Open Access

Regulation of cancer stem cells and immunotherapy of glioblastoma (Review)

  • Authors:
    • Аleksandra Kosianova
    • Oleg Pak
    • Igor Bryukhovetskiy
  • View Affiliations

  • Published online on: December 19, 2023     https://doi.org/10.3892/br.2023.1712
  • Article Number: 24
  • Copyright: © Kosianova 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

Glioblastoma (GB) is one of the most adverse diagnoses in oncology. Complex current treatment results in a median survival of 15 months. Resistance to treatment is associated with the presence of cancer stem cells (CSCs). The present review aimed to analyze the mechanisms of CSC plasticity, showing the particular role of β‑catenin in regulating vital functions of CSCs, and to describe the molecular mechanisms of Wnt‑independent increase of β‑catenin levels, which is influenced by the local microenvironment of CSCs. The present review also analyzed the reasons for the low effectiveness of using medication in the regulation of CSCs, and proposed the development of immunotherapy scenarios with tumor cell vaccines, containing heterogenous cancer cells able of producing a multidirectional antineoplastic immune response. Additionally, the possibility of managing lymphopenia by transplanting hematopoietic stem cells from a healthy sibling and using clofazimine or other repurposed drugs that reduce β‑catenin concentration in CSCs was discussed in the present study.

1. Introduction

Glioblastoma (GB) is a primary glial tumor of astrocytic type, grade IV of malignancy according to the World Health Organization (WHO) classification (1). Compared with other cancer types, the incidence of GB is relatively low, varying from 3 to 5 cases per 100,000 individuals in North America, Europe and Central Asia (2). The large majority of GB cases occur sporadically, and the tumor is found in all age groups, but is more frequently detected during the second half of life in men of the so-called ‘non-Hispanic type’ (3).

Current treatment protocols for GB include neurosurgery, high-dose radiation and chemotherapy (CT), most frequently using such cytostatics as temozolomide (TMZ) and lomustine, as well as the targeted antitumor agent, bevacizumab (4). The effect of treatment is insufficient, with a median relapse-free survival of 4-8 months, and an overall survival of 15 months (5). A total of ~25% of patients are able to survive for 24 months following diagnosis, which makes GB one of the most dismal diseases in oncology.

Resistance to treatment is associated with the heterogeneous nature of the cancer stem cell (CSC) population that dominates in the GB cell hierarchy (6). High β-catenin content is the most important characteristic of functionally active CSCs (7), due to their interaction with the local immunosuppressive microenvironment, thus activating the intracellular Wnt signaling pathway (8), PI3K/AKT/mTOR (9) and other mechanisms (10) that are responsible for plasticity and tumor progression.

Cytostatic drugs fail to effectively destroy CSCs in the body of a patient (11), and attempts to regulate CSCs by combining TMZ, immune checkpoint inhibitors (12) and molecular-targeted drugs (13) have not been unequivocally successful; thus, there is an urgent need to supplement the existing GB treatment protocols with more effective methods of regulating the functional activity of this cell type.

It could be hypothesized that a combination of drugs that inhibit the synthesis and biological activity of β-catenin with immunotherapy may destabilize the interaction between CSCs and the tumor microenvironment; block the mechanisms of CSC phenotypic heterogeneity and plasticity; enhance the antiglioma effect of cytostatics; hinder tumor progression; and prolong the life of a patient.

The present study aimed to investigate the possibility of regulating GB CSCs by combining antagonists of the Wnt/β-catenin signaling pathway with immunotherapy.

2. Principles of GB treatment

Eliminating as many cancer cells (CCs) as possible is the key principle of GB treatment. Surgery is the method of choice for treating patients with GB (14), since it provides one-stage elimination of numerous CCs, reduces intracranial pressure, relieves brain compression, and improves the overall condition of the patient (15). However, the applicability of surgical treatment is limited. The severity of the condition of the patient, a functional status <70 on the Karnofsky scale, or subtentorial, thalamic, multifocal or bihemispheric localization of the tumor often render it impossible to treat the condition surgically without a direct risk to the life of the patient, and make a strong case for the refusal of surgery in patients with newly diagnosed GB (16,17). Moreover, brain tissue infiltration by CCs, even with the use of the most modern surgical technologies, does not allow the radical removal of the tumor without causing severe and irreparable neurological damage to the patient (18,19).

Cytoreduction effectiveness increases significantly when surgery is combined with radiation (20). Radiation therapy damages DNA, induces ageing processes and destroys GB cells (21). Intraoperative irradiation (22) can be used, as well as remote γ-therapy (23), which can be supplemented with brachytherapy (24), proton therapy (25) and neutron-capture therapy (26). The life expectancy of patients with GB is associated with the dose of radiation received, which may reach ≥60 Gy (27), while dose escalation can result in radiation-induced brain necrosis, impaired cerebral circulation, reactive gliosis, sclerosis, cyst formation and development of psychoorganic syndrome (28).

The majority of cases exhibit tumor relapse in 4-8 months after its removal, with ≤30% of patients undergoing reoperation (29), which improves their overall condition and quality of life, reduces their dependence on corticosteroids, and improves the effects of adjuvant CT.

Reoperation requires special skill, as fluid-filled cystic cavities that appear after radiotherapy can be misleading in terms of correctly identifying the necessary extent of resection; thus, careful planning of the surgery is necessary regarding clear anatomical landmarks (such as sulci, ventricles and dura mater boundaries), which is not always possible (30). It is partly for this reason that <10% of patients who are reoperated due to GB progression or recurrence undergo a third surgery, and only 2% of patients undergo ≥3 reoperations (17).

CT is recommended for all patients with GB. CT is the main method of destroying CCs, and it is used in combination with radiotherapy for treating a newly diagnosed GB to prolong remission after standard chemoradiation therapy and to prolong the life of patients with tumor recurrence (6). The drug of choice in CT is a cytostatic alkylating antineoplastic agent called TMZ, which is a tetrazine derivative.

Based on the potent cytotoxic activity of TMZ against glioma and carcinoma cells, the current stage of neuro-oncology development is termed the ‘temozolomide era’ (31). TMZ passes through the blood-brain barrier (BBB), easily penetrates into the cerebrospinal fluid, and, once in the bloodstream, undergoes a chemical transformation into monomethyltriazenoimidazolcarboxamide, the effect of which is attributed to the alkylation of guanine at O6 and N7 positions, with the subsequent triggering of aberrant reduction of methyl residues.

Patients are recommended a TMZ dosage of 75 mg/m2 in combination with radiotherapy, followed by 6-8 cycles of TMZ with an increased dose of ≤150-200 and ≤400 mg/m2. The maximum tolerated dose per treatment cycle is 1,000 mg/m2. TMZ is usually employed in combination with lomustine (32) and bevacizumab, an inhibitor of endothelial growth factor in blood vessels.

Lomustine is an alkylating antineoplastic agent that is a nitrosourea derivative and a second-line CT drug. In addition to DNA alkylation, its antineoplastic effect is achieved via inhibition of genome repair enzymes, DNA matrix damage and suppression of some key enzymatic processes in CCs in the late G1 and early S phases of the cell cycle (32). The recommended dose of lomustine in adults is 130 mg/m2 when administered once orally every 6 weeks, and the total dose for all treatment regimens should not exceed 1,000 mg/m2. Similar to TMZ, further doses of lomustine should be determined based on the therapy effectiveness and hematological response of the patient to the previous dose.

Previous attempts to enhance the antineoplastic treatment effect by combining TMZ or lomustine with the poly(ADP-ribose) polymerase inhibitor, olaparib (33), the histone deacetylase inhibitor, vorinostat (34) and other blockers of DNA repair enzymes (35) have not yet been successful. Procarbazine, vincristine, paclitaxel and platinum-based agents and other cytostatics have no significant advantages over TMZ and lomustine, and their administration has a detrimental effect on hematological status, increases toxicity and does not prolong the life of patients with GB (36). Electromagnetic therapy, which has been widely used in the last decade, expands the possibilities of cytostatics (28), but does not have any strategic advantages.

Thus, cytoreduction, cytotoxic and cytostatic principles define the current paradigm of GB treatment, which is able to ensure an average survival of patients of 15 months, with an average cost of treatment of 62,602$ (37). The main reason for such a cost is CT, the effectiveness of which is rather low despite the large number of antineoplastic agents available. The effect of CT is limited by the BBB permeability (28), but attempts to administer drugs into the removed GB bed, intrathecal/intraventricular administration of CT, or encasing cytostatics into nanocapsules for their targeted delivery to the brain by using monoclonal antibodies have not yet been successful (11), which is usually explained by the phenotypic plasticity (6) and particular properties of CSCs (38).

