
Hundsberger
T
,
Boor
S
,
Kaina
B
. 
Local intracerebral administration of O(6) benzylguanine combined with systemic chemotherapy with temozolomide of a patient suffering from a recurrent glioblastoma
.
J Neurooncol
2007
;
82
:
85
9
.
24
Khan
O
,
Middleton
MR
. 
The therapeutic potential of O6-alkylguanine DNA alkyltransferase inhibitors
.
Expert Opin Investig Drugs
2007
;
16
:
1573
84
.
25
Ribacka
C
,
Hemminki
A
. 
Virotherapy as an approach against cancer stem cells
.
Curr Gene Ther
2008
;
8
:
88
96
.
26
Alonso
MM
,
Gomez-Manzano
C
,
Bekele
BN
,
Yung
WK
,
Fueyo
J
. 
Adenovirus-based strategies overcome temozolomide resistance by silencing the O6-methylguanine-DNA methyltransferase promoter
.
Cancer Res
2007
;
67
:
11499
504
.
27
Eckert
A
,
Kloor
M
,
Giersch
A
, et al
. 
Microsatellite instability in pediatric and adult high-grade gliomas
.
Brain Pathol
2007
;
17
:
146
50
.
28
Vladimirova
V
,
Denkhaus
D
,
Soerensen
N
,
Wagner
S
,
Wolff
JE
,
Pietsch
T
. 
Low level of microsatellite instability in paediatric malignant astrocytomas
.
Neuropathol Appl Neurobiol
2008
;
34
:
547
54
.
29
Hunter
C
,
Smith
R
,
Cahill
DP
, et al
. 
A hypermutation phenotype and somatic MSH6 mutations in recurrent human malignant gliomas after alkylator chemotherapy
.
Cancer Res
2006
;
66
:
3987
91
.
30
Cahill
DP
,
Levine
KK
,
Betensky
RA
, et al
. 
Loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor progression during temozolomide treatment
.
Clin Cancer Res
2007
;
13
:
2038
45
.
31
Maxwell
JA
,
Johnson
SP
,
McLendon
RE
, et al
. 
Mismatch repair deficiency does not mediate clinical resistance to temozolomide in malignant glioma
.
Clin Cancer Res
2008
;
14
:
4859
68
.
32
Gömöri
E
,
Pál
J
,
Mészáros
I
,
Dóczi
T
,
Matolcsy
A
. 
Epigenetic inactivation of the hMLH1 gene in progression of gliomas
.
Diagn Mol Pathol
2007
;
16
:
104
7
.
33
Casorelli
I
,
Russo
MT
,
Bignami
M
. 
Role of mismatch repair and MGMT in response to anticancer therapies
.
Anticancer Agents Med Chem
2008
;
8
:
368
80
.
34
Bodell
WJ
,
Gaikwad
NW
,
Miller
D
,
Berger
MS
. 
Formation of DNA adducts and induction of lacI mutations in Big Blue Rat-2 cells treated with temozolomide: implications for the treatment of low-grade adult and pediatric brain tumors
.
Cancer Epidemiol Biomarkers Prev
2003
;
12
:
545
51
.
35
Trivedi
RN
,
Almeida
KH
,
Fornsaglio
JL
,
Schamus
S
,
Sobol
RW
. 
The role of base excision repair in the sensitivity and resistance to temozolomide-mediated cell death
.
Cancer Res
2005
;
65
:
6394
400
.
36
Trivedi
RN
,
Wang
XH
,
Jelezcova
E
,
Goellner
EM
,
Tang
J
,
Sobol
RW
. 
Human methyl purine DNA glycosylase and DNA polymerase {beta} expression collectively predict sensitivity to temozolomide
.
Mol Pharmacol
2008
;
74
:
505
16
.
37
Bobola
MS
,
Blank
A
,
Berger
MS
,
Stevens
BA
,
Silber
JR
. 
Apurinic/apyrimidinic endonuclease activity is elevated in human adult gliomas
.
Clin Cancer Res
2001
;
7
:
3510
8
.
38
Silber
JR
,
Bobola
MS
,
Blank
A
, et al
. 
