Open access peer-reviewed chapter

Potential Role of Cancer Stem Cells in Glioblastoma: A Therapeutic Aspect

Written By

Meenakshi Tiwari, Lokendra Kumar Sharma and Ajit Kumar Saxena

Submitted: 30 June 2022 Reviewed: 06 July 2022 Published: 02 August 2022

DOI: 10.5772/intechopen.106332

From the Edited Volume

Glioblastoma - Current Evidence

Edited by Amit Agrawal and Daulat Singh Kunwar

Chapter metrics overview

89 Chapter Downloads

View Full Metrics

Abstract

High-grade glioma (HGG) such as glioblastoma multiforme (GBM) is an aggressive brain tumor that is still associated with poor prognosis. With the discovery and advancement in understanding of cancer stem cells (CSC) in glioma, these cells have emerged as seed cells for tumor growth and recurrence and appear as a potential target for therapeutics. Glioma stem cells (GSCs) demonstrate capacity of self-renewal, proliferation, and differentiation into multiple cell types and can contribute to tumor heterogeneity. Their role is established in tumorigenesis, metastasis, chemo- and radio-resistance and appears as a major cause for tumor recurrence. Thus, targeting GSCs by various therapeutics may improve effectiveness of the drugs in use alone or in combination to significantly improve patient survival outcome in GBM cases. In this chapter, we have discussed various mechanisms that drive GSC including signaling pathways and tumor microenvironment. We have also discussed the mechanism behind resistance of GSCs toward therapeutics and the pathways that can be targeted to improve the outcome of the patients.

Keywords

  • Glioblastoma multiforme
  • cancer stem cells
  • glioma stem cells
  • signaling pathways
  • chemotherapeutics
  • tumor microenvironment
  • resistance to therapy

1. Introduction

Glioblastoma multiforme (GBM), classified as grade IV glioma, is highly invasive, heterogeneous, and malignant primary brain tumor. It accounts for ~57% of all gliomas and ~ 48% of all primary malignant central nervous system (CNS) tumors [1, 2]. Such tumors are associated with poor quality of life of the patient due to progressive decline in neurologic function, thus making a huge impact on the patients, care givers, and their families. The standard treatment includes multimodal approach involving maximal surgical resection followed by radiotherapy, systemic therapy (chemotherapy, targeted therapy), and supportive care; however, long-term survival is exceptional. Despite the treatment, these tumors regrow and that too with aggressive phenotype, which worsen the symptoms leading to prognosis with average overall survival time < 14.6 months for primary GBM patients and < 6.9 months for recurrent GBM patients [3]. Understanding the molecular mechanism involved in therapeutic resistance and tumor regrowth despite standard treatment is imperative.

In this regard, researchers have identified existence of cancer stem cells (CSCs) in a variety of cancers that play crucial role in tumor initiation, maintenance, resistance to therapy, recurrence, metastasis, and generation of more aggressive phenotype [4]. These properties of CSC are manifested by their potential to self-renew, proliferate, ability to differentiate in multiple phenotypes, plasticity, quiescence, and dormancy. It is suggested that these CSCs originate either from normal stem cells that were already present in tissue or can be generated from dedifferentiation of somatic cells from bulk of tumor. Based on the properties of CSCs, they pose not only a barrier for anticancer therapy but also are responsible for recurrence into more aggressive phenotype. Various researchers have shown that CSC escape anticancer therapy due to their ability to enter dormancy, plasticity, renewal, and regrowth into heterogeneous group of tumor cells. Of interest, recent evidences have suggested that these CSCs are further enriched in response to standard radio-chemotherapy, which may be responsible for tumor regrowth and aggressive phenotype. These enriched CSCs might be result from the existing population of CSCs that evades the therapy or as per recent evidences, can be generated from non-CSCs from the bulk of tumor in response to therapy. Of note, CSCs have been identified in HGG cases also known as glioma stem cells (GSCs) that contribute to tumor heterogeneity and resistance to therapies, thus a major contributor of tumor recurrence. These GSCs are considered as a potential therapeutic target, therefore understanding the molecular pathways that drive GSCs becomes imperative [5]. In this book chapter, we have discussed about the properties of cancer stem cells, cell surface markers, signaling pathways, and mechanism of resistance to therapies and ways by which these pathways can be targeted using different chemotherapeutic agents.

Advertisement

2. Biology of cancer stem cells

Stem cells are specialized cells present in our body that possess properties such as capacity to self-renew, proliferate, and differentiate into multiple cell types. This quality of self-renewal along with associated signaling pathways is shared between both stem cells and cancer stem cells with added feature of oncogenicity in CSCs. The most common pathways that drive multipotency and self-renewal of stem cells include the Notch, Sonic hedgehog (Shh), and Wnt signaling pathways [4]. Due to activation of oncogenic pathways, CSCs can give rise to tumor mass consisting of heterogeneous cell population. Initially, Bonnet and Dick characterized CSCs in acute myeloid leukemia as leukemia-initiating cell that possessed properties of leukemia stem cell [6]. Later, such cells were also identified in a variety of solid tumors, including prostate [7], colon [8], lung [9], ovarian [10], and brain [11] tumors. It is hypothesized that CSCs are the seed of a tumor that are responsible for tumorigenesis by initiation, maintenance, propagation, resistance to therapy, recurrence as well as progression of the tumor [12].

Advertisement

3. Glioma stem cells

In brain tumors, presence of CSC has been identified and characterized by various groups and are defined as GSCs or glioma initiating cells [11]. When cultured, these cells grown into neurospheres that constitute of cells that express SC markers including Nestin and CD133. When these cells are injected into nude mice, they lead to tumor formation due to their SC properties [13]. To add further, various groups have utilized properties of stem cells that are present in brain predominantly in subventricular zone (SVZ) to initiate tumor by exposure to chemicals (ethylnitrosourea) or viruses (avian sarcoma virus) in animals that strongly support the importance of stem cells in tumor formation [7, 14, 15]. These cells contribute to tumor heterogeneity and plasticity and have shown resistance to therapies and thus have emerged as a major contributor of tumor recurrence [5, 16, 17]. These CSCs are also influenced by micro environmental conditions such as nutrient deprivation, hypoxia, pH, vasculature, radiation, and chemotherapeutic treatment (explained in detail in coming sections) [16, 17, 18, 19].

Several putative GSC surface markers, such as CD133, CD15, and CD44, and GSC transcription factors, such as SRY-box transcription factor 2 (SOX2), octamer-binding transcription factor 4 (OCT4), and NANOG, have been discovered [20, 21]. However, before its clinical implication, higher sensitivity and specificity of these GSC markers need to be established [21, 22].

Advertisement

4. Major signaling pathways that drive glioma stem cells

In order to maintain stemness properties, GSCs depend upon number of signaling pathways that also support them to sustain under adverse conditions during tumorigenesis [23, 24, 25]. To understand the process of stemness in GSC, the signaling pathways that are also a part of normal neuronal stem cells are discussed. These pathways mainly include Notch, bone morphogenic proteins (BMPs), NF-κB, Wnt, epidermal growth factor (EGF), and Shh that determine the property of stemness.

4.1 Notch signaling

Notch signaling pathway is crucial in developmental process and plays a major role during embryonic development. This pathway regulates cellular proliferation, differentiation, apoptosis, and cell lineage decisions. In GSCs, Notch signaling pathways are highly active, which in turn maintain stemness by inhibiting differentiation. Notch signaling is also involved in oncogenic transformation. It has been identified that inhibition of Notch signaling decreases oncogenic potential of GSCs [26, 27].

4.2 Bone morphogenetic proteins (BMPs)

BMP group of molecules belongs to the transforming growth factor-β (TGF-β) superfamily of proteins. BMPs plays role during embryogenesis, development as well as adult tissue homeostasis. It interacts with different signaling molecules including Wnt/β-catenin, basic helix-loop-helix (bHLH), and hypoxia-inducible factor-1α (HIF-1α) to regulate different processes in all the body organs [28, 29]. BMPs have been identified to regulate the niche as well as stem cells residing within. Besides normal functions, BMPs are also involved in tumorigenesis where BMP2 and BMP4 have emerged as key players. It is identified that dysregulation of the BMP pathway results in sustained cell transformation in stem cells and their niche. BMP signaling pathways are also involved in regulation of cellular proliferation, differentiation, and apoptosis in NSCs. NSCs are differentiated to astroglial lineage via Wnt-mediated BMP signaling [30] and antagonist of BMP can inhibit differentiation of GSCs and maintains its self-renewal and tumorigenic potential [31]. Further, it was demonstrated that delivery of BMP4 can inhibit brain tumor growth in in vivo system and decreased the rate of mortality [32].