3. CC plasticity and resistance to treatment

Traditionally, the plasticity of CCs has been defined as their ability to transition into an undifferentiated state and resist the effects of CT (39). A decisive step in understanding GB plasticity was made by Verhaak et al (40), who described proneural, neural, classical and mesenchymal tumor subtypes, showing the possibility of their transformation during treatment. It is noteworthy that the proneural subtype, identified in this study, was the most proliferative, while the mesenchymal subtype of the tumor was poorly proliferative and highly resistant to CT. Subsequently, high genome heterogeneity in the structure of these tumor subtypes (41) was revealed, while the critically low degree of DNA methylation, which was inherent to the mesenchymal GB subtype, predetermined the main trend of the problem development.

Brennan et al (42) further described six classes of DNA methylation in GB cells, with the highest methylation status assigned to isocitrate dehydrogenase (IDH) mutant tumors as the most differentiated and least plastic ones (42). The IDH mutation resulted in an excess of 2-hydroxyglutarate in CCs, which was accompanied by hypermethylation of the promoter regions of the O6-methylguanine-DNA methyltransferase (MGMT) gene, which provided direct DNA repair (43). Reducing the degree of genome methylation made CCs more plastic (44). In light of this, in 2016, the WHO selected IDH mutations as the main criterion for systematization of gliomas, distinguishing IDH-wild-type and mutant types of GB (45).

Theoretically, IDH-wild-type GB cells have great replicative freedom and can use a wide arsenal of mechanisms for repairing single- and double-stranded DNA breaks, including homologous recombination and nonhomologous end joining (43-45). IDH-wild-type GB was divided into seven additional classes on the basis of genome methylation (46), which allowed Neftel et al (47) to describe four types of phenotypic states inherent in CCs: i) Neural progenitor-like cells with amplification of the CDK4 gene; ii) oligodendroglial progenitor-like cells with amplification of the PDGFRA gene; iii) astrocyte-like cells with amplification of the EGFR gene; and iv) mesenchymal-like cells with mutation of the NF1 gene.

Such taxonomy makes the phenotypic plasticity of CCs directly dependent on their local microenvironment (48-50), which not only predetermines the C phenotype, but also appears to be able to switch reproduction programs on and off depending on external conditions (51-53). In light of this, it can be presumed that the majority of phenotypic plasticity is exhibited by CCs with a hypomethylated genome, which are almost insusceptible to CT.

It is commonly considered that cells of this type, which are called CSCs, were described in 1997 by Bonnet and Dick (54) in their study on the hierarchical structure of the cell population in acute myeloid leukemia. However, as early as in 1877, Julius Friedrich Conheim, a student of Rudolf Virchow, indicated the presence of neoplastic elements with embryonic characteristics among the cells of gastric, breast and other aggressive tumors (55).

It is highly probable that CSCs are transformed descendants of normal neural stem cells (NSCs) that inhabit the subventricular zone and other germinative centers of the human brain. This is indicated by: i) The identity of >60% of proteins in the NSC proteome and CSCs of GB (56); ii) complex attractive-permissive interactions between cells of these types (57); iii) the CSC microenvironment, including clones of astrocytic and oligodendroglial progenitor cells that are transcriptomically similar to NSCs of the brain of a patient (58); and iv) the presence of NSC-like elements carrying the same mutations as differentiated CCs at all stages of gliomagenesis (59).

Normal NSCs and CSCs have a number of similar immunocytochemical markers on the cell surface, among which the CD133 antigen (prominin-1) is considered to be the main marker of GB stem cells (13). However, in addition to NSCs, this glycoprotein is present in hematopoietic stem cells (HSCs), endothelial progenitor cells, as well as in kidneys, trachea, salivary and mammary glands, placenta, digestive tract, testes, and other normal cells and tissues (60).

The role of CD133 antigen in the neoplastic process is not fully understood, but its direct association with cancer is clear. This marker is present in CSCs of lung cancer (61), colorectal carcinoma (62) and breast cancer (63). CSCs of the CD133+ immunophenotype rank the highest in the hierarchy of GB cells (64) and are characterized by their tumorigenicity and high proliferative activity. However, the presence of differentiated non-tumorigenic CD133+ cells, progenitor-like CD133+ cells with limited proliferative potential, and CSCs that are tumorigenic and negative to this marker (65) but immunopositive to CD56, SRY-box transcription factor (SOX)2, SOX9, CD15, A2B5 and other antigens (64), allows to assert that the CSC phenomenon is not directly associated with cells of one certain immunophenotype.

This interpretation explains the failure of previous attempts to increase the effect of treating invasive gliomas by combining TMZ or lomustine with monoclonal antibodies against different CSC antigens (64,65). Probably, at the initial stage of a neoplastic process, mutations forming the primary stem lineage occur specifically in NSCs, which have the highest proliferation rate among all the cells of the nervous system. The proliferation of mutant cells leads to an increase in cell mass and competition for oxygen, thus triggering mechanisms that produce new generations of NSCs capable of arbitrarily switching between anaerobic and aerobic types of metabolism by regulating the level of lipid and glutamine utilization, which proliferate or remain in a quiescent state (66).

The functional activity of such cells is determined by the local microenvironment, which activates a number of molecular mechanisms, leading to the proliferation of CCs that have adapted to certain local microconditions. Perivascular localization is a characteristic of proneural CSCs, which are mesenchymal CSCs that are extracted from hypoxic zones and areas of necrosis (67), where local microconditions are unfavorable. The proneural type of GB is characterized by proliferating CSCs with a glycolytic type of metabolism and a low level of lipid utilization, whereas the mesenchymal type of GB is characterized by CSCs that are predominantly in the state of proliferative quiescence and are able to switch between glycolysis and aerobic respiration as well as exhibit a high level of lipid metabolism.

In fact, proneural and mesenchymal tumor subtypes reflect two possible states of CSCs, namely proliferation and survival. The transition to the survival state occurs under the influence of hypoxia (68), cytostatics (69), irradiation (70,71) and anti-angiogenic therapy (72). It can be assumed that the reverse transition, triggering tumor relapse, is also precipitated by the influence of the local microenvironment, which activates numerous molecular mechanisms in CSCs (73), with the canonical Wnt signaling pathway playing a particular role among those mechanisms.

4. Wnt signaling pathway and the microenvironment of CSCs

The canonical Wnt signaling pathway is the most important intracellular signaling pathway, regulating embryogenesis and differentiation of normal stem cells (74). In GB pathogenesis, this pathway controls the balance between symmetric and asymmetric CSC divisions, which predetermines the aggressiveness and fatality of the tumor (75). The central link of this mechanism is β-catenin, which even in the absence of signaling is bound by a multiprotein ‘destructive complex’ represented by the adenomatous polyposis coli (APC), AXIN1/2, CK1 and glycogen synthase kinase (GSK)-3β proteins. GSK-3β protein kinase, which is activated by the ‘destructive complex’, phosphorylates β-catenin, which loses its functionality and undergoes degradation in the proteasome (74).

The Wnt signaling pathway is activated by one of 19 secreted Wnt-proteins interacting with Frizzled and low-density lipoprotein receptor-related protein (LPR)4/5 family receptors of the CSC surface, which activates the intracellular Disheveled protein and triggers a cascade of intermolecular interactions, leading to the blockade of the ‘destructive complex’ and accumulation of β-catenin in the CSC cytoplasm, which enters the nucleus, activates T-cell factor (TCF), and triggers the expression of pluripotency genes (76-78), inluding MYC, CCND1, cellular communication network factor 4 (CCN4 also known as WISP1) and PPARG.

Mutations of the proline, glutamate and leucine rich protein 1 (PELP-1) gene, that is a potential co-activator of the canonical Wnt signaling pathway, are found in the majority of patients with GB, while mutations of the APC gene are identified in 14.5% of cases, and mutations of the FAT1 gene, a negative regulator of Wnt signaling, are detected in 57% of patients (77-79). Mutations of other components are rare (80). A high content of β-catenin is one of the main characteristics of CD133+ CSCs (81,82), which indicates the involvement of other mechanisms that cause its accumulation in this type of cells.

One of the mechanisms previously described (83) is the activation of the PI3K/AKT/mTOR signaling pathway, which enables both the stimulation of PI3K (13) and the immediate activation of intracellular AKT, which directly phosphorylates GSK-3β and increases β-catenin content (12). In turn, mTOR creates two multiprotein complexes, namely mTORC1 and mTORC2, which antagonistically regulate each other's activity: The first one decreases the β-catenin content in CSCs (84), while the second one increases it (85).

The increase in β-catenin content activates telomerases, thus leading to telomere lengthening, immortalization, stabilization of the CSC genome, and survival of CSCs under chemoradiotherapy (CRT) (86-88). The β-catenin content in CSCs increases due to semaphorins. a particular class of secreted and membrane proteins produced by neuroblasts and neurons (89). The synthesis of β-catenin in CSCs is induced by TGF-β secreted by CCs and immunosuppressive microglia cells (90). Through the SMAD and death-associated protein 6 (DAXX) signaling pathways, this cytokine activates the PI3K/AKT/mTOR axis (91), leading to β-catenin accumulation in CSCs, which indicates the strategic role of the microenvironment in CSC regulation.

