Open Access

Regulation of temozolomide resistance via lncRNAs: Clinical and biological properties of lncRNAs in gliomas (Review)

  • Authors:
    • Sui Li
    • Xiaofang Xie
    • Fu Peng
    • Junrong Du
    • Cheng Peng
  • View Affiliations

  • Published online on: July 5, 2022     https://doi.org/10.3892/ijo.2022.5391
  • Article Number: 101
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gliomas are a primary types of intracranial malignancies and are characterized by a poor prognosis due to aggressive recurrence profiles. Temozolomide (TMZ) is an auxiliary alkylating agent that is extensively used in conjunction with surgical resection and forms the mainstay of clinical treatment strategies for gliomas. However, the frequent occurrence of TMZ resistance in clinical practice limits its therapeutic efficacy. Accumulating evidence has demonstrated that long non‑coding RNAs (lncRNAs) can play key and varied roles in glioma progression. lncRNAs have been reported to inhibit glioma progression by targeting various signaling pathways. In addition, the differential expression of lncRNAs has also been found to mediate the resistance of glioma to several chemotherapeutic agents, particularly to TMZ. The present review article therefore summarizes the findings of previous studies in an aim to report the significance and function of lncRNAs in regulating the chemoresistance of gliomas. The present review may provide further insight into the clinical treatment of gliomas.

1. Introduction

The World Health Organization (WHO) currently classifies gliomas into four grades, where a new classification was proposed in 2016 (1). These grades of interest are as follows: i) Diffuse low-grade gliomas (WHO I and II), which include diffuse astrocytoma and oligodendroglioma; ii) diffuse high-grade gliomas (WHO III), which include anaplastic astrocytoma with various mutated genes; and iii) glioblastoma (GBM; WHO IV), which is the most lethal variety of glioma (2). Glioblastoma multiforme (GBM) is a common type of brain malignancy that remains largely incurable. Despite the development of multi-modal therapeutic interventions, such as radiotherapy combined with temozolomide (TMZ) (3) and electrofield therapy combined with TMZ (4), the median survival rate of patients with GBM remains at ≤15 months (5).

TMZ is an orally administered alkylating antitumor drug that can cross the blood-brain barrier with a bioavailability reaching ~100%. It can be used to effectively treat newly diagnosed and relapsed GBM to prolong the survival rate of patients (6,7). Thus, TMZ is the main adjuvant therapeutic agent used in the treatment of glioma in clinical practice (8). However, the development of resistance to TMZ in glioma impairs its therapeutic efficacy, resulting in recurrence (9). A previous study evaluated the responses of 1,035 GBM tumors to therapies using multiple assaying technologies and discovered that gene mutations are largely responsible for resistance to treatment. The mechanisms of action of TMZ involve the alkylation of the O6 site of guanine (O6MeG), resulting in a base mismatch following subsequent DNA replication, causing DNA damage (10). TMZ resistance has become one of the largest obstacles in prolonging the survival of patients with glioma. Several mechanisms have been proposed to underlie the resistance to TMZ. The most common mode lies in the DNA repair system, including the O6MeG DNA methyltransferase (MGMT) pathway (11), DNA mismatch repair and base excision repair (12,13). In addition to the DNA repair system, GBM stemness and autophagy can play critical roles in mediating resistance to TMZ. Other factors, such as the EGFR, PTEN, p53 and PI3K/AKT/mTOR pathways, have all been reported to participate in the resistance process. Due to the high occurrence of TMZ resistance, the development of novel effective treatment methods for gliomas is of utmost urgency.

According to The Encyclopedia of DNA Elements statistics, RNAs account for 75% of the human genome, whilst the ratio of RNA to protein-coding genes is only ~3% (14). Influenced by the central genetic dogma, the untranslated region of genes was previously considered by prejudice to be 'junk' (15). However, at the end of the last century, the discovery of small regulatory non-coding RNAs (ncRNAs) has completely changed the understanding of the role of ncRNAs (16). Among these ncRNAs, there is a class of RNAs with base-pair lengths of ≥200 nucleotides, named long non-coding RNAs (lncRNAs). The function of lncRNAs in protein expression is similar to that of conventional RNAs. Both are transcribed from DNA by RNA polymerase II and are then modified, namely being 5′-capped, polyadenylated and/or spliced, before eventually becoming mature lncRNA (17,18). Compared with mRNAs, lncRNAs contain fewer but longer exons, where their expression levels in different tissues are typically lower (19,20). lncRNAs have been reported to perform a variety of molecular functions. They have been reported to regulate gene expression directly and indirectly by targeting specific proteins, including Polycomb group proteins and enhancer of zeste homolog 2 (EZH2). In addition, lncRNAs can also function as precursors of small RNAs and modulate RNA processing events. lncRNAs can also directly bind to proteins and modulate their function (21). It has been reported that lncRNAs and microRNAs (miRNAs/miRs) typically perform reciprocal functions. miRNAs can promote the degradation of lncRNAs, thereby diminishing the molecular functions of lncRNAs, whereas lncRNAs can also conversely target miRNAs directly to regulate their expression. lncRNAs can compete with miRNAs for binding to the same sites on mRNAs. Furthermore, lncRNAs can be spliced to generate miRNAs (22). Accumulating evidence has indicated that lncRNAs can function as regulatory molecules in cancer development. Han et al (23) previously reported key lncRNAs that can influence glioma pathogenesis using microarrays. However, the most recent network-based algorithms failed to adequately reflect the regulatory role in human tumors. Zhang et al (24) successfully developed a novel global network-based framework, termed 'LncRDNetFlow', to verify that lncRNAs play a key role in regulating cellular processes in different malignancies. Apart from the aforementioned reports, lncRNAs that are associated with cancer have been previously detected in the body fluids of patients with cancer, which suggests the potential of using lncRNAs as biomarkers and therapeutic targets for cancer applications (25). Of note, lncRNAs have been reported to participate in the development of resistance to chemotherapy in various human malignancies, such as head and neck squamous cell carcinoma, hepatocellular carcinoma, gastric cancer, pancreatic cancer, bladder cancer, including glioma (26-32).

In conclusion, lncRNAs are promising therapeutic targets for the treatment of human diseases, particularly glioma. Therefore, the present review article first summarizes research findings from the past 5 years regarding the signaling pathways underlying the resistance of glioma to TMZ, with a focus on oncogenic or tumor suppressive lncRNAs. Moreover, the association between lncRNAs and combination therapy with TMZ and immunosuppressive therapy for glioma is discussed, in an aim to provide further insight into the clinical treatment of glioma.

2. Prominent role of lncRNAs in glioma

In 2012, Zhang et al (33) first associated lncRNA expression with the grade of malignancy in human glioma. Furthermore, lncRNAs have been reported to be associated with the occurrence and progression of various malignancies such as colorectal cancer, gallbladder cancer, ovarian cancer in addition to modulating chemoresistance in glioma (34-38). The expression of a number of lncRNAs has been found to be upregulated in glioma and these lncRNAs have been proposed to function as oncogenes, whilst the expression levels of other lncRNAs have been found to be downregulated in glioma and those lncRNAs have been proposed to function as tumor suppressor genes. From an epigenetic perspective, lncRNAs play a key role in glioma. lncRNA maternally expressed 3 (MEG3) expression was previously reported to be decreased in glioma tissues due to its hypermethylation. Upstream, DNA methyltransferase 1 (DNMT1) was found to be the mediator of MEG3 promoter methylation, which suppressed the transcription of MEG3. In addition, DNMT1 promoted the proliferation and clonogenicity of glioma cells, whilst reducing apoptosis. The inhibition of DNMT1 was demonstrated to activate the p53 signaling pathway in glioma cells. Therefore, MEG3 hypermethylation by DNMT1 was concluded to decrease MEG3 expression, which may represent a molecular mechanism of glioma progression (39). In GBM cells with acquired TMZ resistance, a previous study found that 1,383 lncRNAs were significantly upregulated, whereas 1,309 lncRNAs were significantly downregulated compared with normal GBM cell lines using a human lncRNA microarray; this suggests that lncRNAs are closely associated with TMZ resistance (40). lncRNA long intergenic non-protein coding RNA (LINC)00473, the expression of which was found to be significantly increased in glioma, was found to be associated with a poor prognosis of patients with glioma by functioning as an oncogenic lncRNA (41). In addition, LINC00473 knockdown was revealed to inhibit glioma progression both in vivo and in vitro. The underlying mechanism was proposed to be the inhibition of miR-195-5p by LINC00473 by competing for binding to the same site on the corresponding mRNA. Downstream, Yes-associated protein 1 (YAP1) and TEA domain family member 1 (TEAD1) were found to be downstream targets of miR-195-5p, where both shared a negative correlation. Taken together, LINC00473 downregulation inhibited glioma growth by serving as a competing endogenous RNA (ceRNA) to block miR-195-5p function, thereby promoting YAP1 and TEAD1 expression (41). In another study, lncRNA small nucleolar RNA host gene (SNHG)12 was found to be highly expressed in glioma, where functional analyses revealed that SNHG12 knockdown inhibited the proliferation and promoted the apoptosis of glioma cells (42). Downstream, SNHG12 was demonstrated to interact with Hu antigen R (HuR), an ubiquitous multi-faceted RNA-binding protein, to become stabilized (42). Furthermore, the lncRNA HOX antisense intergenic RNA (HOTAIR) was found to function as an oncogene that is highly expressed in glioma stem cells (GSCs). Functional assays revealed that downregulating HOTAIR expression suppressed GSC development by targeting the programmed cell death 4 protein (43). lncRNA H19 has also been reported to be highly expressed and negatively associated with miR-138 expression in glioma cell lines, according to reverse transcription-quantitative PCR (RT-qPCR) data (44). H19 knockdown was found to suppress angiogenesis in glioma, specifically by functioning as a ceRNA of miR-138 to inhibit its function, thereby promoting the expression of hypoxia-inducible transcription factors and VEGFs (44). In addition, H19 downregulation was also found to inhibit the angiogenesis of glioma by directly binding to miR-29a, which in turn inhibited vasohibin 2 expression, an angiogenic factor in glioma (45). A previous study demonstrated that the expression of the lncRNA colorectal neoplasia differentially expressed (CRNDE) was upregulated in glioma tissues and cells; subsequent functional analyses demonstrated that CRNDE promoted glioma cell proliferation in a mTOR signaling-dependent manner in glioma (46). Downstream, miR-384 expression was also revealed to be downregulated in glioma and inhibited by CRNDE, which increased the expression of piwi-like RNA-mediated gene silencing 4 protein (47). In addition, the lncRNA-MUF, which was induced by TGF-β, was reported to be another oncogene in GBM. The knockdown of lncRNA-MUF expression suppressed tumor growth and promoted apoptosis induced by TMZ by targeting miR-34a (48).

However, lncRNA taurine upregulated 1 (TUG1) was considered to be a suppressor of glioma cell proliferation by promoting apoptosis. The overexpression of TUG1 was found to increase the expression of caspases-3 and -9, whilst decreasing the expression of Bcl-1, suggesting that the suppressive effects of TUG1 were due to the inhibition of the pathways associated with cell apoptosis (49).

Gliomas have also been reported to regulate the expression of certain lncRNAs. Glioma cells retain the ability to secrete exosomes containing mRNAs, miRNAs, lncRNAs and angiogenic proteins into the surrounding microenvironment to regulate the function of target cells, by targeting downstream signaling pathways (50,51). Using electron microscopy and western blot analysis, exosomes secreted by glioma cells were previously found to transport lncRNA-activated by TGF-β (ATB) into astrocytes to activate them. Subsequently, lncRNA-ATB in glioma cell-derived exosomes were demonstrated to promote the proliferation of glioma cells by inhibiting miR-204-3p (52). Angiogenesis also plays a key role in glioma growth. It was reported that glioma-derived exosomes enriched in lncRNA-POU class 3 homeobox 3 promoted angiogenesis in glioma (53). Therefore, these aforementioned findings suggest that gliomas can export lncRNAs by secreting exosomes.