4.3 Wnt/β-catenin signaling

Wnt/β-catenin signaling is a highly conserved pathway that regulates cellular proliferation, differentiation, migration, genetic stability, apoptosis, and stem cell renewal. This pathway also regulates NSC expansion and promotes astroglial lineage differentiation during neural development [33, 34]. In GSC, β-catenin regulates proliferation and differentiation and dysregulated Wnt signaling leads to tumor growth [35, 36, 37]. β-Catenin interacts with FoxM1 to regulate the transcription of various oncogeneic genes such as c-Myc that leads to gliomagenesis [38, 39].

4.4 Epidermal growth factor receptor (EGFR) signaling

The EGFR pathway is one of the most crucial pathways involved in cellular processes including proliferation, differentiation, migration, and apoptosis in a variety of cells including stem cells. Dysregulation of this pathway has been linked to cancer. Critical role of EGFR has been identified in NSCs as well [40, 41, 42]. In GSC EGFR works through activation of β-catenin pathway and promotes self-renewal capacity of GSC and induction of tumorigenic potential [43].

4.5 Sonic hedgehog (Shh) signaling

The Shh signaling pathway is crucial for proper embryonic development as it governs tissue polarity, patterning maintenance, cellular proliferation, intercellular communication, and differentiation [44, 45]. Persistent Shh pathway signaling has been observed in the subventricular zone of adult brain where it plays a critical role in regional specification and maintenance of NSCs [46]. Aberrant regulation of the Shh pathway due to mutation has been identified to cause tumorigenesis in a wide variety of cancer tissues including gliomas and GSCs. This pathway is highly active in GSCs where it regulates stemness genes and thus maintains self-renewal of GSC and promotes tumorigenesis and inhibition of Shh signaling reduces both stemness as well as in vivo tumorigenicity by induction of autophagic cell death [47].

Advertisement

5. Pathways that contributing to resistance of glioma stem cells toward therapies that lead to tumor regrowth

Resistance of CSCs toward therapies resulting in their enrichment and regrowth of tumor due to proliferation of these cells has been suggested by various researchers [48, 49, 50]. In HGG, despite the effectiveness of TMZ in removing the bulk tumor cells, regrowth with a more aggressive phenotype is inevitable, and researchers have identified critical role of CSCs in such regrowth. For instance, in HGG, treatment with therapeutic doses of temozolomide (TMZ) leads to expansion of GSCs pool in both patient-derived and established glioma cell lines. Such expansions are reported not only due to enrichment and proliferation of existing CSCs but also due to interconversion between differentiated tumor cells and GSCs [18]. Similarly, bevacizumab (VEGF antibody) although reduces GBM tumor growth, it is followed by tumor regrowth where the role of autocrine signaling through the VEGF-VEGFR2-Neuropilin-1 (NRP1) axis leads to enrichment of active VEGFR2 GSC subset in human GBM cells [51]. It is evident that the therapeutics evoke enrichment of CSCs involving multiple mechanisms. Thus, understanding various ways by which CSCs escape the radio- and chemotherapy, more effective treatment modalities can be developed. Broadly, in CSCs various different mechanism such as epithelial-mesenchymal transition (EMT), multiple drug resistance (MDR) dormancy, tumor environment contribute to resistance toward therapeutics and other adverse conditions faced by them in tumor microenvironment and are discussed as follows.

5.1 DNA repair systems

GSCs possess better DNA repair capacity as compared with bulk tumor cells [52]. These cells express higher levels of DNA repair enzymes such as O6-methylguanine-methyltransferase (MGMT), which are responsible for therapy resistance against DNA alkylating agents such as TMZ [53, 54, 55, 56]. However, there are contradictory studies that also suggest that TMZ resistance in GSCs is independent of MGMT status and alternate pathways might be involved [57, 58]. Further, preferential expression of DNA checkpoint kinases 1 (Chk1) and 2 (Chk2) lead to more efficient repair of DNA damage in CD133-positive glioma cells than CD133-negative glioma cells [54]. Other transcriptional regulators such as BMI, DNA-PK, poly (ADP-ribose) polymerase-1, hnRNP U, and histone H1, which play a role in DNA double-strand break repair, are highly expressed in CD133-positive GBM cells and play pivotal role in GSCs’ functions [59, 60].

5.2 Epithelial-mesenchymal transition (EMT)

EMT involves phenotypic changes in cells from epithelial to mesenchymal type involving high expression of markers such as N-cadherin and vimentin under various physiological as well as pathological conditions including cancer [61]. Interestingly, CSCs also share the EMT-like cell features [62], and it is believed that the link between EMT and CSCs might be responsible for cancer drug resistance acquisition and plasticity resulting in cancer cells transformation into the malignant cells and vice versa [63]. Circulating tumor cells from patients with metastasis co-express markers of EMT as well as CSCs. Further, induction of EMT confers stem-like features in cancer cells [64, 65]. Various regulators of EMT have been identified that regulate stemness. ZEB1 is one such regulator of EMT that regulates stemness and chemoresistance induction by regulating MGMT via miR-200c and c-MYB in malignant glioma [66]. Therefore, a strong association of EMT and CSCs has been identified that provides not only resistance but also promotes metastasis [67].

5.3 Dormancy of CSCs

As the understanding of CSC biology has improved, it has been identified that CSCs can exist in proliferative or dormant state. Dormant CSCs maintain a low metabolic activity, however, show similarities with the normal proliferative counterpart in terms of stemness and other signaling pathways. For instance, dormant stem cells are low in metabolic activity that preferentially utilize the glycolytic pathway and produce low levels of levels of reactive oxygen species (ROS) [68]. However, these dormant/quiescent cells demonstrate high plasticity and can be reactivated to reenter proliferative stage and lead to tumor formation. Such dormant cells are also chemoresistant due to their dormant nature; interestingly, proliferative CSC can also enter dormancy in response to chemotherapeutics agents. In GBM, existence of a relatively quiescent subset of GSCs has been observed, which is responsible for maintaining the long-term tumor growth and responsible for recurrence by entering into highly proliferative cells upon receiving proper signals [69].

5.4 Anti-apoptosis

Various anti-apoptotic protein such as B-cell lymphoma-2 (BCL-2), BCL2 like 1 (BCL2L1), myeloid cell leukemia-1, MCL1 and BCL-xL are highly expressed in GSCs than differentiated bulk tumor cells. These proteins not only play role in GSCs maintenance but also provide survival advantage to these cells against various chemotherapeutic agents [70]. Other mediator of GSCs resistance includes BMI1, a GSC-enriched protein that inhibits p53-mediated apoptosis against TMZ [71]. Inhibition of such anti-apoptotic pathways can increase sensitivity of GSCs against different therapeutic agents.

5.5 Multidrug resistance

Stem cells express higher levels of several ATP-binding cassette (ABC) transporters resulting in efflux ability for various antineoplastic drugs [72]. In GSCs, increased ABCG1 expression has been documented in the side population cells in flow cytometry that present the GSC phenotype [73]. Further, multidrug resistance 1 (MDR1) overexpression was reported higher in CD133+ GSCs than CD133 bulk tumor cells [74]. ABCG2/BCRP and ABCB1/MDR1 overexpression in GSCs has also been correlated with resistance of GSCs to chemotherapeutic drugs and use of an ABC transporter inhibitor, such as verapamil, can help in increasing sensitivity of GSCs toward chemotherapeutic agents such as temozolomide, doxorubicin, and mitoxantrone in GSCs [75]. Similarly, methylation of ABC transporter ABCG2/BCRP promoter by melatonin (N-acetyl-5-methoxytryptamine) promotes toxic effect of TMZ on GSCs [75]. Interestingly, treatment with chemotherapeutic agents can further increase expression of these MDR proteins conferring resistance to these cells against chemotherapeutic agents [76]. Thus, inhibition of drug efflux proteins such as MDR proteins appears as a potential target for increasing sensitivity of GSCs toward various chemotherapeutic agents [75, 77].

5.6 Metabolism

GSCs show metabolic adaptations to survive adverse conditions of tumor microenvironment that includes low pH, hypoxia, and low nutrient supply; at the same time they proliferate at a high rate to maintain their stemness [16, 17]. Majority of GSCs rely on glucose uptake via high-affinity glucose transporter 3 (GLUT3) to provide carbon source for nucleotide biosynthesis for rapid proliferating cells along with high energy demands [78, 79, 80]. These cells also highly express glutamine synthetase as compared with differentiated glioma cells for higher glutamine uptake, which acts as preferential source for de-novo purine biosynthesis [81]. Further studies demonstrate that in therapy-resistant GSCs expression of glucose uptake associated genes is downregulated, and they preferentially use fatty acids as a major ATP source [82]. Additionally, slow-cycling GSCs rely on oxidative phosphorylation and lipid metabolism than fast-cycling GSCs which prefer glycolysis [83]. These anabolic advantages of GSCs may contribute to their chemoresistant phenotype and can be targeted to improve sensitivity of GBM treatment.