Thus, β-catenin is one of the most significant regulators of CSC proliferation, with the local microenvironment being the main regulator of the intracellular β-catenin content in CSCs. The microenvironment regulates CSCs by triggering two divergent processes, namely autophagy and epithelial-mesenchymal transition (EMT) (73). The inflammatory microenvironment triggers micro- and macro-autophagy, which degrades intracellular proteins (92), thus providing energy-independent utilization of β-catenin, critically reducing CSC activity, and destabilizing the interaction with the extracellular matrix, which allows CSCs to migrate, and survive hypoxia, metabolic acidosis and CRT.

On the contrary, an immunosuppressive microenvironment (93) produces Wnt3a and Wnt5 ligands that activate the Wnt signaling pathway, with WISP1 as one of the target genes (94), the protein product of which increases the synthesis of other Wnt ligands, as well as that of IL-10 and IL-35, thus suppressing autophagy, and leading to an increase in β-catenin content and enhanced interaction with the extracellular matrix. In turn, the activation of AKT, which is mediated by α6β1 and other components of the integrin signaling pathway (95), activates the production of Wnt ligands, immunosuppressive cytokines, programmed cell death (PD)-1 and cytotoxic T-lymphocyte-associated protein 4(94), which creates an ‘autocrine loop’, leading to increased immunosuppression, niche development and proliferation of CSCs (96).

Thus, the plasticity of CCs is caused by the influence of an immunosuppressive microenvironment, contributing to the increase of β-catenin content, which provides the transition from survival to proliferation in CSCs. Numerous attempts have been made to regulate these processes using targeted therapy.

5. Targeted therapy and CSC plasticity

Differentially activated signaling pathways differ for proneural and mesenchymal types of CSCs (97). In the former, the main role belongs to the signaling pathways of tyrosine kinase (TRK) β-receptor of platelet-derived growth factor (PDGF) and Notch; in the latter, the TGF-β, NF-κB and glycolysis signaling pathways are dominant. Pharmacologists have paid close attention to these molecular mechanisms; however, important breakthroughs have yet to emerge.

PDGF has become one of the priority targets for the inhibition of CSC proliferation. However, imatinib, an inhibitor of the TRK activity of PDGF from a new class of targeted cytostatics, failed trials in patients with GB (98). The multikinase inhibitor, sorafenib, was marginally effective when delivered directly to the tumor and only together with alternating electromagnetic field therapy (99), while sunitinib showed no effect at all in patients with GB (100).

Proliferation suppression of CSCs via the TRK-signaling domain of epidermal growth factor receptor (EGFR) in patients with GB has been attempted and failed. The peptide vaccine, rindopepimut, against the EGFRvIII antigen failed to meet expectations in combination with TMZ and bevacizumab (101). Depatuxizumab, a monoclonal antibody against EGFR, loaded with the monomethyl auristatin F and displaying antimitotic effects, has demonstrated modest results both in combination with TMZ (102) and without it (103).

Onartuzumab, a monoclonal antibody directed against TRK-receptor of hepatocyte growth factor (HGF), was not successful in treating patients with GB in combination with TMZ and bevacizumab (104). The multikinase inhibitor cabozantinib, by blocking HGF signaling among other mechanisms, was ineffective in treating patients with GB (105). Only bevacizumab, by inhibiting TRK-receptor of vascular endothelial growth factor (VEGF) (106), in combination with TMZ, increased the life expectancy of patients with GB by 4-6 months, which is a remarkable achievement for targeted therapy.

Poor efficiency of TRK receptor inhibitors has spawned numerous unsuccessful attempts to target downstream components of the PI3K/AKT/mTOR signaling pathway. Buparlisib, a PI3K-kinase inhibitor, proved to be no more advantageous than monotherapy with TMZ (107), carboplatin and lomustine (108). The inhibitors of AKT kinase (perifosine) (109) and mTOR kinase (temsirolimus) failed to meet expectations both in combination with TMZ and without it (110).

The BRAF-V600E mutation is found in 20% of tumors, and it involves the substitution of valine-600 for glutamic acid, permanently activating the RAS/RAF/MEK/ERK signaling pathway. However, the inhibitors of the mutant RAF protein kinase, sorafenib (99) and vemurafenib (111), have not exhibited any effect in patients with GB.

The Notch signaling pathway is the oldest reported mechanism regulating the processes of stem cell differentiation (112). Activation of this pathway occurs when one of four Delta-like ligands or Jag1-2 ligands, binds to the membrane of one cell, directly interacts with one of four Notch receptors on the membrane of the cell, receiving the signal that triggers a cascade of intermolecular interactions, thus leading to the activation of target genes. Suppression of these mechanisms can block the proliferative functions of CSCs, but none of the Notch-signaling inhibitors has shown any effect in GB (113).

Attempts to affect CSCs via pharmacological suppression of the EMT-related mechanisms were also ineffective. The inhibitors of TGF-β, trabedersen (114) and galunisertib (115), did not produce a significant increase in survival rates when combined with TMZ, procarbazine, lomustine and vincristine.

Theories on a possible pharmacological regulation of CSCs via the PI3K/AKT/NF-κB axis are inconsistent (116). There have been some reports on the apoptosis of GB cells under the influence of this target (117) and the ability of cannabidiol to inhibit NF-κB activity (118), but this method has not been used for the regulation of CSCs in the complex treatment of GB.

Therefore, attempts of regulating CSC with the help of targeted therapy have been shown to be practically ineffective, but certain prospects are associated with the suppression of the Wnt/β-catenin signaling pathway.

6. β-catenin inhibitors and CT

β-catenin is a central factor that ensures the transition of CSCs from survival to proliferation mode. The strategy of counteracting this transition is based on (78) inhibiting the Wnt-ligand bind to the Frizzled receptor complex, suppressing the antagonists of the β-catenin destruction complex, and blocking the interaction between β-catenin and TCF/LEF. Most inhibitors of the Wnt signaling pathway have not yet undergone clinical trials; in this regard, repurposed drugs are of particular interest (Fig. 1 and Table I).

Table I

Repurposed drugs targeting the Wnt/β-catenin signaling pathway.

Table I

Repurposed drugs targeting the Wnt/β-catenin signaling pathway.

Name of the drugMechanism of Wnt signaling inhibitionResults of preclinical/clinical studies(Refs.)
Aspirin-Prostaglandin E2/cyclooxygenase-dependent-Suppression of proliferation in almost any Wnt-dependent cancer(119)
 -Inactivation of PP2A and phosphorylation of β-catenin-Reduction of tumor formation in the FAP mouse model, reduction of β-catenin levels in the tumor 
 -Cross-talk between other pathways (such as NF-κB)-Retrospective studies, especially for the prevention of colon cancer 
  -Recommended for the prevention of CRC in individuals 50 to 69 years of age 
Celecoxib-Prostaglandin E2/cyclooxygenase-dependent-Violation of proliferation in CRC, hepatoma, osteosarcoma and GB Decreased CD133+ colon cancer stem cells(120,121)
 -Promotes proteasomal degradation of TCF1 and TCF4-Inhibition of β-catenin-positive precancerous lesions in the colon of mice and in a model of colon cancer in rats 
 -Cross-linking of the c-Met/AKT pathway promoting GSK-3β phosphorylation-Reduction of polyps in patients with FAP after 6 months of treatment 
Indomethacin -PGE2/COX-dependent-Growth suppression of CRC cell lines(122)
 -Degradation of β-catenin through transcription inhibition-Decreased tumor burden in chemically-induced colon cancer 
 -Impaired formation of the β-catenin/TCF4 complex  
Sulindac -PGE2/COX-dependentGrowth suppression of colorectal cancer cell lines(122)
 -Inactivation of PP2A and β-catenin phosphorylation  
Tigecycline-Decrease in the content of β-catenin in the cytoplasm of CCs-Suppression of the growth of cervical cancer cells(125)
 -Increased synthesis of AXIN1-Inhibition of growth of cervical cancer xenografts 
Niclosamide-Promotion of FZD 1 endocytosis-Antiproliferative activity against osteosarcoma, CRC, breast cancer, lung cancer, hepatoma and GB.(126)
 -Inhibition of DVL2-Reduces the levels of β-catenin in mouse models of colorectal and basal breast cancer 
 -LPR6 degradation  
Pyrvinium pamoate-Activation of the isoform of casein kinase 1α, part of the Wnt pathway destruction complex-Suppresses tumor growth in a colon cancer model(127)
Ivermectin-Deactivation of β-catenin by reducing C-terminal phosphory-lation due to overactivation of phosphatases PP2A and PP1-Antiproliferative against colon cancer (including stem cells) and lung cancer(128)
  -Reduced tumor growth in colon cancer xenograft models 
Clofazimine-Participation in the inhibition of the TCF transcription complex-Suppression of the growth of squamous hepatocellular cancer and lung cancer(129)
 -Cross-talk between other pathways-Suppression of the growth of glioblastoma, lung and breast cancer 
  -Combination and monotherapy for hepatocellular carcinoma with moderate positive results 

[i] PP, protein phosphatase; GB, glioblastoma; CRC, colorectal cancer; TCF, transcription factor; CC, colon cancer; FZD1, Frizzled class receptor 1; DVL2, dishevelled segment polarity protein 2; LRP, low-density lipoprotein receptor-related protein.