3. Oncogenic lncRNAs promote TMZ resistance in gliomas

Oncogenic lncRNAs (Table I) were previously found to be overexpressed or upregulated in gliomas, and this can promote TMZ resistance by targeting a multitude of signaling pathways.

Table I

The function and target of oncogenic lncRNAs affecting TMZ resistance in glioma.

Table I

The function and target of oncogenic lncRNAs affecting TMZ resistance in glioma.

lncRNACell lines Targets/regulatorsSignaling pathwaysFunctional impact(Refs.)
LINC01410GBM/TMZ-resistant GBM cellsmiR-370-3pPTEN/AKT TMZ resistancePromotes glioma growth and(62)
MIR155HG--PD-1/PD-L1 axisIndicates a poorer overall survival in GBM(63,64)
FoxD2-AS1-MGMT miR-98-5pPI3K/PTEN/AktImpairs glioma cell sensitivity to TMZ, induces MGMT promoter hypermethylation, and inhibits invasion, proliferation, migration and the TMZ resistance of drug-resistant glioma cells(67)
BCYRN1/BC200-miR-218-5pp53 sitePromotes the proliferation, migration, invasion and TMZ resistance(68)
lnc-TALC-miR-20b-3pp38/MAPK c-Met pathwayInfluences the sensitivity of glioma to TMZ(55,70)
LINC00174-miR-138-5pWnt/β-cateninInhibits glioma cell proliferation in TMZ- resistant glioma cells(71)
CCAT2SHG44/U251miR-424P53 sitePromotes the proliferation, invasion and migration of glioma cells(69)
H19U87MG/U251MDR, MRP, and ABCG2 genesWnt/β-Catenin NF-κBEnhances the resistance of TMZ-resistant cells(72-74)
LINC00021-E2F1p21/NotchImproves TMZ resistance and apoptosis in TMZ- resistant cells(75)

[i] lncRNAs, long non-coding RNAs; TMZ, temozolomide; GBM, glioblastoma multiforme; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; FoxD2-AS1, forkhead box D2-antisense RNA1; BCYRN1, brain cytoplasmic RNA1; TALC, TMZ-associated lncRNA; CCAT, colon cancer-associated transcript; MDR, multi-drug resistance gene; MRP, multidrug resistance-associated protein; ABCG2, ATP binding cassette subfamily G member 2.

c-Met pathway

c-Met is a receptor tyrosine kinase, the expression of which has been found to be upregulated in TMZ-resistant GBM (54). MGMT is an enzyme that removes the methyl group at O6MeG, thereby affecting the activity of TMZ (11). Wu et al (55) reported that lncRNA-TMZ-associated lncRNA (TALC), a novel lncRNA associated with GBM recurrence, upregulated the expression of MGMT and promoted TMZ resistance by activating c-Met signaling.

TGF-β1/Smad signaling pathway

TGF-β1 is a cytokine that plays a crucial role in promoting tumor growth (56). Smad proteins are phosphorylated by TGF-β1 receptors, which relays the TGF-β1 signal into the nucleus (57,58). TGF-β has been reported to be a novel player in mediating resistance against both targeted and conventional therapeutic agents (59). The mechanism underlying this process appears to be associated with epithelial-mesenchymal transition (EMT) and the activity of cancer stem cells (60). However, the effects of TGF-β signaling on TMZ resistance in glioma remain to be a subject of further exploration.

The knockdown of K-homology splicing regulatory protein (KSRP) expression has been demonstrated to inhibit the expression of miR-198 (61). In addition, miR-198 may enhance sensitivity to TMZ by inhibiting the expression of MGMT, which is abrogated by TGF-β1 through the suppression of KSRP expression in GBM cells. TGF-β1 may also upregulate the expression of lncRNAs H19 and HOXD cluster antisense RNA 2 by activating Smad, which promotes GBM resistance to TMZ by activating MGMT and inhibiting miR-198. However, KSRP knockdown was shown to abolished the effects of TGF-β1 and lncRNA H19 on miR-198 and MGMT. In the clinic, TMZ tended to be more effective in patients with lower expression levels of TGF-β1 or lncRNA H19. Collectively, these observations suggest that TGF-β1 can confer resistance to TMZ by regulating the processing of lncRNAs and increasing the expression of MGMT (61).

A literature search revealed various lncRNAs affecting TMZ resistance in glioma, as well as their functions and targets (62-82). This list of lncRNAs is presented in Table I.

Wnt/β-catenin signaling pathway

Using transcriptome sequencing, a previous study found that the Wnt/β-catenin pathway was significantly activated in TMZ-treated glioma cells in a PI3K/Akt pathway-dependent manner. Mechanistically, Akt activation promoted β-catenin translocation into the nucleus, resulting in the transcriptional activation of Wnt/β-catenin signaling to suppress apoptosis whilst promoting TMZ resistance (83). Sex-determining region Y-box9 protein (SOX9) has been found to be overexpressed in GBM, where it can stimulate GBM growth by activating Wnt/β-catenin signaling (84). In another study, Li et al (71) explored the effects of LINC00174 on glioma cells. They found that LINC00174 expression was increased in glioma tissues, and that LINC00174 knockdown significantly inhibited the proliferation of TMZ-resistant glioma cells. Mechanistically, the knockdown of LINC00174 expression suppressed the growth of TMZ-resistant glioma by sponging miR-138-5p, which negatively targets SOX9. Therefore, LINC00174 was proposed to activate Wnt/β-catenin signaling by targeting the miR-138-5p/SOX9 axis. Furthermore, in vivo experiments also revealed consistent findings (71).

It has been previously demonstrated that the expression level of lncRNA H19 is significantly increased in patients with glioma resistant to TMZ (73). An in vitro TMZ drug-resistant cell model was established and the model was verified using RT-qPCR. Silencing H19 expression enhanced the sensitivity of TMZ-resistant cells to TMZ by downregulating the expression of a number of genes, including multidrug resistance mutation, multidrug resistance-associated protein and ATP binding cassette subfamily G member 2. Therefore, H19 was proposed to be a novel therapeutic target for TMZ-resistant gliomas (73). In addition, other studies have previously investigated the specific mechanisms involved in lncRNA H19-mediated regulation. H19 knockdown was found to inhibit EMT by promoting the expression of the epithelial marker, E-cadherin, whilst inhibiting that of the mesenchymal markers, vimentin and zinc finger E-box binding homeobox (ZEB)1. Furthermore, H19 downregulation was found to decrease the expression of β-catenin and its downstream targets, c-Myc and surviving, in TMZ-treated glioma cells. However, H19 reversed the resistance of glioma cells to TMZ by repressing EMT and targeting Wnt/β-Catenin pathway (72). A recent study also reported that lncRNA PSMG3-AS1 was significantly upregulated in GBM. PSMG3-AS1 was observed to promote TMZ resistance in glioma cells by binding to c-Myc in the nucleus (74).

The silencing of lncRNA miR-155 host gene (MIR155HG) expression was previously reported to promote glioma sensitivity to TMZ through the Wnt/β catenin pathway by inhibiting poly-pyrimidine tract-binding protein 1 expression (64). Furthermore, the knockdown of LINC00511 expression enhanced the sensitivity of U87-R glioma cells to TMZ by targeting miR-126-5p and activating Wnt/β-catenin signaling (82). It has also been previously reported that EGFR signaling plays a key role in GBM (85), by regulating lncRNA nuclear enriched abundant transcript 1 (NEAT1) expression and in turn promoting glioma cell growth by targeting the Wnt/β-catenin pathway. Taken together, these results suggest that NEAT1 is a potential therapeutic target in GBM (86).

p21/Notch pathway

TMZ has been shown to induce GBM cell senescence and autophagy, which promote GBM cell survival without affecting proliferation (87). TMZ-induced senescence has been found to be dependent on p21 activity. GSCs are a main cause of TMZ resistance. MMP14 is an enzyme that is localized to the cell membrane (88). It was previously reported that TMZ treatment increased MMP14 expression in GSCs, which activated Notch signaling (89). In another study, the expression of lncRNA LINC00021 was revealed to be markedly upregulated in TMZ-resistant GBM cells, and the silencing LINC00021 expression promoted TMZ sensitivity, whilst suppressing the apoptosis of TMZ-resistant cells (75). Additionally, the expression of p21 was found to be downregulated by LINC00021 by targeting EZH2. Western blot analysis revealed that LINC00021 knockdown inhibited Notch expression. Therefore, LINC00021 was concluded to promote the malignant features of GSCs and the chemoresistant phenotype through the p21/Notch axis (75).

EMT signaling pathway

EMT has been observed to not only promote cell migration, but also chemoresistance in tumor cells. Wen et al (90) reported that the expression of EMT markers was upregulated in oxaliplatin-resistant human gastric cancer cell lines. In addition, EMT has been found to be associated with the resistance of pancreatic cancer to treatment (91). In another study, Peng et al (92) discussed that EMT can be regulated through miRNAs and summarized a list of drug candidates that may suppress EMT in cancer. EMT was also identified to be part of the mechanism underlying cancer resistance to EGFR tyrosine kinase inhibitors in both mouse models and in clinical non-small cell lung cancer (NSCLC) specimens (92). However, the effects of EMT on TMZ resistance in glioma cells remain unclear and require further investigation. Previously, a novel lncRNA TCONA_00004099 was identified through the analysis of sequencing data, the expression levels of which were high in glioma tissues and cell lines (79). In both in vitro and in vivo models, TCONA_00004099 knockdown was found to inhibit glioma progression and promote TMZ-induced U87 and U251 cell apoptosis. This mechanism of resistance associated with TCONA_00004099 in glioma was then examined using miRNA mimics. TCONA_00004099 was demonstrated to sponge the miRNA TCONA_00004099 and its target protein tyrosine phosphatase, receptor type F, which is involved in EGF signaling (79). Therefore, these findings reveal potentially novel therapeutic targets for TMZ-resistant glioma. lncRNA EGFR-AS1 was previously observed to promote the proliferation, migration and invasion, whilst inhibiting the apoptosis of glioma cells by regulating miR-133b/ribosomal receptor for activated C-kinase 1 (RACK1) (80). However, the effects of lnc-EGFR-AS1 on the sensitivity of GBM cells to TMZ requires further investigation both in vivo and in vitro (80). lncRNA HOTTIP expression was reported to be significantly elevated in SF268 glioma cells, particularly resistant to TMZ (93). Moreover, the expression of miR-10b and the mesenchymal markers, ZEB1/ZEB2, was increased, while the expression of E-cadherin was decreased in SF268, indicating that EMT plays a crucial role in TMZ resistance. Moreover, EMT was reversed in HOTTIP overexpressing cell lines by silencing miR-10b. Thus, HOTTIP regulated TMZ resistance in glioma cells via the EMT signaling pathway by targeting miR-10b (93).

FGFR3, platelet-derived growth factor receptor (PDGFR) and EGFR have all been found to be among the upregulated receptor tyrosine kinases involved in GBM tumorigenesis (94,95), which are also associated with the EMT process. The expression profile of FGFR3 was consistent with that of EGFR, whereby FGFR inhibition enhanced NSCLC sensitivity to EGFR inhibitors (96). Furthermore, PDGFR was found to promote the EMT process in glioma by promoting ZEB1 expression, an inducer of EMT (97). Cui et al (76) previously reported that lncRNA colon cancer-associated transcript 1 (CCAT1) expression was increased in glioma tissues, whilst that of miR-181b was downregulated. CCAT1 silencing was observed to significantly suppress the malignant biological behavior of glioma cells, which was reversed with miR-181b inhibitors. In addition, CCAT1 was found to function as a ceRNA with miR-181b to promote the expression of FGFR3 and PDGFR. Collectively, CCAT1 downregulation suppressed glioma growth by targeting miR-181b, resulting in the downregulation of the endogenous targets, FGFR3 and PDGFR (76). However, the effects of CCAT1 on the resistance of gliomas to TMZ and EMT require further in vivo and in vitro investigations.

p38/MAPK pathway

The function of p38 MAPK signaling is to relay extracellular stimuli through the cell (98). The activation of the p38 MAPK pathway has been reported to promote glioma invasiveness (99). Nuclear transcription factor erythroid 2p45 (NF-E2)-related factor 2 (Nrf2) has also been shown to promote the resistance of glioma cells to the combined treatment of TMZ and radiotherapy (100). In addition, another previous study found that TMZ increased Nrf2 expression by activating the p38 MAPK signaling pathway, leading to the development of TMZ resistance (101).

