Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Dec 1.
Published in final edited form as: Pharmacol Ther. 2021 Jun 23;228:107922. doi: 10.1016/j.pharmthera.2021.107922

Genotoxic therapy and resistance mechanism in gliomas

Fengchao Lang 1, Yang Liu 1, Fu-Ju Chou 1, Chunzhang Yang 1
PMCID: PMC8848306  NIHMSID: NIHMS1778823  PMID: 34171339

Abstract

Glioma is one of the most common and lethal brain tumors. Surgical resection followed by radiotherapy plus chemotherapy is the current standard of care for patients with glioma. The existence of resistance to genotoxic therapy, as well as the nature of tumor heterogeneity greatly limits the efficacy of glioma therapy. DNA damage repair pathways play essential roles in many aspects of glioma biology such as cancer progression, therapy resistance, and tumor relapse. O6-methylguanine-DNA methyltransferase (MGMT) repairs the cytotoxic DNA lesion generated by temozolomide (TMZ), considered as the main mechanism of drug resistance. In addition, mismatch repair, base excision repair, and homologous recombination DNA repair also play pivotal roles in treatment resistance as well. Furthermore, cellular mechanisms, such as cancer stem cells, evasion from apoptosis, and metabolic reprogramming, also contribute to TMZ resistance in gliomas. Investigations over the past two decades have revealed comprehensive mechanisms of glioma therapy resistance, which has led to the development of novel therapeutic strategies and targeting molecules.

Keywords: Genotoxic therapy, glioma, cancer, therapy resistance

Introduction

Gliomas account for over 70% of primary brain tumors. Traditionally, gliomas are histologically classified into four grades. The World Health Organization (WHO) grades I and II are characterized as low-grade gliomas, while grades III and IV are high-grade gliomas. Several pioneering studies have shown that mutations in isocitrate dehydrogenase (IDH1/2) are highly frequent genetic abnormalities in glioma, identified in approximately 80% of all WHO grade II/III gliomas and secondary glioblastomas (Cohen et al., 2013). Glioblastoma (GBM, WHO grade IV) is the most common primary brain tumor, accounting for 54% of all gliomas and 14.5% of all brain tumors diagnosed in the United States (Ostrom et al., 2020). GBM is a highly aggressive disease with a poor median survival of approximately 12–15 months, and only 4.7% of patients survive longer than 5 years (Oike et al., 2013; Stupp et al., 2009). The 2016 revised WHO classification of tumors in the CNS combines biology-driven molecular marker diagnostics with classical histological cancer diagnosis. The new classification includes major restructuring of the diffuse gliomas, medulloblastomas, and other embryonal tumors and incorporates new entities that are defined by both histology and molecular features such as glioblastoma (IDH-wild-type and IDH-mutant types), and diffuse midline glioma (Louis et al., 2016).

The chemotherapy for gliomas includes TMZ and other alkylating agents such as carmustine (BCNU), nimustine (ACNU), and lomustine (CCNU) (Fukushima et al., 2009). The clinical benefit of TMZ in patients with gliomas remains limited, primarily due to intrinsic or acquired chemoresistance mechanisms (Sarkaria et al., 2008a). Genomic heterogeneity, a highly infiltrative nature, and distinct mechanisms also enable cancer cells to escape chemotherapy and radiotherapy (Annovazzi et al., 2017). As the standards of care for glioma are mostly based on introducing DNA damage to the cancer genome, the intrinsic DNA repair pathways play essential roles in protecting tumor cells and determining disease outcomes. The activation of DNA repair machinery has been extensively investigated. In the present review, we have summarized the current understanding of DNA repair pathways in glioma therapy resistance. We focused on DNA repair enzymes while including other resistance mechanisms such as programmed cell death, cancer metabolism, and glioma stem cells. We also provide an overview of novel molecular targeting strategies that assist genotoxic therapy in glioma.

DNA damage and repair

The integrity of genomic DNA is maintained by a balance between DNA damage and repair. Cells are constantly exposed to genotoxic factors such as oxidative damage, DNA strand breaks, and replication errors. In eukaryotic cells, many DNA repair mechanisms are established through evolution to limit the impact of DNA damage. Radiotherapy and chemotherapy are commonly used glioma therapies. They trigger the formation of both single- and double-strand breaks. The intrinsic DNA repair pathways are frequently exploited by glioma to handle the impact of DNA damage. Owing to the comprehensive anatomic structure of the blood-brain barrier (BBB), many anti-cancer therapeutics can hardly access intracranial malignancies; hence, genotoxic therapies remain the major treatment options. In the following section, we will discuss several DNA repair machineries that are related to glioma therapy resistance.

1. MGMT in glioma resistance to TMZ therapies

In 2005, Stupp et al. (Stupp et al., 2005) demonstrated that the addition of TMZ to radiotherapy resulted in clinical benefit for patients with newly diagnosed GBM with prolonged overall survival, which led to the approval of TMZ as part of the standard of care for glioma by the United States Food and Drug Administration (FDA). TMZ is almost completely absorbed through the gut, penetrates the BBB, and affects glioma cells. It achieves sufficient concentrations against tumors within the brain, with a concentration between 14.95 and 34.54 μM in brain tumors, about 20% of its plasma concentration (Kaina, 2019). TMZ does not require hepatic metabolism for activation, but is spontaneously hydrolyzed to 3-methyl-(triazen-1-yl) imidazole-4-carboxamide (MTIC), and then splits into monomethylhydrazine and 5-aminoimidazole-4-carboxamide (AIC). These drug metabolites lead to rapid methyl adducts on nucleosides. TMZ results in DNA methylation specifically at the N7 positions of guanine in guanine-rich regions, the N3 positions in adenine or guanine, and the O6 position in guanine. The ratio of these lesions are N7-methylguanine (N7-meG) (80%–85%), N3-methyladenine (N3-meA) or N3-methylguanine (N3-meG) (8%–20%), and O6-methylguanine (O6-meG) (8%). The formation of O6-meG is the major molecular mechanism for TMZ-associated toxicity, as O6-meG-associated nucleotide mispairs result in inhibition of DNA synthesis, and DSB formation (Zhang et al., 2012). Severe DNA damage subsequently activates the ATM/Chk2 signaling cascade and induces p53-associated G2/M cycle arrest (Caporali et al., 2004). Owing to DNA damage, tumor cells eventually undergo apoptosis, autophagy, or senescence (Aasland et al., 2019; Wurstle et al., 2017).

MGMT (O-6-Methylguanine-DNA Methyltransferase) is an enzyme that repairs naturally occurring O6-meG lesions, preventing DNA mismatch and replication errors. In glioma, MGMT plays a major role in the resistance to TMZ and other alkylating agents through the removal of the O6-meG lesion. MGMT repairs the O6-meG lesion in a “suicide” way by transferring the methyl group from the adduct to its cysteine residue, resulting in the degradation of the enzyme (Fig. 1). MGMT knockout mice exhibited higher sensitivity to the toxic effects of TMZ than MGMT wild-type counterparts (Glassner et al., 1999). Inhibition of MGMT with O6-benzylguanine promotes the anti-tumoral activity of TMZ both in vitro (Bobola et al., 1996) and in vivo (Friedman et al., 1995).

Figure 1. TMZ induced DNA damage repair.

Figure 1.

TMZ is hydrolyzed to its active metabolite, 3-methyl-(triazen-1-yl) imidazole-4-carboxamide (MTIC), and then splits into monomethyl diazonium ions and 5-aminoimidazole-4-carboxamide (AIC). TMZ causes methyl adducts specifically at the N1-methyladenine (N1meA) and N3-methylcytosine (N3meC) (2%); N7-methylguanine (N7-meG) (80%–85%); N3-methyladenine (N3-meA) or N3-methylguanine (N3-meG) (8%), and O6-methylguanine (O6-meG) (8%). ALKBH2 and ALKBH3 remove N1meA and N3meC, accompanying with oxidative decarboxylation of 2-oxoglutarate (α-ketoglutarate, αKG) to succinate (Suc). MGMT is responsible for repairing O6-meG lesions. MGMT transfers the methyl group from O6-meG to its cysteine residue, resulting in the methylation and degradation of itself. If O6-meG adducts fail to be removed by MGMT, it will mispair with thymine. O6-meG-T mismatches are recognized by MMR components MSH2 and MSH6. MMR components MLH1 and PMS2 further removes the thymine residue and leave the O6-meG adduct unrepaired. The futile repair cycles lead to the accumulation of DNA strand breaks and eventually apoptosis. N7-meG and N3-meA adducts account for about 80% of TMZ-induced DNA adducts. BER system efficiently fixes these lesions. MPG is responsible for the detection and removal of aberrantly N7-meG and N3-meA. APE recognizes the abasic site, cleaves the 5’ end of the DNA, and triggers PARP1 to undergo patch repair. Patch repair can be completed by DNA polymerase, DNA ligase III, and XRCC1 (short patch) or FEN1 and DNA ligase I (long patch).

Owing to the essential role of MGMT in TMZ resistance, the expression level of MGMT is closely related to the efficacy of cancer therapies. Hypermethylation of the promoter region of the MGMT gene epigenetically silences the protein expression. Low expression of MGMT correlates with a better clinical response and prolonged survival (Hegi et al., 2005). The promoter methylation status determines the expression level of MGMT, which could be considered as a biomarker for TMZ responses in patients with glioma (Hermisson et al., 2006; Stupp et al., 2007). Indeed, a clinical data analysis showed that low MGMT methylation could confer some sensitivity to TMZ treatment, providing cut-offs that allow treatment decisions for personalized therapy (Villano et al., 2009).

Besides primary glioma, MGMT also plays critical roles during the disease recurrence. Higher expression of MGMT was found in clinical samples from recurrent GBM compared to matched primary lesions (Storey et al., 2019). The altered expression levels of MGMT in recurrent GBM could be resulted from changes in its promoter methylation status. Lower MGMT promoter methylation correlates with recurrent stage compared to primary tumors. Additionally, MGMT promoter is prone to establish unmethylated status when the cancers proceed to recurrence (Choi et al., 2021). On the other hand, the aberrantly high expression MGMT could also be induced by other factors and further contributes to GBM recurrence. A recent study reveals that MGMT genomic rearrangements lead to MGMT overexpression in recurrent gliomas while MGMT promoter methylation is unaltered (Oldrini et al., 2020).

Since the discovery of MGMT in cancer therapy resistance, molecular targeting strategies have been pursued to overcome MGMT-associated DNA repair. For example, lomeguatrib (a potent MGMT inhibitor) has shown promising efficacy and safety profile in sensitizing solid tumors for TMZ therapy in Phase I and II clinical trials (Middleton et al., 2002). Decitabine (DAC), an inhibitor of DNA methyltransferase that depletes MGMT, resulted in a 12.4-month median overall survival when combined with TMZ therapy in patients with metastatic melanoma, suggesting possible superiority over the historical 1-year overall survival (Tawbi et al., 2013). Moreover, O6-benzylguanine (O6 BG), a nucleotide analog of O6-meG that can be recognized by MGMT, inactivates MGMT enzyme by transferring a benzyl group to the active site of MGMT. O6 BG showed exciting anti-tumoral effects in preclinical studies, but the combination of O6 BG and TMZ in pediatric patients did not improve treatment response in clinical studies (Warren et al., 2005). A recent study by Rahman et al. (Rahman et al., 2019) showed that bortezomib sensitizes glioblastoma to TMZ treatment through the depletion of MGMT mRNA and protein with prolonged animal survival.