Wnt-inhibitory activity has been described for a number of agents from the group of non-steroidal anti-inflammatory drugs. Aspirin is one of the most popular types of medication from this group; its pharmacological effect is based on the inhibition of cyclooxygenase enzymes that reduce the levels of β-catenin in CCs, inhibit the expression of Wnt-target genes, enhance the cytotoxic effect of TMZ and bevacizumab (119), and prevent the development of colorectal cancer and a number of Wnt-dependent neoplasms (Fig. 1A).

Celecoxib, another non-steroidal anti-inflammatory agent, reactivates GSK-3β, eliminates the effects of the activation of the ‘β-catenin degradation complex’, inhibits TCF at a dose of 1,200-1,600 mg/day, and suppresses cyclooxygenase 2 and carboanhydrase, which impairs the adaptation of CCs to hypoxia (120) and enhances the cytotoxic effect of TMZ. In light of this, the administration of celecoxib preoperatively and before the end of CRT (121) has been described as a low-risk and justifiable procedure, but the ability of celecoxib to inhibit platelet aggregation combined with the hematological toxicity of TMZ raises some doubts concerning such claims (Fig. 1B).

The ability to inhibit the intracellular Wnt/β-catenin signaling pathway has also been observed in other drugs of this group. Indomethacin (Fig. 1C) is an indoleacetic acid derivative and cyclooxygenase 2 inhibitor, which disrupts the formation of the β-catenin/TCF complex with DNA and inhibits gene expression (122). Sulindac, a non-steroidal anti-inflammatory drug (Fig. 1D) from the group of acetic acid derivatives (123), enhances the degradation of β-catenin and prevents its translocation to the nucleus, thus inhibiting the expression of Wnt-target genes in CCs.

Despite the antiglioma potential of cyclooxygenase inhibitors, there are practically no data concerning their effect on CSCs. It should not be dismissed that inhibition of cyclooxygenase enzymes (124) may suppress the synthesis of Wnt-ligands by microglia cells, fibroblasts and endothelial cells from the microenvironment of CSCs. However, these drugs are not the only ones to hinder the activity of the Wnt signaling pathway in CSCs.

The antibiotic tigecycline (Fig. 1E) stimulates AXIN1 β gene expression and reduces β-catenin levels in CCs (125). The antiparasitic drug niclosamide (Fig. 1F) causes LRP6 degradation, reduces β-catenin content in the nucleus, and inhibits TCF/LEF factor activity (126). Pyrvinium pamoate (Fig. 1G) regulates MGMT gene expression in GB cells, reactivates GSK-3β and increases the sensitivity of GB cells to TMZ (127). Ivermectin (Fig. 1H) binds to the telomere maintenance 2 (TELO2) protein, which regulates PI3K activity and decreases β-catenin content in CCs (128).

Particular interest should be paid to a drug from the riminophenazine group called clofazimine (CFZ), N,5-bis-(4-chlorophenyl)-3,5-dihydro-3-[(1-methylethyl)imino]-2-phenazinamine, which was synthesized by Vincent Barry in 1957. CFZ was originally used as an antimycobacterial agent with proven bactericidal activity against Hansen's bacillus (Fig. 1I). CFZ inhibits mycobacterial growth, promotes the formation of reactive oxygen species, interacts with phospholipids of cell membranes, and disrupts the ionic equilibrium and energy metabolism of bacterial cells (129). Its anti-inflammatory properties combined with its ability to induce the release of prostaglandins and inhibit phospholipase A2 made it applicable in the complex treatment of erythema nodosum leprosum (130).

The antineoplastic properties of CFZ against triple-negative breast cancer are associated with the inhibited expression of Wnt signaling pathway-related genes, reduction of the cytoplasmic β-catenin level, and triggering of apoptosis due to cell cycle arrest in the G2/M-phase in CCs (131). The antineoplastic effect of CFZ against CCs of different cell lines from colorectal adenocarcinoma and ovarian cancer is exhibited by the IC50 of the drug, in the range of 2-10 µm/l, while against different cell lines of human GB this value is 20-40 µm/l, which may be associated with both Wnt inhibition and other metabolic effects of the drug (132).

CFZ partially penetrates into the cerebrospinal fluid through the intact BBB, but by being a lipophilic substance, the drug accumulates well in adipose tissue and in monocytes, and is captured by macrophages (133). Phagocytosis of CFZ crystals is not accompanied by obvious toxic manifestations, it suppresses NF-κB, enhances the synthesis of IL-1 receptor antagonist, and induces the M2 activation of macrophages (134). CFZ, captured and transported by monocytes, accumulates in the lungs, liver, and spleen (135), and partially in the bone marrow. Such findings suggest that CFZ may be a promising medical agent to be delivered into a tumor with the help of mononuclear cells, which constitute an important part of the CSC microenvironment (136).

There is a reasonable assumption that the need for specific transport through the BBB could decrease the antiglioma potential of the drug. However, the antiglioma Wnt-inhibitory effect has been described for valproic acid derivatives, phenothiazine neuroleptics, olanzapine, amisulpiride and other drugs (28), which pass through the BBB with ease. Nevertheless (120), since 2005, >100 studies on the combination of CT with different drugs have failed. Identifying the reasons of such outcomes is directly dependent on the need to regulate the CSC microenvironment, which is, the main factor contributing to the lower antioglioma potential of pharmacological agents. However, the development of practical approaches to solving this problem requires rethinking a number of its important theoretical aspects.

7. CSC microenvironment, immunotherapy and immunodeficiency

Since the 1950s, the brain has been considered to be an immune privileged organ. The microenvironment of CSCs was associated only with resident microglia cells, which were considered to originate from cells of the embryonic yolk sac and not to interact with the immune system after the BBB closure (137), and mononuclear cells and other bone marrow immunocytes were not considered to interact with microglia in any way. However, numerous data suggest otherwise.

It has been demonstrated that microglial cells can enter deep cervical lymph nodes and interact with T and B lymphocytes. Each T cell can recognize several hundred fragments of a single antigen on the antigen-presenting cell membrane together with class I and class II major histocompatibility complex molecules. At the same time, B cells can bind intact antigens with their native structure, indicating an active informational interaction between bone marrow cells and microglia. Simultaneously, immunocytes are able to penetrate the brain through the tumor bloodstream, vascular plexuses, cranial microchannels and sinuses, and cerebrospinal and interstitial fluid (138), and kill cells containing the presented antigen. Such findings indicate an important role of the immune system in the formation of the CSC microenvironment.

Experimental studies conducted in the last decade have expanded the concept with regard to the participation of immunocytes in GB pathogenesis. Tumor development in the brain is accompanied by migration and homing of red bone marrow cells to the tumor nidus (139). To date, >80 chemoattractants have been described to draw bone marrow cells to the tumor nidus through different types of receptors, including the recognition of stromal cell factor (SDF)1 or chemokine C-X-C motif ligand 12 (CXCL12), a chemokine of the CXC subfamily that binds to the CXCR4 receptor on the membrane of CD45+ cells in the bone marrow, inducing their migration to the tumor (140). Numerous studies (141,142) suggest that the involvement of bone marrow cells in the neoplastic process enriches the population of immunosuppressive tumor microglia, and is accompanied by a stronger resistance of CCs to cytostatics.

Production of numerous immunosuppressive cytokines by neoplastic cells determines the microglia polarization vector, with a significant proportion of CCs being completely removed during surgery or destroyed by CRT. It is safe to assume that vaccination with dead tumor tissue can enhance the antineoplastic immune response, and the production of exosome-containing microRNAs and other antitumor factors that can significantly alter the properties of the CSC microenvironment, resulting in increased effectiveness of CT. However, the local microenvironment in tumor recurrence is formed in conditions of overall immunodeficiency that results from the debilitating effect of PD-ligands, glucocorticosteroids, radiation and CT (143), and this fact predetermines its characteristics.

In fact, the exhausted phenotype of immunocytes is developed due to the GB cells producing PD-L1 and 2, CTLA-4 and other proteins that inhibit T-cell activity receptors (144). The emergence of a new class of drugs that prevent PD-ligands from binding to T-cell receptors has attracted interest (145), but the effectiveness of nivolumab and other immune checkpoint inhibitors in the complex treatment of GB was revealed to be low (146), thus suggesting the existence of other causes of systemic immunosuppression.