The role of lnc-TALC in promoting the resistance of GBM to TMZ has been observed. Furthermore, lnc-TALC can be incorporated into exosomes and exported to tumor-associated macrophages to promote M2 polarization. M2 macrophage polarization has been shown to be associated with the secretion of complement components C5/C5a, which occurs after the binding of lnc-TALC to enolase 1 to promote the phosphorylation of p38 MAPK. Furthermore, C5/C5a can promote the resistance to chemotherapy of GBM. Taken together, lnc-TALC can potentially regulate the microenvironment of GBM to improve GBM resistance to TMZ chemotherapy through the p38 MAPK pathway (70).

PTEN/PI3K/Akt pathway

PTEN is a protein phosphatase that targets the lipid phosphatidylinositol-3,4,5-triphosphate (PIP3), which is the product of PI3K. PTEN mutation and PI3K activation promote the accumulation of PIP3, resulting in the activation of AKT (102). PI3K/AKT signaling activation is associated with the expression of MGMT, the main factor of TMZ resistance. Supporting this, the pan-PI3K inhibitor, PX-866, has been found to block TMZ-induced autophagy, whilst promoting GBM cell death (103). In addition, the activation of PTEN/PI3K/AKT signaling has been reported to be an essential step in the development of TMZ resistance in GBM cells in a MGMT-dependent manner (104-106).

In glioma tissues and cells resistant to TMZ, the expression of the lncRNA musculin antisense RNA1 (MSC-AS1) has been demonstrated to be increased. MSC-AS1 knockdown has been shown to induce cell apoptosis and promote TMZ sensitivity by targeting the PI3K/AKT pathway (65). Cytoplasmic polyadenylation element binding protein (CPEB4) is an RNA-binding protein that participates in autophagy. The increased expression of CPEB4 has been found to be associated with migration, tumor growth, angiogenesis, metastasis and invasion (66). Additionally, the suppression of lncRNA-forkhead box D2-antisense RNA1 (FoxD2-AS1) expression has been found to inhibit the invasion, proliferation, migration and of TMZ-resistant glioma cells by promoting miR-98-5p expression, which inhibits CPEB4 expression (67). These data suggest that FoxD2-AS1 can inhibit glioma chemoresistance to TMZ by modulating the miR-98-5p/CPEB4 axis (58). Furthermore, the expression of CPEB4, as a target of miR-373-3p, was also found to be regulated by MSC-AS1. Specifically, MSC-AS1 knockdown inhibited the malignant behavior of glioma cells and increased their sensitivity to TMZ by sponging miR-373-3p, which activated the PI3K/Akt pathway both in vitro or in vivo (65). Similarly, the expression of the non-catalytic region of tyrosine kinase adaptor protein 1-antisense RNA1 was reported to be increased in glioma tissues and cells resistant to TMZ, which facilitated the resistance to TMZ by activating the miR-137/tripartite motif containing 24 pathways (81). In addition, in a previous study, the expression of the lncRNA LINC01410 was found to be upregulated in GBM and in GBM cells resistant to TMZ. LINC01410 silencing promoted the sensitivity of TMZ-resistant cells whilst also upregulating the expression of PTEN but reducing the phosphorylation of AKT. Inhibition of miR-370-3p reversed the effects of LINC01410 silencing on the malignant biological behavior of GBM cells. Based on these findings, LINC01410 was concluded to promote glioma growth and TMZ resistance by sponging miR-370-3p to inhibit the PTEN/AKT pathway (62).

p53 signaling

p53 is a key transcription factor that can regulate the transcription of target genes in response to DNA damage. p53 suppresses the gene expression of O6-alkylguanine DNA alkyltransferase. In addition, Y-box binding protein 1 and mouse double minute 2 homolog were found to be overexpressed in glioma cells and were previously found to be associated with TMZ resistance by directly targeting p53 (107,108). Therefore, p53 may be associated with the resistance of glioma cells to TMZ.

The levels of lncRNA brain cytoplasmic RNA1 (BCYRN1) were previously found to be increased in the blood and tumor tissues from patients with GBM. Furthermore, the expression of BCYRN1 differed significantly from that of isocitrate dehydrogenase 1 (IDH1) and the p53 status and was markedly higher compared with both p53 and Ki-67 expression. The silencing of BCYRN1 expression was previously reported to inhibit the proliferation, migration, invasion and reverse resistance to TMZ in GB cells by sponging miR-218-5p (68).

Compared with normal tissues, higher expression levels of lncRNA CCAT2 were previously found in glioma tissues and cells and were found to negatively correlate with those of miR-424 (69). In addition, different glioma cell lines exhibited varying degrees of resistance to chemotherapeutic agents. Specifically, SHG44 was the most resistant cell line whereas U251 cells were more sensitive to agents, such as TMZ. However, regardless of the cell line tested, the knockdown of CCAT2 expression or the overexpression of miR-424 markedly inhibited the proliferation, invasion and migration of glioma cells (69). Furthermore, checkpoint kinase 1 (CHK1), which is activated by DNA damage, was reported to be a target of miR-424. The overexpression of CHK1 reversed the effects of miR-424 overexpression in both p53 wild-type and p53 mutant glioma cells (109). Taken together, these findings suggest that lncRNA CCAT2 expression is upregulated in glioma tissues and cells, which in turn promotes tumor growth and resistance to TMZ possibly through the miR-424/CHK1 axis (69).

PIM1/survivin pathway

PIM1 is a serine/threonine kinase that plays a key role in promoting cell proliferation and conferring chemoresistance in various malignancies, such as prostate cancer, HER2-positive breast cancer, high-risk neuroblastoma, triple negative breast cancer and so on (110-113). The expression of survivin can be decreased by the MGMT inhibitor (114). In addition, TMZ-induced senescence results in tumor cell cycle arrest, promoting TMZ resistance. Song et al (115) previously indicated that the knockdown of survivin promoted glioma cell sensitivity to TMZ treatment by inducing the apoptosis of senescent cells. Furthermore, survivin nuclear trapping has been found to facilitate GBM response to TMZ (116).

The lncRNA potassium voltage-gated channel subfamily Q member 1 opposite strand/antisense transcript one gene (KCNQ1OT1) was previously reported to promote the proliferation and colony formation of glioma cells by negatively regulating miRNA-761 expression. It was also found to impair the effects of TMZ on the promotion of the apoptosis of glioma cells in vitro. Furthermore, the overexpression of KCNQ1OT1 was found to increase survivin expression by promoting PIM1 expression in glioma cells. In vivo, the experimental results were consistent with their in vitro counterparts. Taken together, it was found that lncRNA KCNQ1OT1 played a significant role in glioma sensitivity to TMZ by sponging miR-761 to activate PIM-1/survivin signaling, which may be exploited as a potential prognostic target (78).

Programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) signaling pathway

Over the past decade, immune checkpoint inhibitors have become a topic of extensive research for tumor treatment. Among these, PD-1/PD-L1 have been found to exert promising therapeutic effects against various malignancies. PD-L1 has also been shown to be strongly expressed in a wide range of tumor cells, including breast cancer, oral squamous cell carcinoma and nasopharyngeal carcinoma (117-119). The PD-1/PD-L1 signaling pathway plays a constructive role in tumor immunosuppression by inducing T-cell death and activating tumor immune escape (120).

Furthermore, several studies have demonstrated that a number of lncRNAs are associated with PD-1/PD-L1 in different types of cancer (Fig. 1). A previous study demonstrated that human antisense lncRNA-NK-2 homeobox 1 antisense RNA1 (NKX2-1-AS1) negatively regulated the expression of endogenous CD274/PD-L1, and that NKX2-1-AS1 knockdown was able to upregulate the expression of genes that are involved in cell adhesion and checkpoints (121). In addition, PD-1/PD-L1 was found to exert promoting effects on diffuse large-B-cell lymphoma (DLBCL), and lncRNA SNHG14 expression negatively correlated with miR-152-3p, the expression of which was decreased in DLBCL tissues and cell lines. Furthermore, SNHG14/miR-152-3p inhibited apoptosis and promoted cell proliferation in cytotoxic T-lymphocytes (CTLs) in DLBCL by sponging the PD-1/PD-L1 checkpoint (122). Similarly, SNHG15 lncRNA, which is highly expressed in gastric cancer cells, was reported to increase the expression of PD-L1 (123). Zhou et al (124) found that in pancreatic cancer tissues and cell lines, the expression of LINC00473 was increased, whilst that of miR-195-5p was decreased, and the expression of PD-L1 was increased. Mechanistically, LINC00473 upregulated PD-L1 expression by targeting miR-195-5p. Furthermore, CD8+ T-cells could be activated either by the silencing of LINC00473 expression or increasing miR-195-5p expression, which inhibited PC progression (124). In addition to these aforementioned pre-clinical findings, a database analyses have also revealed the potential of lncRNAs in regulating PD-1/PD-L1 signaling. According to The Cancer Genome Atlas-liver hepatocellular carcinoma (HCC) dataset, PC-esterase domain-containing 1B-antisense RNA1 promoted PD-L1 and PD-L2 expression by inhibiting hsa-miR-194-5p expression, resulting in immunosuppression in HCC (125). Similarly, the association between lncRNA-myocardial infarction associated transcript (MIAT) and immunomodulatory regulation in HCC was also investigated. The results revealed that the expression of MIAT in HCC was positively associated with that of PD-1/PD-L1. Furthermore, MIAT reduced the sensitivity of HCC to sorafenib, an anticancer drug, mediating immune escape (126).