5.7 Autophagy

Autophagy is a catabolic pathway which is a cellular stress response under physiological as well as pathological conditions. This pathways acts by removal of damaged macromolecules such as proteins, nucleic acid, and lipids and recycles them for cellular processes and thus promotes cell survival; however, defect or dysregulation of such pathway may lead to cell death [84]. Role of autophagy is well established in a variety of cancers including GBM where it can play a role in cell survival or cell death [84, 85]. Autophagy also contributes to the maintenance of stemness characteristics of GSCs as well as provides chemoresistance to CSC against therapeutic agents [19, 86]. Inhibition of autophagy sensitizes GSCs towards a variety of therapeutic agents [19, 87, 88, 89]. Interestingly, other studies demonstrated that induction of autophagy by mammalian target of rapamycin (mTOR) inhibitors as well as curcumin-induced autophagy shows anti proliferative effect, induces differentiation and also improves sensitivity of GSC towards DNA damaging agents [90, 91, 92]. Together, these results suggest that GSCs require a balanced level of autophagy, too much or too little can significantly affect their stemness potential and resistance toward therapeutics. Further, role of autophagy has also been shown to support motility/migration capacity of GSCs [93]. However, role of autophagy in suppression of the self-renewal ability and tumorigenicity of GSCs has also been demonstrated where autophagy mediates Notch1 degradation [94]. Thus, role of autophagy in GSCs is crucial for maintenance of stemness as well as chemotherapeutic agents; targeting such pathway appears as a p These cells also highly express glutamine synthetase otential strategy to make the existing treatment more effective.

5.8 Extrinsic chemoresistance

Besides the signaling pathways and genetic signature of GSCs, extracellular environment also called as microenvironment in which these cells resides also plays crucial role in its functions and determines response towards therapeutic agents [95]. It has been identified that GSCs reside in inner tumor mass where rapid growth and high energy requirement of these cells along with neovasculature result in hypoxic conditions as well as low pH [29, 96]. These adverse conditions further promote expression of GSC markers and associated phenotype [97]. Various hypoxia and acidic pH-induced genes such as hypoxia-inducible factor (HIF) 1 and 2alpha and vascular endothelial growth factor (VEGF) are highly expressed in GSCs that contribute to GSC functions [98, 99]. It has been shown that in GSCs Notch signaling and MGMT expression are also regulated by HIF-1α, resulting in GSC stemness and also resistance toward TMZ [100, 101]. Further, hypoxic GSCs release extracellular vesicles that deliver HIF-1α induced miR-30b-3p that further activates STAT3 pathway and promotes TMZ resistance [102]. Further, it has been identified that TMZ increases the GSC pool in non-GSC subpopulations, indicating that non-GSC shows plasticity and can be converted to GSCs that might be responsible for resistance as well as regrowth of the tumor [18, 19]. Together, these findings suggest that stemness of GSCs may be regulated by tumor microenvironment as well as cellular plasticity; TMZ can stimulate the dedifferentiation of non-GSCs, which might contribute to resistance and recurrence after therapy [18, 19].

5.9 Role of Notch and sonic hedgehog pathways in mediating chemoresistance

Various signaling pathways such as Notch and SHH are active in GSCs compared with bulk tumor cells [103]. Further, in response to TMZ treatment of GSCs from primary GBM cells resulted in upregulation of NOTCH 1, NCOR2, HES1, HES5, and GLI1 genes, suggesting resistance of these cells and increase in the population of such stem cells in glioma, which could be reversed by inhibitors of Notch or SHH inhibitors [104]. Epithelial-mesenchymal transition (EMT) mediates GBM chemoresistance. Another fact contributing to resistance of GSCs is the potential of epithelial-mesenchymal transition. It has been shown that EMT mediator gene ZEB1 can regulate stemness and SOX2 and OLIG2 in gliomas [105].

Advertisement

6. Strategies targeting glioma stem cells

Despite extensive research in oncology, the target is being missed leading to recurrence in a variety of high-grade tumors including malignant gliomas. With advancement in understanding of GSCs and its capacity of initiation, progression, resistance as well as recurrence of tumor, they appear as most promising target to treat cancers such as HGG. The drugs that can target GSC are being developed using multiple strategist including molecular targeting, autophagy inhibition, drug repositioning, and indirect targeting of GSC niches [17, 21, 106].

6.1 Targeting GSC markers and related signaling pathways

GSCs are regulated by various pathways involving differential expression of epidermal growth factor receptor (EGFR), Notch1, sonic hedgehog (Shh), and STAT3, as well as related signaling pathways.

As discussed earlier, CD133 is the most well-characterized cell surface marker for GSCs, which has become a potential target for antibody-based therapy. Different immunotherapeutic approaches such use of synthetic monoclonal antibody, dual-antigen T cell engager, and chimeric antigen receptor (CAR) T cell have been utilized to target CD133+ GSCs. RW03 (anti-CD133 antibody) targets self-renewal ability of GSCs without effecting its proliferative capacity and could be a promising strategy in targeting GSCs [107]. Further, photothermal therapy has also shown selective efficacy in diminishing CD133-positive cells both in-vitro and in-vivo [108].

EGFR, a receptor tyrosine kinase, which is highly expressed in GSCs, is crucial for its survival, self-renewal, and tumorigenicity. Of importance, EGFR variant III (EGFRvIII) mutation is most commonly detectable (25–33%) in GBM cases [109]. Thus, EGFR inhibition becomes a potential target to inhibit GSCs proliferation, self-renewal, and induction of apoptosis [110]. EGFR inhibition in fact enhanced chemo- and radio-sensitivity of human glioma CSCs. Various reversible and irreversible inhibits of EGFR are available that can bind EGFR alone or along with its co-receptor HER2 [111, 112]. First-generation EGFR TKIs include gefitinib and erlotinib that can reversibly bind EGFR along with HER2; however, less than 20% of patients presented a response to these treatments [112, 113]. Irreversible inhibitor of EGFR, osimertinib, has shown efficiency in crossing the blood-brain barrier (BBB) and significantly inhibits GBM tumorigenesis in-vivo [114]. It has also entered phase II clinical studies [115, 116], however, has shown to be marginally effective, which could be due to heterogeneity of GBM [117, 118]. Further, combined treatment of antibodies against EGFRvIII and CD133 showed higher effectivity in elimination of GSCs compared with the antibody against either EGFRvIII or CD133 [111].

Various signaling pathways such as Notch and SHH are active in GSCs compared with bulk tumor cells [114]. Further, in response to TMZ treatment of GSCs from primary GBM cells resulted in upregulation of NOTCH 1, NCOR2, HES1, HES5, and GLI1 genes, suggesting resistance of these cells and increase in the population of such stem cells in glioma, which could be reversed by inhibitors of Notch or SHH inhibitors [118]. Notch1 signaling that contributes to regulation of GSC can be blocked by γ-secretase inhibitor [119]. RO4929097, a γ-secretase inhibitor, reduces the viability of GSCs [120]. Further, Notch1 also regulates VEGF activity in GSCs, and co-inhibition of Notch1 and VEGF have shown synergistic effects in GBM [121]; however, their combined inhibition (RO4929097 with bevacizumab) in phase I clinical trial did not show much improvement in overall survival (OS) and progression-free survival (PFS) in GBM cases [122]. Other studies also identify the role of γ-secretase inhibitor, N-[N-(3, 5-difluorophenacetyl-L-alanyl)]-S-phenylglycine t-butyl ester (DAPT) in improving TMZ sensitivity [123].

Shh/Gli signaling that regulates GSCs cell proliferation, stem cell fate determination, and differentiation has also appeared as potential target for GSCs therapeutics [124]. Inhibition of hedgehog pathway by LDE225 induces autophagic cell death in GSCs with higher sensitivity of CD133-positive cells than CD133-negative cells [125]. LDE225 inhibits expression and nuclear translocation of Gli proteins, a transcriptional effectors of the Shh signaling pathway [126]. Casein kinase 2 (CK2) is another target to inhibit Shh/Gli signaling via transcriptional activation of β-catenin [127] and inhibition of CK2 by, CX-4945 (silmitasertib), reduces MGMT expression and sensitized tumor cells to TMZ [128].

Signal transducer and activator of transcription 3 (STAT3) that regulates multiple processes such as cell cycle and survival, regulation, immune response, and differentiation, tumorigenic transformation has also been implicated in GSC maintenance [129, 130]. Resveratrol (RV), a polyphenol present in grapes, a tumor preventive agent targets STAT3 signaling. In glioma, RV has shown antineoplastic actions by apoptosis induction and improving radio sensitivity of GSCs CD133+ cell population along with reducing of tumorigenic potential. Furthermore, RV also modulates Wnt signaling pathway and EMT activators, thereby regulating stemness of GSCs and reducing cellular motility [131, 132]. Another molecule that inhibits STAT pathway is WP1066, which is an analog of the natural product caffeic acid benzyl ester and targets GSCs. This molecule has shown promising results in clinical trial for patients with recurrent malignant glioma [133]. Other STAT3 inhibitors, STX-0119 and WP1066 have shown ability to suppress GSC proliferation in-vitro; however, inhibition of tumor growth in subcutaneous xenograft model of GSCs was shown only by STX-0119. STX-0119 further demonstrated ability to downregulate expression of GSCs markers [134]. Another small-molecule STAT3 inhibitor, ODZ10117, also decreased the stem cell properties of GSCs and reduced tumor growth in vivo [130].