2. DNA damage responses in in glioma resistance to TMZ therapies

2.1. Deficient mismatch repair tolerates glioma to TMZ treatment

Mismatch repair (MMR), base excision repair (BER), and nucleotide excision repair (NER) are SSB repair mechanisms. The SSB repair mechanism recognizes and fixes the damaged DNA nucleotide by using the other DNA strand as a template (Jackson and Bartek, 2009). MMR and BER mechanisms have been extensively studied in glioma resistance.

MMR is a molecular mechanism that recognizes erroneous insertion, deletion, and mis-incorporations of nucleobases through DNA replication, recombination, and damage. MMR is a highly conserved mechanism that is governed by mismatch repair proteins MutL homolog 1 (MLH1), MutS homolog 2 (MSH2), MSH3, MSH6, and mismatch repair endonuclease PMS2 (PMS2). Alkylating agents such as TMZ result in methylated nucleosides, and mispairs between O6-meG with thymine or cytosine, causing O6-meG:T or O6-meG:C mismatch in the progeny DNA (Sarkaria et al., 2008b) (Fig. 1). Although MMR recognizes these mispairs and removes the thymine residue, the enzymes are not able to remove the O6-meG. Thus, the O6-meG persists in the genomic DNA, mispairs with thymine repeatedly, leading to repetitive rounds of MMR. It has been proposed that the repeating ‘futile repair’ cycle eventually leads to replication forks collapse, DNA double-strand breakages and cell death via apoptosis or autophagy (Villano et al., 2009). A recent study using CRISPR/Cas9 screening confirmed the role of the MMR system in TMZ therapeutic sensitivity (MacLeod et al., 2019). The results highlighted that three weeks after high doses of TMZ treatment in patient-derived glioblastoma stem cells, there is an enrichment for gRNAs targeting four genes involved in the MMR (MLH1, MSH2, MSH6, and PMS2) in resistant cells (MacLeod et al., 2019). It was also reported that a significant decrease in the MSH2, MSH6, and PMS2 protein expression levels was observed in recurrent GBM (Felsberg et al., 2011).

Tumors deficient in the MMR pathway are relatively resistant to genotoxic therapies. Karran et al. (Karran, 2001) proposed that MMR-deficient cells exhibited a 50-fold lower sensitivity towards methylating agents compared to their MMR-proficient counterparts. Cahill et al. (Cahill et al., 2007) reported that loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor progression during TMZ treatment. In addition, McFaline-Figueroa et al. (McFaline-Figueroa et al., 2015) also showed that loss of MMR genes MSH2 and MSH6 provided substantial survival benefits to glioma cells. Moreover, mutations in the MMR pathway are specifically enriched in hypermutant glioma recurrence (Wang et al., 2016a). Mutations in MMR proteins may contribute to disease relapse and the hypermutated genotype through error-prone DNA replication, the disabled futile repair cycle, and tolerance of apoptotic changes.

2.2. BER promotes glioma resistance to genotoxic therapies

BER is responsible for correcting small base lesions such as nucleotides with methylation, oxidation, or deamination. At the beginning of BER, a specific DNA glycosylase recognizes the lesion and removes the affected base (Fig. 1). There are at least 11 distinct DNA glycosylases divided into four structurally distinct super families: uracil DNA glycosylases (UDGs), helix-hairpin-helix (HhH) glycosylases, 3-methylpurine glycosylase (MPG), and endonuclease VIII-like (NEIL) glycosylases (Jacobs and Schar, 2012). Apurinic/apyrimidinic endonuclease 1 (APE1) recognizes the abasic site and cleaves the 5’ end of the DNA. There are two repair pathways following cleavage. Short patch (one nucleotide) repair is conducted by DNA polymerase, DNA ligase III, and X-ray repair cross complementing 1 (XRCC1). Long-patch (2–10 nucleotide) repair requires flap structure-specific endonuclease 1 (FEN1) and DNA ligase I (Krokan and Bjoras, 2013).

Although N7-meG and N3-meA adducts account for about 80% of TMZ-induced DNA adducts (Wirtz et al., 2010), they rarely cause cytotoxic effects on cells, as the BER system efficiently fixes these lesions. Several lines of evidence have suggested that the inhibition of BER is synergistic with TMZ treatment. For example, MPG is responsible for the removal of aberrantly N7-meG and N3-meA in mammalian cells (Montaldo et al., 2019). Agnihotri et al. (Agnihotri et al., 2012) found a significant correlation between the expression levels of the MPG and the survival of adult GBM cells under TMZ treatment, indicating a supportive role of MPG in TMZ resistance. Moreover, methoxyamine, an inhibitor of MPG, sensitizes tumor cells to alkylating agents in vitro and in vivo (Agnihotri et al., 2014). It was also shown that the down-regulation of APE1 expression by small interfering RNA or nitroxoline affects the resistance of GBM to TMZ (Cho et al., 2019; Montaldi et al., 2015). Inhibition of DNA polymerase beta (Goellner et al., 2012; Trivedi et al., 2008) or endonuclease non-catalytic subunit 1 (ERCC1) also enhances TMZ efficacy in glioma treatment (Boccard et al., 2015). Goellner et al. (Goellner et al., 2011) found that poly(ADP-ribose) polymerase 1 (PARP1), which functions as a participator and sensor of incomplete BER, is hyperactivated when BER is inhibited. Aberrantly activated PARP1 continuously consumes NAD+ to synthesize poly (ADP-ribose) (PAR), leading to ATP depletion and energy-dependent cell apoptosis (Goellner et al., 2011).

PARP1 is thought to play a vital role in the BER pathway by interacting with BER proteins such as DNA ligase III, DNA polymerase beta, and XRCC1 (Dantzer et al., 2000; Heale et al., 2006). These proteins can be modified by PARylation through the action of PARP1. The PAR chain facilitates the recruitment of DNA repair complexes by providing a docking platform (Wei and Yu, 2016). PARP1 is highly expressed in GBM but not in normal brain tissues (Bao et al., 2006; Galia et al., 2012; Rasmussen et al., 2016), indicating that PARP1 is relevant to treatment resistance in glioma (Bao et al., 2006; Galia et al., 2012; Rasmussen et al., 2016). PARP inhibitors have been extensively studied in the past three decades and have shown promising results with increased TMZ sensitivity in GBM and ideal BBB penetrations. There are several PARP inhibitors in Phase I/II clinical trials on GBM patients, including niraparib (NCT04221503), veliparib (NCT03581292), and BGB-290 (NCT03749187).

2.3. Homologous repair in gliomas resistance to genotoxic therapy

Several lines of evidence suggest that TMZ resistance in recurrent glioma can be acquired without the up-regulation of MGMT or deficient MMR (Gil Del Alcazar et al., 2016; Happold et al., 2012). This indicates that there are alternative pathways for recurrent glioma tumors to handle genotoxic agents. Increased double-strand breaks (DSB) repair is one of the newly identified mechanisms contributing to acquire TMZ resistance in GBM.

TMZ-induced DNA lesions lead to the stall of replication forks and eventually MMR-dependent DSB (Fan et al., 2013). DSB is among the most severe and lethal types of DNA damage. DSB requires complicated repair mechanisms because of extensive damage and lack of a template to generate the new strand (Cannan and Pederson, 2016). There are two major repair mechanisms involved in DSB repair: homologous recombination (HR) and non-homologous end-joining (NHEJ). HR occurs when DNA strands are damaged during the S/G2 phase of the cell cycle. NHEJ exerts its role when cells lack a second copy of the DNA, and throughout most of the cell cycle, it predominantly acts in the G1 phase. ATM, ATR, DNA-PK, and KU70/80 kinases are the first proteins recruited by the DSB and play important roles in sensing the damage and initiating a downstream signaling cascade (Blackford and Jackson, 2017). The DNA damage response activates subsequent cell cycle checkpoints, which either repair damaged DSB or trigger cell death pathways.

HR DNA repair plays a vital role in glioma resistance genotoxic therapy (Fig. 2). An investigation of the Cancer Genome Atlas (TCGA) revealed that HR deficiency strongly correlated with progression-free interval and better clinical outcomes, indicating an adverse effect of HR DNA repair on GBM treatment (Knijnenburg et al., 2018). Studies have shown more efficient HR DNA repair in glioma-initiating cells than in non-tumor forming neural progenitor cells (Lim et al., 2012). Higher HR DNA repair efficiency was observed in glioma cells compared to neural progenitor cells. Carlos et al. (Buis et al., 2012; Esashi et al., 2005; Tomimatsu et al., 2014) reported that augmented HR DNA repair confers TMZ resistance in recurrent tumors. CDK1/2 facilitates the HR process by phosphorylating BRCA2 and exonuclease 1 (EXO1), or interacting with Double-strand break repair protein MRE11 and nibrin (NBS1). CDK1 and CDK2 are highly expressed in GBM and they induce radioresistance in GBM (Wang et al., 2016b; Zhang et al., 2018). Furthermore, CDK1/2 inhibitors suppress HR efficiency in recurrent glioma tumors and sensitize tumor cells to TMZ (Gil Del Alcazar et al., 2016).

Figure 2. TMZ induced double strands breaks addressed by Homologous Repair.

Figure 2.

Unrepaired O6-meG adducts will lead to DSB, which is the most lethal DNA damage. DSB can be sensed by the MRE11-RAD50-NBS1 (MRN complexes), which set in motion a numbers of processes collectively called the DNA damage response to coordinate DNA repair, These sensor complexes recruit and activate kinases including ATM, ATR, DNA-PK and subsequently other modifying enzymes which, through cascades of phosphorylation and ubiquitination events, activate and mobilize a large number of proteins, such as p53, BRCA1, 53BP1 and H2AX. Homologous Repair requires resection by BRCA1, CtIP and BARD1 to generate single-stranded DNA (ssDNA), which immediately becomes coated by RPA and subsequently replaced by Rad51 preparing for strand invasion. Failure of Homologous Repair will result in cell death.

3. Aberrant cell cycle regulation in gliomas resistance to genotoxic therapy

High RNA expression levels of the DNA repair/cell-cycle genes including AURKA, CDC25C, CDC6, CDK1, CENPA, RAD51, RAD54, were revealed in relapsed glioma after radiotherapy alone or radiotherapy plus TMZ (Gobin et al., 2019). Dysregulated activity of cell cycle regulation proteins has been observed in human malignancies, and their aberrant activation is tightly associated with resistance to genotoxic therapy in cancer cells (Ferri et al., 2020; Huang and Zhou, 2020).