The Apurinic/Apyrimidinic endonuclease activity of Ape/Ref-1 contributes to human glioma cell resistance to alkylating agents and is elevated by oxidative stress
.
Clin Cancer Res
2002
;
8
:
3008
18
.
39
Bobola
MS
,
Emond
MJ
,
Blank
A
, et al
. 
Apurinic endonuclease activity in adult gliomas and time to tumor progression after alkylating agent-based chemotherapy and after radiotherapy
.
Clin Cancer Res
2004
;
10
:
7875
83
.
40
Bobola
MS
,
Finn
LS
,
Ellenbogen
RG
, et al
. 
Apurinic/apyrimidinic endonuclease activity is associated with response to radiation and chemotherapy in medulloblastoma and primitive neuroectodermal tumors
.
Clin Cancer Res
2005
;
11
:
7405
14
.
41
Adhikari
S
,
Choudhury
S
,
Mitra
PS
,
Dubash
JJ
,
Sajankila
SP
,
Roy
R
. 
Targeting base excision repair for chemosensitization
.
Anticancer Agents Med Chem
2008
;
8
:
351
7
.
42
Bobola
MS
,
Varadarajan
S
,
Smith
NW
, et al
. 
Human glioma cell sensitivity to the sequence-specific alkylating agent methyl-lexitropsin
.
Clin Cancer Res
2007
;
13
:
612
20
.
43
Tentori
L
,
Portarena
I
,
Torino
F
,
Scerrati
M
,
Navarra
P
,
Graziani
G
. 
Poly(ADP-ribose) polymerase inhibitor increases growth inhibition and reduces G(2)/M cell accumulation induced by temozolomide in malignant glioma cells
.
Glia
2002
;
40
:
44
54
.
44
Curtin
NJ
,
Wang
LZ
,
Yiakouvaki
A
, et al
. 
Novel poly(ADP-ribose) polymerase-1 inhibitor, AG14361, restores sensitivity to temozolomide in mismatch repair-deficient cells
.
Clin Cancer Res
2004
;
10
:
881
9
.
45
Russell
J.S.
,
Brady
K.
,
Burgan
W.E.
, et al
. 
Gleevec-mediated inhibition of Rad51 expression and enhancement of tumor cell radiosensitivity
.
Cancer Res
2003
;
63
:
7377
83
.
46
Short
SC
,
Martindale
C
,
Bourne
S
,
Brand
G
,
Woodcock
M
,
Johnston
P
. 
DNA repair after irradiation in glioma cells and normal human astrocytes
.
Neuro-oncol
2007
;
9
:
404
11
.
47
Kil
WJ
,
Cerna
D
,
Burgan
WE
, et al
. 
In vitro and in vivo radiosensitization induced by the DNA methylating agent temozolomide
.
Clin Cancer Res
2008
;
14
:
931
8
.
48
Bencokova
Z
,
Pauron
L
,
Devic
C
, et al
. 
Molecular and cellular response of the most extensively used rodent glioma models to radiation and/or cisplatin
.
J Neurooncol
2008
;
86
:
13
21
.
49
Fischer
U
,
Meese
E
. 
Glioblastoma multiforme: the role of DSB repair between genotype and phenotype
.
Oncogene
2007
;
26
:
7809
15
.
50
Shao
CJ
,
Fu
J
,
Shi
HL
,
Mu
YG
,
Chen
ZP
. 
Activities of DNA-PK and Ku86, but not Ku70, may predict sensitivity to cisplatin in human gliomas
.
J Neurooncol
2008
;
89
:
27
35
.
51
Chen
CC
,
Taniguchi
T
,
D'Andrea
A
. 
The Fanconi anemia (FA) pathway confers glioma resistance to DNA alkylating agents
.
J Mol Med
2007
;
85
:
497
509
.
52
Chiu
J
,
Khan
ZA
,
Farhangkhoee
H
,
Chakrabarti
S
. 
Curcumin prevents diabetes-associated abnormalities in the kidneys by inhibiting p300 and nuclear factor-kappaB
.