6.2 Targeting tumor microenvironment

GSCs are localized in specific niches, which have been identified as protective microenvironments in GBM. Five types of GSC niches have been identified where different cell types exists and have specific singling pathways: peri-vascular, peri-arteriolar, peri-hypoxic, peri-immune, and extracellular matrix out of which peri-vascular niche is the most frequently described GSC [135]. GSC microenvironment lacks organizations and has compromised BBB, higher levels of hypoxia, and excessive angiogenesis making it a target for anti-angiogenic therapy [136, 137].

6.3 Drugs targeting metabolic pathways

Drug repositioning also called as repurpose drugs is a growing concept that explores pre-existing a well-established drug to treat diseases aside from the intended ones. This concept results in lowering the overall developmental cost, time and risk assessments, as the efficacy and safety of the original drug have already been well accessed and approved by regulatory authorities [138]. In case of GSCs, repurpose drugs are being tested and have shown encouraging results. Especially, anti-diabetes drugs have been most well studied with promising results in GSCs targeting. Metformin, successfully used for type 2 diabetes mellitus, has entered phase I clinical trial for GBM in combination with TMZ [139]. Metformin preferentially acts in GSCs by inhibiting Akt activation and also induces conversion on GSCs to non-GSCs [140, 141]. Similarly glimepiride, another anti-diabetes drug, impairs GSCs by targeting glycolytic flux and increases its radio sensitivity to GBM [142]. Further, more repurpose drugs need to be identified that can effectively target GSCs along with its associated mechanism before it can be used in clinical application [138].

6.4 Targeting autophagy pathways

Autophagy is a cellular stress response, which can either promote survival or cell death. Our laboratory along with others has identified that autophagy is required for maintenance of GSCs and also plays a role in resistance of GSCs toward chemotherapy [19, 142, 143]. Targeting autophagy by commonly used agent chloroquine (CQ), which blocks the fusion of autophagosomes with lysosomes, has been shown to inhibit GSCs as well as sensitized them toward chemotherapeutic agents [19, 144]. This drug has also entered multiple clinical trials as an adjuvant treatment for GBM; where its antitumor effects of CQ are not limited to GSCs [145]. Further, combination of autophagy inhibitors with radiation effectively induced apoptosis and inhibited tumorosphere formation in GSCs [146, 147]. More selective autophagy inhibitor NSC185058, antagonist of autophagy-related 4B, inhibits tumorigenic potential of GSCs and enhances GBM sensitivity to radiotherapy in xenograft mouse models [148].

Advertisement

7. Conclusions and future directions

Treatment of HGG remains challenging. With identification of GSCs and their properties to resist treatment and repopulate the original tumor, a big momentum has been created in the development of novel therapeutics. Such therapeutic will involve a combination of drugs that controls the bulk tumor mass along with CSCs-directed agents. It has been identified that GSCs are responsible for tumorigenesis, therapeutic resistance, and tumor recurrence, and thus GSC-targeting drugs are being developed for improvement of treatment regime. These GSCs can survive cancer treatment by activating multiple mechanisms such as EMT, signaling pathways to regulate self-renewal, its interaction with tumor microenvironment, higher expression of drug transporters or detoxification proteins, plasticity, autophagy induction, anti-apoptotic mechanism, induction of dormant phenotype, and many others to overcome the toxic effects of therapeutics. With knowledge of these pathways, anticancer therapeutics are targeted against GSCs, which includes directing specific and pathways that regulate GSCs and protect them from therapeutic stress. Such GSC-directed drugs can be combined with agents that are currently in use to achieve better survival rates of cancer patients.

Identification of bioactive products and their molecular mechanisms that can modulate GSCs needs to be incorporated in treatment regime of HGG patients. With recent advancements in the field of high-throughput screening and genetic and epigenetic signatures, specific targeted drugs that can target bulk tumor with minimal generation of induced GSCs along with combination of drug that can target GSCs can be developed. Furthermore, tumor microenvironment that significantly regulates GSCs is also a potential target to prevent rate of dedifferentiation. It is important to consider that current therapeutic can result in conversion of non-GSC to GSCs; therefore, newer drugs or combinations need to be developed that can prevent this detrimental conversion. More stringent strategies involving GSC-targeted therapy along with glioma molecular subtypes need to be designed for selective and effective clinical trials.

However, most therapeutic agents have failed to be approved for clinical application or during clinical trials due to lack of understanding of the underlying mechanisms or failure to consider individual characteristics of the tumor. Further investigation of the molecular pathways that drive GSCs and make them resistant to therapies along with subtype-specific pathways of GSCs is required. Such studies will significantly improve not only the understanding of disease but will also direct the development of highly specific drugs with minimal side effects along with improved patient outcome.

Advertisement

Acknowledgments

The authors would like to acknowledge the funding support from Department of Biotechnology Bio-CARe grant, Govt. of India (BT/P19357/BIC/101/927/2016 to M.T.) and Intramural Research Grant by SGPGIMS (PGI/DIR/RC/36/2021) to LKS.

Advertisement

Conflict of interest

The authors declare that they have no conflict of interest.