3.1. ATM/CHK2 pathway

The G1/S and intra-S checkpoints are controlled by the ATM/p53/CHK2 signaling cascade. ATM/p53/CHK2 induced cell cycle arrest and suppressed tumor formation in a glioma mouse model (Squatrito et al., 2010). Chk2−/− tumor-bearing mice showed increased GBM resistance to genotoxic therapy with a lower apoptotic index and higher proliferation rate. Consistently, inhibition of checkpoint kinases or ATM sensitized glioma stem cells (GSCs) to ionizing radiation (IR) in vitro (Bao et al., 2006) (Carruthers et al., 2015). The ATM inhibitors AZ32 and AZD1390 result in promising results in glioma xenograft models (Durant et al., 2018; Karlin et al., 2018). They abolish ATM-dependent DNA damage response (DDR) pathway activity, thereby resensitizing radiotherapy-resistant glioma cells to G2/M checkpoint arrest and apoptosis. Both AZ32 and AZD1390 have been specifically designed for effective BBB penetration, granting their future clinical application. A Phase I study is currently conducted to assess the safety and tolerability of AZD1390 administered with radiotherapy in patients with brain cancer (NCT03423628).

3.2. ATR/CHK1 pathway

ATR acts primarily through CHK1 and controls the intra-S and G2/M checkpoints. ATR-CHK1 checkpoints were found to be upregulated in glioblastoma stem-like cells, suggesting a more effective DNA repair reaction. Eich et al. (Eich et al., 2013) have shown that ATR contributes to the resistance of GBM cells to TMZ in vitro. Knockdown ATR sensitizes GBM cells to TMZ, while ATR depletion has a significantly stronger effect. Knockdown of ATR, but not ATM, abolished phosphorylation of H2A.X, the DSB repair marker, and CHK1 and CHK2.

Ahmed et al. (Ahmed et al., 2015) showed that the ATR-CHK1 pathway is involved in the radioresistance of GSC by enhancing G2 checkpoint arrest. A combination of ATR-CHK1 inhibition and IR decreased the colony-forming ability of GSCs through catastrophic chromosome segregation during mitosis. Gil et al. (Gil del Alcazar et al., 2014) reported that the ATR inhibitor, NVP-BEZ235, potently inhibited the repair of IR-induced DNA damage in the glioma cell line U87vIII derived tumors. The attenuated DDR sensitizes glioma tumors and extends the survival of brain tumor-bearing mice (Gil del Alcazar et al., 2014). It has been reported that another BBB-penetrating ATR inhibitor AZD6738 enhances the therapeutic efficacy of cisplatin in non-small cell lung cancer (NSCLC) xenograft models, and AZD6738 can effectively penetrate the brain, indicating its potential use for treating brain tumors (Frosina et al., 2018; Vendetti et al., 2015). Currently, a Phase II clinical trial aimed at examining the efficacy of NVP-BEZ235 in glioblastoma treatment (NCT02430363). There are multiple clinical studies on AZD6738 in relapsed/refractory cancers. For example, AZD6738 in IDH1 and IDH2 mutated, refractory cholangiocarcinoma, or other solid malignant tumors are under investigation in an ongoing Phase II clinical trial (NCT03878095). Another Phase II trial of AZD6738 alone and in combination with olaparib aims to treat participants with renal cell carcinoma, urothelial carcinoma, all pancreatic cancers, or other solid tumors (NCT03682289). However, primary central nervous system tumors are rarely included in such studies.

4. Resistance of Glioma Stem Cells to genotoxic therapy

Cancer stem cells (CSCs) are a minority population in tumor cells characterized by self-renewal, proliferation, and differentiation (Clarke et al., 2006). These properties confer CSCs the ability to be relatively resistant to conventional chemotherapeutic agents. Similarly, glioma stem cells (GSCs) share many common characteristics with normal neural stem cells (NSCs) such as self-renewal, expression of molecular markers CD133, CD15, L1CAM, and SOX2, and the ability to differentiate into neurons, astrocytes, and oligodendrocytes (Vescovi et al., 2006). GSCs promote tumor heterogeneity and therapy resistance to genotoxic stresses and are thus a key therapeutic target (Prager et al., 2020).

The major players of DDR proteins (ATM, ATR, Chk1, Chk2, PARP1, and RAD51) are upregulated in GSCs (King et al., 2017; Ning et al., 2019). Aberrant G2-M cell-cycle checkpoint activation and increased DNA repair efficiency were found in GSCs. Inhibition of ATM, CHK1, ATR, or PARP abrogated G2-M checkpoint function and resulted in a profound radiosensitization of GSCs (Ahmed et al., 2015). Carruthers et al. (Carruthers et al., 2018) recently revealed high levels of DNA replication stress as a novel mechanism responsible for up-regulating DDR response and radiotherapy resistance in GSCs. Elevated levels of DNA replication stress evoked constitutive activation of the DNA damage response, characterized by increased phosphorylation of ATR, CHK1, and RPA32 in GSC-enriched cultures. Inhibition of the replication stress response with ATR and PARP inhibitors in GSCs reduces neurosphere formation and abrogates radiation resistance (Carruthers et al., 2018).

In addition, high metabolic demands are required to support maintenance of stemness, self-renewal, and proliferation in GSCs. GSCs also exhibit reprogrammed metabolism pathways such as glycolysis, oxidative phosphorylation, and glutaminolysis to support their cell activities more than their differentiated counterparts. For example, GSCs increase glucose uptake by co-opting the neuronal glucose transporter, Glut3 (Cosset et al., 2017; Flavahan et al., 2013). Differentiated tumor cells mainly rely on glycolysis, while GSCs consume less glucose and heavily depend on oxidative phosphorylation through IGF2BP2 regulation and FGFR3-TACC3 gene fusion mechanisms (Frattini et al., 2018; Janiszewska et al., 2012; Vlashi et al., 2011). GSCs can also acquire nutrients from glutamine and acetate as their bioenergetic and growth resources (Tardito et al., 2015). These additional metabolic pathways confer GSCs with an increased radiation resistance when challenged in hostile conditions.

5. Cell death regulation facilitates TMZ resistance in gliomas

Genotoxic agents efficiently clear tumor cells because the treatment-induced DNA damage is commonly translated into programmed cell death, such as apoptosis, senescence, autophagy and ferroptosis. Here we have briefly introduced several major regulated cell death pathways related to chemotherapy resistance. More details could be reached in the review articles which are focused on cell death and chemotherapies (Mohammad et al., 2015; Ricci and Zong, 2006; Sui et al., 2013).

Apoptosis is a central cell death program that can be induced by genotoxic therapies. Unrepaired DNA damage changes mitochondrial membrane permeabilization through activation of p53 and B cell lymphoma 2 (BCL-2) family proteins such as BAD, BAX, and BID (Fang et al., 2016).

p53 is stabilized and phosphorylated by ATM or ATR pathways in response to TMZ induced DNA damage (Caporali et al., 2004). Activated p53 induces expression of its major transcription target p21. p21 leads to G1 cell cycle arrest through its universal cyclin-dependent kinase inhibitor function (including CDK1 and CDK2) (Georgakilas et al., 2017). p16INK4a, an inhibitor of CDK 4/6, prevents phosphorylation of pRb through inactivation of CDK4/6 complex, and thus stops cell cycle progression. The p53-p21 and p16INK4a-Rb axis work together to pause G1 to S phase transition, allowing cells to repair DNA breaks or enter apoptosis process (Georgakilas et al., 2017). Up to 50% of GBM possess p53 inactivation mutations (Liu et al., 2020a). The p53 mutation or loss of functional p53 sensitizes glioma cells to TMZ and BCNU due to the unrepaired DNA breaks (Blough et al., 2011; Dinca et al., 2008; Xu et al., 2005). Similarly, about 78% of GBM tumors are deficient in p16INK4a-Rb pathway (Cen et al., 2012), suggesting that genotoxic therapy could be effective for GBM therapy.

The BCL-2 family, which plays a key role in regulating apoptosis, balances between pro- and anti-apoptotic processes. The BCL-2 family is divided into three groups: 1) pro-apoptotic proteins such as BAX, BAK, and BCL-Xs; 2) anti-apoptotic proteins BCL-2, BCL-XL, and MCL-1; and 3) pro-apoptotic Bcl-2 homology domain 3 (BH3)-only proteins such as BAD, BID, BIK, and BIM. Numerous studies have revealed the roles of dysregulation of apoptosis pathways in chemoresistance in gliomas. BCL-2 and BCL-XL were highly expressed in GBM (Kouri et al., 2012; Nagane et al., 1998). A study has shown that there is a correlation between BH3-only pro-apoptotic protein expression and overall survival in GBM patients, and the BCL-2/BCL-XL pathway confers resistance in GBM cells against TMZ treatment (Cartron et al., 2012). Apoptotic GBM cells also promote therapy resistance by secreting apoptotic extracellular vesicles (apoEVs) enriched with various components of spliceosomes. Cell cycle gene RNA splicing can be altered in recipient cells, thereby promoting their resistance to radiotherapy or TMZ therapy (Pavlyukov et al., 2018).

Autophagy, senescence, and ferroptosis are several recently identified programmed cell death mechanisms shown to be relevant in glioma resistance under genotoxic stress. Cells segregate damaged cytoplasmic constituents into autophagosomes for lysosomal degradation through autophagy, serving as a pro-survival response to TMZ therapy. Chen et al. (Chen et al., 2015b) found that the autophagy-related protein VAMP8 is elevated in glioma tissues and silencing autophagy-related gene 5 (ATG5) or syntaxin 17 (STX17) can reverse TMZ resistance in VAMP8 overexpressed glioma cells. Filippi-Chiela et al. (Filippi-Chiela et al., 2015) reported that TMZ induces a transient induction of autophagy and cell resistance of the therapy through inhibition of the Akt-mTOR pathway. The inhibition of autophagy can largely increase the level of apoptosis following TMZ treatment (Knizhnik et al., 2013). Targeting autophagy with chloroquine, a classic inhibitor of autophagy, has now emerged as a promising adjuvant therapy for patients with GBM (NCT00486603, NCT02378532, NCT01430351).

Senescence is frequently induced in brain tumors when treated with TMZ, and this is attributed to clinically achievable low TMZ concentrations within the brain (14.95–34.54 μM in brain tumors). TMZ-induced senescence is triggered by the unrepaired DNA lesion O6-meG (Aasland et al., 2019). The massive “senescence-like dormant phase”, rather than efficient apoptosis, can partially explain the resistance of glioma cells, since the senescent cells can re-enter the normal cell cycle when genotoxic stress is withdrawn (Beausejour et al., 2003).