Previous studies have demonstrated that various lncRNAs can regulate the PD-1/PD-L1 axis in gliomas. The lncRNA interferon (IFN)-stimulated non-coding RNA 1 (INCR1) can be transcribed from the PD-L1 locus following stimulation by tumor IFNs and can promote checkpoint blockade resistance in GBM. INCR1 knockdown was found to inhibit PD-L1 expression and enhance GBM cell sensitivity to cytotoxic T-cell-mediated cell death (127). Mechanistically, INCR1 was found to promote the expression of PD-L1 genes by binding to heterogeneous nuclear ribonucleoprotein H1 (HNRNPH1), a nuclear ribonucleoprotein, to negatively regulate the transcription of PD-L1. In summary, INCR1 lncRNA can regulate PD-L1 expression and responses to immune checkpoint therapy by targeting HNRNPH1 after IFNγ signaling (127). Similarly, the lncRNA HOTAIR was also found to be highly expressed in glioma cells (77). Bioinformatics analysis revealed that HOTAIR activated the expression of proteins in the NF-κB signaling pathway. Furthermore, the downregulation of HOTAIR expression was found to inhibit PD-L1 expression in vivo, suggesting that HOTAIR can improve the sensitivity of glioma cells to immune checkpoint inhibitor therapy. In summary, HOTAIR positively regulated PD-L1 expression by targeting the NF-κB signaling pathway in gliomas (77). According to the existing public bioinformatics database, higher levels of the lncRNA MIR155HG have been shown to be associated with a poorer overall survival of patients with GBM. In addition, the levels of MIR155HG expression have been demonstrated to be significantly consistent with those of PD-L1, suggesting that MIR155HG can be used predict the efficacy of immune checkpoint inhibitor therapy (63). Taken together, lncRNAs may represent a therapeutic target that can regulate the effects of PD-1/PD-L1 treatment in various types of cancer, particularly gliomas. However, T-cells in GBM are very unresponsive to immunotherapy, and PD-1 checkpoint blockade cannot be overcome through intracellular regulation. Further research on combination therapy is still in progress (128). Due to chemo-immunotherapy resistance, Miyazaki et al (129) used a TMZ-treated glioma model to examine the effects of anti-PD-L1 antibody on the tumor microenvironment. They also established TMZ-resistant TS (TMZRTS) cells to examine the effects of anti-PD-L1 anti-body on glioma growth. The results of their study revealed that anti-PD-L1 antibody significantly suppressed tumor growth and markedly decreased CD163-positive Mϕ infiltration into tumors. However, the safety and effectiveness of clinical therapy need to be further studied (129). Nivolumab is a type of PD-1 inhibitor. Recent research has reported that a patient with GBM received nivolumab treatment for almost 2 years following standard radiotherapy and TMZ therapy. Magnetic resonance imaging revealed a continuous shrinking of the tumor (130), suggesting that the combined application with PD-1 inhibitor may be effective for the treatment of gliomas. Clinically, TMZ is used as an adjuvant therapy for patients with glioma, while the prognosis is poor. Thus, exploring the role of PD-1/PD-L1 combined with TMZ is of therapeutic significance. Notably, it was demonstrated that stereotactic radiation, which to date is a conventional treatment for glioma, combined with anti-PD-1 blockade enhanced the long-term survival rate of tumor-bearing mice (131), which provided further evidence of the potential of combining TMZ with PD-1 inhibitors. In a previous clinical study, 47 patients were divided into two subgroups (132). One subgroup of 27 patients, which was named as ADCTA, was treated with concomitant chemo-radiotherapy (CCRT). Moreover, the patients also received post-surgical adjuvant immunotherapy with an autologous dendritic cell/tumor antigen vaccine. The other subgroup was only treated with CCRT and TMZ without immunotherapy as the control group. The results of that study demonstrated that the ADCTA group exhibited a markedly low TIL PD-1+/CD8+ ratio within the GBM tumor compared with the control group, which prolonged the overall survival rate of the patients. On the whole, that study suggested that adjuvant immune therapy for the immune system combined with conventional therapies, such as TMZ has great potential for the treatment of GBM (132). In conclusion, the com-bination of immune checkpoint inhibitors and anti-tumor chemical drugs, such as TMZ, has a promising therapeutic effect on glioma, which further provides new insight into the treatment of gliomas in clinical practice.

4. Tumor suppressive lncRNAs influence the sensitivity of gliomas to TMZ

The expression of a number of tumor suppressor lncRNAs (Table II) has been found to be downregulated in gliomas compared with normal brain tissues (38). The re-expression of these tumor suppressor lncRNAs can enhance the therapeutic effects of TMZ by regulating the mTOR, PTEN/AKT and p53/NF-κB signaling pathways.

Table II

Functions and targets of tumor suppressive lncRNAs affecting TMZ resistance in glioma.

Table II

Functions and targets of tumor suppressive lncRNAs affecting TMZ resistance in glioma.

lncRNACell lines Targets/regulatorsSignaling pathwaysFunctional impact(Refs.)
CASC2- miR-181a/miR-193a-5pPTEN/p-AKT mTORImpairs TMZ resistance and proliferation of glioma.(131,137)
GAS5U251 U87 miR-18a-5p/miR-196a-5pmTORSuppresses the proliferation, migration, and invasion of humanglioma cells; attenuates gliomacell resistance(132,135)
ZNF281U251s-NF-κB1Suppresses the self-renewal capacity and invasion of glioma stem-like cells(139)
PR-LncRNA-SOXp53Inhibits the malignant behavior of glioma cells(139)
LIFR-AS1-miR-4262NF-κBInhibits glioma cell growth and enhance glioma cell sensitivity to TMZ(140)

[i] lncRNAs, long non-coding RNAs; TMZ, temozolomide; CASC2, cancer susceptibility candidate 2; GAS5, growth arrest specific 5; ZNF281, zinc finger protein 281; LIFR-AS1, leukemia inhibitory factor receptor-antisense RNA1.

mTOR signaling pathway

mTOR is a serine/threonine kinase that can regulate cell proliferation (133). Tomar et al (83) previously observed that mTOR signaling was activated following TMZ treatment. A novel inhibitor (Rapalink-1) targeting the mTOR pathway was previously identified to function synergistically with TMZ to reduce cell proliferation (134). Autophagy is a process that has been previously associated with TMZ resistance. Zou et al (135) reported that the upregulation of autophagy was mediated by transient receptor potential cation channel subfamily C member 5 in glioma cells, which promoted TMZ resistance through the mTOR signaling pathway. Based on this finding, it can be hypothesized that the mTOR pathway is associated with autophagy, thereby affecting TMZ resistance.

In vitro, the expression of the lncRNA cancer susceptibility candidate 2 (CASC2) was found to be decreased in glioma tissues and cell lines, the upregulation of which was associated with a longer survival time (136). Clinically, TMZ treatment significantly inhibited glioma proliferation, whilst the upregulation of CACS2 expression resulted in the opposite effect. Furthermore, a downstream target of CASC2 was determined to be miR-193a-5p, with which negatively correlated (136). In TMZ-resistant glioma cells, CASC2 upregulation impaired autophagy, which was reduced by TMZ by inhibiting miR-193a-5p expression, thereby activating the mTOR pathway. These data suggest that upregulating the expression of CASC2 may be a potential therapeutic target for patients with TMZ-resistant glioma. However, further clinical trials are required to examine the feasibility of this target (136).

lncRNA growth arrest specific 5 (GAS5) has been reported to play a constructive role in various tumor types, such as gastric cancer and NSCLC. It has been observed to inhibit the proliferation, migration and invasion of human glioma cells by targeting miR-18a-5p or miR-196a-5p (137,138). In addition, GAS5 can reverse glioma cell resistance to cisplatin by preventing excessive autophagy through activation of mTOR signaling (139). Similarly, in another previous study (140), which investigated the U251 and U87 cell lines, the expression of GAS5 was found to be decreased, and the upregulation of GAS5 was able to inhibit tumor growth by sponging glutathione-S-transferase M3, the overexpression of which has been associated with chemoresistance. However, the effects of GAS5 on the resistance of glioma cells to TMZ require further examination in vitro and in vivo (140).

PTEN/p-AKT pathway

lncRNAs can also regulate resistance to TMZ by targeting the PTEN, p53 and PI3K/AKT/mTOR pathways (141). In a previous study, the overexpression of lncRNA CACS2 was found to reverse the resistance of glioma cells to TMZ by suppressing miR-181a expression (142). In addition, CASC2 promoted PTEN protein expression and inhibited AKT phosphorylation by targeting miR-181a in a reversible manner by miR-181a overexpression. Therefore, CASC2 may be a clinically relevant lncRNA that can mediate TMZ resistance by regulating miR-181a expression (142).

p53/NF-κB pathway

NF-κB is a transcription factor and plays a key role in cell survival, inflammation and immune responses. In particular, NF-κB has been found to be associated with cellular senescence in response to chemotherapy. Accordingly, senescence induced by chemotherapy and mediated by NF-κB is a key characteristic of superior therapeutic outcomes (143). Therefore, NF-κB will likely affect TMZ resistance by modulating cellular senescence.

The p53-regulated tumor suppressor signature of lncRNAs (PR-lncRNAs) was first identified in colorectal cancer (144). In glioma, the expression of PR-lncRNAs was found to be negatively associated with its grade (145). A previous functional study reported that PR-lncRNA knockdown aggravated glioma malignancy by inhibiting SOX activity. This suggests that the downregulation of PR-lncRNAs can abrogate the beneficial effects of PR-lncRNAs on cell proliferation, migration, invasion and resistance to TMZ in vitro (145). Similarly, the expression of leukemia inhibitory factor receptor-antisense RNA1 (LIFR-AS1) has also been found to be decreased, whereas that of miR-4262 was found to be increased in glioma tissues and cell lines. The overexpression of LIFR-AS1 inhibited glioma cell proliferation and enhanced glioma cell sensitivity to TMZ by inactivating the miR-4262/NF-κB pathway (146). lncRNA-zinc finger protein 281 (ZNF281) was previously reported to be expressed at lower levels in glioma stem-like cells (U251s) compared with those in normal brain tissues. Furthermore, lncRNA-ZNF281 was able to suppress the self-renewal capacity and invasion of glioma stem cells by activating the NF-κB signaling pathway in vitro and in vivo (147).

The various mechanisms of lncRNAs in the regulation of TMZ resistance in glioma are illustrated in Fig. 2.

5. Conclusions and future perspectives

GBM, which is considered to originate from neural stem cells or progenitors, remains to be one of the greatest threats to human health to date. Regrettably, even with the traditional treatment methods of gliomas, including surgery, radiation therapy and alkylating agent chemotherapy, such as TMZ, the overall survival of patients with gliomas has not improved satisfactorily (148). Additionally, >50% patients with glioma treated with TMZ develop resistance to this drug, which has become a major obstacle in the clinical treatment efficacy of glioma (9). Apart from DNA repair systems, the mechanism underlying chemotherapy resistance also included translesion synthesis (TLS). When DNA lesions occur due to chemotherapy and/or radiation, such as following treatment with cisplatin and TMZ, TLS can bypass these types of lesions at the cost of increasing the risk of mutations (149), which in turn increases the risk of developing chemotherapeutic resistance. The TLS process involves several key regulators, including RAD18 (150), DNA polymerases (POL)κ, POLι, POLη or REV1 (which introduce a nucleotide opposite the lesion) and TLS DNA polymerase, such as POLζ (comprising of the Rev3L/Rev7/Pold2d/Pold3 complex) (149,151). Peng et al (152) previously reported that the overexpression of POLκ increased the TMZ resistance of GBM cells which were previously TMZ-sensitive. The higher expression of POLκ significantly promoted TMZ sensitivity in vitro and in orthotopic xenograft mouse models. Furthermore, other TLS polymerases, such as Rev1 and Rev7, have been proposed as targets for developing agents for reversing chemoresistance (151,153). RAD18 was previously found to be overexpressed in GBM cell lines, where it promoted resistance to radiotherapy by inhibiting the expression of p53 (154).

lncRNAs have been demonstrated to regulate the survival, proliferation, apoptosis and invasion of GBM cells (155). Furthermore, lncRNAs play a key role in regulating the resistance of gliomas to TMZ by sponging miRNAs and various signaling pathways. However, TMZ can conversely regulate the levels of lncRNA expression to enhance the effectiveness of TMZ treatment. Recently, lncRNA ATXN8OS was found to be mainly located in the cytoplasm and to be downregulated in glioma cell lines (156). Functional experiments proved that ATXN8OS enhanced TMZ sensitivity of glioma in vivo and in vitro. Mechanistically, ATXN8OS stabilized glutaminase 2 mRNA to promote the ferroptosis of glioma cell lines, which may become a new target of TMZ resistance (154). High mobility group box 1 protein (HMGB1), which is a highly conserved protein, has been reported to be expressed in a variety of cell types and can be secreted into the tumor micro-environment to function as a tumor-derived signal. HMGB1 expression has been found to be upregulated in GBM and to influence GBM growth. GSCs play a crucial role in patients with TMZ-resistant glioma, and HMGB1 expression has been found to be upregulated in GSCs (157). HMGB1 can then alter the expression profile of mRNAs, lncRNAs and miRNAs in GSCs. Since TMZ-induced HMGB1 expression can promote the formation of GSCs, this process is inhibited when the secretion of HMGB1 is decreased. In terms of the mechanism, TMZ-induced upregulation of HMGB1 has been found to promote the formation of GSCs through the Toll-like receptor 2/NEAT1/Wnt pathway. Taken together, TMZ-induced HMGB1 may serve as a potential therapeutic target for the treatment of patients with TMZ-resistant GBM (157). In addition, a number of traditional Chinese medicine ingredients have also been reported to affect TMZ resistance by regulating the expression of lncRNAs. Isoliquiritigenin has been found to inhibit the expression of metabolic enzymes of arachidonic acid (AA), such as cyclooxygenase-2 (COX-2), microsomal prostaglandin E synthase-1 (mPGES-1) and cytochrome P450 (CYP) 4A11, all of which serve significant roles in glioma angiogenesis. Furthermore, isoliquiritigenin can enhance the therapeutic effects of TMZ in a rat C6 glioma model. In terms of the mechanism, isoliquiritigenin can reprogram COX-2-, mPGES-1- and CYP4A-mediated AA metabolism in glioma by suppressing angiogenic Akt/fibroblast growth factor 2/TGFβ/VEGF signaling, which was achieved by sponging miR-194-5p and lncRNA NEAT1 (158). To conclude, lncRNAs may serve as potential therapeutic and prognostic targets to improve the sensitivity of glioma to TMZ. However, enormous research efforts remain necessary to transform this basic concept into a feasible clinical strategy.