References

  1. 1. Dolecek TA, Propp JM, Stroup NE, Kruchko C. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2005-2009 [published correction appears in Neuro Oncol. 2013 May;15(5):646-7]. Neuro-Oncology. 2012;14(Suppl 5):v1-v49. DOI: 10.1093/neuonc/nos218
  2. 2. Davis ME. Glioblastoma: Overview of disease and treatment. Clinical Journal of Oncology Nursing. 2016;20(5 Suppl):S2-S8. DOI: 10.1188/16.CJON.S1.2-8
  3. 3. Hanif F, Muzaffar K, Perveen K, Malhi SM, Simjee SU. Glioblastoma Multiforme: A review of its epidemiology and pathogenesis through clinical presentation and treatment. Asian Pacific Journal of Cancer Prevention. 2017;18(1):3-9. Published 2017 Jan 1. DOI: 10.22034/APJCP.2017.18.1.3
  4. 4. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105-111. DOI: 10.1038/35102167
  5. 5. Kang H, Lee H, Kim D, et al. Targeting glioblastoma stem cells to overcome Chemoresistance: An overview of current therapeutic strategies. Biomedicine. 2022;10(6):1308. Published 2022 Jun 2. DOI: 10.3390/biomedicines10061308
  6. 6. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nature Medicine. 1997;3(7):730-737. DOI: 10.1038/nm0797-730
  7. 7. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Research. 2005;65(23):10946-10951. DOI: 10.1158/0008-5472.CAN-05-2018
  8. 8. Ricci-Vitiani L, Lombardi DG, Pilozzi E, et al. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445(7123):111-115. DOI: 10.1038/nature05384
  9. 9. Kim CF, Jackson EL, Woolfenden AE, et al. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell. 2005;121(6):823-835. DOI: 10.1016/j.cell.2005.03.032
  10. 10. Zhang S, Balch C, Chan MW, et al. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Research. 2008;68(11):4311-4320. DOI: 10.1158/0008-5472.CAN-08-0364
  11. 11. Singh SK, Clarke ID, Terasaki M, et al. Identification of a cancer stem cell in human brain tumors. Cancer Research. 2003;63(18):5821-5828
  12. 12. Sundar SJ, Hsieh JK, Manjila S, Lathia JD, Sloan A. The role of cancer stem cells in glioblastoma. Neurosurgical Focus. 2014;37(6):E6. DOI: 10.3171/2014.9.FOCUS14494
  13. 13. Yuan X, Curtin J, Xiong Y, et al. Isolation of cancer stem cells from adult glioblastoma multiforme. Oncogene. 2004;23(58):9392-9400. DOI: 10.1038/sj.onc.1208311
  14. 14. Copeland DD, Bigner DD. The role of the subependymal plate in avian sarcoma virus brain tumor induction: Comparison of incipient tumors in neonatal and adult rats. Acta Neuropathologica. 1977;38(1):1-6. DOI: 10.1007/BF00691268
  15. 15. Vick NA, Lin MJ, Bigner DD. The role of the subependymal plate in glial tumorigenesis. Acta Neuropathologica. 1977;40(1):63-71. DOI: 10.1007/BF00688574
  16. 16. Gimple RC, Bhargava S, Dixit D, Rich JN. Glioblastoma stem cells: Lessons from the tumor hierarchy in a lethal cancer. Genes & Development. 2019;33(11-12):591-609. DOI: 10.1101/gad.324301.119
  17. 17. Rodriguez SMB, Staicu GA, Sevastre AS, et al. Glioblastoma stem cells-useful tools in the Battle against cancer. International Journal of Molecular Sciences. 2022;23(9):4602. Published 2022 Apr 21. DOI: 10.3390/ijms23094602
  18. 18. Auffinger B, Tobias AL, Han Y, et al. Conversion of differentiated cancer cells into cancer stem-like cells in a glioblastoma model after primary chemotherapy. Cell Death and Differentiation. 2014;21(7):1119-1131. DOI: 10.1038/cdd.2014.31
  19. 19. Abbas S, Singh SK, Saxena AK, Tiwari S, Sharma LK, Tiwari M. Role of autophagy in regulation of glioma stem cells population during therapeutic stress. Journal of Stem Cells & Regenerative Medicine. 2020;16(2):80-89. Published 2020 Dec 11. DOI: 10.46582/jsrm.1602012
  20. 20. Suvà ML, Tirosh I. The glioma stem cell model in the era of single-cell genomics. Cancer Cell. 2020;37(5):630-636. DOI: 10.1016/j.ccell.2020.04.001
  21. 21. Tang X, Zuo C, Fang P, et al. Targeting glioblastoma stem cells: A review on biomarkers, signal pathways and targeted therapy. Frontiers in Oncology. 2021;11:701291. Published 2021 Jul 8. DOI: 10.3389/fonc.2021.701291
  22. 22. Saygin C, Matei D, Majeti R, Reizes O, Lathia JD. Targeting cancer Stemness in the clinic: From hype to Hope. Cell Stem Cell. 2019;24(1):25-40. DOI: 10.1016/j.stem.2018.11.017
  23. 23. Hemmati HD, Nakano I, Lazareff JA, et al. Cancerous stem cells can arise from pediatric brain tumors. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(25):15178-15183. DOI: 10.1073/pnas.2036535100
  24. 24. Rich JN, Eyler CE. Cancer stem cells in brain tumor biology. Cold Spring Harbor Symposia on Quantitative Biology. 2008;73:411-420. DOI: 10.1101/sqb.2008.73.060
  25. 25. Vescovi AL, Galli R, Reynolds BA. Brain tumour stem cells. Nature Reviews. Cancer. 2006;6(6):425-436. DOI: 10.1038/nrc1889
  26. 26. Kanamori M, Kawaguchi T, Nigro JM, et al. Contribution of Notch signaling activation to human glioblastoma multiforme. Journal of Neurosurgery. 2007;106(3):417-427. DOI: 10.3171/jns.2007.106.3.417
  27. 27. Purow BW, Haque RM, Noel MW, et al. Expression of Notch-1 and its ligands, Delta-like-1 and Jagged-1, is critical for glioma cell survival and proliferation. Cancer Research. 2005;65(6):2353-2363. DOI: 10.1158/0008-5472.CAN-04-1890
  28. 28. Kurooka H, Nakahiro T, Mori K, Sano K, Yokota Y. BMP signaling is responsible for serum-induced Id2 expression. Biochemical and Biophysical Research Communications. 2012;420(2):281-287. DOI: 10.1016/j.bbrc.2012.02.150
  29. 29. Pistollato F, Chen HL, Rood BR, et al. Hypoxia and HIF1alpha repress the differentiative effects of BMPs in high-grade glioma. Stem Cells. 2009;27(1):7-17. DOI: 10.1634/stemcells.2008-0402
  30. 30. Kasai M, Satoh K, Akiyama T. Wnt signaling regulates the sequential onset of neurogenesis and gliogenesis via induction of BMPs. Genes to Cells. 2005;10(8):777-783. DOI: 10.1111/j.1365-2443.2005.00876.x
  31. 31. Yan K, Wu Q , Yan DH, et al. Glioma cancer stem cells secrete Gremlin1 to promote their maintenance within the tumor hierarchy. Genes & Development. 2014;28(10):1085-1100. DOI: 10.1101/gad.235515.113
  32. 32. Piccirillo SG, Reynolds BA, Zanetti N, et al. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature. 2006;444(7120):761-765. DOI: 10.1038/nature05349
  33. 33. Paina S, Garzotto D, DeMarchis S, et al. Wnt5a is a transcriptional target of dlx homeogenes and promotes differentiation of interneuron progenitors in vitro and in vivo. The Journal of Neuroscience. 2011;31(7):2675-2687. DOI: 10.1523/JNEUROSCI.3110-10.2011
  34. 34. Pei Y, Brun SN, Markant SL, et al. WNT signaling increases proliferation and impairs differentiation of stem cells in the developing cerebellum. Development. 2012;139(10):1724-1733. DOI: 10.1242/dev.050104
  35. 35. Grigoryan T, Wend P, Klaus A, Birchmeier W. Deciphering the function of canonical Wnt signals in development and disease: Conditional loss- and gain-of-function mutations of beta-catenin in mice. Genes & Development. 2008;22(17):2308-2341. DOI: 10.1101/gad.1686208
  36. 36. Hatten ME, Roussel MF. Development and cancer of the cerebellum. Trends in Neurosciences. 2011;34(3):134-142. DOI: 10.1016/j.tins.2011.01.002
  37. 37. Munji RN, Choe Y, Li G, Siegenthaler JA, Pleasure SJ. Wnt signaling regulates neuronal differentiation of cortical intermediate progenitors. The Journal of Neuroscience. 2011;31(5):1676-1687. DOI: 10.1523/JNEUROSCI.5404-10.2011
  38. 38. Bowman A, Nusse R. Location, location, location: FoxM1 mediates β-catenin nuclear translocation and promotes glioma tumorigenesis. Cancer Cell. 2011;20(4):415-416. DOI: 10.1016/j.ccr.2011.10.003
  39. 39. Zhang N, Wei P, Gong A, et al. FoxM1 promotes β-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis. Cancer Cell. 2011;20(4):427-442. DOI: 10.1016/j.ccr.2011.08.016
  40. 40. Ayuso-Sacido A, Moliterno JA, Kratovac S, et al. Activated EGFR signaling increases proliferation, survival, and migration and blocks neuronal differentiation in post-natal neural stem cells. Journal of Neuro-Oncology. 2010;97(3):323-337. DOI: 10.1007/s11060-009-0035-x