Ferroptosis is an iron-dependent cell death process that is relevant to TMZ-induced cytotoxicity (Xie et al., 2016). The accumulation of reactive oxygen species, lipid peroxidation, and suppression of glutathione peroxidase is regarded as a characteristic of ferroptosis. NADPH oxidase (NOX) and p53 act as positive regulators of ferroptosis by increasing ROS levels and inhibiting the expression of xCT (SLC7A11), a plasma membrane transporter that facilitates glutathione biosynthesis. Glutathione peroxidase 4 (GPX4), heat shock protein beta-1 (HSPB1), and nuclear factor erythroid 2-related factor 2 (NRF2) function as negative regulators of ferroptosis by limiting cellular iron uptake and reducing ROS production (Xie et al., 2016). Zhao et al. (Zhao et al., 2017) reported a strong correlation between high expression of GPX4 and poor prognosis of glioma patients. Downregulation of GPX4 by ALZ003, a curcumin analog, enhances sensitivity to chemotherapy through ferroptosis in TMZ-resistant glioblastoma cells (Chen et al., 2020b). SLC7A11 contributes to TMZ resistance in glioma by repressing ferroptosis. Overexpression of SLC7A11 in tumor cells inhibits ROS-induced ferroptosis, which is induced by salicylic acid sulfapyridine (SAS) inhibition of SLC7A11 activity (Dixon et al., 2012; Jiang et al., 2015). A study showed that the ferroptosis-inducing agent erastin can synlethalize xCT-depleted glioma cells, suggesting that SLC7A11 contributes to TMZ glioma resistance to TMZ by suppressing ferroptosis (Sehm et al., 2016).

6. Cancer metabolism in genotoxic resistance

Gliomas are characterized by altered metabolism. Rewriting cellular metabolism facilitates glioma cell survival, growth, and resistance to therapies. Abnormal cancer metabolism includes antioxidant regeneration, macromolecule synthesis, and NAD metabolism.

Elevated ROS levels are one of the mechanisms of TMZ-mediated cytotoxicity. Glutathione (GSH) and GSH-related enzymes are among the most important antioxidant components that protect cells from toxicity due to chemotherapy. Cysteine is the rate-limiting substrate for GSH synthesis, and its uptake is regulated by xCT (encoded by SLC7A11 gene). GSH levels are highly correlated with drug resistance in gliomas. Zhu et al. (Zhu et al., 2018) reported higher levels of GSH and glutathione reductase in TMZ-resistant glioma cells than in sensitive cells. Glutathione depletion by the glutathione inhibitor, buthionine sulfoximine sensitized TMZ-resistant glioma cells to TMZ sensitive, both in vitro and in vivo (Rocha et al., 2014). It was shown that xCT is highly expressed in glioma cells (Polewski et al., 2016). Suppression of xCT increased ROS accumulation and compromised glutathione generation, leading to sensitivity to oxidative and genotoxic TMZ stress (Chen et al., 2015a). NRF2 is a transcriptional factor that regulates antioxidant-related genes and cellular redox (Liu et al., 2020c; Yu et al., 2020). High expression of NRF2 induced by TMZ also confers TMZ resistance to gliomas, and inhibition of NRF2 resensitizes glioma cells to TMZ (Cai et al., 2019; Liu et al., 2020b; Rocha et al., 2016; Zhang and Wang, 2017).

Glutamate can be catalyzed into α-ketoglutarate (αKG) and contributes to redox homeostasis with antioxidant GSH production (Wang et al., 2010). Glutamine is converted to an ammonium ion and glutamate by glutaminase (GLS). GLS has been proposed to mediate resistance to GBM therapy. Elevated GLS and glutamate levels were found to promote resistance of GBM cells under mTOR inhibition (Tanaka et al., 2015). Gu et al. (Gu et al., 2017) found that mTORC2 protects glioma cells from cellular stress by repressing cystine uptake and glutathione synthesis in gliomas via inhibition of xCT. The diversity of metabolic adaptations contributes to therapeutic resistance and promotes survival or proliferation. Indeed, mTORC2 inhibition can effectively repress TMZ-resistant gliomas (Miyata et al., 2017).

NAD is an essential co-factor in DDR through PARP DNA repair. PARP1 recruits downstream DDR proteins by synthesizing poly ADP-ribose (PAR) chains, requiring NAD as a substrate (Zhou and Wahl, 2019). PARP inhibitors compete with NAD for the catalytic pocket of PARPs, increasing the radio-sensitivity of gliomas (Hurtado-Bages et al., 2020). NAMPT, the NAD biosynthetic enzyme, is highly expressed in glioblastoma tumors and is associated with overall poor survival in such patients (Gujar et al., 2016). Treatment with NAMPT inhibitors sensitizes glioblastoma cells to TMZ (Feng et al., 2016; Sampath et al., 2015). Aberrantly activated PARP1 by BER inhibition continuously consume NAD to synthesize PAR chains. Overconsumption of NAD leads to ATP depletion and energy-dependent cell apoptosis (Goellner et al., 2011).

7. IDH mutation associated TMZ sensitivity

Isocitrate dehydrogenase enzymes (IDH) are essential enzymes that participate in several major metabolic processes such as the Krebs cycle, glutamine metabolism, lipogenesis, and redox regulation (Tang et al., 2020). Approximately 80% of WHO grade II/III gliomas are IDH mutations (Han et al., 2020; Liu et al., 2019). IDH mutations cluster in the active sites of the enzymes (R132 for IDH1, R140, or R172 for IDH2) (Cohen et al., 2013). Mutant IDH heterodimers catalyze α-ketoglutarate (α-KG) into D-2-hydroxyglutarate (D-2-HG). D-2-HG disrupts the demethylation process in histones and DNA by inhibiting the activities of Lysine-specific demethylase 4 (KDM4) or Ten-eleven translocation methylcytosine dioxygenase (TET). IDH mutations predict better disease outcomes and greater sensitivity to chemotherapy in low-grade gliomas and secondary glioblastomas (Minniti et al., 2014).

Recent investigations have revealed functional changes in DNA repair pathways in the context of IDH mutants and D-2-HG. For example, D-2-HG leads to a genome-wide hypermethylation phenotype, which may trigger MGMT promoter methylation, thereby sensitizing glioma cells to radiotherapy and chemotherapy (Minniti et al., 2014). In addition, D-2-HG inhibits ALKBH2 and ALKBH3, the enzymes mainly responsible for the removal of N1-meA and N3-meC induced by TMZ. Deletion of mutant IDH allele or overexpression of ALKBH2 or AKLBH3 would rescue D-2-HG-induced DNA damage and sensitization to alkylating agents (Wang et al., 2015). The clinical relevance of these findings is supported by a previous report that ALKBH2 confers resistance to TMZ in GBM cells (Johannessen et al., 2013). Moreover, a recent investigation showed that D-2-HG compromises homologous repair through epigenetic regulation. Inhibition of KDM4A and KDM4B by D-2-HG blocks DNA repair protein recruitment to sites of DNA damage (Sulkowski et al., 2017). Inhibition of KDM4A induces hypermethylated H3K9me3 and the widespread hypermethylation of H3K9 masks the specific local H3K9me3, thus blocking the recruitment of factors required for the homologous repair process (Sulkowski et al., 2020). Consequently, impaired homologous repair increases IDH mutant glioma cell sensitivity to genotoxic therapies. Overall, glioma with the IDH mutant appears to exhibit a distinctive biological pattern. More effort is urgently needed to identify the unique therapy response and resistance mechanisms in this disease cluster.

8. Epigenetic factors in glioma pathogenesis and treatment

Epigenetic regulations are tightly linked to glioma pathogenesis. The unique epigenetic patterns in glioma, such as DNA/histone methylation and chromatin remodeling, have been studied for novel molecular targeting strategies to reverse glioma-associated epigenetic shifts. The epigenetics in glioma has been covered by several elegant review articles recently (Cheng et al., 2019; Dong and Cui, 2019; Gusyatiner and Hegi, 2018). Here we highlight several known mechanisms that glioma-associated epigenetic shift that affects genotoxic therapy.

Glioma-CpG Island Methylator Phenotype (G-CIMP) is a glioma-specific DNA methylation pattern (Ruiz-Rodado et al., 2020). G-CIMP is featured with genome-wide hypermethylated CpG islands. G-CIMP mostly occurs in IDH-mutated gliomas. IDH mutation is sufficient to establish the unique DNA methylation pattern in gliomas and most IDH-mutant gliomas are characterized with G-CIMP (M and Wojtas, 2019; Turcan et al., 2012). One of the mechanism is IDH mutant gliomas produce oncometablites such as 2-hydroxyglutarate (2-HG) and these oncometablites represses TETs-mediated DNA demethylation process (Xu et al., 2011). Hypermethylated CpG islands play important roles in GBM development and could be a prognostic factor for treatment. Most G-CIMP+ patients exhibit a better prognosis compared to G-CIMP− patients (Malta et al., 2018). As previously described, the hypermethylation status of MGMT promoter has been extensively studied in glioma. MGMT methylation is related to a better prognosis of the tumor. Tumor suppressors, such as p16INK4a, p14ARF and RUNX3 are also inactivated due to DNA methylation and thus promote GBM tumor development (Kanu et al., 2009).

Since DNA methylation causes tumor suppressor inactivation, the DNA Methyltransferases (DNMTs) are targeted for treatment of gliomas. The DNMT inhibitors 5-Azacitidine (5-Aza-CR) and 5-aza-2′-deoxycytidine (5-Aza-CdR) are two promising drugs targeting DNMTs (Palii et al., 2008). They incorporate into DNA during replication as analogues of cytosine. Their binding with DNMTs will reduce soluble DNMT levels and reverse DNA methylation status. Currently there are several ongoing clinical trials using these two DNMT inhibitors to treat gliomas (NCT03572530, NCT02332889, and NCT02940483).

Histone modifications mediated by histone deacetylases and histone demethylases are dysregulated in Glioma. Several histone deacetylases such as HDAC1, HDAC3, HDAC5 and HDAC9 (Staberg et al., 2017; Yang et al., 2015) have been revealed to be overexpressed in adult GBM. The high expression of HDACs is indicated to be correlated with a poor patient prognosis (Lee et al., 2017; Wang et al., 2017). Inhibition of HDAC has shown effective anti-cancer effects. Several clinical studies are currently ongoing to evaluate the therapeutic value of HDAC inhibitors including Valproic acid (NCT01817751 and NCT03048084), and Entinostat (NCT03048084) (Chen et al., 2020a). Histone demethylases are also shown abnormal expression in gliomas and could contribute to tumor development. The histone demethylase KDM1 (Amente et al., 2015), KDM2B (Staberg et al., 2018), JMJD2A (Li et al., 2020) is overexpressed in glioma samples. The high expression of these Histone demethylases promotes the proliferation of glioma cells and indicating shorter overall survival.

Interestingly, the oncometablite D-2HG produced by IDH mutation have been reported to repress several histone demethylases including KDM4A, KDM7A, and KDM2B (Liu et al., 2020a). KDM4A repression by IDH mutation leads to impaired DNA damage response to genotoxtic agents (Sulkowski et al., 2020). The histone demethylase inhibition effects can partially explain glioma patients with IDH mutation have a better prognosis.