Nutrition
2009
;,
Epub 2009 Mar 4
.
53
Iqbal
M
,
Okazaki
Y
,
Okada
S
. 
Curcumin attenuates oxidative damage in animals treated with a renal carcinogen, ferric nitrilotriacetate (Fe-NTA): implications for cancer prevention
.
Mol Cell Biochem
2009
;
324
:
157
64
.
54
Dhandapani
KM
,
Mahesh
VB
,
Brann
DW
. 
Curcumin suppresses growth and chemoresistance of human glioblastoma cells via AP-1 and NFkappaB transcription factors
.
J Neurochem
2007
;
102
:
522
38
.
55
Batista
LF
,
Roos
WP
,
Christmann
M
,
Menck
CF
,
Kaina
B
. 
Differential sensitivity of malignant glioma cells to methylating and chloroethylating anticancer drugs: p53 determines the switch by regulating xpc, ddb2, and DNA double-strand breaks
.
Cancer Res
2007
;
67
:
11886
95
.
56
Ianzini
F
,
Domann
FE
,
Kosmacek
EA
,
Phillips
SL
,
Mackey
MA
. 
Human glioblastoma U87MG cells transduced with a dominant negative p53 (TP53) adenovirus construct undergo radiation-induced mitotic catastrophe
.
Radiat Res
2007
;
168
:
183
92
.
57
Harrison
JF
,
Rinne
ML
,
Kelley
MR
,
Druzhyna
NM
,
Wilson
GL
,
Ledoux
SP
. 
Altering DNA base excision repair: use of nuclear and mitochondrial-targeted N-methylpurine DNA glycosylase to sensitize astroglia to chemotherapeutic agents
.
Glia
2007
;
55
:
1416
25
.
58
Lu
C
,
Shervington
A
. 
Chemoresistance in gliomas
.
Mol Cell Biochem
2008
;
312
:
71
80
.
59
Martinez
R
,
Setien
F
,
Voelter
C
, et al
. 
CpG island promoter hypermethylation of the pro-apoptotic gene caspase-8 is a common hallmark of relapsed glioblastoma multiforme
.
Carcinogenesis
2007
;
28
:
1264
8
.
60
Auger
N
,
Thillet
J
,
Wanherdrick
K
, et al
. 
Genetic alterations associated with acquired temozolomide resistance in SNB-19, a human glioma cell line
.
Mol Cancer Ther
2006
;
5
:
2182
92
.
61
Rosenblum
ML
,
Gerosa
M
,
Dougherty
DV
, et al
. 
Age-related chemosensitivity of stem cells from human malignant brain tumors
.
Lancet
1982
;
1
:
885
7
.
62
Barami
K
. 
Biology of the subventricular zone in relation to gliomagenesis
.
J Clin Neurosci
2007
;
14
:
1143
9
.
63
Dirks
PB
. 
Brain tumor stem cells: bringing order to the chaos of brain cancer
.
J Clin Oncol
2008
;
26
:
2916
24
.
64
Xie
Z
,
Chin
LS
. 
Molecular and cell biology of brain tumor stem cells: lessons from neural progenitor/stem cells
.
Neurosurg Focus
2008
;
24
:
E25
.
65
Das
S
,
Srikanth
M
,
Kessler
JA
. 
Cancer stem cells and glioma
.
Nat Clin Pract Neurol
2008
;
4
:
427
35
.
66
Ma
YH
,
Mentlein
R
,
Knerlich
F
,
Kruse
ML
,
Mehdorn
HM
,
Held-Feindt
J
. 
Expression of stem cell markers in human astrocytomas of different WHO grades
.
J Neurooncol
2008
;
86
:
31
45
.
67
Colleoni
F
,
Torrente
Y
. 
The new challenge of stem cell: Brain tumour therapy
.
Cancer Lett
2008
;
272
:
1
11
.
68
Strojnik
T
,
Rosland
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
43
.
69
Maderna
E
,
Salmaggi
A
,
Calatozzolo
C
,
Limido
L
,
Pollo
B
. 
Nestin, PDGFRbeta, CXCL12 and VEGF in glioma patients: different profiles of (pro-angiogenic) molecule expression are related with tumor grade and may provide prognostic information
.