Corticosteroids in the complex treatment of GB are used to counteract cerebral oedema, but their use (147) is associated with inhibition of lymphocyte proliferation, suppression of migration and interaction of macrophages with T and B lymphocytes, inhibition of interferon gamma (IFN-γ) release from macrophages, and reduced antibody formation. The use of corticosteroids leads to increased blood glucose levels and dexamethasone-induced leukocytosis (148), which exhausts the red bone marrow (149) and reduces overall survival.

The maximum possible reduction of the number of CCs in brain tissue is achieved by active use of chemoradiation therapy. According to a previous study, >30 fractions of γ-therapy allow lymphocytes to accumulate an average radiation dose of 2.3 Gy, and the number of CD4+ cells in the organism decreases by half and remains low for >1 year (150), which is time a patient may not have. It is likely that higher radiation doses, which are often used in treatment regimens such as in the case of radiosurgery, may be accompanied by accumulation of a significantly higher radiation dose in immunocytes, which induces a bystander effect in the bone marrow and causes the death of a significant number of immune cells.

In turn, cytostatics suppress hematopoiesis and immunopoiesis, contributing to the development of clinically significant thrombocytopenia and leukopenia, which is generally considered the only criterion (151) limiting TMZ dose escalation. Notably, a neutrophil count ≥40% below the norm is a criterion for a satisfactory prognosis in IDH-wild-type GB (152), while reaching grades 3 and 4 neutropenia is considered a positive prognosis for CT outcome, with a neutrophil count of <1-109 cells/l, warranting dose reduction during the subsequent treatment stage.

In light of this, the completion of the main regimen of GB treatment results in the immunodeficiency of the patient, while tumor relapse is accompanied by the recruitment of immunocytes with an ‘exhausted phenotype’ and the formation of an immunosuppressive CSC microenvironment, which supports tumor growth, promotes further involvement of the bone marrow in the neoplastic process and prevents immunotherapy from fulfilling its antineoplastic potential completely (Fig. 2).

8. HSCs and immunotherapy

Reprogramming of HSCs in the bone marrow during tumor growth largely contributes to the process of immunosuppression. Participation of normal stem cells in the tumor process became a focus of attention after the publication of the study by Aboody et al (152), which showed the migration of NSCs to glioma cells. Snyder et al (153) reported the existence of an association between GB cells and normal stem cells of other types, in particular with HSCs. HSCs have been demonstrated to have high mobility towards GB cells (154) and, when injected into the bloodstream of an animal with glioma, they migrate to the tumor nidus, where they accumulate in the blood vessels of the tumor (155), spread throughout the invasion area, and penetrate into the necrotic zones of neoplastic tissue.

While migrating into a tumor, HSCs are capable of interacting with CCs and can exchange a fluorescent stain, which becomes inextricably connected with intracellular proteins in the process of staining (156), and this fact indicates the exchange of information between stem cells and CCs. The reported mechanisms of such exchange (157) include the fusion of stem cells and CCs, horizontal transfer of information through tights intercellular junctions with areas of partial membrane fusion and cytoplasm unification (158), and the production of exosomes and other microvesicles containing fragments of DNA, microRNA and proteins (159) into the external environment. All this is accompanied by the reprogramming of interacting cells and their development of new properties.

The significance of reprogramming of HSCs in GB pathogenesis has barely been studied, but there are reasons to consider that it is the most important factor of the immune system inactivation in the tumor process (160). Bone marrow stem cells are known to be a heterogeneous mixture of subpopulations, having cell elements with different degrees of maturity, lifespan, gene expression profiles and epigenetic programs of differentiation (161). Notably, a part of HSCs, reprogrammed by the tumor, are able to gain advantage over other HSC clones in bone marrow (162), which can be accompanied by the expansion of mutant immunocytes that are tumor tolerant (163).

Theoretically speaking, this is the most important issue predetermining the final effect of all existing glioma immunotherapy methods, the scenarios of which are based on the use of mononuclear CD45+ cells recruited from the bone marrow into the systemic bloodstream when granulocytic (G-CSF) or granulocytic-macrophage (GM-CSF) colony-stimulating factors are administered to patients. According to experimental data (164,165), the introduction of G-CSF into the organism of experimental animals with injected glial brain tumor fills the tumor tissue with markers of anti-inflammatory microglia (166), while the subsequent introduction of bacterial lipopolysaccharide and IFN-γ (167) promotes the inflammatory M1 activation of macrophages. An even greater antiglioma effect is achieved when HSCs of a healthy sibling are transplanted into the organism of an animal with glioma (161).

This fact directly indicates the essential role of HSCs in the formation of the CSC microenvironment. Reprogramming of HSCs during their interaction with CSCs and severe leukopenia from chemoradiation therapy form a vicious circle, creating conditions for β-catenin accumulation in CSCs and enhancing the lethal potential of this cell type. In this regard, β-catenin content in tumors is an important criterion of immunotherapy effectiveness, and the existing protocols of immunotherapy should be supplemented with drugs that reduce the level of β-catenin (Fig. 3). At the same time, the construction of immunotherapy scenarios using HSCs of a healthy relative or autologous HSCs prepared before the disease, suggests positive future prospects for the complex treatment of gliomas.

The existing algorithms of adaptive cell-based immunotherapy of gliomas are still far from reaching this goal. Adaptive immunotherapy involves the use of autologous immunocytes, the source of which is a ‘leucoconcentrate’ of mononuclear cells expressing the leukocyte common antigen CD45+, with subsequent separation of T-lymphocytes ex vivo, their additional activation by IL-2 and the targeting of GB cells with proteins against key antigens, including IL13Rα2, HER2 and EGFRvIII. The disadvantages of such therapy are clear, since administration of G-CSF to the patient and stimulation with antigens of Bacillus Calmette-Guérin, Staphylococcus aureus or other suppurative microflora may lead to increased brain oedema, which is a life-threatening condition.

An alternative strategy involves the stimulation of the immunocytes of a patient ex vivo with antigens of suppurative microflora and CCs; however, after CT, the bone marrow of the patient is exhausted, and it is practically impossible to accumulate the required number of immunocytes during one stage of leucopheresis. At the same time, repeated administration of G-CSF to the patient further exhausts the bone marrow and can promote the formation of an immunosuppressive environment of CSCs.

Attempts have been made to use genetically modified T cells equipped with chimeric antigen receptors (CARs), in which sections of antigen-recognition domains, consisting of monoclonal antibodies, are connected to sections of intracellular T-lymphocyte signaling domains. The main problem with this technology is the lack of absolutely perfect target antigens that are completely tumor-specific, since the antigens IL13Rα2, HER2, EGFRvIII, B7-H3 and CSPG4, which are traditionally used as targets for CAR-T cell creation, are not specific to CSCs, but much less homogeneously expressed in tumors.

Attempts to create multivalent CAR-T cells (167) targeted against the synNotch receptor and other molecular targets in CSCs have not yet produced the desired effect, since local immunosuppression remains the main problem limiting the antitumor potential of CAR-T cells. Attempts to combine CAR-T cells with microRNAs that locally inhibit immunosuppressive genes (168) have not yet exhibited any specific benefits and are associated with toxicity and BBB penetrability problems (169). Attempts to increase the efficiency of the targeted delivery of CAR-T cells by binding them to chlorotoxin, a peptide particularly tropic to CCs (170) that is found in the venom of the scorpion Leiurus quinquestriatus, as well as to combine CAR-T therapy with intratumoral delivery of IL-12(171) have not been successful either.

The main disadvantage of adaptive immunotherapy methods is the use of autologous exhausted immunocytes. Exhaustion is a state of T-cell dysfunction that occurs in cases of chronic infections and cancer, and is characterized by decreased effector function and self-renewal capacity, sustained expression of inhibitory receptors and a transcriptional state (171) distinct from that of functionally active effector cells or memory T cells. Exhaustion prevents optimal control of infections and tumors, but serves as a mechanism to protect cells from death when they are hyperstimulated by tumor antigens.

It can be assumed that a high proliferation rate and a dynamic change of the antigen spectrum of GB cells are aggression factors that inactivate the immune system. Attempts to solve this problem using only immune checkpoint inhibitors have not been successful thus far; in this regard, the use of allogeneic cytotoxic lymphocytes derived from a healthy donor has been reported (172), which requires increasing doses of dexamethasone, revealing the typical consequences of such therapy. However, this approach deserves special attention, particularly taking into account the possibility of regulating the local microenvironment of CSCs by using a combination of CAR-T cell technology with oncolytic adenoviruses (173) and immune checkpoint inhibitors (174).