Availability of data and materials

Not applicable.

Authors' contributions

The present review article was conceptualized by all the authors (SL, XX, FP, JD and CP). SL was involved in the study design and in the preparation of the original draft. FP, XX and JD edited the manuscript. FP, JD and CP were involved in funding-related administration. SL and FP were involved in manuscript revision. All authors have read and agreed to the published version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 82003879 and U19A2010) the Key Project of Science and Technology Department of Sichuan Province (grant nos. 2020YFS0053 and 2021YFS0044), and the Youth Talent Promotion Project of China Association for Science and Technology (grant no. CACM-2020-QNRC1-01) and the Open Research Fund of Chengdu University of Traditional Chinese Medicine Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China (grant no. 2021LF1026).

References

1 

Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P and Ellison DW: The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 131:803–820. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Rynkeviciene R, Simiene J, Strainiene E, Stankevicius V, Usinskiene J, Miseikyte Kaubriene E, Meskinyte I, Cicenas J and Suziedelis K: Non-coding RNAs in glioma. Cancers (Basel). 11:172018. View Article : Google Scholar

3 

De Sanctis V, Mazzarella G, Osti MF, Valeriani M, Alfó M, Salvati M, Banelli E, Tombolini V and Enrici RM: Radiotherapy and sequential temozolomide compared with radiotherapy with concomitant and sequential temozolomide in the treatment of newly diagnosed glioblastoma multiforme. Anticancer Drugs. 17:969–975. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Stupp R, Taillibert S, Kanner A, Read W, Steinberg D, Lhermitte B, Toms S, Idbaih A, Ahluwalia MS, Fink K, et al: Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: A randomized clinical trial. JAMA. 318:2306–2316. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Strobel H, Baisch T, Fitzel R, Schilberg K, Siegelin MD, Karpel-Massler G, Debatin KM and Westhoff MA: Temozolomide and other alkylating agents in glioblastoma therapy. Biomedicines. 7:692019. View Article : Google Scholar :

7 

Zhang J, Stevens MF and Bradshaw TD: Temozolomide: Mechanisms of action, repair and resistance. Curr Mol Pharmacol. 5:102–114. 2012. View Article : Google Scholar

8 

Stupp R, Brada M, van den Bent MJ and Tonn JC: High-grade glioma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 25(Suppl 3): iii93–iii101. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Lee SY: Temozolomide resistance in glioblastoma multiforme. Genes Dis. 3:198–210. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Kanzawa T, Bedwell J, Kondo Y, Kondo S and Germano IM: Inhibition of DNA repair for sensitizing resistant glioma cells to temozolomide. J Neurosurg. 99:1047–1052. 2003. View Article : Google Scholar

11 

Jiang G, Li LT, Xin Y, Zhang L, Liu YQ and Zheng JN: Strategies to improve the killing of tumors using temozolomide: Targeting the DNA repair protein MGMT. Curr Med Chem. 19:3886–3892. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Perazzoli G, Prados J, Ortiz R, Caba O, Cabeza L, Berdasco M, Gónzalez B and Melguizo C: Temozolomide resistance in glioblastoma cell lines: Implication of MGMT, MMR, P-glycoprotein and CD133 expression. PLoS One. 10:e01401312015. View Article : Google Scholar : PubMed/NCBI

13 

Tang JB, Svilar D, Trivedi RN, Wang XH, Goellner EM, Moore B, Hamilton RL, Banze LA, Brown AR and Sobol RW: N-methylpurine DNA glycosylase and DNA polymerase beta modulate BER inhibitor potentiation of glioma cells to temozolomide. Neuro Oncol. 13:471–486. 2011. View Article : Google Scholar : PubMed/NCBI

14 

ENCODE Project Consortium: An integrated encyclopedia of DNA elements in the human genome. Nature. 489:57–74. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Comings DE: The structure and function of chromatin. Adv Hum Genet. 3:237–431. 1972. View Article : Google Scholar : PubMed/NCBI

16 

Hombach S and Kretz M: Non-coding RNAs: Classification, biology and functioning. Non-coding RNAs in Colorectal Cancer. Slaby O and Calin GA: Springer International Publishing; Cham: pp. 3–17. 2016, View Article : Google Scholar

17 

Ling H, Vincent K, Pichler M, Fodde R, Berindan-Neagoe I, Slack FJ and Calin GA: Junk DNA and the long non-coding RNA twist in cancer genetics. Oncogene. 34:5003–5011. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, et al: The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 22:1775–1789. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Schmitz SU, Grote P and Herrmann BG: Mechanisms of long noncoding RNA function in development and disease. Cell Mol Life Sci. 73:2491–2509. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Yu B and Wang S: Angio-LncRs: LncRNAs that regulate angiogenesis and vascular disease. Theranostics. 8:3654–3675. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Wilusz JE, Sunwoo H and Spector DL: Long noncoding RNAs: Functional surprises from the RNA world. Genes Dev. 23:1494–1504. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Yoon JH, Abdelmohsen K and Gorospe M: Functional interactions among microRNAs and long noncoding RNAs. Semin Cell Dev Biol. 34:9–14. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Han L, Zhang K, Shi Z, Zhang J, Zhu J, Zhu S, Zhang A, Jia Z, Wang G, Yu S, et al: LncRNA profile of glioblastoma reveals the potential role of lncRNAs in contributing to glioblastoma pathogenesis. Int J Oncol. 40:2004–2012. 2012.PubMed/NCBI

24 

Zhang J, Zhang Z, Chen Z and Deng L: Integrating multiple heterogeneous networks for novel LncRNA-disease association inference. IEEE/ACM Trans Comput Biol Bioinform. 16:396–406. 2019. View Article : Google Scholar

25 

Bolha L, Ravnik-Glavač M and Glavač D: Long noncoding RNAs as biomarkers in cancer. Dis Markers. 2017:72439682017. View Article : Google Scholar : PubMed/NCBI

26 

Mahinfar P, Baradaran B, Davoudian S, Vahidian F, Cho WC and Mansoori B: Long Non-coding RNAs in multidrug resistance of glioblastoma. Genes (Basel). 12:4552021. View Article : Google Scholar

27 

Jiang Y, Guo H, Tong T, Xie F, Qin X, Wang X, Chen W and Zhang J: lncRNA lnc-POP11 upregulated by VN1R5 promotes cisplatin resistance in head and neck squamous cell carcinoma through interaction with MCM5. Mol Ther. 30:448–467. 2022. View Article : Google Scholar

28 

Chen KY, Zhu SG, He JW and Duan XP: LncRNA CRNDE is involved in radiation resistance in hepatocellular carcinoma via modulating the SP1/PDK1 axis. Neoplasma. 211230N18532022.Epub ahead of print. PubMed/NCBI

29 

Wu J, Xu S, Li W, Lu Y, Zhou Y, Xie M, Luo Y, Cao Y, He Y, Zeng T and Ling H: lncRNAs as hallmarks for individualized treatment of gastric cancer. Anticancer Agents Med Chem. 22:1440–1457. 2022. View Article : Google Scholar

30 

Ye X, Wang LP, Han C, Hu H, Ni CM, Qiao GL, Ouyang L and Ni JS: Increased m6A modification of lncRNA DBH-AS1 suppresses pancreatic cancer growth and gemcitabine resistance via the miR-3163/USP44 axis. Ann Transl Med. 10:3042022. View Article : Google Scholar

31 

Jiang X, Li H, Fang Y and Xu C: LncRNA PVT1 contributes to invasion and doxorubicin resistance of bladder cancer cells through promoting MDM2 expression and AURKB-mediated p53 ubiquitination. Environ Toxicol. 37:1495–1508. 2022. View Article : Google Scholar : PubMed/NCBI

32 

Cheng M, Wang Q, Chen L, Zhao D, Tang J, Xu J and He Z: LncRNA UCA1/miR-182-5p/MGMT axis modulates glioma cell sensitivity to temozolomide through MGMT-related DNA damage pathways. Hum Pathol. 123:59–73. 2022. View Article : Google Scholar : PubMed/NCBI

33 

Zhang X, Sun S, Pu JK, Tsang AC, Lee D, Man VO, Lui WM, Wong ST and Leung GK: Long non-coding RNA expression profiles predict clinical phenotypes in glioma. Neurobiol Dis. 48:1–8. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Lin X, Zhuang S, Chen X, Du J, Zhong L, Ding J, Wang L, Yi J, Hu G, Tang G, et al: lncRNA ITGB8-AS1 functions as a ceRNA to promote colorectal cancer growth and migration through integrin-mediated focal adhesion signaling. Mol Ther. 30:688–702. 2022. View Article : Google Scholar

35 

Li DQ, Ding YR, Che JH, Su Z, Yang WZ, Xu L, Li YJ, Wang HH and Zhou WY: Tumor suppressive lncRNA MEG3 binds to EZH2 and enhances CXCL3 methylation in gallbladder cancer. Neoplasma. 69:538–549. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Yuan D, Guo T, Zhu D, Ge H, Zhao Y, Huang A, Wang X, Cao X, He C, Qian H and Yu H: Exosomal lncRNA ATB derived from ovarian cancer cells promotes angiogenesis via regulating miR-204-3p/TGFβR2 axis. Cancer Manag Res. 14:327–337. 2022. View Article : Google Scholar :

37 

Yan Y, Xu Z, Li Z, Sun L and Gong Z: An insight into the increasing role of LncRNAs in the pathogenesis of gliomas. Front Mol Neurosci. 10:532017. View Article : Google Scholar : PubMed/NCBI

38 

Peng Z, Liu C and Wu M: New insights into long noncoding RNAs and their roles in glioma. Mol Cancer. 17:612018. View Article : Google Scholar : PubMed/NCBI

39 

Li J, Bian EB, He XJ, Ma CC, Zong G, Wang HL and Zhao B: Epigenetic repression of long non-coding RNA MEG3 mediated by DNMT1 represses the p53 pathway in gliomas. Int J Oncol. 48:723–733. 2016. View Article : Google Scholar

40 

Zeng H, Xu N, Liu Y, Liu B, Yang Z, Fu Z, Lian C and Guo H: Genomic profiling of long non-coding RNA and mRNA expression associated with acquired temozolomide resistance in glioblastoma cells. Int J Oncol. 51:445–455. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Wang X, Li XD, Fu Z, Zhou Y, Huang X and Jiang X: Long non-coding RNA LINC00473/miR-195-5p promotes glioma progression via YAP1-TEAD1-Hippo signaling. Int J Oncol. 56:508–521. 2020.PubMed/NCBI

42 

Lei W, Wang ZL, Feng HJ, Lin XD, Li CZ and Fan D: Long non-coding RNA SNHG12promotes the proliferation and migration of glioma cells by binding to HuR. Int J Oncol. 53:1374–1384. 2018.PubMed/NCBI