Cancer Biol Ther
2007
;
6
:
1018
24
.
70
Kong
DS
,
Kim
MH
,
Park
WY
, et al
. 
The progression of gliomas is associated with cancer stem cell phenotype
.
Oncol Rep
2008
;
19
:
639
43
.
71
Beier
D
,
Wischhusen
J
,
Dietmaier
W
, et al
. 
CD133 expression and cancer stem cells predict prognosis in high-grade oligodendroglial tumors
.
Brain Pathol
2008
;
18
:
370
7
.
72
Christensen
K
,
Schrøder
HD
,
Kristensen
BW
. 
CD133 identifies perivascular niches in grade II-IV astrocytomas
.
J Neurooncol
2008
;
90
:
157
70
.
73
Rebetz
J
,
Tian
D
,
Persson
A
, et al
. 
Glial progenitor-like phenotype in low-grade glioma and enhanced CD133-expression and neuronal lineage differentiation potential in high-grade glioma
.
PLoS ONE
2008
;
3
:
e1936
.
74
Zeppernick
F
,
Ahmadi
R
,
Campos
B
, et al
. 
Stem cell marker CD133 affects clinical outcome in glioma patients
.
Clin Cancer Res
2008
;
14
:
123
9
.
75
Thon
N
,
Damianoff
K
,
Hegermann
J
, et al
. 
Presence of pluripotent CD133(+) cells correlates with malignancy of gliomas
.
Mol Cell Neurosci
2008
.
Epub 2008 Aug 7
.
76
Shu
Q
,
Wong
KK
,
Su
JM
, et al
. 
Direct orthotopic transplantation of fresh surgical specimen preserves CD133+ tumor cells in clinically relevant mouse models of medulloblastoma and glioma
.
Stem Cells
2008
;
26
:
1414
24
.
77
Shen
G
,
Shen
F
,
Shi
Z
, et al
. 
Identification of cancer stem-like cells in the C6 glioma cell line and the limitation of current identification methods
.
In Vitro Cell Dev Biol Anim
2008
;
44
:
280
9
.
78
Beier
D
,
Hau
P
,
Proescholdt
M
, et al
. 
CD133(+) and CD133(−) glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles
.
Cancer Res
2007
;
67
:
4010
5
.
79
Günther
HS
,
Schmidt
NO
,
Phillips
HS
, et al
. 
Glioblastoma-derived stem cell-enriched cultures form distinct subgroups according to molecular and phenotypic criteria
.
Oncogene
2008
;
27
:
2897
909
.
80
Bidlingmaier
S
,
Zhu
X
,
Liu
B
. 
The utility and limitations of glycosylated human CD133 epitopes in defining cancer stem cells
.
J Mol Med
2008
;
86
:
1025
32
.
81
Joo
KM
,
Kim
SY
,
Jin
X
, et al
. 
Clinical and biological implications of CD133-positive and CD133-negative cells in glioblastomas
.
Lab Invest
2008
;
88
:
808
15
.
82
Yao
Y
,
Wang
X
,
Jin
K
, et al
. 
B7–4 is preferentially expressed in non-dividing brain tumor cells and in a subset of brain tumor stem-like cells
.
J Neurooncol
2008
;
89
:
121
9
.
83
Kang
MK
,
Kang
SK
. 
Tumorigenesis of chemotherapeutic drug-resistant cancer stem-like cells in brain glioma
.
Stem Cells Dev
2007
;
16
:
837
48
.
84
Kang
MK
,
Her
BI
,
Ko
MH
,
Kim
CH
,
Cha
SH
,
Kang
SK
. 
Potential identity of multi-potential cancer stem-like subpopulation after radiation of cultured brain glioma
.
BMC Neurosci
2008
;
9
:
15
.
85
Ogden
AT
,
Waziri
AE
,
Lochhead
RA
, et al
. 
Identification of A2B5+CD133- tumor-initiating cells in adult human gliomas
.
Neurosurgery
2008
;
62
:
505
14
.