  41. 41. Wong RW. Transgenic and knock-out mice for deciphering the roles of EGFR ligands. Cellular and Molecular Life Sciences. 2003;60(1):113-118. DOI: 10.1007/s000180300007
  42. 42. Ayuso-Sacido A, Graham C, Greenfield JP, Boockvar JA. The duality of epidermal growth factor receptor (EGFR) signaling and neural stem cell phenotype: Cell enhancer or cell transformer? Current Stem Cell Research & Therapy. 2006;1(3):387-394. DOI: 10.2174/157488806778226849
  43. 43. Yang W, Xia Y, Ji H, et al. Nuclear PKM2 regulates β-catenin transactivation upon EGFR activation [published correction appears in nature. 2017 Sep 20]. Nature. 2011;480(7375):118-122. DOI: 10.1038/nature10598
  44. 44. Agarwala S, Sanders TA, Ragsdale CW. Sonic hedgehog control of size and shape in midbrain pattern formation. Science. 2001;291(5511):2147-2150. DOI: 10.1126/science.1058624
  45. 45. Roelink H, Porter JA, Chiang C, et al. Floor plate and motor neuron induction by different concentrations of the amino-terminal cleavage product of sonic hedgehog autoproteolysis. Cell. 1995;81(3):445-455. DOI: 10.1016/0092-8674(95)90397-6
  46. 46. Ihrie RA, Shah JK, Harwell CC, et al. Persistent sonic hedgehog signaling in adult brain determines neural stem cell positional identity. Neuron. 2011;71(2):250-262. DOI: 10.1016/j.neuron.2011.05.018
  47. 47. Clement V, Sanchez P, de Tribolet N, Radovanovic I, Ruiz i Altaba A. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity [published correction appears in Curr Biol. 2007 Jan 23;17(2):192]. Current Biology. 2007;17(2):165-172. DOI: 10.1016/j.cub.2006.11.033
  48. 48. Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nature Reviews. Cancer. 2005;5(4):275-284. DOI: 10.1038/nrc1590
  49. 49. Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: Accumulating evidence and unresolved questions. Nature Reviews. Cancer. 2008;8(10):755-768. DOI: 10.1038/nrc2499
  50. 50. Alison MR, Lim SM, Nicholson LJ. Cancer stem cells: Problems for therapy? [published correction appears in J Pathol. 2012 Feb;226(3):e1]. The Journal of Pathology. 2011;223(2):147-161. DOI: 10.1002/path.2793
  51. 51. Hamerlik P, Lathia JD, Rasmussen R, et al. Autocrine VEGF-VEGFR2-Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. The Journal of Experimental Medicine. 2012;209(3):507-520. DOI: 10.1084/jem.20111424
  52. 52. Ropolo M, Daga A, Griffero F, et al. Comparative analysis of DNA repair in stem and nonstem glioma cell cultures. Molecular Cancer Research. 2009;7(3):383-392. DOI: 10.1158/1541-7786.MCR-08-0409
  53. 53. Liu G, Yuan X, Zeng Z, et al. Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Molecular Cancer. 2006;5:67. Published 2006 Dec 2. DOI: 10.1186/1476-4598-5-67
  54. 54. Bao S, Wu Q , McLendon RE, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756-760. DOI: 10.1038/nature05236
  55. 55. Pistollato F, Abbadi S, Rampazzo E, et al. Intratumoral hypoxic gradient drives stem cells distribution and MGMT expression in glioblastoma. Stem Cells. 2010;28(5):851-862. DOI: 10.1002/stem.415
  56. 56. Gong X, Schwartz PH, Linskey ME, Bota DA. Neural stem/progenitors and glioma stem-like cells have differential sensitivity to chemotherapy. Neurology. 2011;76(13):1126-1134. DOI: 10.1212/WNL.0b013e318212a89f
  57. 57. Beier D, Röhrl S, Pillai DR, et al. Temozolomide preferentially depletes cancer stem cells in glioblastoma. Cancer Research. 2008;68(14):5706-5715. DOI: 10.1158/0008-5472.CAN-07-6878
  58. 58. Blough MD, Westgate MR, Beauchamp D, et al. Sensitivity to temozolomide in brain tumor initiating cells. Neuro-Oncology. 2010;12(7):756-760. DOI: 10.1093/neuonc/noq032
  59. 59. Abdouh M, Facchino S, Chatoo W, Balasingam V, Ferreira J, Bernier G. BMI1 sustains human glioblastoma multiforme stem cell renewal. The Journal of Neuroscience. 2009;29(28):8884-8896. DOI: 10.1523/JNEUROSCI.0968-09.2009
  60. 60. Facchino S, Abdouh M, Chatoo W, Bernier G. BMI1 confers radioresistance to normal and cancerous neural stem cells through recruitment of the DNA damage response machinery. The Journal of Neuroscience. 2010;30(30):10096-10111. DOI: 10.1523/JNEUROSCI.1634-10.2010
  61. 61. Noh MG, Oh SJ, Ahn EJ, et al. Prognostic significance of E-cadherin and N-cadherin expression in gliomas. BMC Cancer. 2017;17(1):583. Published 2017 Aug 29. DOI: 10.1186/s12885-017-3591-z
  62. 62. Shibue T, Weinberg RA. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nature Reviews. Clinical Oncology. 2017;14(10):611-629. DOI: 10.1038/nrclinonc.2017.44
  63. 63. Loret N, Denys H, Tummers P, Berx G. The role of epithelial-to-mesenchymal plasticity in ovarian cancer progression and therapy resistance. Cancers (Basel). 2019;11(6):838. Published 2019 Jun 17. DOI: 10.3390/cancers11060838
  64. 64. Puisieux A, Brabletz T, Caramel J. Oncogenic roles of EMT-inducing transcription factors. Nature Cell Biology. 2014;16(6):488-494. DOI: 10.1038/ncb2976
  65. 65. Mani SA, Guo W, Liao MJ, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704-715. DOI: 10.1016/j.cell.2008.03.027
  66. 66. Siebzehnrubl FA, Silver DJ, Tugertimur B, et al. The ZEB1 pathway links glioblastoma initiation, invasion and chemoresistance. EMBO Molecular Medicine. 2013;5(8):1196-1212. DOI: 10.1002/emmm.201302827
  67. 67. Polyak K, Weinberg RA. Transitions between epithelial and mesenchymal states: Acquisition of malignant and stem cell traits. Nature Reviews. Cancer. 2009;9(4):265-273. DOI: 10.1038/nrc2620
  68. 68. Chen W, Dong J, Haiech J, Kilhoffer MC, Zeniou M. Cancer stem cell quiescence and plasticity as major challenges in cancer therapy. Stem Cells International. 2016;2016:1740936. DOI: 10.1155/2016/1740936
  69. 69. Chen J, Li Y, Yu TS, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488(7412):522-526. DOI: 10.1038/nature11287
  70. 70. Fanfone D, Idbaih A, Mammi J, Gabut M, Ichim G. Profiling anti-apoptotic BCL-xL protein expression in glioblastoma Tumorspheres. Cancers (Basel). 2020;12(10):2853. Published 2020 Oct 2. DOI: 10.3390/cancers12102853
  71. 71. Alajez NM, Shi W, Hui AB, et al. Targeted depletion of BMI1 sensitizes tumor cells to P53-mediated apoptosis in response to radiation therapy. Cell Death and Differentiation. 2009;16(11):1469-1479. DOI: 10.1038/cdd.2009.85
  72. 72. Begicevic RR, Falasca M. ABC transporters in cancer stem cells: Beyond chemoresistance. International Journal of Molecular Sciences. 2017;18(11):2362. Published 2017 Nov 8. DOI: 10.3390/ijms18112362
  73. 73. Chua C, Zaiden N, Chong KH, et al. Characterization of a side population of astrocytoma cells in response to temozolomide. Journal of Neurosurgery. 2008;109(5):856-866. DOI: 10.3171/JNS/2008/109/11/0856
  74. 74. Yamada K, Tso J, Ye F, et al. Essential gene pathways for glioblastoma stem cells: Clinical implications for prevention of tumor recurrence. Cancers (Basel). 2011;3(2):1975-1995. Published 2011 Apr 18. DOI: 10.3390/cancers3021975
  75. 75. Martín V, Sanchez-Sanchez AM, Herrera F, et al. Melatonin-induced methylation of the ABCG2/BCRP promoter as a novel mechanism to overcome multidrug resistance in brain tumour stem cells. British Journal of Cancer. 2013;108(10):2005-2012. DOI: 10.1038/bjc.2013.188
  76. 76. Nakai E, Park K, Yawata T, et al. Enhanced MDR1 expression and chemoresistance of cancer stem cells derived from glioblastoma. Cancer Investigation. 2009;27(9):901-908. DOI: 10.3109/07357900801946679
  77. 77. Tivnan A, Zakaria Z, O'Leary C, et al. Inhibition of multidrug resistance protein 1 (MRP1) improves chemotherapy drug response in primary and recurrent glioblastoma multiforme. Frontiers in Neuroscience. 2015;9:218. Published 2015 Jun 16. DOI: 10.3389/fnins.2015.00218
  78. 78. Flavahan WA, Wu Q , Hitomi M, et al. Brain tumor initiating cells adapt to restricted nutrition through preferential glucose uptake. Nature Neuroscience. 2013;16(10):1373-1382. DOI: 10.1038/nn.3510
  79. 79. Kang H, Lee S, Kim K, et al. Downregulated CLIP3 induces radioresistance by enhancing stemness and glycolytic flux in glioblastoma. Journal of Experimental & Clinical Cancer Research. 2021;40(1):282. Published 2021 Sep 6. DOI: 10.1186/s13046-021-02077-4