Chromatin remodeling factors, such as lymphoid-specific helicase (LSH) and E2F1 are also found up-regulated in GBM and promote the development of GBM tumors (Xiao et al., 2017). SWI/SNF complex is reported involved in the maintenance of stemness in glioma cells (Hiramatsu et al., 2017).

9. Conclusions

The development of new treatments to improve the outcomes of patients with glioma has been heavily pursued in recent decades. TMZ has been used as the standard chemotherapy for glioma for over a decade. However, its efficacy is limited by a series of resistance mechanisms. Understanding these mechanisms, developing novel modalities of therapy, and exploring new combinations of TMZ to overcome these resistances is an important topic in glioma therapy.

The mechanisms of glioma drug resistance to TMZ are highly comprehensive and require further investigation. The different resistance mechanisms such as DNA damage repair pathways, heterogeneity of cancer cell populations, newly discovered death mode, and reprogrammed cancer metabolism indicate the need for combinatorial therapies targeting multiple signaling pathways. In addition, targeting altered metabolism might be a promising strategy for this challenging disease. Mutated IDH and its metabolic product, D-2-HG, lead to a distinctive pattern of the Krebs cycle, redox homeostasis, energy metabolism, and resistance mechanism, suggesting that selective therapy could be possible for this subtype of glioma.

Acknowledgement

This research was supported by the Intramural Research Program of the NIH, NCI.

List of nonstandard abbreviations used in the text

BCL-2

B cell lymphoma 2

BER

base excision repair

D-2-HG

D-2-hydroxyglutarate

DDR

DNA damage response

DSB

double-strand breaks

GBM

Glioblastoma

GSCs

glioma stem cells

GSH

Glutathione

HR

Homologous repair

IDH

Isocitrate dehydrogenase enzymes

MGMT

O6-methylguanine-DNA methyltransferase

MMR

Mismatch repair

N3-meA

N3-methyladenine

N3-meG

N3-methylguanine

N7-meG

N7-methylguanine

NHEJ

non-homologous end-joining

NRF2

nuclear factor erythroid 2-related factor 2

O6-meG

O6-methylguanine

PARP1

poly(ADP-ribose) polymerase 1

SLC7A11

Solute Carrier Family 7 Member 11

SSB

single-strand breaks

TMZ

temozolomide

α-KG

α-ketoglutarate

Footnotes

The authors declare no competing financial interests

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References:

  1. Aasland D, Gotzinger L, Hauck L, Berte N, Meyer J, Effenberger M, Schneider S, Reuber EE, Roos WP, Tomicic MT, et al. (2019). Temozolomide Induces Senescence and Repression of DNA Repair Pathways in Glioblastoma Cells via Activation of ATR-CHK1, p21, and NF-kappaB. Cancer research 79, 99–113. [DOI] [PubMed] [Google Scholar]
  2. Agnihotri S, Burrell K, Buczkowicz P, Remke M, Golbourn B, Chornenkyy Y, Gajadhar A, Fernandez NA, Clarke ID, Barszczyk MS, et al. (2014). ATM regulates 3-methylpurine-DNA glycosylase and promotes therapeutic resistance to alkylating agents. Cancer Discov 4, 1198–1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Agnihotri S, Gajadhar AS, Ternamian C, Gorlia T, Diefes KL, Mischel PS, Kelly J, McGown G, Thorncroft M, Carlson BL, et al. (2012). Alkylpurine-DNA-N-glycosylase confers resistance to temozolomide in xenograft models of glioblastoma multiforme and is associated with poor survival in patients. The Journal of clinical investigation 122, 253–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ahmed SU, Carruthers R, Gilmour L, Yildirim S, Watts C, and Chalmers AJ (2015). Selective Inhibition of Parallel DNA Damage Response Pathways Optimizes Radiosensitization of Glioblastoma Stem-like Cells. Cancer research 75, 4416–4428. [DOI] [PubMed] [Google Scholar]
  5. Amente S, Milazzo G, Sorrentino MC, Ambrosio S, Di Palo G, Lania L, Perini G, and Majello B (2015). Lysine-specific demethylase (LSD1/KDM1A) and MYCN cooperatively repress tumor suppressor genes in neuroblastoma. Oncotarget 6, 14572–14583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Annovazzi L, Mellai M, and Schiffer D (2017). Chemotherapeutic Drugs: DNA Damage and Repair in Glioblastoma. Cancers 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, and Rich JN (2006). Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444, 756–760. [DOI] [PubMed] [Google Scholar]
  8. Beausejour CM, Krtolica A, Galimi F, Narita M, Lowe SW, Yaswen P, and Campisi J (2003). Reversal of human cellular senescence: roles of the p53 and p16 pathways. The EMBO journal 22, 4212–4222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Blackford AN, and Jackson SP (2017). ATM, ATR, and DNA-PK: The Trinity at the Heart of the DNA Damage Response. Molecular cell 66, 801–817. [DOI] [PubMed] [Google Scholar]
  10. Blough MD, Beauchamp DC, Westgate MR, Kelly JJ, and Cairncross JG (2011). Effect of aberrant p53 function on temozolomide sensitivity of glioma cell lines and brain tumor initiating cells from glioblastoma. J Neurooncol 102, 1–7. [DOI] [PubMed] [Google Scholar]
  11. Bobola MS, Tseng SH, Blank A, Berger MS, and Silber JR (1996). Role of O6-methylguanine-DNA methyltransferase in resistance of human brain tumor cell lines to the clinically relevant methylating agents temozolomide and streptozotocin. Clin Cancer Res 2, 735–741. [PubMed] [Google Scholar]
  12. Boccard SG, Marand SV, Geraci S, Pycroft L, Berger FR, and Pelletier LA (2015). Inhibition of DNA-repair genes Ercc1 and Mgmt enhances temozolomide efficacy in gliomas treatment: a pre-clinical study. Oncotarget 6, 29456–29468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Buis J, Stoneham T, Spehalski E, and Ferguson DO (2012). Mre11 regulates CtIP-dependent double-strand break repair by interaction with CDK2. Nat Struct Mol Biol 19, 246–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cahill DP, Levine KK, Betensky RA, Codd PJ, Romany CA, Reavie LB, Batchelor TT, Futreal PA, Stratton MR, Curry WT, et al. (2007). Loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor progression during temozolomide treatment. Clin Cancer Res 13, 2038–2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cai SJ, Liu Y, Han S, and Yang C (2019). Brusatol, an NRF2 inhibitor for future cancer therapeutic. Cell Biosci 9, 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Cannan WJ, and Pederson DS (2016). Mechanisms and Consequences of Double-Strand DNA Break Formation in Chromatin. J Cell Physiol 231, 3–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Caporali S, Falcinelli S, Starace G, Russo MT, Bonmassar E, Jiricny J, and D’Atri S (2004). DNA damage induced by temozolomide signals to both ATM and ATR: Role of the mismatch repair system. Mol Pharmacol 66, 478–491. [DOI] [PubMed] [Google Scholar]
  18. Carruthers R, Ahmed SU, Strathdee K, Gomez-Roman N, Amoah-Buahin E, Watts C, and Chalmers AJ (2015). Abrogation of radioresistance in glioblastoma stem-like cells by inhibition of ATM kinase. Mol Oncol 9, 192–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Carruthers RD, Ahmed SU, Ramachandran S, Strathdee K, Kurian KM, Hedley A, Gomez-Roman N, Kalna G, Neilson M, Gilmour L, et al. (2018). Replication Stress Drives Constitutive Activation of the DNA Damage Response and Radioresistance in Glioblastoma Stem-like Cells. Cancer research 78, 5060–5071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Cartron PF, Loussouarn D, Campone M, Martin SA, and Vallette FM (2012). Prognostic impact of the expression/phosphorylation of the BH3-only proteins of the BCL-2 family in glioblastoma multiforme. Cell death & disease 3, e421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Cen L, Carlson BL, Schroeder MA, Ostrem JL, Kitange GJ, Mladek AC, Fink SR, Decker PA, Wu W, Kim JS, et al. (2012). p16-Cdk4-Rb axis controls sensitivity to a cyclin-dependent kinase inhibitor PD0332991 in glioblastoma xenograft cells. Neuro-oncology 14, 870–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chen L, Li X, Liu L, Yu B, Xue Y, and Liu Y (2015a). Erastin sensitizes glioblastoma cells to temozolomide by restraining xCT and cystathionine-gamma-lyase function. Oncol Rep 33, 1465–1474. [DOI] [PubMed] [Google Scholar]
  23. Chen R, Zhang MX, Zhou YM, Guo WJ, Yi M, Zhang ZY, Ding YP, and Wang YL (2020a). The application of histone deacetylases inhibitors in glioblastoma. J Exp Clin Canc Res 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Chen TC, Chuang JY, Ko CY, Kao TJ, Yang PY, Yu CH, Liu MS, Hu SL, Tsai YT, Chan H, et al. (2020b). AR ubiquitination induced by the curcumin analog suppresses growth of temozolomide-resistant glioblastoma through disrupting GPX4-Mediated redox homeostasis. Redox biology 30, 101413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Chen Y, Meng D, Wang H, Sun R, Wang D, Wang S, Fan J, Zhao Y, Wang J, Yang S, et al. (2015b). VAMP8 facilitates cellular proliferation and temozolomide resistance in human glioma cells. Neuro-oncology 17, 407–418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cheng Y, He C, Wang M, Ma X, Mo F, Yang S, Han J, and Wei X (2019). Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther 4, 62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Cho HR, Kumari N, Thakur N, Vu HT, Kim H, and Choi SH (2019). Decreased APE-1 by Nitroxoline Enhances Therapeutic Effect in a Temozolomide-resistant Glioblastoma: Correlation with Diffusion Weighted Imaging. Sci Rep 9, 16613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Choi HJ, Choi SH, You SH, Yoo RE, Kang KM, Yun TJ, Kim JH, Sohn CH, Park CK, and Park SH (2021). MGMT Promoter Methylation Status in Initial and Recurrent Glioblastoma: Correlation Study with DWI and DSC PWI Features. AJNR Am J Neuroradiol. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL, and Wahl GM (2006). Cancer stem cells--perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer research 66, 9339–9344. [DOI] [PubMed] [Google Scholar]
  30. Cohen AL, Holmen SL, and Colman H (2013). IDH1 and IDH2 mutations in gliomas. Curr Neurol Neurosci Rep 13, 345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Cosset E, Ilmjarv S, Dutoit V, Elliott K, von Schalscha T, Camargo MF, Reiss A, Moroishi T, Seguin L, Gomez G, et al. (2017). Glut3 Addiction Is a Druggable Vulnerability for a Molecularly Defined Subpopulation of Glioblastoma. Cancer cell 32, 856–868 e855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Dantzer F, de La Rubia G, Menissier-De Murcia J, Hostomsky Z, de Murcia G, and Schreiber V (2000). Base excision repair is impaired in mammalian cells lacking Poly(ADP-ribose) polymerase-1. Biochemistry 39, 7559–7569. [DOI] [PubMed] [Google Scholar]
  33. Dinca EB, Lu KV, Sarkaria JN, Pieper RO, Prados MD, Haas-Kogan DA, Vandenberg SR, Berger MS, and James CD (2008). p53 Small-molecule inhibitor enhances temozolomide cytotoxic activity against intracranial glioblastoma xenografts. Cancer research 68, 10034–10039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al. (2012). Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Dong Z, and Cui H (2019). Epigenetic modulation of metabolism in glioblastoma. Semin Cancer Biol 57, 45–51. [DOI] [PubMed] [Google Scholar]
  36. Durant ST, Zheng L, Wang Y, Chen K, Zhang L, Zhang T, Yang Z, Riches L, Trinidad AG, Fok JHL, et al. (2018). The brain-penetrant clinical ATM inhibitor AZD1390 radiosensitizes and improves survival of preclinical brain tumor models. Sci Adv 4, eaat1719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Eich M, Roos WP, Nikolova T, and Kaina B (2013). Contribution of ATM and ATR to the resistance of glioblastoma and malignant melanoma cells to the methylating anticancer drug temozolomide. Mol Cancer Ther 12, 2529–2540. [DOI] [PubMed] [Google Scholar]
  38. Esashi F, Christ N, Gannon J, Liu Y, Hunt T, Jasin M, and West SC (2005). CDK-dependent phosphorylation of BRCA2 as a regulatory mechanism for recombinational repair. Nature 434, 598–604. [DOI] [PubMed] [Google Scholar]
  39. Fan CH, Liu WL, Cao H, Wen C, Chen L, and Jiang G (2013). O6-methylguanine DNA methyltransferase as a promising target for the treatment of temozolomide-resistant gliomas. Cell death & disease 4, e876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Fang EF, Scheibye-Knudsen M, Chua KF, Mattson MP, Croteau DL, and Bohr VA (2016). Nuclear DNA damage signalling to mitochondria in ageing. Nat Rev Mol Cell Biol 17, 308–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Felsberg J, Thon N, Eigenbrod S, Hentschel B, Sabel MC, Westphal M, Schackert G, Kreth FW, Pietsch T, Loffler M, et al. (2011). Promoter methylation and expression of MGMT and the DNA mismatch repair genes MLH1, MSH2, MSH6 and PMS2 in paired primary and recurrent glioblastomas. Int J Cancer 129, 659–670. [DOI] [PubMed] [Google Scholar]
  42. Feng J, Yan PF, Zhao HY, Zhang FC, Zhao WH, and Feng M (2016). Inhibitor of Nicotinamide Phosphoribosyltransferase Sensitizes Glioblastoma Cells to Temozolomide via Activating ROS/JNK Signaling Pathway. Biomed Res Int 2016, 1450843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Ferri A, Stagni V, and Barila D (2020). Targeting the DNA Damage Response to Overcome Cancer Drug Resistance in Glioblastoma. Int J Mol Sci 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Filippi-Chiela EC, Bueno e Silva MM, Thome MP, and Lenz G (2015). Single-cell analysis challenges the connection between autophagy and senescence induced by DNA damage. Autophagy 11, 1099–1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Flavahan WA, Wu Q, Hitomi M, Rahim N, Kim Y, Sloan AE, Weil RJ, Nakano I, Sarkaria JN, Stringer BW, et al. (2013). Brain tumor initiating cells adapt to restricted nutrition through preferential glucose uptake. Nat Neurosci 16, 1373–1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Frattini V, Pagnotta SM, Tala, Fan JJ, Russo MV, Lee SB, Garofano L, Zhang J, Shi P, Lewis G, et al. (2018). A metabolic function of FGFR3-TACC3 gene fusions in cancer. Nature 553, 222–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Friedman HS, Dolan ME, Pegg AE, Marcelli S, Keir S, Catino JJ, Bigner DD, and Schold SC Jr. (1995). Activity of temozolomide in the treatment of central nervous system tumor xenografts. Cancer research 55, 2853–2857. [PubMed] [Google Scholar]
  48. Frosina G, Profumo A, Marubbi D, Marcello D, Ravetti JL, and Daga A (2018). ATR kinase inhibitors NVP-BEZ235 and AZD6738 effectively penetrate the brain after systemic administration. Radiat Oncol 13, 76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Fukushima T, Takeshima H, and Kataoka H (2009). Anti-glioma therapy with temozolomide and status of the DNA-repair gene MGMT. Anticancer Res 29, 4845–4854. [PubMed] [Google Scholar]
  50. Galia A, Calogero AE, Condorelli R, Fraggetta F, La Corte A, Ridolfo F, Bosco P, Castiglione R, and Salemi M (2012). PARP-1 protein expression in glioblastoma multiforme. Eur J Histochem 56, e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Georgakilas AG, Martin OA, and Bonner WM (2017). p21: A Two-Faced Genome Guardian. Trends Mol Med 23, 310–319. [DOI] [PubMed] [Google Scholar]
  52. Gil del Alcazar CR, Hardebeck MC, Mukherjee B, Tomimatsu N, Gao X, Yan J, Xie XJ, Bachoo R, Li L, Habib AA, et al. (2014). Inhibition of DNA double-strand break repair by the dual PI3K/mTOR inhibitor NVP-BEZ235 as a strategy for radiosensitization of glioblastoma. Clin Cancer Res 20, 1235–1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Gil Del Alcazar CR, Todorova PK, Habib AA, Mukherjee B, and Burma S (2016). Augmented HR Repair Mediates Acquired Temozolomide Resistance in Glioblastoma. Mol Cancer Res 14, 928–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Glassner BJ, Weeda G, Allan JM, Broekhof JL, Carls NH, Donker I, Engelward BP, Hampson RJ, Hersmus R, Hickman MJ, et al. (1999). DNA repair methyltransferase (Mgmt) knockout mice are sensitive to the lethal effects of chemotherapeutic alkylating agents. Mutagenesis 14, 339–347. [DOI] [PubMed] [Google Scholar]