86
Fan
X
,
Salford
LG
,
Widegren
B
. 
Glioma stem cells: evidence and limitation
.
Semin Cancer Biol
2007
;
17
:
214
8
.
87
Dietrich
J
,
Imitola
J
,
Kesari
S
. 
Mechanisms of disease: the role of stem cells in the biology and treatment of gliomas
.
Nat Clin Pract Oncol
2008
;
5
:
393
404
.
88
Tabatabai
G
,
Bahr
O
,
Mohle
R
, et al
. 
Lessons from bone marrow: how malignant glioma cells attract adult haematopoietic progenitor cells
.
Brain
2005
;
128
:
2200
11
.
89
Serfozo
P
,
Schlarman
MS
,
Pierret
C
,
Maria
BL
,
Kirk
MD
. 
Selective migration of neuralized embryonic stem cells to stem cell factor and media conditioned by glioma cell lines
.
Cancer Cell Int
2006
;
6
:
1
.
90
Gutova
M
,
Najbauer
J
,
Frank
RT
, et al
. 
uPA and uPAR mediate human stem cell tropism to malignant solid tumors
.
Stem Cells
2008
;
26
:
1406
13
.
91
Jeon
JY
,
An
JH
,
Kim
SU
,
Park
HG
,
Lee
MA
. 
Migration of human neural stem cells toward an intracranial glioma
.
Exp Mol Med
2008
;
40
:
84
91
.
92
Jurvansuu
J
,
Zhao
Y
,
Leung
DS
, et al
. 
Transmembrane protein 18 enhances the tropism of neural stem cells for glioma cells
.
Cancer Res
2008
;
68
:
4614
22
.
93
Kendall
SE
,
Najbauer
J
,
Johnston
HF
, et al
. 
Neural stem cell targeting of glioma is dependent on PI3K signaling
.
Stem Cells
2008
;
26
:
1575
86
.
94
Xu
F
,
Zhu
JH
. 
Stem cells tropism for malignant gliomas
.
Neurosci Bull
2007
;
23
:
363
9
.
95
Li
XB
,
Guan
YQ
,
Ren
P
, et al
. 
The tropism of embryoid body cells for glioma cells
.
Neuroreport
2007
;
18
:
1821
5
.
96
Liu
JM
,
Mao
BY
,
Hong
S
,
Liu
YH
,
Wang
XJ
. 
The postoperative brain tumour stem cell (BTSC) niche and cancer recurrence
.
Adv Ther
2008
;
25
:
389
98
.
97
Tang
C
,
Ang
BT
,
Pervaiz
S
. 
Cancer stem cell: target for anti-cancer therapy
.
FASEB J
2007
;
21
:
3777
85
.
98
Sauvageot
CM
,
Kesari
S
,
Stiles
CD
. 
Molecular pathogenesis of adult brain tumors and the role of stem cells
.
Neurol Clin
2007
;
25
:
891
924
.
99
Lee da
Y
,
Gutmann
DH
. 
Cancer stem cells and brain tumors: uprooting the bad seeds
.
Expert Rev Anticancer Ther
2007
;
7
:
1581
90
.
100
Rajan
P
,
Srinivasan
R
. 
Targeting cancer stem cells in cancer prevention and therapy
.
Stem Cell Rev
2008
;
4
:
211
6
.
101
Rubin
LL
. 
Stem cells and drug discovery: the beginning of a new era?
Cell
2008
;
132
:
549
52
.
102
Yang
YM
,
Chang
JW
. 
Current status and issues in cancer stem cell study
.
Cancer Invest
2008
;
5
:
1
15
.
103
Zhao
RC
,
Zhu
YS
,
Shi
Y
. 
New hope for cancer treatment: Exploring the distinction between normal adult stem cells and cancer stem cells
.
Pharmacol Ther
2008
;
119
:
74
82
.
104
Setoguchi
T
,
Taga
T
,
Kondo
T
. 
Cancer stem cells persist in many cancer cell lines
.
Cell Cycle
2004
;
3
:
414
5
.
105
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
.
106
Kondo
T
. 
Brain cancer stem-like cells
.