  80. 80. Wang X, Yang K, Xie Q , et al. Purine synthesis promotes maintenance of brain tumor initiating cells in glioma. Nature Neuroscience. 2017;20(5):661-673. DOI: 10.1038/nn.4537
  81. 81. Tardito S, Oudin A, Ahmed SU, et al. Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastoma. Nature Cell Biology. 2015;17(12):1556-1568. DOI: 10.1038/ncb3272
  82. 82. Ye F, Zhang Y, Liu Y, et al. Protective properties of radio-chemoresistant glioblastoma stem cell clones are associated with metabolic adaptation to reduced glucose dependence. PLoS One. 2013;8(11):e80397. Published 2013 Nov 18. DOI: 10.1371/journal.pone.0080397
  83. 83. Hoang-Minh LB, Siebzehnrubl FA, Yang C, et al. Infiltrative and drug-resistant slow-cycling cells support metabolic heterogeneity in glioblastoma. The EMBO Journal. 2018;37(23):e98772. DOI: 10.15252/embj.201798772
  84. 84. White E. Deconvoluting the context-dependent role for autophagy in cancer. Nature Reviews. Cancer. 2012;12(6):401-410. Published 2012 Apr 26. DOI: 10.1038/nrc3262
  85. 85. Yang K, Niu L, Bai Y, Le W. Glioblastoma: Targeting the autophagy in tumorigenesis. Brain Research Bulletin. 2019;153:334-340. DOI: 10.1016/j.brainresbull.2019.09.012
  86. 86. Ryskalin L, Gaglione A, Limanaqi F, et al. The autophagy status of cancer stem cells in Gliobastoma Multiforme: From cancer promotion to therapeutic strategies. International Journal of Molecular Sciences. 2019;20(15):3824. Published 2019 Aug 5. DOI: 10.3390/ijms20153824
  87. 87. Taylor MA, Das BC, Ray SK. Targeting autophagy for combating chemoresistance and radioresistance in glioblastoma. Apoptosis. 2018;23(11-12):563-575. DOI: 10.1007/s10495-018-1480-9
  88. 88. Golden EB, Cho HY, Jahanian A, et al. Chloroquine enhances temozolomide cytotoxicity in malignant gliomas by blocking autophagy. Neurosurgical Focus. 2014;37(6):E12. DOI: 10.3171/2014.9.FOCUS14504
  89. 89. Dolma S, Selvadurai HJ, Lan X, et al. Inhibition of dopamine receptor D4 impedes Autophagic flux, proliferation, and survival of glioblastoma stem cells. Cancer Cell. 2016;29(6):859-873. DOI: 10.1016/j.ccell.2016.05.002
  90. 90. Zhuang WZ, Long LM, Ji WJ, Liang ZQ. Rapamycin induces differentiation of glioma stem/progenitor cells by activating autophagy. Chinese Journal of Cancer. 2011;30(10):712-720. DOI: 10.5732/cjc.011.10234
  91. 91. Zhuang W, Li B, Long L, Chen L, Huang Q , Liang Z. Induction of autophagy promotes differentiation of glioma-initiating cells and their radiosensitivity. International Journal of Cancer. 2011;129(11):2720-2731. DOI: 10.1002/ijc.25975
  92. 92. Zhuang W, Long L, Zheng B, et al. Curcumin promotes differentiation of glioma-initiating cells by inducing autophagy. Cancer Science. 2012;103(4):684-690. DOI: 10.1111/j.1349-7006.2011.02198.x
  93. 93. Galavotti S, Bartesaghi S, Faccenda D, et al. The autophagy-associated factors DRAM1 and p62 regulate cell migration and invasion in glioblastoma stem cells. Oncogene. 2013;32(6):699-712. DOI: 10.1038/onc.2012.111
  94. 94. Tao Z, Li T, Ma H, et al. Autophagy suppresses self-renewal ability and tumorigenicity of glioma-initiating cells and promotes Notch1 degradation. Cell Death & Disease. 2018;9(11):1063. Published 2018 Oct 18. DOI: 10.1038/s41419-018-0957-3
  95. 95. Beier D, Schulz JB, Beier CP. Chemoresistance of glioblastoma cancer stem cells--much more complex than expected. Molecular Cancer. 2011;10:128. Published 2011 Oct 11. DOI: 10.1186/1476-4598-10-128
  96. 96. Monteiro AR, Hill R, Pilkington GJ, Madureira PA. The role of hypoxia in glioblastoma invasion. Cell. 2017;6(4):45. Published 2017 Nov 22. DOI: 10.3390/cells6040045
  97. 97. Colwell N, Larion M, Giles AJ, et al. Hypoxia in the glioblastoma microenvironment: Shaping the phenotype of cancer stem-like cells. Neuro-Oncology. 2017;19(7):887-896. DOI: 10.1093/neuonc/now258
  98. 98. Li Z, Bao S, Wu Q , et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell. 2009;15(6):501-513. DOI: 10.1016/j.ccr.2009.03.018
  99. 99. Hjelmeland AB, Wu Q , Heddleston JM, et al. Acidic stress promotes a glioma stem cell phenotype. Cell Death and Differentiation. 2011;18(5):829-840. DOI: 10.1038/cdd.2010.150
  100. 100. Qiang L, Wu T, Zhang HW, et al. HIF-1α is critical for hypoxia-mediated maintenance of glioblastoma stem cells by activating Notch signaling pathway. Cell Death and Differentiation. 2012;19(2):284-294. DOI: 10.1038/cdd.2011.95
  101. 101. Persano L, Pistollato F, Rampazzo E, et al. BMP2 sensitizes glioblastoma stem-like cells to Temozolomide by affecting HIF-1α stability and MGMT expression. Cell Death & Disease. 2012;3(10):e412. Published 2012 Oct 18. DOI: 10.1038/cddis.2012.153
  102. 102. Yin J, Ge X, Shi Z, et al. Extracellular vesicles derived from hypoxic glioma stem-like cells confer temozolomide resistance on glioblastoma by delivering miR-30b-3p. Theranostics. 2021;11(4):1763-1779. Published 2021 Jan 1. DOI: 10.7150/thno.47057
  103. 103. Cochrane CR, Szczepny A, Watkins DN, Cain JE. Hedgehog signaling in the maintenance of cancer stem cells. Cancers (Basel). 2015;7(3):1554-1585. Published 2015 Aug 11. DOI: 10.3390/cancers7030851
  104. 104. Ulasov IV, Nandi S, Dey M, Sonabend AM, Lesniak MS. Inhibition of sonic hedgehog and Notch pathways enhances sensitivity of CD133(+) glioma stem cells to temozolomide therapy. Molecular Medicine. 2011;17(1-2, 112):103. DOI: 10.2119/molmed.2010.00062
  105. 105. Wellner U, Schubert J, Burk UC, et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nature Cell Biology. 2009;11(12):1487-1495. DOI: 10.1038/ncb1998
  106. 106. Alves ALV, Gomes INF, Carloni AC, et al. Role of glioblastoma stem cells in cancer therapeutic resistance: A perspective on antineoplastic agents from natural sources and chemical derivatives. Stem Cell Research & Therapy. 2021;12(1):206. Published 2021 Mar 24. DOI: 10.1186/s13287-021-02231-x
  107. 107. Piper K, DePledge L, Karsy M, Cobbs C. Glioma stem cells as immunotherapeutic targets: Advancements and challenges. Front. Oncologia. 2021;11:615704. Published 2021 Feb 24. DOI: 10.3389/fonc.2021.615704
  108. 108. Wang CH, Chiou SH, Chou CP, Chen YC, Huang YJ, Peng CA. Photothermolysis of glioblastoma stem-like cells targeted by carbon nanotubes conjugated with CD133 monoclonal antibody. Nanomedicine. 2011;7(1):69-79. DOI: 10.1016/j.nano.2010.06.010
  109. 109. Aldape KD, Ballman K, Furth A, et al. Immunohistochemical detection of EGFRvIII in high malignancy grade astrocytomas and evaluation of prognostic significance. Journal of Neuropathology and Experimental Neurology. 2004;63(7):700-707. DOI: 10.1093/jnen/63.7.700
  110. 110. Pang LY, Saunders L, Argyle DJ. Epidermal growth factor receptor activity is elevated in glioma cancer stem cells and is required to maintain chemotherapy and radiation resistance. Oncotarget. 2017;8(42):72494-72512. Published 2017 Aug 3. DOI: 10.18632/oncotarget.19868
  111. 111. Emlet DR, Gupta P, Holgado-Madruga M, et al. Targeting a glioblastoma cancer stem-cell population defined by EGF receptor variant III. Cancer Research. 2014;74(4):1238-1249. DOI: 10.1158/0008-5472.CAN-13-1407
  112. 112. Karachaliou N, Fernandez-Bruno M, Bracht JWP, Rosell R. EGFR first- and second-generation TKIs-there is still place for them in EGFR-mutant NSCLC patients. Translational Cancer Research. 2019;8(Suppl. 1):S23-S47. DOI: 10.21037/tcr.2018.10.06
  113. 113. Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nature Reviews. Cancer. 2007;7(3):169-181. DOI: 10.1038/nrc2088
  114. 114. Liu X, Chen X, Shi L, et al. The third-generation EGFR inhibitor AZD9291 overcomes primary resistance by continuously blocking ERK signaling in glioblastoma. Journal of Experimental & Clinical Cancer Research. 2019;38(1):219. Published 2019 May 23. DOI: 10.1186/s13046-019-1235-7
  115. 115. Colclough N, Chen K, Johnström P, et al. Preclinical comparison of the blood-brain barrier permeability of Osimertinib with other EGFR TKIs. Clinical Cancer Research. 2021;27(1):189-201. DOI: 10.1158/1078-0432.CCR-19-1871