According to a previous study (175), CAR-T cell therapy poses serious problems in the treatment of central nervous system tumors, which emphasizes the problem of the hostile immunosuppressive microenvironment of CSCs. However, in addition to CAR-T cells, attempts to use CAR-T natural killer cells or CAR macrophages and to generate active antineoplastic immunity in patients with GB are of great interest.

Active immunotherapy involves the development of antineoplastic immunity in a patient with GB by vaccination with tumor cell vaccines or incubation of immunocytes with CC lysates ex vivo, with the subsequent return to the patient in the form of a dendritic cell vaccine (176). The use of tumor cell vaccines has numerous advantages, including the fact that systematic vaccination activates CD8-lymphocytes, the processes of antigen presentation by macrophages, synthesis of pro-inflammatory cytokines and modification of the tumor microenvironment (177), which increases the survival rate of patients with both newly diagnosed and relapsed GB (178).

Autologous or allogeneic CSCs are used as antigens to create tumor cell vaccines, which can stimulate an immune response considering the heterogeneity of the neoplastic cell population (179), which is particularly relevant for GB. In light of this, a tumor cell vaccine should include a combination of dead CSCs (for example, after repeated freezing and thawing of proneural-type CSCs derived from the first removed tumor) and CSCs of mesenchymal phenotype obtained by sequential irradiation of autologous CCs, as well as CSC derivatives from other patients. This approach allows not only the destruction of CCs, but can also significantly increase antineoplastic immunity (180) and allow the formation of local criss-cross intratumor interactions between CD4+ lymphocytes and other T cells (181), thus leading to increased production of proinflammatory cytokines and modifying the microenvironment of CSCs.

It is possible to create vaccines (182) using live genetically modified CCs producing GM-CSF. Antigen-specific antineoplastic vaccines producing GM-CSF have been used for tumor treatment for >20 years, and their use predominantly reveals the problem of leucopenia and immunodeficiency, actually limiting the antiglioma potential of immunotherapy. A possible method to solve this issue is the combination of a tumor cell vaccine with transplantation of HSCs from a haploidentical donor, ideally a sibling.

The common disadvantages of using tumor cell vaccines are characteristic of the immunotherapy method with dendritic cell vaccines. Dendritic cells are professional antigen-presenting cells with high functional plasticity, which originate from HSCs and show immunostimulatory or immunosuppressive potential depending on the sequence and combination of microenvironmental stimuli. The technology for the preparation of dendritic cell vaccines includes the administration of G-CSF or GM-CSF to the patient in order to recruit immunocytes into the bloodstream, subsequent isolation of a fraction of mononuclear cells containing a sufficient number of CD34+ HSCs, ex vivo stimulation with CC antigens combined with IL-2 or IL-4, multiplication in the presence of pro-inflammatory cytokines, and return to the patient (182). Antigens for the creation of dendritic cell vaccines include CSCs, CC lysates, produced CCs and CSCs, exosomes, glioma-associated peptides, and DNA and RNA fragments (183), but their efficacy in the treatment of GB is relatively low.

Despite increased individual survival periods of >23 months that have been reported (184), the overall situation has not changed even after combining dendritic cell vaccines with immune checkpoint inhibitors and other immunotherapy methods (185). Therapy with dendritic cell vaccines results in general immunostimulatory effects in the form of increased levels of immunosuppressive cytokines and local infiltration of the tumor stroma by immunocytes. However, the main challenge of therapy with dendritic cell vaccines is the problem of immunosuppression, the solution of which is possible only after creating such vaccines on the basis of healthy HSCs or developing methods of restoring the patient's own HSCs.

It is the condition of HSCs that predetermines the final effectiveness of almost all immunotherapy methods, but the official ClinicalTrails website describes only one study (namely NCT00014573) suggesting the use of immunotherapy methods while supporting HSC transplantation in treatment-refractory brain tumors. The use of autologous HSCs or stem cells of a sibling, and pharmacological regulation of β-catenin level in HSCs together with immunotherapy has not been discussed to date.

9. Conclusion

CSCs present a major challenge in GB treatment. Destruction of these CCs with irradiation and cytostatics appears to be impossible, and requires certain adjustments of existing GB treatment strategies towards regulation of CSCs rather than destruction of CCs. Numerous attempts to solve this problem with the help of targeted medication have not been successful thus far, which is usually explained by the plasticity of CSCs, with β-catenin being the central link in the system of intracellular signaling pathways, regulating the plasticity of this cell type.

Pharmacological regulation of the β-catenin level in CSCs using repurposed drugs opens new horizons for the regulation of the proliferative potential of this cell type. The antileprosy drug CFZ has the ability to inhibit the intracellular Wnt/β-catenin signaling pathway. It is characterized by its good tolerability and has demonstrated antineoplastic activity against CSCs of a number of aggressive carcinomas, while its antiglioma potential is virtually unexplored. The ability of CFZ to accumulate in monocytes makes it very promising to be used as a drug for targeted delivery to the tumor nidus, due to the transportation potential of this cell type (Fig. 4).

Particular attention should be paid to the fact that the β-catenin level in CSCs directly increases under the influence of an immunosuppressive microenvironment, which is formed with participation of microgliocytes and monocytes of the exhausted phenotype, caused by radiation, cytostatics and the reprogramming effect of the tumor on bone marrow HSCs. Indeed, all existing GB treatment protocols lead to immunodeficiency, which actually limits the therapeutic potential of all drugs and technologies, since it is almost impossible to reduce the β-catenin level without suppressing Wnt ligand production by the local microenvironment of HSCs.

The use of CFZ and other repurposed drugs with demonstrated Wnt-inhibitory activity can reduce the level of β-catenin in CSCs. In turn, immunotherapy can regulate the microenvironment of CSCs, resulting in a decrease in Wnt-ligand synthesis and breaking the aforementioned vicious circle, which would allow to create a technology of CSC management as part of the complex immunotherapy of GB. One of the most important tasks in the creation of such technology is developing immunotherapy scenarios with the use of tumor cell vaccines containing a heterogeneous composition of autologous and allogeneic, irradiated and non-irradiated CCs, which will allow to induce a multidirectional antineoplastic immune response. This should be enhanced by the transplantation of healthy sibling HSCs and the administration of CFZ or other drugs, thus reducing the β-catenin content in CSCs. Pharmacological regulation of β-catenin content in CSCs and cellular immunotherapy should be used together, since they are two sides of the same coin.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

AK, OP and IB conceived the study and searched for relevant articles. AK and IB wrote the original draft, and reviewed and edited the final manuscript. Data authentication is not applicable. All authors have read and agreed to the final manuscript.

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.

References

1 

Herrlinger U: News on the horizon in glioblastoma therapy. ESMO Open. 5(e000601)2020.PubMed/NCBI View Article : Google Scholar

2 

Schaff LR and Mellinghoff IK: Glioblastoma and other primary brain malignancies in adults: A review. JAMA. 329:574–587. 2023.PubMed/NCBI View Article : Google Scholar

3 

Delavar A, Wali AR, Santiago-Dieppa DR, Al Jammal OM, Kidwell RL and Khalessi AA: Racial and ethnic disparities in brain tumor survival by age group and tumor type. Br J Neurosurg. 36:705–711. 2022.PubMed/NCBI View Article : Google Scholar

4 

Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al: Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-Year analysis of the EORTC-NCIC trial. Lancet Oncol. 10:459–466. 2009.PubMed/NCBI View Article : Google Scholar

5 

Luo C, Song K, Wu S, Hameed NUF, Kudulaiti N, Xu H, Qin ZY and Wu JS: The prognosis of glioblastoma: A large, multifactorial study. Br J Neurosurg. 35:555–561. 2021.PubMed/NCBI View Article : Google Scholar

6 

Yabo YA, Niclou SP and Golebiewska A: Cancer cell heterogeneity and plasticity: A paradigm shift in glioblastoma. Neuro Oncol. 24:669–682. 2022.PubMed/NCBI View Article : Google Scholar

7 

Bikfalvi A, da Costa CA, Avril T, Barnier JV, Bauchet L, Brisson L, Cartron PF, Castel H, Chevet E, Chneiweiss H, et al: Challenges in glioblastoma research: Focus on the tumor microenvironment. Trends Cancer. 9:9–27. 2023.PubMed/NCBI View Article : Google Scholar

8 

Huang M, Zhang D, Wu JY, Xing K, Yeo E, Li C, Zhang L, Holland E, Yao L, Qin L, et al: Wnt-mediated endothelial transformation into mesenchymal stem cell-like cells induces chemoresistance in glioblastoma. Sci Transl Med. 12(eaay7522)2020.PubMed/NCBI View Article : Google Scholar

9 

Barzegar Behrooz A, Talaie Z, Jusheghani F, Łos MJ, Klonisch T and Ghavami S: Wnt and PI3K/Akt/mTOR survival pathways as therapeutic targets in glioblastoma. Int J Mol Sci. 23(1353)2022.PubMed/NCBI View Article : Google Scholar