Eur J Cancer
2006
;
42
:
1237
42
.
107
Kondo
T
. 
Stem cell-like cancer cells in cancer cell lines
.
Cancer Biomark
2007
;
3
:
245
50
.
108
Rappa
G
,
Mercapide
J
,
Anzanello
F
, et al
. 
Growth of cancer cell lines under stem cell-like conditions has the potential to unveil therapeutic targets
.
Exp Cell Res
2008
;
314
:
2110
22
.
109
Jin
F
,
Zhao
L
,
Zhao
HY
, et al
. 
Paradoxical expression of anti-apoptotic and MRP genes on cancer stem-like cell isolated from TJ905 glioblastoma multiforme cell line
.
Cancer Invest
2008
;
26
:
338
43
.
110
Yu
SC
,
Ping
YF
,
Yi
L
, et al
. 
Isolation and characterization of cancer stem cells from a human glioblastoma cell line U87
.
Cancer Lett
2008
;
265
:
124
34
.
111
Ropolo
M
,
Daga
A
,
Griffero
F
, et al
. 
Comparative analysis of DNA repair in stem and non-stem glioma cell cultures
.
Mol Cancer Res
2009
;
7
:
383
92
.
112
Medrano
S
,
Burns-Cusato
M
,
Atienza
MB
,
Rahimi
D
,
Scrable
H
. 
Regenerative capacity of neural precursors in the adult mammalian brain is under the control of p53
.
Neurobiol Aging
2009
;
30
:
483
97
.
113
Meletis
K
,
Wirta
V
,
Hede
SM
,
Nistér
M
,
Lundeberg
J
,
Frisén
J
. 
p53 suppresses the self-renewal of adult neural stem cells
.
Development
2006
;
133
:
363
9
.
114
Hulleman
E
,
Helin
K
. 
Molecular mechanisms in gliomagenesis
.
Adv Cancer Res
2005
;
94
:
1
27
.
115
Boyer
MJ
,
Cheng
T
. 
The CDK inhibitors: potential targets for therapeutic stem cell manipulations?
Gene Ther
2008
;
15
:
117
25
.
116
Jackson
EL
,
Alvarez-Buylla
A
. 
Characterization of adult neural stem cells and their relation to brain tumors
.
Cells Tissues Organs
2008
;
188
:
212
24
.
117
Lee
JS
,
Gil
JE
,
Kim
JH
, et al
. 
Brain cancer stem-like cell genesis from p53-deficient mouse astrocytes by oncogenic Ras
.
Biochem Biophys Res Commun
2008
;
365
:
496
502
.
118
Dellovade
T
,
Romer
JT
,
Curran
T
,
Rubin
LL
. 
The hedgehog pathway and neurological disorders
.
Annu Rev Neurosci
2006
;
29
:
539
63
.
119
Cai
C
,
Thorne
J
,
Grabel
L
. 
Hedgehog serves as a mitogen and survival factor during embryonic stem cell neurogenesis
.
Stem Cells
2008
;
26
:
1097
108
.
120
Galvin
KE
,
Ye
H
,
Erstad
DJ
,
Feddersen
R
,
Wetmore
C
. 
Gli1 induces G2/M arrest and apoptosis in hippocampal but not tumor-derived neural stem cells
.
Stem Cells
2008
;
26
:
1027
36
.
121
Clark
PA
,
Treisman
DM
,
Ebben
J
,
Kuo
JS
. 
Developmental signaling pathways in brain tumor-derived stem-like cells
.
Dev Dyn
2007
;
236
:
3297
308
.
122
Mimeault
M
,
Hauke
R
,
Mehta
PP
,
Batra
SK
. 
Recent advances in cancer stem/progenitor cell research: therapeutic implications for overcoming resistance to the most aggressive cancers
.
J Cell Mol Med
2007
;
11
:
981
1011
.
123
Oka
N
,
Soeda
A
,
Inagaki
A
, et al
. 
VEGF promotes tumorigenesis and angiogenesis of human glioblastoma stem cells
.
Biochem Biophys Res Commun
2007
;
360
:
553
9
.