43 

Fang K, Liu P, Dong S, Guo Y, Cui X, Zhu X, Li X, Jiang L, Liu T and Wu Y: Magnetofection based on superparamagnetic iron oxide nanoparticle-mediated low lncRNA HOTAIR expression decreases the proliferation and invasion of glioma stem cells. Int J Oncol. 49:509–518. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Liu ZZ, Tian YF, Wu H, Ouyang SY and Kuang WL: LncRNA H19 promotes glioma angiogenesis through miR-138/HIF-1α/VEGF axis. Neoplasma. 67:111–118. 2020. View Article : Google Scholar

45 

Jia P, Cai H, Liu X, Chen J, Ma J, Wang P, Liu Y, Zheng J and Xue Y: Long non-coding RNA H19 regulates glioma angiogenesis and the biological behavior of glioma-associated endothelial cells by inhibiting microRNA-29a. Cancer Lett. 381:359–369. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Wang Y, Wang Y, Li J, Zhang Y, Yin H and Han B: CRNDE, a long-noncoding RNA, promotes glioma cell growth and invasion through mTOR signaling. Cancer Lett. 367:122–128. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Zheng J, Liu X, Wang P, Xue Y, Ma J, Qu C and Liu Y: CRNDE promotes malignant progression of glioma by attenuating miR-384/PIWIL4/STAT3 axis. Mol Ther. 24:1199–1215. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Shree B, Tripathi S and Sharma V: Transforming growth factor-beta-regulated LncRNA-MUF promotes invasion by modulating the miR-34a snail1 axis in glioblastoma multiforme. Front Oncol. 11:7887552022. View Article : Google Scholar : PubMed/NCBI

49 

Li J, Zhang M, An G and Ma Q: LncRNA TUG1 acts as a tumor suppressor in human glioma by promoting cell apoptosis. Exp Biol Med (Maywood). 241:644–649. 2016. View Article : Google Scholar

50 

Arscott WT, Tandle AT, Zhao S, Shabason JE, Gordon IK, Schlaff CD, Zhang G, Tofilon PJ and Camphausen KA: Ionizing radiation and glioblastoma exosomes: Implications in tumor biology and cell migration. Transl Oncol. 6:638–648. 2013. View Article : Google Scholar

51 

Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, Curry WT Jr, Carter BS, Krichevsky AM and Breakefield XO: Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 10:1470–1476. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Bian EB, Chen EF, Xu YD, Yang ZH, Tang F, Ma CC, Wang HL and Zhao B: Exosomal lncRNA-ATB activates astrocytes that promote glioma cell invasion. Int J Oncol. 54:713–721. 2019.

53 

Lang HL, Hu GW, Chen Y, Liu Y, Tu W, Lu YM, Wu L and Xu GH: Glioma cells promote angiogenesis through the release of exosomes containing long non-coding RNA POU3F3. Eur Rev Med Pharmacol Sci. 21:959–972. 2017.PubMed/NCBI

54 

Li MY, Yang P, Liu YW, Zhang CB, Wang KY, Wang YY, Yao K, Zhang W, Qiu XG, Li WB, et al: Low c-Met expression levels are prognostic for and predict the benefits of temozolomide chemotherapy in malignant gliomas. Sci Rep. 6:211412016. View Article : Google Scholar : PubMed/NCBI

55 

Wu P, Cai J, Chen Q, Han B, Meng X, Li Y, Li Z, Wang R, Lin L, Duan C, et al: Lnc-TALC promotes O6-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p. Nat Commun. 10:20452019. View Article : Google Scholar :

56 

Wesolowska A, Kwiatkowska A, Slomnicki L, Dembinski M, Master A, Sliwa M, Franciszkiewicz K, Chouaib S and Kaminska B: Microglia-derived TGF-beta as an important regulator of glioblastoma invasion-an inhibition of TGF-beta-dependent effects by shRNA against human TGF-beta type II receptor. Oncogene. 27:918–930. 2008. View Article : Google Scholar

57 

Han J, Alvarez-Breckenridge CA, Wang QE and Yu J: TGF-β signaling and its targeting for glioma treatment. Am J Cancer Res. 5:945–955. 2015.

58 

Miyazawa K and Miyazono K: Regulation of TGF-β family signaling by inhibitory smads. Cold Spring Harb Perspect Biol. 9:a0220952017. View Article : Google Scholar

59 

Brunen D, Willems SM, Kellner U, Midgley R, Simon I and Bernards R: TGF-β: An emerging player in drug resistance. Cell Cycle. 12:2960–2968. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Oshimori N, Oristian D and Fuchs E: TGF-β promotes heterogeneity and drug resistance in squamous cell carcinoma. Cell. 160:963–976. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Nie E, Jin X, Miao F, Yu T, Zhi T, Shi Z, Wang Y, Zhang J, Xie M and You Y: TGF-β1 modulates temozolomide resistance in glioblastoma via altered microRNA processing and elevated MGMT. Neuro Oncol. 23:435–446. 2021. View Article : Google Scholar

62 

Fu T, Yang Y, Mu Z, Sun R, Li X and Dong J: Silencing lncRNA LINC01410 suppresses cell viability yet promotes apoptosis and sensitivity to temozolomide in glioblastoma cells by inactivating PTEN/AKT pathway via targeting miR-370-3p. Immunopharmacol Immunotoxicol. 43:680–692. 2021. View Article : Google Scholar : PubMed/NCBI

63 

Peng L, Chen Z, Chen Y, Wang X and Tang N: MIR155HG is a prognostic biomarker and associated with immune infiltration and immune checkpoint molecules expression in multiple cancers. Cancer Med. 8:7161–7173. 2019. View Article : Google Scholar : PubMed/NCBI

64 

He X, Sheng J, Yu W, Wang K, Zhu S and Liu Q: LncRNA MIR155HG promotes temozolomide resistance by activating the Wnt/β-catenin pathway via binding to PTBP1 in glioma. Cell Mol Neurobiol. 41:1271–1284. 2021. View Article : Google Scholar

65 

Li C, Feng S and Chen L: MSC-AS1 knockdown inhibits cell growth and temozolomide resistance by regulating miR-373-3p/CPEB4 axis in glioma through PI3K/Akt pathway. Mol Cell Biochem. 476:699–713. 2021. View Article : Google Scholar :

66 

Boustani MR, Mehrabi F, Yahaghi E, Khoshnood RJ, Shahmohammadi M, Darian EK and Goudarzi PK: Somatic CPEB4 and CPEB1 genes mutations spectrum on the prognostic predictive accuracy in patients with high-grade glioma and their clinical significance. J Neurol Sci. 363:80–83. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Gu N, Wang X, Di Z, Xiong J, Ma Y, Yan Y, Qian Y, Zhang Q and Yu J: Silencing lncRNA FOXD2-AS1 inhibits proliferation, migration, invasion and drug resistance of drug-resistant glioma cells and promotes their apoptosis via microRNA-98-5p/CPEB4 axis. Aging (Albany NY). 11:10266–10283. 2019. View Article : Google Scholar

68 

Su YK, Lin JW, Shih JW, Chuang HY, Fong IH, Yeh CT and Lin CM: Targeting BC200/miR218-5p signaling axis for overcoming temozolomide resistance and suppressing glioma stemness. Cells. 9:18592020. View Article : Google Scholar :

69 

Ding J, Zhang L, Chen S, Cao H, Xu C and Wang X: lncRNA CCAT2 enhanced resistance of glioma cells against chemodrugs by disturbing the normal function of miR-424. Onco Targets Ther. 13:1431–1445. 2020. View Article : Google Scholar : PubMed/NCBI

70 

Li Z, Meng X, Wu P, Zha C, Han B, Li L, Sun N, Qi T, Qin J, Zhang Y, et al: Glioblastoma cell-derived lncRNA-containing exosomes induce microglia to produce complement C5, promoting chemotherapy resistance. Cancer Immunol Res. 9:1383–1399. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Li B, Zhao H, Song J, Wang F and Chen M: LINC00174 down-regulation decreases chemoresistance to temozolomide in human glioma cells by regulating miR-138-5p/SOX9 axis. Hum Cell. 33:159–174. 2020. View Article : Google Scholar

72 

Jia L, Tian Y, Chen Y and Zhang G: The silencing of LncRNA-H19 decreases chemoresistance of human glioma cells to temozolomide by suppressing epithelial-mesenchymal transition via the Wnt/β-catenin pathway. Onco Targets Ther. 11:313–321. 2018. View Article : Google Scholar :

73 

Jiang P, Wang P, Sun X, Yuan Z, Zhan R, Ma X and Li W: Knockdown of long noncoding RNA H19 sensitizes human glioma cells to temozolomide therapy. Onco Targets Ther. 9:3501–3509. 2016.PubMed/NCBI

74 

Zhou L, Huang X, Zhang Y, Wang L, Li H and Huang H: PSMG3-AS1 enhances glioma resistance to temozolomide via stabilizing c-Myc in the nucleus. Brain Behav. 12:e25312022. View Article : Google Scholar : PubMed/NCBI

75 

Zhang S, Guo S, Liang C and Lian M: Long intergenic noncoding RNA 00021 promotes glioblastoma temozolomide resistance by epigenetically silencing p21 through Notch pathway. IUBMB Life. 72:1747–1756. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Cui B, Li B, Liu Q and Cui Y: lncRNA CCAT1 promotes glioma tumorigenesis by sponging miR-181b. J Cell Biochem. 118:4548–4557. 2017. View Article : Google Scholar : PubMed/NCBI

77 

Wang Y, Yi K, Liu X, Tan Y, Jin W, Li Y, Zhou J, Wang F and Kang C: HOTAIR up-regulation activates NF-κB to induce immunoescape in gliomas. Front Immunol. 12:7854632021. View Article : Google Scholar

78 

Wang W, Han S, Gao W, Feng Y, Li K and Wu D: Long noncoding RNA KCNQ1OT1 confers gliomas resistance to temozolomide and enhances cell growth by retrieving PIM1 from miR-761. Cell Mol Neurobiol. 42:695–708. 2022. View Article : Google Scholar

79 

Wang Y, Shan A, Zhou Z, Li W, Xie L, Du B and Lei B: LncRNA TCONS_00004099-derived microRNA regulates oncogenesis through PTPRF in gliomas. Ann Transl Med. 9:10232021. View Article : Google Scholar : PubMed/NCBI

80 

Dong ZQ, Guo ZY and Xie J: The lncRNA EGFR-AS1 is linked to migration, invasion and apoptosis in glioma cells by targeting miR-133b/RACK1. Biomed Pharmacother. 118:1092922019. View Article : Google Scholar : PubMed/NCBI

81 

Chen M, Cheng Y, Yuan Z, Wang F, Yang L and Zhao H: NCK1-AS1 increases drug resistance of glioma cells to temozolomide by modulating miR-137/TRIM24. Cancer Biother Radiopharm. 35:101–108. 2020. View Article : Google Scholar

82 

Lu Y, Tian M, Liu J and Wang K: LINC00511 facilitates temozolomide resistance of glioblastoma cells via sponging miR-126-5p and activating Wnt/β-catenin signaling. J Biochem Mol Toxicol. 35:e228482021. View Article : Google Scholar

83 

Tomar VS, Patil V and Somasundaram K: Temozolomide induces activation of Wnt/β-catenin signaling in glioma cells via PI3K/Akt pathway: Implications in glioma therapy. Cell Biol Toxicol. 36:273–278. 2020. View Article : Google Scholar

84 

Liu H, Liu Z, Jiang B, Peng R, Ma Z and Lu J: SOX9 overexpression promotes glioma metastasis via Wnt/β-catenin signaling. Cell Biochem Biophys. 73:205–212. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, et al: The somatic genomic landscape of glioblastoma. Cell. 155:462–477. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Chen Q, Cai J, Wang Q, Wang Y, Liu M, Yang J, Zhou J, Kang C, Li M and Jiang C: Long noncoding RNA NEAT1, regulated by the EGFR pathway, contributes to glioblastoma progression through the WNT/β-catenin pathway by scaffolding EZH2. Clin Cancer Res. 24:684–695. 2018. View Article : Google Scholar