  55. Gobin M, Nazarov PV, Warta R, Timmer M, Reifenberger G, Felsberg J, Vallar L, Chalmers AJ, Herold-Mende CC, Goldbrunner R, et al. (2019). A DNA Repair and Cell-Cycle Gene Expression Signature in Primary and Recurrent Glioblastoma: Prognostic Value and Clinical Implications. Cancer research 79, 1226–1238. [DOI] [PubMed] [Google Scholar]
  56. Goellner EM, Grimme B, Brown AR, Lin YC, Wang XH, Sugrue KF, Mitchell L, Trivedi RN, Tang JB, and Sobol RW (2011). Overcoming temozolomide resistance in glioblastoma via dual inhibition of NAD+ biosynthesis and base excision repair. Cancer research 71, 2308–2317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Goellner EM, Svilar D, Almeida KH, and Sobol RW (2012). Targeting DNA polymerase ss for therapeutic intervention. Curr Mol Pharmacol 5, 68–87. [PMC free article] [PubMed] [Google Scholar]
  58. Gu Y, Albuquerque CP, Braas D, Zhang W, Villa GR, Bi J, Ikegami S, Masui K, Gini B, Yang H, et al. (2017). mTORC2 Regulates Amino Acid Metabolism in Cancer by Phosphorylation of the Cystine-Glutamate Antiporter xCT. Mol Cell 67, 128–138 e127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Gujar AD, Le S, Mao DD, Dadey DY, Turski A, Sasaki Y, Aum D, Luo J, Dahiya S, Yuan L, et al. (2016). An NAD+-dependent transcriptional program governs self-renewal and radiation resistance in glioblastoma. Proc Natl Acad Sci U S A 113, E8247–E8256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Gusyatiner O, and Hegi ME (2018). Glioma epigenetics: From subclassification to novel treatment options. Semin Cancer Biol 51, 50–58. [DOI] [PubMed] [Google Scholar]
  61. Han S, Liu Y, Cai SJ, Qian M, Ding J, Larion M, Gilbert MR, and Yang C (2020). IDH mutation in glioma: molecular mechanisms and potential therapeutic targets. Br J Cancer 122, 1580–1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Happold C, Roth P, Wick W, Schmidt N, Florea AM, Silginer M, Reifenberger G, and Weller M (2012). Distinct molecular mechanisms of acquired resistance to temozolomide in glioblastoma cells. Journal of neurochemistry 122, 444–455. [DOI] [PubMed] [Google Scholar]
  63. Heale JT, Ball AR Jr., Schmiesing JA, Kim JS, Kong X, Zhou S, Hudson DF, Earnshaw WC, and Yokomori K (2006). Condensin I interacts with the PARP-1-XRCC1 complex and functions in DNA single-strand break repair. Molecular cell 21, 837–848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, et al. (2005). MGMT gene silencing and benefit from temozolomide in glioblastoma. The New England journal of medicine 352, 997–1003. [DOI] [PubMed] [Google Scholar]
  65. Hermisson M, Klumpp A, Wick W, Wischhusen J, Nagel G, Roos W, Kaina B, and Weller M (2006). O6-methylguanine DNA methyltransferase and p53 status predict temozolomide sensitivity in human malignant glioma cells. Journal of neurochemistry 96, 766–776. [DOI] [PubMed] [Google Scholar]
  66. Hiramatsu H, Kobayashi K, Kobayashi K, Haraguchi T, Ino Y, Todo T, and Iba H (2017). The role of the SWI/SNF chromatin remodeling complex in maintaining the stemness of glioma initiating cells. Sci Rep 7, 889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Huang RX, and Zhou PK (2020). DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther 5, 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Hurtado-Bages S, Knobloch G, Ladurner AG, and Buschbeck M (2020). The taming of PARP1 and its impact on NAD(+) metabolism. Mol Metab 38, 100950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Jackson SP, and Bartek J (2009). The DNA-damage response in human biology and disease. Nature 461, 1071–1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Jacobs AL, and Schar P (2012). DNA glycosylases: in DNA repair and beyond. Chromosoma 121, 1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Janiszewska M, Suva ML, Riggi N, Houtkooper RH, Auwerx J, Clement-Schatlo V, Radovanovic I, Rheinbay E, Provero P, and Stamenkovic I (2012). Imp2 controls oxidative phosphorylation and is crucial for preserving glioblastoma cancer stem cells. Genes Dev 26, 1926–1944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R, and Gu W (2015). Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520, 57–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Johannessen TC, Prestegarden L, Grudic A, Hegi ME, Tysnes BB, and Bjerkvig R (2013). The DNA repair protein ALKBH2 mediates temozolomide resistance in human glioblastoma cells. Neuro-oncology 15, 269–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Kaina B (2019). Temozolomide in Glioblastoma Therapy: Role of Apoptosis, Senescence and Autophagy. Comment on Strobel et al., Temozolomide and Other Alkylating Agents in Glioblastoma Therapy. Biomedicines 2019, 7, 69. Biomedicines 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Kanu OO, Hughes B, Di CH, Lin NJ, Fu JR, Bigner DD, Yan H, and Adamson C (2009). Glioblastoma Multiforme Oncogenomics and Signaling Pathways. Clin Med Insights-On 3, 39–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Karlin J, Allen J, Ahmad SF, Hughes G, Sheridan V, Odedra R, Farrington P, Cadogan EB, Riches LC, Garcia-Trinidad A, et al. (2018). Orally Bioavailable and Blood-Brain Barrier-Penetrating ATM Inhibitor (AZ32) Radiosensitizes Intracranial Gliomas in Mice. Mol Cancer Ther 17, 1637–1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Karran P (2001). Mechanisms of tolerance to DNA damaging therapeutic drugs. Carcinogenesis 22, 1931–1937. [DOI] [PubMed] [Google Scholar]
  78. King HO, Brend T, Payne HL, Wright A, Ward TA, Patel K, Egnuni T, Stead LF, Patel A, Wurdak H, et al. (2017). RAD51 Is a Selective DNA Repair Target to Radiosensitize Glioma Stem Cells. Stem Cell Reports 8, 125–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Knijnenburg TA, Wang L, Zimmermann MT, Chambwe N, Gao GF, Cherniack AD, Fan H, Shen H, Way GP, Greene CS, et al. (2018). Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas. Cell Rep 23, 239–254 e236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Knizhnik AV, Roos WP, Nikolova T, Quiros S, Tomaszowski KH, Christmann M, and Kaina B (2013). Survival and death strategies in glioma cells: autophagy, senescence and apoptosis triggered by a single type of temozolomide-induced DNA damage. PloS one 8, e55665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Kouri FM, Jensen SA, and Stegh AH (2012). The role of Bcl-2 family proteins in therapy responses of malignant astrocytic gliomas: Bcl2L12 and beyond. ScientificWorldJournal 2012, 838916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Krokan HE, and Bjoras M (2013). Base excision repair. Cold Spring Harb Perspect Biol 5, a012583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Lee DH, Ryu HW, Won HR, and Kwon SH (2017). Advances in epigenetic glioblastoma therapy. Oncotarget 8, 18577–18589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Li M, Cheng J, Ma Y, Guo H, Shu H, Huang H, Kuang Y, and Yang T (2020). The histone demethylase JMJD2A promotes glioma cell growth via targeting Akt-mTOR signaling. Cancer Cell Int 20, 101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Lim YC, Roberts TL, Day BW, Harding A, Kozlov S, Kijas AW, Ensbey KS, Walker DG, and Lavin MF (2012). A role for homologous recombination and abnormal cell-cycle progression in radioresistance of glioma-initiating cells. Mol Cancer Ther 11, 1863–1872. [DOI] [PubMed] [Google Scholar]
  86. Liu Y, Lang F, Chou FJ, Zaghloul KA, and Yang C (2020a). Isocitrate Dehydrogenase Mutations in Glioma: Genetics, Biochemistry, and Clinical Indications. Biomedicines 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Liu Y, Lang F, and Yang C (2020b). NRF2 in human neoplasm: Cancer biology and potential therapeutic target. Pharmacology & therapeutics, 107664. [DOI] [PubMed] [Google Scholar]
  88. Liu Y, Lu Y, Celiku O, Li A, Wu Q, Zhou Y, and Yang C (2019). Targeting IDH1-Mutated Malignancies with NRF2 Blockade. J Natl Cancer Inst 111, 1033–1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Liu Y, Pang Y, Caisova V, Ding J, Yu D, Zhou Y, Huynh TT, Ghayee H, Pacak K, and Yang C (2020c). Targeting NRF2-Governed Glutathione Synthesis for SDHB-Mutated Pheochromocytoma and Paraganglioma. Cancers (Basel) 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, and Ellison DW (2016). The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol 131, 803–820. [DOI] [PubMed] [Google Scholar]
  91. M JD, and Wojtas B (2019). Global DNA Methylation Patterns in Human Gliomas and Their Interplay with Other Epigenetic Modifications. Int J Mol Sci 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. MacLeod G, Bozek DA, Rajakulendran N, Monteiro V, Ahmadi M, Steinhart Z, Kushida MM, Yu H, Coutinho FJ, Cavalli FMG, et al. (2019). Genome-Wide CRISPR-Cas9 Screens Expose Genetic Vulnerabilities and Mechanisms of Temozolomide Sensitivity in Glioblastoma Stem Cells. Cell Reports 27, 971-+. [DOI] [PubMed] [Google Scholar]
  93. Malta TM, de Souza CF, Sabedot TS, Silva TC, Mosella MS, Kalkanis SN, Snyder J, Castro AVB, and Noushmehr H (2018). Glioma CpG island methylator phenotype (G-CIMP): biological and clinical implications. Neuro-oncology 20, 608–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. McFaline-Figueroa JL, Braun CJ, Stanciu M, Nagel ZD, Mazzucato P, Sangaraju D, Cerniauskas E, Barford K, Vargas A, Chen Y, et al. (2015). Minor Changes in Expression of the Mismatch Repair Protein MSH2 Exert a Major Impact on Glioblastoma Response to Temozolomide. Cancer research 75, 3127–3138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Middleton MR, Thatcher N, McMurry TB, McElhinney RS, Donnelly DJ, and Margison GP (2002). Effect of O6-(4-bromothenyl)guanine on different temozolomide schedules in a human melanoma xenograft model. Int J Cancer 100, 615–617. [DOI] [PubMed] [Google Scholar]
  96. Minniti G, Scaringi C, Arcella A, Lanzetta G, Di Stefano D, Scarpino S, Bozzao A, Pace A, Villani V, Salvati M, et al. (2014). IDH1 mutation and MGMT methylation status predict survival in patients with anaplastic astrocytoma treated with temozolomide-based chemoradiotherapy. Journal of neuro-oncology 118, 377–383. [DOI] [PubMed] [Google Scholar]
  97. Miyata H, Ashizawa T, Iizuka A, Kondou R, Nonomura C, Sugino T, Urakami K, Asai A, Hayashi N, Mitsuya K, et al. (2017). Combination of a STAT3 Inhibitor and an mTOR Inhibitor Against a Temozolomide-resistant Glioblastoma Cell Line. Cancer Genomics Proteomics 14, 83–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Mohammad RM, Muqbil I, Lowe L, Yedjou C, Hsu HY, Lin LT, Siegelin MD, Fimognari C, Kumar NB, Dou QP, et al. (2015). Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol 35 Suppl, S78–S103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Montaldi AP, Godoy PR, and Sakamoto-Hojo ET (2015). APE1/REF-1 down-regulation enhances the cytotoxic effects of temozolomide in a resistant glioblastoma cell line. Mutat Res Genet Toxicol Environ Mutagen 793, 19–29. [DOI] [PubMed] [Google Scholar]
  100. Montaldo NP, Bordin DL, Brambilla A, Rosinger M, Fordyce Martin SL, Bjoras KO, Bradamante S, Aas PA, Furrer A, Olsen LC, et al. (2019). Alkyladenine DNA glycosylase associates with transcription elongation to coordinate DNA repair with gene expression. Nat Commun 10, 5460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Nagane M, Levitzki A, Gazit A, Cavenee WK, and Huang HJ (1998). Drug resistance of human glioblastoma cells conferred by a tumor-specific mutant epidermal growth factor receptor through modulation of Bcl-XL and caspase-3-like proteases. Proceedings of the National Academy of Sciences of the United States of America 95, 5724–5729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Ning JF, Stanciu M, Humphrey MR, Gorham J, Wakimoto H, Nishihara R, Lees J, Zou L, Martuza RL, Wakimoto H, et al. (2019). Myc targeted CDK18 promotes ATR and homologous recombination to mediate PARP inhibitor resistance in glioblastoma. Nat Commun 10, 2910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Oike T, Suzuki Y, Sugawara K, Shirai K, Noda SE, Tamaki T, Nagaishi M, Yokoo H, Nakazato Y, and Nakano T (2013). Radiotherapy plus concomitant adjuvant temozolomide for glioblastoma: Japanese mono-institutional results. PLoS One 8, e78943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Oldrini B, Vaquero-Siguero N, Mu Q, Kroon P, Zhang Y, Galan-Ganga M, Bao Z, Wang Z, Liu H, Sa JK, et al. (2020). MGMT genomic rearrangements contribute to chemotherapy resistance in gliomas. Nat Commun 11, 3883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, and Barnholtz-Sloan JS (2020). CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013–2017. Neuro Oncol 22, iv1–iv96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Palii SS, Van Emburgh BO, Sankpal UT, Brown KD, and Robertson KD (2008). DNA methylation inhibitor 5-aza-2 ‘-deoxycytidine induces reversible genome-wide DNA damage that is distinctly influenced by DNA methyltransferases 1 and 3B. Mol Cell Biol 28, 752–771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Pavlyukov MS, Yu H, Bastola S, Minata M, Shender VO, Lee Y, Zhang S, Wang J, Komarova