124
Knizetova
P
,
Darling
JL
,
Bartek
J
. 
Vascular endothelial growth factor in astroglioma stem cell biology and response to therapy
.
J Cell Mol Med
2008
;
12
:
111
25
.
125
Diabira
S
,
Morandi
X
. 
Gliomagenesis and neural stem cells: Key role of hypoxia and concept of tumor “neo-niche”
.
Med Hypotheses
2008
;
70
:
96
104
.
126
Yao
XH
,
Ping
YF
,
Chen
JH
, et al
. 
Glioblastoma stem cells produce vascular endothelial growth factor by activation of a G-protein coupled formylpeptide receptor FPR
.
J Pathol
2008
;
215
:
369
76
.
127
Bao
S
,
Wu
Q
,
Sathronsumetee
S
, et al
. 
Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor
.
Cancer Res
2006
;
66
:
7843
8
.
128
Wong
ET
,
Brem
S
. 
Antiangiogenesis treatment for glioblastoma multiforme: challenges and opportunities
.
J Natl Compr Canc Netw
2008
;
6
:
515
22
.
129
Martinez
R
,
Schackert
G
. 
Epigenetic aberrations in malignant gliomas: an open door leading to better understanding and treatment
.
Epigenetics
2007
;
2
:
147
50
.
130
Castellino
RC
,
Durden
DL
. 
Mechanisms of disease: The PI3K-Akt-PTEN signaling node-an intercept point for the control of angiogenesis in brain tumors
.
Nat Clin Pract Neurol
2007
;
3
:
682
93
.
131
Kao
GD
,
Jiang
Z
,
Fernandes
AM
,
Gupta
AK
,
Maity
A
. 
Inhibition of phosphatidylinositol-3-OH kinase/AKT signaling impairs DNA repair in glioblastoma cells following ionizing radiation
.
J Biol Chem
2007
;
282
:
21206
12
.
132
Kwon
CH
,
Zhao
D
,
Chen
J
, et al
. 
Pten haploinsufficiency accelerates formation of high-grade astrocytomas
.
Cancer Res
2008
;
68
:
3286
94
.
133
Jiang
BH
,
Liu
LZ
. 
PI3K/PTEN signaling in tumorigenesis and angiogenesis
.
Biochim Biophys Acta
2008
;
1784
:
150
8
.
134
Hambardzumyan
D
,
Squatrito
M
,
Carbajal
E
,
Holland
EC
. 
Glioma formation, cancer stem cells, and Akt signaling
.
Stem Cell Rev
2008
;
4
:
203
10
.
135
Eyler
CE
,
Foo
WC
,
LaFiura
KM
,
McLendon
RE
,
Hjelmeland
AB
,
Rich
JN
. 
Brain cancer stem cells display preferential sensitivity to Akt inhibition
.
Stem Cells
2008
;
26
:
3027
36
.
136
Bao
S
,
Wu
Q
,
McLendon
RE
, et al
. 
Glioma stem cells promote radioresistance by preferential activation of the DNA damage response
.
Nature
2006
;
444
:
756
60
.
137
Rich
JN
. 
Cancer stem cells in radiation resistance
.
Cancer Res
2007
;
67
:
8980
4
.
138
Kato
T
,
Kanemura
Y
,
Shiraishi
K
,
Miyake
J
,
Kodama
S
,
Hara
M
. 
Early response of neural stem/progenitor cells after X-ray irradiation in vitro
.
Neuroreport
2007
;
18
:
895
900
.
139
Eramo
A
,
Ricci-Vitiani
L
,
Zeuner
A
, et al
. 
Chemotherapy resistance of glioblastoma stem cells
.
Cell Death Differ
2006
;
13
:
1238
41
.
140
Beier
D
,
Röhrl
S
,
Pillai
DR
, et al
. 
Temozolomide preferentially depletes cancer stem cells in glioblastoma
.
Cancer Res
2008
;
68
:
5706
15
.
141
Murat
A
,
Migliavacca
E
,
Gorlia
T
, et al
. 
Stem cell-related "self-renewal" signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma
.