87 

Knizhnik AV, Roos WP, Nikolova T, Quiros S, Tomaszowski KH, Christmann M and Kaina B: Survival and death strategies in glioma cells: Autophagy, senescence and apoptosis triggered by a single type of temozolomide-induced DNA damage. PLoS One. 8:e556652013. View Article : Google Scholar : PubMed/NCBI

88 

Linder S, Wiesner C and Himmel M: Degrading devices: Invadosomes in proteolytic cell invasion. Annu Rev Cell Dev Biol. 27:185–211. 2011. View Article : Google Scholar : PubMed/NCBI

89 

Ulasov IV, Mijanovic O, Savchuk S, Gonzalez-Buendia E, Sonabend A, Xiao T, Timashev P and Lesniak MS: TMZ regulates GBM stemness via MMP14-DLL4-Notch3 pathway. Int J Cancer. 146:2218–2228. 2020. View Article : Google Scholar

90 

Wen Q, Chen Z, Chen Z, Chen J, Wang R, Huang C and Yuan W: EphA2 affects the sensitivity of oxaliplatin by inducing EMT in oxaliplatin-resistant gastric cancer cells. Oncotarget. 8:47998–48011. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Gaianigo N, Melisi D and Carbone C: EMT and treatment resistance in pancreatic cancer. Cancers (Basel). 9:1222017. View Article : Google Scholar

92 

Peng F, Fan H, Li S, Peng C and Pan X: MicroRNAs in epithelial-mesenchymal transition process of cancer: potential targets for chemotherapy. Int J Mol Sci. 22:75262021. View Article : Google Scholar : PubMed/NCBI

93 

Li Z, Li M, Xia P and Lu Z: HOTTIP mediated therapy resistance in glioma cells involves regulation of EMT-related miR-10b. Front Oncol. 12:8735612022. View Article : Google Scholar : PubMed/NCBI

94 

Loilome W, Joshi AD, ap Rhys CM, Piccirillo S, Vescovi AL, Gallia GL and Riggins GJ: Glioblastoma cell growth is suppressed by disruption of fibroblast growth factor pathway signaling. J Neurooncol. 94:359–366. 2009. View Article : Google Scholar : PubMed/NCBI

95 

Snuderl M, Fazlollahi L, Le LP, Nitta M, Zhelyazkova BH, Davidson CJ, Akhavanfard S, Cahill DP, Aldape KD, Betensky RA, et al: Mosaic amplification of multiple receptor tyrosine kinase genes in glioblastoma. Cancer Cell. 20:810–817. 2011. View Article : Google Scholar : PubMed/NCBI

96 

Raoof S, Ruddy D, Timonia D, Damon L, Engelman J and Hata A: Abstract A142: Targeting FGFR to overcome EMT-related resistance in EGFR-mutated non-small cell lung cancer. Mol Cancer Ther. 17(1 Suppl): A1422018. View Article : Google Scholar

97 

Zhang L, Zhang W, Li Y, Alvarez A, Li Z, Wang Y, Song L, Lv D, Nakano I, Hu B, et al: SHP-2-upregulated ZEB1 is important for PDGFRα-driven glioma epithelial-mesenchymal transition and invasion in mice and humans. Oncogene. 35:5641–5652. 2016. View Article : Google Scholar : PubMed/NCBI

98 

Brichkina A, Nguyen NT, Baskar R, Wee S, Gunaratne J, Robinson RC and Bulavin DV: Proline isomerisation as a novel regulatory mechanism for p38MAPK activation and functions. Cell Death Differ. 23:1592–1601. 2016. View Article : Google Scholar : PubMed/NCBI

99 

Park CM, Park MJ, Kwak HJ, Lee HC, Kim MS, Lee SH, Park IC, Rhee CH and Hong SI: Ionizing radiation enhances matrix metalloproteinase-2 secretion and invasion of glioma cells through Src/epidermal growth factor receptor-mediated p38/Akt and phosphatidylinositol 3-kinase/Akt signaling pathways. Cancer Res. 66:8511–8519. 2006. View Article : Google Scholar : PubMed/NCBI

100 

Cong ZX, Wang HD, Zhou Y, Wang JW, Pan H, Zhang DD, Zhang L and Zhu L: Temozolomide and irradiation combined treatment-induced Nrf2 activation increases chemoradiation sensitivity in human glioblastoma cells. J Neurooncol. 116:41–48. 2014. View Article : Google Scholar

101 

Ma L, Liu J, Zhang X, Qi J, Yu W and Gu Y: p38 MAPK-dependent Nrf2 induction enhances the resistance of glioma cells against TMZ. Med Oncol. 32:692015. View Article : Google Scholar : PubMed/NCBI

102 

Carnero A, Blanco-Aparicio C, Renner O, Link W and Leal JF: The PTEN/PI3K/AKT signalling pathway in cancer, therapeutic implications. Curr Cancer Drug Targets. 8:187–198. 2008. View Article : Google Scholar : PubMed/NCBI

103 

Harder BG, Peng S, Sereduk CP, Sodoma AM, Kitange GJ, Loftus JC, Sarkaria JN and Tran NL: Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells. Mol Med. 25:492019. View Article : Google Scholar : PubMed/NCBI

104 

Pridham KJ, Shah F, Hutchings KR, Sheng KL, Guo S, Liu M, Kanabur P, Lamouille S, Lewis G, Morales M, et al: Connexin 43 confers chemoresistance through activating PI3K. Oncogenesis. 11:22022. View Article : Google Scholar : PubMed/NCBI

105 

Zając A, Sumorek-Wiadro J, Langner E, Wertel I, Maciejczyk A, Pawlikowska-Pawlęga B, Pawelec J, Wasiak M, Hułas-Stasiak M, Bądziul D, et al: Involvement of PI3K pathway in glioma cell resistance to temozolomide treatment. Int J Mol Sci. 22:51552021. View Article : Google Scholar

106 

Zhang LH, Yin AA, Cheng JX, Huang HY, Li XM, Zhang YQ, Han N and Zhang X: TRIM24 promotes glioma progression and enhances chemoresistance through activation of the PI3K/Akt signaling pathway. Oncogene. 34:600–610. 2015. View Article : Google Scholar

107 

Cao X, Hou J, An Q, Assaraf YG and Wang X: Towards the overcoming of anticancer drug resistance mediated by p53 mutations. Drug Resist Updat. 49:1006712020. View Article : Google Scholar

108 

Hientz K, Mohr A, Bhakta-Guha D and Efferth T: The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget. 8:8921–8946. 2017. View Article : Google Scholar :

109 

Hirose Y, Berger MS and Pieper RO: Abrogation of the Chk1-mediated G(2) checkpoint pathway potentiates temozolomide-induced toxicity in a p53-independent manner in human glioblastoma cells. Cancer Res. 61:5843–5849. 2001.PubMed/NCBI

110 

Holder SL and Abdulkadir SA: PIM1 kinase as a target in prostate cancer: Roles in tumorigenesis, castration resistance, and docetaxel resistance. Curr Cancer Drug Targets. 14:105–114. 2014. View Article : Google Scholar

111 

Wang BW, Huang CH, Liu LC, Cheng FJ, Wei YL, Lin YM, Wang YF, Wei CT, Chen Y, Chen YJ and Huang WC: Pim1 kinase inhibitors exert anti-cancer activity against HER2-positive breast cancer cells through downregulation of HER2. Front Pharmacol. 12:6146732021. View Article : Google Scholar : PubMed/NCBI

112 

Trigg RM, Lee LC, Prokoph N, Jahangiri L, Reynolds CP, Amos Burke GA, Probst NA, Han M, Matthews JD, Lim HK, et al: The targetable kinase PIM1 drives ALK inhibitor resistance in high-risk neuroblastoma independent of MYCN status. Nat Commun. 10:54282019. View Article : Google Scholar : PubMed/NCBI

113 

Wein L and Loi S: Mechanisms of resistance of chemotherapy in early-stage triple negative breast cancer (TNBC). Breast. 34(Suppl 1): S27–S30. 2017. View Article : Google Scholar : PubMed/NCBI

114 

Bobustuc GC, Kassam AB, Rovin RA, Jeudy S, Smith JS, Isley B, Singh M, Paranjpe A, Srivenugopal KS and Konduri SD: MGMT inhibition in ER positive breast cancer leads to CDC2, TOP2A, AURKB, CDC20, KIF20A, Cyclin A2, cyclin B2, cyclin D1, ERα and survivin inhibition and enhances response to temozolomide. Oncotarget. 9:29727–29742. 2018. View Article : Google Scholar : PubMed/NCBI

115 

Song Z, Pan Y, Ling G, Wang S, Huang M, Jiang X and Ke Y: Escape of U251 glioma cells from temozolomide-induced senescence was modulated by CDK1/survivin signaling. Am J Transl Res. 9:2163–2180. 2017.PubMed/NCBI

116 

Reich TR, Schwarzenbach C, Vilar JB, Unger S, Mühlhäusler F, Nikolova T, Poplawski A, Baymaz HI, Beli P, Christmann M and Tomicic MT: Localization matters: Nuclear-trapped survivin sensitizes glioblastoma cells to temozolomide by elevating cellular senescence and impairing homologous recombination. Cell Mol Life Sci. 78:5587–5604. 2021. View Article : Google Scholar : PubMed/NCBI

117 

Li Z, Wu X, Zhao Y, Xiao Y, Zhao Y, Zhang T, Li H, Sha F, Wang Y, Deng L and Ma X: Clinical benefit of neoadjuvant anti-PD-1/PD-L1 utilization among different tumors. MedComm (2020). 2:60–68. 2021.

118 

Zhou Y, Miao J, Wu H, Tang H, Kuang J, Zhou X, Peng Y, Hu D, Shi D, Deng W, et al: PD-1 and PD-L1 expression in 132 recurrent nasopharyngeal carcinoma: The correlation with anemia and outcomes. Oncotarget. 8:51210–51223. 2017. View Article : Google Scholar : PubMed/NCBI

119 

Qin T, Zeng YD, Qin G, Xu F, Lu JB, Fang WF, Xue C, Zhan JH, Zhang XK, Zheng QF, et al: High PD-L1 expression was associated with poor prognosis in 870 Chinese patients with breast cancer. Oncotarget. 6:33972–33981. 2015. View Article : Google Scholar : PubMed/NCBI

120 

Jiang X, Wang J, Deng X, Xiong F, Ge J, Xiang B, Wu X, Ma J, Zhou M, Li X, et al: Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer. 18:102019. View Article : Google Scholar : PubMed/NCBI

121 

Kathuria H, Millien G, McNally L, Gower AC, Tagne JB, Cao Y and Ramirez MI: NKX21-AS1 negatively regulates CD274/PD-L1, cell-cell interaction genes, and limits human lung carcinoma cell migration. Sci Rep. 8:144182018. View Article : Google Scholar

122 

Tian Y, Li L, Lin G, Wang Y, Wang L, Zhao Q, Hu Y, Yong H, Wan Y and Zhang Y: lncRNA SNHG14 promotes oncogenesis and immune evasion in diffuse large-B-cell lymphoma by sequestering miR-152-3p. Leuk Lymphoma. 62:1574–1584. 2021. View Article : Google Scholar : PubMed/NCBI

123 

Dang S, Malik A, Chen J, Qu J, Yin K, Cui L and Gu M: LncRNA SNHG15 contributes to immuno-escape of gastric cancer through targeting miR141/PD-L1. Onco Targets Ther. 13:8547–8556. 2020. View Article : Google Scholar : PubMed/NCBI

124 

Zhou WY, Zhang MM, Liu C, Kang Y, Wang JO and Yang XH: Long noncoding RNA LINC00473 drives the progression of pancreatic cancer via upregulating programmed death-ligand 1 by sponging microRNA-195-5p. J Cell Physiol. 234:23176–23189. 2019. View Article : Google Scholar : PubMed/NCBI