S, Wang J, et al. (2018). Apoptotic Cell-Derived Extracellular Vesicles Promote Malignancy of Glioblastoma Via Intercellular Transfer of Splicing Factors. Cancer cell 34, 119–135 e110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Polewski MD, Reveron-Thornton RF, Cherryholmes GA, Marinov GK, Cassady K, and Aboody KS (2016). Increased Expression of System xc- in Glioblastoma Confers an Altered Metabolic State and Temozolomide Resistance. Mol Cancer Res 14, 1229–1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Prager BC, Bhargava S, Mahadev V, Hubert CG, and Rich JN (2020). Glioblastoma Stem Cells: Driving Resilience through Chaos. Trends Cancer 6, 223–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Rahman MA, Navarro AG, Brekke J, Engelsen A, Bindesboll C, Sarowar S, Bahador M, Bifulco E, Goplen D, Waha A, et al. (2019). Bortezomib administered prior to temozolomide depletes MGMT, chemosensitizes glioblastoma with unmethylated MGMT promoter and prolongs animal survival. British journal of cancer 121, 545–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Rasmussen RD, Gajjar MK, Jensen KE, and Hamerlik P (2016). Enhanced efficacy of combined HDAC and PARP targeting in glioblastoma. Mol Oncol 10, 751–763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Ricci MS, and Zong WX (2006). Chemotherapeutic approaches for targeting cell death pathways. Oncologist 11, 342–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Rocha CR, Garcia CC, Vieira DB, Quinet A, de Andrade-Lima LC, Munford V, Belizario JE, and Menck CF (2014). Glutathione depletion sensitizes cisplatin- and temozolomide-resistant glioma cells in vitro and in vivo. Cell Death Dis 5, e1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Rocha CR, Kajitani GS, Quinet A, Fortunato RS, and Menck CF (2016). NRF2 and glutathione are key resistance mediators to temozolomide in glioma and melanoma cells. Oncotarget 7, 48081–48092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Ruiz-Rodado V, Malta TM, Seki T, Lita A, Dowdy T, Celiku O, Cavazos-Saldana A, Li A, Liu Y, Han S, et al. (2020). Metabolic reprogramming associated with aggressiveness occurs in the G-CIMP-high molecular subtypes of IDH1mut lower grade gliomas. Neuro-oncology 22, 480–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Sampath D, Zabka TS, Misner DL, O’Brien T, and Dragovich PS (2015). Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) as a therapeutic strategy in cancer. Pharmacol Ther 151, 16–31. [DOI] [PubMed] [Google Scholar]
  117. Sarkaria JN, Kitange GJ, James CD, Plummer R, Calvert H, Weller M, and Wick W (2008a). Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clin Cancer Res 14, 2900–2908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Sarkaria JN, Kitange GJ, James CD, Plummer R, Calvert H, Weller M, and Wick W (2008b). Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clinical Cancer Research 14, 2900–2908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Sehm T, Rauh M, Wiendieck K, Buchfelder M, Eyupoglu IY, and Savaskan NE (2016). Temozolomide toxicity operates in a xCT/SLC7a11 dependent manner and is fostered by ferroptosis. Oncotarget 7, 74630–74647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Squatrito M, Brennan CW, Helmy K, Huse JT, Petrini JH, and Holland EC (2010). Loss of ATM/Chk2/p53 pathway components accelerates tumor development and contributes to radiation resistance in gliomas. Cancer cell 18, 619–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Staberg M, Michaelsen SR, Rasmussen RD, Villingshoj M, Poulsen H, and Hamerlik P (2017). Inhibition of histone deacetylases sensitizes glioblastoma cells to lomustine. Cell Oncol 40, 21–32. [DOI] [PubMed] [Google Scholar]
  122. Staberg M, Rasmussen RD, Michaelsen SR, Pedersen H, Jensen KE, Villingshoj M, Skjoth-Rasmussen J, Brennum J, Vitting-Seerup K, Poulsen HS, et al. (2018). Targeting glioma stem-like cell survival and chemoresistance through inhibition of lysine-specific histone demethylase KDM2B. Mol Oncol 12, 406–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Storey K, Leder K, Hawkins-Daarud A, Swanson K, Ahmed AU, Rockne RC, and Foo J (2019). Glioblastoma Recurrence and the Role of O(6)-Methylguanine-DNA Methyltransferase Promoter Methylation. JCO Clin Cancer Inform 3, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Stupp R, Hegi ME, Gilbert MR, and Chakravarti A (2007). Chemoradiotherapy in malignant glioma: standard of care and future directions. J Clin Oncol 25, 4127–4136. [DOI] [PubMed] [Google Scholar]
  125. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al. (2009). Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10, 459–466. [DOI] [PubMed] [Google Scholar]
  126. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al. (2005). Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352, 987–996. [DOI] [PubMed] [Google Scholar]
  127. Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, Han W, Lou F, Yang J, Zhang Q, et al. (2013). Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell death & disease 4, e838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Sulkowski PL, Corso CD, Robinson ND, Scanlon SE, Purshouse KR, Bai H, Liu Y, Sundaram RK, Hegan DC, Fons NR, et al. (2017). 2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivity. Sci Transl Med 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Sulkowski PL, Oeck S, Dow J, Economos NG, Mirfakhraie L, Liu Y, Noronha K, Bao X, Li J, Shuch BM, et al. (2020). Oncometabolites suppress DNA repair by disrupting local chromatin signalling. Nature 582, 586–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Tanaka K, Sasayama T, Irino Y, Takata K, Nagashima H, Satoh N, Kyotani K, Mizowaki T, Imahori T, Ejima Y, et al. (2015). Compensatory glutamine metabolism promotes glioblastoma resistance to mTOR inhibitor treatment. J Clin Invest 125, 1591–1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Tang X, Fu X, Liu Y, Yu D, Cai SJ, and Yang C (2020). Blockade of Glutathione Metabolism in IDH1-Mutated Glioma. Mol Cancer Ther 19, 221–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Tardito S, Oudin A, Ahmed SU, Fack F, Keunen O, Zheng L, Miletic H, Sakariassen PO, Weinstock A, Wagner A, et al. (2015). Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastoma. Nat Cell Biol 17, 1556–1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Tawbi HA, Beumer JH, Tarhini AA, Moschos S, Buch SC, Egorin MJ, Lin Y, Christner S, and Kirkwood JM (2013). Safety and efficacy of decitabine in combination with temozolomide in metastatic melanoma: a phase I/II study and pharmacokinetic analysis. Ann Oncol 24, 1112–1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Tomimatsu N, Mukherjee B, Catherine Hardebeck M, Ilcheva M, Vanessa Camacho C, Louise Harris J, Porteus M, Llorente B, Khanna KK, and Burma S (2014). Phosphorylation of EXO1 by CDKs 1 and 2 regulates DNA end resection and repair pathway choice. Nat Commun 5, 3561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Trivedi RN, Wang XH, Jelezcova E, Goellner EM, Tang JB, and Sobol RW (2008). Human methyl purine DNA glycosylase and DNA polymerase beta expression collectively predict sensitivity to temozolomide. Mol Pharmacol 74, 505–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Turcan S, Rohle D, Goenka A, Walsh LA, Fang F, Yilmaz E, Campos C, Fabius AWM, Lu C, Ward PS, et al. (2012). IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483, 479–U137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Vendetti FP, Lau A, Schamus S, Conrads TP, O’Connor MJ, and Bakkenist CJ (2015). The orally active and bioavailable ATR kinase inhibitor AZD6738 potentiates the anti-tumor effects of cisplatin to resolve ATM-deficient non-small cell lung cancer in vivo. Oncotarget 6, 44289–44305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Vescovi AL, Galli R, and Reynolds BA (2006). Brain tumour stem cells. Nat Rev Cancer 6, 425–436. [DOI] [PubMed] [Google Scholar]
  139. Villano JL, Seery TE, and Bressler LR (2009). Temozolomide in malignant gliomas: current use and future targets. Cancer Chemother Pharmacol 64, 647–655. [DOI] [PubMed] [Google Scholar]
  140. Vlashi E, Lagadec C, Vergnes L, Matsutani T, Masui K, Poulou M, Popescu R, Della Donna L, Evers P, Dekmezian C, et al. (2011). Metabolic state of glioma stem cells and nontumorigenic cells. Proceedings of the National Academy of Sciences of the United States of America 108, 16062–16067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Wang J, Cazzato E, Ladewig E, Frattini V, Rosenbloom DI, Zairis S, Abate F, Liu Z, Elliott O, Shin YJ, et al. (2016a). Clonal evolution of glioblastoma under therapy. Nat Genet 48, 768–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Wang J, Yang T, Xu G, Liu H, Ren C, Xie W, and Wang M (2016b). Cyclin-Dependent Kinase 2 Promotes Tumor Proliferation and Induces Radio Resistance in Glioblastoma. Transl Oncol 9, 548–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, Wilson KF, Ambrosio AL, Dias SM, Dang CV, et al. (2010). Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell 18, 207–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Wang P, Wu J, Ma S, Zhang L, Yao J, Hoadley KA, Wilkerson MD, Perou CM, Guan KL, Ye D, et al. (2015). Oncometabolite D-2-Hydroxyglutarate Inhibits ALKBH DNA Repair Enzymes and Sensitizes IDH Mutant Cells to Alkylating Agents. Cell Rep 13, 2353–2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Wang XQ, Bai HM, Li ST, Sun H, Min LZ, Tao BB, Zhong J, and Li B (2017). Knockdown of HDAC1 expression suppresses invasion and induces apoptosis in glioma cells. Oncotarget 8, 48027–48040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Warren KE, Aikin AA, Libucha M, Widemann BC, Fox E, Packer RJ, and Balis FM (2005). Phase I study of O-6-benzylguanine and temozolomide administered daily for 5 days to pediatric patients with solid tumors. Journal of Clinical Oncology 23, 7646–7653. [DOI] [PubMed] [Google Scholar]
  147. Wei H, and Yu X (2016). Functions of PARylation in DNA Damage Repair Pathways. Genomics Proteomics Bioinformatics 14, 131–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Wirtz S, Nagel G, Eshkind L, Neurath MF, Samson LD, and Kaina B (2010). Both base excision repair and O-6-methylguanine-DNA methyltransferase protect against methylation-induced colon carcinogenesis. Carcinogenesis 31, 2111–2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Wurstle S, Schneider F, Ringel F, Gempt J, Lammer F, Delbridge C, Wu W, and Schlegel J (2017). Temozolomide induces autophagy in primary and established glioblastoma cells in an EGFR independent manner. Oncol Lett 14, 322–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Xiao D, Huang J, Pan Y, Li H, Fu C, Mao C, Cheng Y, Shi Y, Chen L, Jiang Y, et al. (2017). Chromatin Remodeling Factor LSH is Upregulated by the LRP6-GSK3beta-E2F1 Axis Linking Reversely with Survival in Gliomas. Theranostics 7, 132–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, Kang R, and Tang D (2016). Ferroptosis: process and function. Cell death and differentiation 23, 369–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Xu GW, Mymryk JS, and Cairncross JG (2005). Pharmaceutical-mediated inactivation of p53 sensitizes U87MG glioma cells to BCNU and temozolomide. Int J Cancer 116, 187–192. [DOI] [PubMed] [Google Scholar]
  153. Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Wang P, Xiao MT, et al. (2011). Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer cell 19, 17–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Yang R, Wu YN, Wang M, Sun ZF, Zou JH, Zhang YD, and Cui HJ (2015). HDAC9 promotes glioblastoma growth via TAZ-mediated EGFR pathway activation. Oncotarget 6, 7644–7656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Yu D, Liu Y, Zhou Y, Ruiz-Rodado V, Larion M, Xu G, and Yang C (2020). Triptolide suppresses IDH1-mutated malignancy via Nrf2-driven glutathione metabolism. Proc Natl Acad Sci U S A 117, 9964–9972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Zhang J, Stevens MF, and Bradshaw TD (2012). Temozolomide: mechanisms of action, repair and resistance. Curr Mol Pharmacol 5, 102–114. [DOI] [PubMed] [Google Scholar]
  157. Zhang L, and Wang H (2017). FTY720 inhibits the Nrf2/ARE pathway in human glioblastoma cell lines and sensitizes glioblastoma cells to temozolomide. Pharmacol Rep 69, 1186–1193. [DOI] [PubMed] [Google Scholar]
  158. Zhang Y, Xia Q, and Lin J (2018). Identification of the potential oncogenes in glioblastoma based on bioinformatic analysis and elucidation of the underlying mechanisms. Oncology reports 40, 715–725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Zhao H, Ji B, Chen J, Huang Q, and Lu X (2017). Gpx 4 is involved in the proliferation, migration and apoptosis of glioma cells. Pathology, research and practice 213, 626–633. [DOI] [PubMed] [Google Scholar]
  160. Zhou W, and Wahl DR (2019). Metabolic Abnormalities in Glioblastoma and Metabolic Strategies to Overcome Treatment Resistance. Cancers (Basel) 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Zhu Z, Du S, Du Y, Ren J, Ying G, and Yan Z (2018). Glutathione reductase mediates drug resistance in glioblastoma cells by regulating redox homeostasis. Journal of neurochemistry 144, 93–104. [DOI] [PubMed] [Google Scholar]

RESOURCES