J Clin Oncol
2008
;
26
:
3015
24
.
142
Vescovi
AL
,
Galli
R
,
Reynolds
BA
. 
Brain tumour stem cells
.
Nat Rev Cancer
2006
;
6
:
425
36
.
143
Salmaggi
A
,
Boiardi
A
,
Gelati
M
, et al
. 
Glioblastoma-derived tumorospheres identify a population of tumor stem-like cells with angiogenic potential and enhanced multidrug resistance phenotype
.
Glia
2006
;
54
:
850
60
.
144
Jin
F
,
Zhao
L
,
Zhao
HY
, et al
. 
Comparison between cells and cancer stem-like cells isolated from glioblastoma and astrocytoma on expression of anti-apoptotic and multidrug resistance-associated protein genes
.
Neuroscience
2008
;
154
:
541
50
.
145
Shervington
A
,
Lu
C
. 
Expression of multidrug resistance genes in normal and cancer stem cells
.
Cancer Invest
2008
;
26
:
535
42
.
146
Tagscherer
KE
,
Fassl
A
,
Campos
B
, et al
. 
Apoptosis-based treatment of glioblastomas with ABT-737, a novel small molecule inhibitor of Bcl-2 family proteins
.
Oncogene
2008
;
27
:
6646
56
.
147
Mammolenti
M
,
Gajavelli
S
,
Tsoulfas
P
,
Levy
R
. 
Absence of major histocompatibility complex class I on neural stem cells does not permit natural killer cell killing and prevents recognition by alloreactive cytotoxic T lymphocytes in vitro
.
Stem Cells
2004
;
22
:
1101
10
.
148
Hori
J
,
Ng
TF
,
Shatos
M
,
Klassen
H
,
Streilein
JW
,
Young
MJ
. 
Neural progenitor cells lack immunogenicity and resist destruction as allografts
.
Ocul Immunol Inflamm
2007
;
15
:
261
73
.
149
Viale
A
,
De Franco
F
,
Orleth
A
, et al
. 
Cell-cycle restriction limits DNA damage and maintains self-renewal of leukaemia stem cells
.
Nature
2009
;
457
:
51
6
.
150
Cimprich
KA
,
Cortez
D
. 
ATR: an essential regulator of genome integrity
.
Nat Rev Mol Cell Biol
2008
;
9
:
616
27
.
151
Reinhardt
HC
,
Yaffe
MB
. 
Kinases that control the cell cycle in response to DNA damage: Chk1, Chk2, and MK2
.
Curr Opin Cell Biol
2009
;
21
:
245
55
.
152
Enders
GH
. 
Expanded roles for Chk1 in genome maintenance
.
J Biol Chem
2008
;
283
:
17749
52
.
153
Sharma
V
,
Hupp
CD
,
Tepe
JJ
. 
Enhancement of chemotherapeutic efficacy by small molecule inhibition of NF-kappaB and checkpoint kinases
.
Curr Med Chem
2007
;
14
:
1061
74
.
154
Bucher
N
,
Britten
CD
. 
G2 checkpoint abrogation and checkpoint kinase-1 targeting in the treatment of cancer
.
Br J Cancer
2008
;
98
:
523
8
.
155
Zabludoff
SD
,
Deng
C
,
Grondine
MR
, et al
. 
AZD7762, a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies
.
Mol Cancer Ther
2008
;
7
:
2955
66
.
156
Rainey
MD
,
Charlton
ME
,
Stanton
RV
,
Kastan
MB
. 
Transient inhibition of ATM kinase is sufficient to enhance cellular sensitivity to ionizing radiation
.
Cancer Res
2008
;
68
:
7466
74
.
157
Glaab
WE
,
Tindall
KR
. 
Mutation rate at the hprt locus in human cancer cell lines with specific mismatch repair-gene defects
.
Carcinogenesis
1997
;
18
:
1
8
.
158
Roos
WP
,
Batista
LF
,
Naumann
SC
, et al
. 
Apoptosis in malignant glioma cells triggered by the temozolomide-induced DNA lesion O6-methylguanine
.
Oncogene
2007
;
26
:
186
97
.