  116. 116. Peled N, Kian W, Inbar E, et al. Osimertinib in advanced EGFR-mutant lung adenocarcinoma with asymptomatic brain metastases: An open-label, 3-arm, phase II pilot study. Neurooncoly Advances. 2021;4(1):vdab188. Published 2021 Dec 27. DOI: 10.1093/noajnl/vdab188
  117. 117. Cardona AF, Jaramillo-Velásquez D, Ruiz-Patiño A, et al. Efficacy of osimertinib plus bevacizumab in glioblastoma patients with simultaneous EGFR amplification and EGFRvIII mutation. Journal of Neuro-Oncology. 2021;154(3):353-364. DOI: 10.1007/s11060-021-03834-3
  118. 118. Makhlin I, Salinas RD, Zhang D, et al. Clinical activity of the EGFR tyrosine kinase inhibitor osimertinib in EGFR-mutant glioblastoma. CNS. Oncologia. 2019;8(3):CNS43. DOI: 10.2217/cns-2019-0014
  119. 119. Fan X, Khaki L, Zhu TS, et al. NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts. Stem Cells. 2010;28(1):5-16. DOI: 10.1002/stem.254
  120. 120. Saito N, Fu J, Zheng S, et al. A high Notch pathway activation predicts response to γ secretase inhibitors in proneural subtype of glioma tumor-initiating cells. Stem Cells. 2014;32(1):301-312. DOI: 10.1002/stem.1528
  121. 121. Saito N, Aoki K, Hirai N, et al. Effect of Notch expression in glioma stem cells on therapeutic response to chemo-radiotherapy in recurrent glioblastoma. Brain Tumor Pathology. 2015;32(3):176-183. DOI: 10.1007/s10014-015-0215-7
  122. 122. Pan E, Supko JG, Kaley TJ, et al. Phase I study of RO4929097 with bevacizumab in patients with recurrent malignant glioma. Journal of Neuro-Oncology. 2016;130(3):571-579. DOI: 10.1007/s11060-016-2263-1
  123. 123. Gilbert CA, Daou MC, Moser RP, Ross AH. Gamma-secretase inhibitors enhance temozolomide treatment of human gliomas by inhibiting neurosphere repopulation and xenograft recurrence. Cancer Research. 2010;70(17):6870-6879. DOI: 10.1158/0008-5472.CAN-10-1378
  124. 124. Liebelt BD, Shingu T, Zhou X, Ren J, Shin SA, Hu J. Glioma stem cells: Signaling, microenvironment, and therapy. Stem Cells International. 2016;2016:7849890. DOI: 10.1155/2016/7849890
  125. 125. Torrisi F, Alberghina C, Lo Furno D, et al. Connexin 43 and sonic hedgehog pathway interplay in glioblastoma cell proliferation and migration. Biology (Basel). 2021;10(8):767. Published 2021 Aug 12. DOI: 10.3390/biology10080767
  126. 126. Fu J, Rodova M, Nanta R, et al. NPV-LDE-225 (Erismodegib) inhibits epithelial mesenchymal transition and self-renewal of glioblastoma initiating cells by regulating miR-21, miR-128, and miR-200. Neuro-Oncology. 2013;15(6):691-706. DOI: 10.1093/neuonc/not011
  127. 127. Nitta RT, Gholamin S, Feroze AH, et al. Casein kinase 2α regulates glioblastoma brain tumor-initiating cell growth through the β-catenin pathway. Oncogene. 2015;34(28):3688-3699. DOI: 10.1038/onc.2014.299
  128. 128. Nitta RT, Bolin S, Luo E, et al. Casein kinase 2 inhibition sensitizes medulloblastoma to temozolomide [published correction appears in oncogene. 2019 Oct 28;]. Oncogene. 2019;38(42):6867-6879. DOI: 10.1038/s41388-019-0927-y
  129. 129. Sai K, Wang S, Balasubramaniyan V, et al. Induction of cell-cycle arrest and apoptosis in glioblastoma stem-like cells by WP1193, a novel small molecule inhibitor of the JAK2/STAT3 pathway. Journal of Neuro-Oncology. 2012;107(3):487-501. DOI: 10.1007/s11060-011-0786-z
  130. 130. Kim BH, Lee H, Park CG, et al. STAT3 inhibitor ODZ10117 suppresses glioblastoma malignancy and prolongs survival in a glioblastoma xenograft model. Cell. 2020;9(3):722. Published 2020 Mar 15. DOI: 10.3390/cells9030722
  131. 131. Yang YP, Chang YL, Huang PI, et al. Resveratrol suppresses tumorigenicity and enhances radiosensitivity in primary glioblastoma tumor initiating cells by inhibiting the STAT3 axis. Journal of Cellular Physiology. 2012;227(3):976-993. DOI: 10.1002/jcp.22806
  132. 132. Cilibrasi C, Riva G, Romano G, et al. Resveratrol impairs glioma stem cells proliferation and motility by modulating the Wnt signaling pathway. PLoS One. 2017;12(1):e0169854. Published 2017 Jan 12. DOI: 10.1371/journal.pone.0169854
  133. 133. Groot J, Ott M, Wei J, et al. A first-in-human phase I trial of the oral p-STAT3 inhibitor WP1066 in patients with recurrent malignant glioma. CNS. Oncologia. 2022;11(2):CNS87. DOI: 10.2217/cns-2022-0005
  134. 134. Ashizawa T, Miyata H, Iizuka A, et al. Effect of the STAT3 inhibitor STX-0119 on the proliferation of cancer stem-like cells derived from recurrent glioblastoma. International Journal of Oncology. 2013;43(1):219-227. DOI: 10.3892/ijo.2013.1916
  135. 135. Aderetti DA, Hira VVV, Molenaar RJ, van Noorden CJF. The hypoxic peri-arteriolar glioma stem cell niche, an integrated concept of five types of niches in human glioblastoma. Biochimica Et Biophysica Acta. Reviews on Cancer. 2018;1869(2):346-354. DOI: 10.1016/j.bbcan.2018.04.008
  136. 136. Reardon DA, Turner S, Peters KB, et al. A review of VEGF/VEGFR-targeted therapeutics for recurrent glioblastoma. Journal of the National Comprehensive Cancer Network. 2011;9(4):414-427. DOI: 10.6004/jnccn.2011.0038
  137. 137. Ameratunga M, Pavlakis N, Wheeler H, Grant R, Simes J, Khasraw M. Anti-angiogenic therapy for high-grade glioma. Cochrane Database of Systematic Reviews. 2018;11(11):CD008218. Published 2018 Nov 22. DOI: 10.1002/14651858.CD008218.pub4
  138. 138. Alomari S, Zhang I, Hernandez A, et al. Drug repurposing for glioblastoma and current advances in drug delivery-a comprehensive review of the literature. Biomolecules. 2021;11(12):1870. Published 2021 Dec 13. DOI: 10.3390/biom11121870
  139. 139. Maraka S, Groves MD, Mammoser AG, et al. Phase 1 lead-in to a phase 2 factorial study of temozolomide plus memantine, mefloquine, and metformin as postradiation adjuvant therapy for newly diagnosed glioblastoma [published correction appears in Cancer. 2019 Apr 15;125(8):1387]. Cancer. 2019;125(3):424-433. DOI: 10.1002/cncr.31811
  140. 140. Sato A, Sunayama J, Okada M, et al. Glioma-initiating cell elimination by metformin activation of FOXO3 via AMPK. Stem Cells Translational Medicine. 2012;1(11):811-824. DOI: 10.5966/sctm.2012-0058
  141. 141. Würth R, Pattarozzi A, Gatti M, et al. Metformin selectively affects human glioblastoma tumor-initiating cell viability: A role for metformin-induced inhibition of Akt. Cell Cycle. 2013;12(1):145-156. DOI: 10.4161/cc.23050
  142. 142. Kim W, Lee S, Seo D, et al. Cellular stress responses in radiotherapy. Cell. 2019;8(9):1105. Published 2019 Sep 18. DOI: 10.3390/cells8091105
  143. 143. Feng F, Zhang M, Yang C, Heng X, Wu X. The dual roles of autophagy in gliomagenesis and clinical therapy strategies based on autophagic regulation mechanisms. Biomedicine & Pharmacotherapy. 2019;120:109441. DOI: 10.1016/j.biopha.2019.109441
  144. 144. Jia B, Xue Y, Yan X, et al. Autophagy inhibitor chloroquine induces apoptosis of cholangiocarcinoma cells via endoplasmic reticulum stress. Oncology Letters. 2018;16(3):3509-3516. DOI: 10.3892/ol.2018.9131
  145. 145. Verbaanderd C, Maes H, Schaaf MB, et al. Repurposing drugs in oncology (ReDO)-chloroquine and hydroxychloroquine as anti-cancer agents. Ecancermedicalscience. 2017;11:781. Published 2017 Nov 23. DOI: 10.3332/ecancer.2017.781
  146. 146. Firat E, Weyerbrock A, Gaedicke S, Grosu AL, Niedermann G. Chloroquine or chloroquine-PI3K/Akt pathway inhibitor combinations strongly promote γ-irradiation-induced cell death in primary stem-like glioma cells. PLoS One. 2012;7(10):e47357. DOI: 10.1371/journal.pone.0047357
  147. 147. Lomonaco SL, Finniss S, Xiang C, et al. The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells. International Journal of Cancer. 2009;125(3):717-722. DOI: 10.1002/ijc.24402
  148. 148. Huang T, Kim CK, Alvarez AA, et al. MST4 phosphorylation of ATG4B regulates Autophagic activity, Tumorigenicity, and Radioresistance in glioblastoma. Cancer Cell. 2017;32(6):840-855.e8. DOI: 10.1016/j.ccell.2017.11.005

Written By

Meenakshi Tiwari, Lokendra Kumar Sharma and Ajit Kumar Saxena

Submitted: 30 June 2022 Reviewed: 06 July 2022 Published: 02 August 2022