10 

Behrooz AB and Syahir A: Could we address the interplay between CD133, Wnt/β-catenin, and TERT signaling pathways as a potential target for glioblastoma therapy? Front Oncol. 11(642719)2021.PubMed/NCBI View Article : Google Scholar

11 

Crunkhorn S: Targeting drug-resistant glioblastoma. Nat Rev Drug Discov. 21(711)2022.PubMed/NCBI View Article : Google Scholar

12 

Precilla DS, Kuduvalli SS, Purushothaman M, Marimuthu P, Muralidharan AR and Anitha TS: Wnt/β-catenin antagonists: Exploring new avenues to trigger old drugs in alleviating glioblastoma multiforme. Curr Mol Pharmacol. 15:338–360. 2022.PubMed/NCBI View Article : Google Scholar

13 

Yuan B, Wang G, Tang X, Tong A and Zhou L: Immunotherapy of glioblastoma: Recent advances and future prospects. Hum Vaccin Immunother. 18(2055417)2022.PubMed/NCBI View Article : Google Scholar

14 

Youngblood MW, Stupp R and Sonabend AM: Role of Resection in glioblastoma management. Neurosurg Clin N Am. 32:9–22. 2021.PubMed/NCBI View Article : Google Scholar

15 

De Biase G, Garcia DP, Bohnen A and Quiñones-Hinojosa A: Perioperative management of patients with glioblastoma. Neurosurg Clin N Am. 32:1–8. 2021.PubMed/NCBI View Article : Google Scholar

16 

Lu VM, Goyal A, Graffeo CS, Perry A, Burns TC, Parney IF, Quinones-Hinojosa A and Chaichana KL: Survival benefit of maximal resection for glioblastoma reoperation in the temozolomide era: A meta-analysis. World Neurosurg. 127:31–37. 2019.PubMed/NCBI View Article : Google Scholar

17 

Robin AM, Lee I and Kalkanis SN: Reoperation for recurrent glioblastoma multiforme. Neurosurg Clin N Am. 28:407–428. 2017.PubMed/NCBI View Article : Google Scholar

18 

Liang C, Gong J, Zhang B, Meng Z, Li M and Guo Y: Multiple subtentorial metastasis in diffuse midline glioma receiving tumor treating fields: A case report and literature review. Ann Transl Med. 9(1604)2021.PubMed/NCBI View Article : Google Scholar

19 

Chen J, Shi Q, Li S, Zhao Y and Huang H: Clinical characteristics of glioblastoma with metastatic spinal dissemination. Ann Palliat Med. 11:506–512. 2022.PubMed/NCBI View Article : Google Scholar

20 

Shah AH, Mahavadi A, Di L, Sanjurjo A, Eichberg DG, Borowy V, Figueroa J, Luther E, de la Fuente MI, Semonche A, et al: Survival benefit of lobectomy for glioblastoma: Moving towards radical supramaximal resection. J Neurooncol. 148:501–508. 2020.PubMed/NCBI View Article : Google Scholar

21 

Ryan JT, Nakayama M, Gleeson I, Mannion L, Geso M, Kelly J, Ng SP and Hardcastle N: Functional brain imaging interventions for radiation therapy planning in patients with glioblastoma: A systematic review. Radiat Oncol. 17(178)2022.PubMed/NCBI View Article : Google Scholar

22 

Ylanan AMD, Pascual JSG, Cruz-Lim EMD, Ignacio KHD, Cañal JPA and Khu KJO: Intraoperative radiotherapy for glioblastoma: A systematic review of techniques and outcomes. J Clin Neurosci. 93:36–41. 2021.PubMed/NCBI View Article : Google Scholar

23 

Khan L, Soliman H, Sahgal A, Perry J, Xu W and Tsao MN: External beam radiation dose escalation for high grade glioma. Cochrane Database Syst Rev. 5(CD011475)2020.PubMed/NCBI View Article : Google Scholar

24 

Barbarite E, Sick JT, Berchmans E, Bregy A, Shah AH, Elsayyad N and Komotar RJ: The role of brachytherapy in the treatment of glioblastoma multiforme. Neurosurg Rev. 40:195–211. 2017.PubMed/NCBI View Article : Google Scholar

25 

Vogelius IR and Bentzen SM: Proton vs photon radiation therapy for glioblastoma: Maximizing information from trial. Neuro Oncol. 24:849–850. 2022.PubMed/NCBI View Article : Google Scholar

26 

Malouff TD, Seneviratne DS, Ebner DK, Stross WC, Waddle MR, Trifiletti DM and Krishnan S: Boron Neutron capture therapy: A review of clinical applications. Front Oncol. 11(601820)2021.PubMed/NCBI View Article : Google Scholar

27 

Laprie A, Tensaouti F and Cohen-Jonathan Moyal E: Radiation dose intensification for glioblastoma. Cancer Radiother. 26:894–898. 2022.PubMed/NCBI View Article : Google Scholar : (In French).

28 

Tan AC, Ashley DM, López GY, Malinzak M, Friedman HS and Khasraw M: Management of glioblastoma: State of the art and future directions. CA Cancer J Clin. 70:299–312. 2020.PubMed/NCBI View Article : Google Scholar

29 

Woodroffe RW, Zanaty M, Soni N, Mott SL, Helland LC, Pasha A, Maley J, Dhungana N, Jones KA, Monga V and Greenlee JDW: Survival after reoperation for recurrent glioblastoma. J Clin Neurosci. 73:118–124. 2020.PubMed/NCBI View Article : Google Scholar

30 

Zakaria R and Weinberg JS: Challenges associated with reoperation in patients with glioma. Neurosurg Clin N Am. 32:129–135. 2021.PubMed/NCBI View Article : Google Scholar

31 

Mathen P, Rowe L, Mackey M, Smart D, Tofilon P and Camphausen K: Radiosensitizers in the temozolomide era for newly diagnosed glioblastoma. Neurooncol Pract. 7:268–276. 2020.PubMed/NCBI View Article : Google Scholar

32 

Herrlinger U, Tzaridis T, Mack F, Steinbach JP, Schlegel U, Sabel M, Hau P, Kortmann RD, Krex D, Grauer O, et al: Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): A randomised, open-label, phase 3 trial. Lancet. 393:678–688. 2019.PubMed/NCBI View Article : Google Scholar

33 

Hwang K, Lee JH, Kim SH, Go KO, Ji SY, Han JH and Kim CY: The combination PARP inhibitor olaparib with temozolomide in an experimental glioblastoma model. In Vivo. 35:2015–2023. 2021.PubMed/NCBI View Article : Google Scholar

34 

Nguyen TTT, Zhang Y, Shang E, Shu C, Torrini C, Zhao J, Bianchetti E, Mela A, Humala N, Mahajan A, et al: HDAC inhibitors elicit metabolic reprogramming by targeting super-enhancers in glioblastoma models. J Clin Invest. 130:3699–3716. 2020.PubMed/NCBI View Article : Google Scholar

35 

Bindra RS: Penetrating the brain tumor space with DNA damage response inhibitors. Neuro Oncol. 22:1718–1720. 2020.PubMed/NCBI View Article : Google Scholar

36 

Zhao J, Yang S, Cui X, Wang Q, Yang E, Tong F, Hong B, Xiao M, Xin L, Xu C, et al: A novel compound EPIC-0412 reverses temozolomide resistance via inhibiting DNA repair/MGMT in glioblastoma. Neuro Oncol. 25:857–870. 2023.PubMed/NCBI View Article : Google Scholar

37 

Goel NJ, Bird CE, Hicks WH and Abdullah KG: Economic implications of the modern treatment paradigm of glioblastoma: An analysis of global cost estimates and their utility for cost assessment. J Med Econ. 24:1018–1024. 2021.PubMed/NCBI View Article : Google Scholar

38 

Lauko A, Lo A, Ahluwalia MS and Lathia JD: Cancer cell heterogeneity & plasticity in glioblastoma and brain tumors. Semin Cancer Biol. 82:162–175. 2022.PubMed/NCBI View Article : Google Scholar

39 

Oliver L, Lalier L, Salaud C, Heymann D, Cartron PF and Vallette FM: Drug resistance in glioblastoma: Are persisters the key to therapy? Cancer Drug Resist. 3:287–301. 2020.PubMed/NCBI View Article : Google Scholar

40 

Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, et al: Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 17:98–110. 2010.PubMed/NCBI View Article : Google Scholar

41 

Steponaitis G and Tamasauskas A: Mesenchymal and proneural subtypes of glioblastoma disclose branching based on GSC associated signature. Int J Mol Sci. 22(4964)2021.PubMed/NCBI View Article : Google Scholar

42 

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.PubMed/NCBI View Article : Google Scholar