125 

Fan F, Chen K, Lu X, Li A, Liu C and Wu B: Dual targeting of PD-L1 and PD-L2 by PCED1B-AS1 via sponging hsa-miR-194-5p induces immunosuppression in hepatocellular carcinoma. Hepatol Int. 15:444–458. 2021. View Article : Google Scholar

126 

Peng L, Chen Y, Ou Q, Wang X and Tang N: LncRNA MIAT correlates with immune infiltrates and drug reactions in hepatocellular carcinoma. Int Immunopharmacol. 89:1070712020. View Article : Google Scholar : PubMed/NCBI

127 

Mineo M, Lyons SM, Zdioruk M, von Spreckelsen N, Ferrer-Luna R, Ito H, Alayo QA, Kharel P, Giantini Larsen A, Fan WY, et al: Tumor interferon signaling is regulated by a lncRNA INCR1 transcribed from the PD-L1 locus. Mol Cell. 78:1207–1223.e8. 2020. View Article : Google Scholar : PubMed/NCBI

128 

Wagle N, Nguyen M, Carrillo J, Truong J, Dobrawa L and Kesari S: Characterization of molecular pathways for targeting therapy in glioblastoma. Chin Clin Oncol. 9:772020. View Article : Google Scholar : PubMed/NCBI

129 

Miyazaki T, Ishikawa E, Matsuda M, Sugii N, Kohzuki H, Akutsu H, Sakamoto N, Takano S and Matsumura A: Infiltration of CD163-positive macrophages in glioma tissues after treatment with anti-PD-L1 antibody and role of PI3Kγ inhibitor as a combination therapy with anti-PD-L1 antibody in in vivo model using temozolomide-resistant murine glioma-initiating cells. Brain Tumor Pathol. 37:41–49. 2020. View Article : Google Scholar : PubMed/NCBI

130 

Roth P, Valavanis A and Weller M: Long-term control and partial remission after initial pseudoprogression of glioblastoma by anti-PD-1 treatment with nivolumab. Neuro Oncol. 19:454–456. 2017.PubMed/NCBI

131 

Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, Durham N, Meyer C, Harris TJ, Albesiano E, et al: Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys. 86:343–349. 2013. View Article : Google Scholar : PubMed/NCBI

132 

Jan CI, Tsai WC, Harn HJ, Shyu WC, Liu MC, Lu HM, Chiu SC and Cho DY: Predictors of response to autologous dendritic cell therapy in glioblastoma multiforme. Front Immunol. 9:7272018. View Article : Google Scholar : PubMed/NCBI

133 

Hua H, Kong Q, Zhang H, Wang J, Luo T and Jiang Y: Targeting mTOR for cancer therapy. J Hematol Oncol. 12:712019. View Article : Google Scholar : PubMed/NCBI

134 

Vargas-Toscano A, Nickel AC, Li G, Kamp MA, Muhammad S, Leprivier G, Fritsche E, Barker RA, Sabel M, Steiger HJ, et al: Rapalink-1 targets glioblastoma stem cells and acts synergistically with tumor treating fields to reduce resistance against temozolomide. Cancers (Basel). 12:38592020. View Article : Google Scholar

135 

Zou Y, Chen M, Zhang S, Miao Z, Wang J, Lu X and Zhao X: TRPC5-induced autophagy promotes the TMZ-resistance of glioma cells via the CAMMKβ/AMPKα/mTOR pathway. Oncol Rep. 41:3413–3423. 2019.PubMed/NCBI

136 

Jiang C, Shen F, Du J, Fang X, Li X, Su J, Wang X, Huang X and Liu Z: Upregulation of CASC2 sensitized glioma to temozolomide cytotoxicity through autophagy inhibition by sponging miR-193a-5p and regulating mTOR expression. Biomed Pharmacother. 97:844–850. 2018. View Article : Google Scholar

137 

Liu Q, Yu W, Zhu S, Cheng K, Xu H, Lv Y, Long X, Ma L, Huang J, Sun S and Wang K: Long noncoding RNA GAS5 regulates the proliferation, migration, and invasion of glioma cells by negatively regulating miR-18a-5p. J Cell Physiol. 234:757–768. 2018. View Article : Google Scholar : PubMed/NCBI

138 

Zhao X, Liu Y, Zheng J, Liu X, Chen J, Liu L, Wang P and Xue Y: GAS5 suppresses malignancy of human glioma stem cells via a miR-196a-5p/FOXO1 feedback loop. Biochim Biophys Acta Mol Cell Res. 1864:1605–1617. 2017. View Article : Google Scholar : PubMed/NCBI

139 

Huo JF and Chen XB: Long noncoding RNA growth arrest-specific 5 facilitates glioma cell sensitivity to cisplatin by suppressing excessive autophagy in an mTOR-dependent manner. J Cell Biochem. 120:6127–6136. 2019. View Article : Google Scholar

140 

Li G, Cai Y, Wang C, Huang M and Chen J: LncRNA GAS5 regulates the proliferation, migration, invasion and apoptosis of brain glioma cells through targeting GSTM3 expression. The effect of LncRNA GAS5 on glioma cells. J Neurooncol. 143:525–536. 2019. View Article : Google Scholar : PubMed/NCBI

141 

Yan Y, Xu Z, Dai S, Qian L, Sun L and Gong Z: Targeting autophagy to sensitive glioma to temozolomide treatment. J Exp Clin Cancer Res. 35:232016. View Article : Google Scholar : PubMed/NCBI

142 

Liao Y, Shen L, Zhao H, Liu Q, Fu J, Guo Y, Peng R and Cheng L: LncRNA CASC2 interacts with miR-181a to modulate glioma growth and resistance to TMZ through PTEN pathway. J Cell Biochem. 118:1889–1899. 2017. View Article : Google Scholar : PubMed/NCBI

143 

Jing H and Lee S: NF-κB in cellular senescence and cancer treatment. Mol Cells. 37:189–195. 2014. View Article : Google Scholar : PubMed/NCBI

144 

Sánchez Y, Segura V, Marín-Béjar O, Athie A, Marchese FP, González J, Bujanda L, Guo S, Matheu A and Huarte M: Genome-wide analysis of the human p53 transcriptional network unveils a lncRNA tumour suppressor signature. Nat Commun. 5:58122014. View Article : Google Scholar : PubMed/NCBI

145 

Torres-Bayona S, Aldaz P, Auzmendi-Iriarte J, Saenz-Antoñanzas A, Garcia I, Arrazola M, Gerovska D, Undabeitia J, Querejeta A, Egaña L, et al: PR-LncRNA signature regulates glioma cell activity through expression of SOX factors. Sci Rep. 8:127462018. View Article : Google Scholar : PubMed/NCBI

146 

Ding H, Cui L and Wang C: Long noncoding RNA LIFR-AS1 suppresses proliferation, migration and invasion and promotes apoptosis through modulating miR-4262/NF-κB pathway in glioma. Neurol Res. 43:210–219. 2021. View Article : Google Scholar

147 

Li XT, Li JC, Feng M, Zhou YX and Du ZW: Novel lncRNA-ZNF281 regulates cell growth, stemness and invasion of glioma stem-like U251s cells. Neoplasma. 66:118–127. 2019. View Article : Google Scholar

148 

Weller M, Wick W, Aldape K, Brada M, Berger M, Pfister SM, Nishikawa R, Rosenthal M, Wen PY, Stupp R and Reifenberger G: Glioma. Nat Rev Dis Primers. 1:150172015. View Article : Google Scholar : PubMed/NCBI

149 

Yang W and Gao Y: Translesion and repair DNA polymerases: Diverse structure and mechanism. Annu Rev Biochem. 87:239–261. 2018. View Article : Google Scholar : PubMed/NCBI

150 

Bailly V, Lamb J, Sung P, Prakash S and Prakash L: Specific complex formation between yeast RAD6 and RAD18 proteins: A potential mechanism for targeting RAD6 ubiquitin-conjugating activity to DNA damage sites. Genes Dev. 8:811–820. 1994. View Article : Google Scholar : PubMed/NCBI

151 

Wojtaszek JL, Chatterjee N, Najeeb J, Ramos A, Lee M, Bian K, Xue JY, Fenton BA, Park H, Li D, et al: A small molecule targeting mutagenic translesion synthesis improves chemotherapy. Cell. 178:152–159.e11. 2019. View Article : Google Scholar : PubMed/NCBI

152 

Peng C, Chen Z, Wang S, Wang HW, Qiu W, Zhao L, Xu R, Luo H, Chen Y, Chen D, et al: The error-prone DNA polymerase κ promotes temozolomide resistance in glioblastoma through Rad17-dependent activation of ATR-Chk1 signaling. Cancer Res. 76:2340–2353. 2016. View Article : Google Scholar : PubMed/NCBI

153 

Vassel FM, Bian K, Walker GC and Hemann MT: Rev7 loss alters cisplatin response and increases drug efficacy in chemotherapy-resistant lung cancer. Proc Natl Acad Sci USA. 117:28922–28924. 2020. View Article : Google Scholar : PubMed/NCBI

154 

Wu B, Wang H, Zhang L, Sun C, Li H, Jiang C and Liu X: High expression of RAD18 in glioma induces radiotherapy resistance via down-regulating P53 expression. Biomed Pharmacother. 112:1085552019. View Article : Google Scholar : PubMed/NCBI

155 

Rezaei O, Tamizkar KH, Sharifi G, Taheri M and Ghafouri-Fard S: Emerging role of long non-coding RNAs in the pathobiology of glioblastoma. Front Oncol. 10:6258842021. View Article : Google Scholar : PubMed/NCBI

156 

Luo J, Bai R, Liu Y, Bi H, Shi X and Qu C: Long non-coding RNA ATXN8OS promotes ferroptosis and inhibits the temozolomide-resistance of gliomas through the ADAR/GLS2 pathway. Brain Res Bull. 186:27–37. 2022.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

157 

Gao XY, Zang J, Zheng MH, Zhang YF, Yue KY, Cao XL, Cao Y, Li XX, Han H, Jiang XF and Liang L: Temozolomide treatment induces HMGB1 to promote the formation of glioma stem cells via the TLR2/NEAT1/Wnt pathway in glioblastoma. Front Cell Dev Biol. 9:6208832021. View Article : Google Scholar : PubMed/NCBI

158 

Wang C, Chen Y, Wang Y, Liu X, Liu Y, Li Y, Chen H, Fan C, Wu D and Yang J: Inhibition of COX-2, mPGES-1 and CYP4A by isoliquiritigenin blocks the angiogenic Akt signaling in glioma through ceRNA effect of miR-194-5p and lncRNA NEAT1. J Exp Clin Cancer Res. 38:3712019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2022
Volume 61 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li S, Xie X, Peng F, Du J and Peng C: Regulation of temozolomide resistance via lncRNAs: Clinical and biological properties of lncRNAs in gliomas (Review). Int J Oncol 61: 101, 2022
APA
Li, S., Xie, X., Peng, F., Du, J., & Peng, C. (2022). Regulation of temozolomide resistance via lncRNAs: Clinical and biological properties of lncRNAs in gliomas (Review). International Journal of Oncology, 61, 101. https://doi.org/10.3892/ijo.2022.5391
MLA
Li, S., Xie, X., Peng, F., Du, J., Peng, C."Regulation of temozolomide resistance via lncRNAs: Clinical and biological properties of lncRNAs in gliomas (Review)". International Journal of Oncology 61.3 (2022): 101.
Chicago
Li, S., Xie, X., Peng, F., Du, J., Peng, C."Regulation of temozolomide resistance via lncRNAs: Clinical and biological properties of lncRNAs in gliomas (Review)". International Journal of Oncology 61, no. 3 (2022): 101. https://doi.org/10.3892/ijo.2022.5391