43 

Melhem JM, Detsky J, Lim-Fat MJ and Perry JR: Updates in IDH-wildtype glioblastoma. Neurotherapeutics. 19:1705–1723. 2022.PubMed/NCBI View Article : Google Scholar

44 

French R and Pauklin S: Epigenetic regulation of cancer stem cell formation and maintenance. Int J Cancer. 148:2884–2897. 2021.PubMed/NCBI View Article : Google Scholar

45 

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.PubMed/NCBI View Article : Google Scholar

46 

Capper D, Stichel D, Sahm F, Jones DTW, Schrimpf D, Sill M, Schmid S, Hovestadt V, Reuss DE, Koelsche C, et al: Practical implementation of DNA methylation and copy-number-based CNS tumor diagnostics: The Heidelberg experience. Acta Neuropathol. 136:181–210. 2018.PubMed/NCBI View Article : Google Scholar

47 

Neftel C, Laffy J, Filbin MG, Hara T, Shore ME, Rahme GJ, Richman AR, Silverbush D, Shaw ML, Hebert CM, et al: An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell. 178:835–849.e21. 2019.PubMed/NCBI View Article : Google Scholar

48 

Garofano L, Migliozzi S, Oh YT, D'Angelo F, Najac RD, Ko A, Frangaj B, Caruso FP, Yu K, Yuan J, et al: Pathway-based classification of glioblastoma uncovers a mitochondrial subtype with therapeutic vulnerabilities. Nat Cancer. 2:141–156. 2021.PubMed/NCBI View Article : Google Scholar

49 

Hubert CG and Lathia JD: Seeing the GBM diversity spectrum. Nat Cancer. 2:135–137. 2021.PubMed/NCBI View Article : Google Scholar

50 

Richards LM, Whitley OKN, MacLeod G, Cavalli FMG, Coutinho FJ, Jaramillo JE, Svergun N, Riverin M, Croucher DC, Kushida M, et al: Gradient of developmental and injury response transcriptional states defines functional vulnerabilities underpinning glioblastoma heterogeneity. Nat Cancer. 2:157–173. 2021.PubMed/NCBI View Article : Google Scholar

51 

Ensenyat-Mendez M, Íñiguez-Muñoz S, Sesé B and Marzese DM: iGlioSub: An integrative transcriptomic and epigenomic classifier for glioblastoma molecular subtypes. BioData Min. 14(42)2021.PubMed/NCBI View Article : Google Scholar

52 

Wang LB, Karpova A, Gritsenko MA, Kyle JE, Cao S, Li Y, Rykunov D, Colaprico A, Rothstein JH, Hong R, et al: Proteogenomic and metabolomic characterization of human glioblastoma. Cancer Cell. 39:509–528.e20. 2021.PubMed/NCBI View Article : Google Scholar

53 

Drakulic D, Schwirtlich M, Petrovic I, Mojsin M, Milivojevic M, Kovacevic-Grujicic N and Stevanovic M: Current opportunities for targeting dysregulated neurodevelopmental signaling pathways in glioblastoma. Cells. 11(2530)2022.PubMed/NCBI View Article : Google Scholar

54 

Bonnet D and Dick JE: Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 3:730–737. 1997.PubMed/NCBI View Article : Google Scholar

55 

Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, Blaudszun AR, Yevsa T, Fricke S and Kossatz-Boehlert U: Cancer stem cells-origins and biomarkers: Perspectives for targeted personalized therapies. Front Immunol. 11(1280)2020.PubMed/NCBI View Article : Google Scholar

56 

Bryukhovetskiy A, Shevchenko V, Kovalev S, Chekhonin V, Baklaushev V, Bryukhovetskiy I and Zhukova M: To the novel paradigm of proteome-based cell therapy of tumors: Through comparative proteome mapping of tumor stem cells and tissue-specific stem cells of humans. Cell Transplant. 23 (Suppl 1):S151–S170. 2014.PubMed/NCBI View Article : Google Scholar

57 

Bryukhovetskiy IS, Dyuizen IV, Shevchenko VE, Bryukhovetskiy AS, Mischenko PV, Milkina EV and Khotimchenko YS: Hematopoietic stem cells as a tool for the treatment of glioblastoma multiforme. Mol Med Rep. 14:4511–4520. 2016.PubMed/NCBI View Article : Google Scholar

58 

Couturier CP, Nadaf J, Li Z, Baig S, Riva G, Le P, Kloosterman DJ, Monlong J, Nkili Meyong A, Allache R, et al: Glioblastoma scRNA-seq shows treatment-induced, immune-dependent increase in mesenchymal cancer cells and structural variants in distal neural stem cells. Neuro Oncol. 24:1494–1508. 2022.PubMed/NCBI View Article : Google Scholar

59 

Wang X, Zhou R, Xiong Y, Zhou L, Yan X, Wang M, Li F, Xie C, Zhang Y, Huang Z, et al: Sequential fate-switches in stem-like cells drive the tumorigenic trajectory from human neural stem cells to malignant glioma. Cell Res. 31:684–702. 2021.PubMed/NCBI View Article : Google Scholar

60 

Mizrak D, Brittan M and Alison M: CD133: Molecule of the moment. J Pathol. 214:3–9. 2008.PubMed/NCBI View Article : Google Scholar

61 

Zheng Y, Wang L, Yin L, Yao Z, Tong R, Xue J and Lu Y: Lung cancer stem cell markers as therapeutic targets: An update on signaling pathways and therapies. Front Oncol. 12(873994)2022.PubMed/NCBI View Article : Google Scholar

62 

Park J, Kim SK, Hallis SP, Choi BH and Kwak MK: Role of CD133/NRF2 axis in the development of colon cancer stem cell-like properties. Front Oncol. 11(808300)2022.PubMed/NCBI View Article : Google Scholar

63 

Hefni AM, Sayed AM, Hussien MT, Abdalla AZ and Gabr AG: CD133 is an independent predictive and prognostic marker in metastatic breast cancer. Cancer Biomark. 35:207–215. 2022.PubMed/NCBI View Article : Google Scholar

64 

Gimple RC, Bhargava S, Dixit D and Rich JN: Glioblastoma stem cells: Lessons from the tumor hierarchy in a lethal cancer. Genes Dev. 33:591–609. 2019.PubMed/NCBI View Article : Google Scholar

65 

Beier CP and Beier D: CD133 negative cancer stem cells in glioblastoma. Front Biosci (Elite Ed). 3:701–710. 2011.PubMed/NCBI View Article : Google Scholar

66 

Wang Z, Zhang H, Xu S, Liu Z and Cheng Q: The adaptive transition of glioblastoma stem cells and its implications on treatments. Signal Transduct Target Ther. 6(124)2021.PubMed/NCBI View Article : Google Scholar

67 

Xie XP, Laks DR, Sun D, Ganbold M, Wang Z, Pedraza AM, Bale T, Tabar V, Brennan C, Zhou X and Parada LF: Quiescent human glioblastoma cancer stem cells drive tumor initiation, expansion, and recurrence following chemotherapy. Dev Cell. 57:32–46.e8. 2022.PubMed/NCBI View Article : Google Scholar

68 

Liu J, Gao L, Zhan N, Xu P, Yang J, Yuan F, Xu Y, Cai Q, Geng R and Chen Q: Hypoxia induced ferritin light chain (FTL) promoted epithelia mesenchymal transition and chemoresistance of glioma. J Exp Clin Cancer Res. 39(137)2020.PubMed/NCBI View Article : Google Scholar

69 

Gao L, Tong S, Liu J, Cai J, Ye Z, Zhou L, Song P, Li Z, Lei P, Wei H, et al: TMEM2 induces epithelial-mesenchymal transition and promotes resistance to temozolomide in GBM cells. Heliyon. 9(e16559)2023.PubMed/NCBI View Article : Google Scholar

70 

Zhang X, Wang X, Xu R, Ji J, Xu Y, Han M, Wei Y, Huang B, Chen A, Zhang Q, et al: YM155 decreases radiation-induced invasion and reverses epithelial-mesenchymal transition by targeting STAT3 in glioblastoma. J Transl Med. 16(79)2018.PubMed/NCBI View Article : Google Scholar

71 

Zhang X, Wang X, Xu R, Ji J, Xu Y, Han M, Wei Y, Huang B, Chen A, Zhang Q, et al: Correction to: YM155 decreases radiation-induced invasion and reverses epithelial-mesenchymal transition by targeting STAT3 in glioblastoma. J Transl Med. 19(407)2021.PubMed/NCBI View Article : Google Scholar

72 

Huang W, Zhang C, Cui M, Niu J and Ding W: Inhibition of bevacizumab-induced epithelial-mesenchymal transition by BATF2 overexpression involves the suppression of Wnt/β-catenin signaling in glioblastoma cells. Anticancer Res. 37:4285–4294. 2017.PubMed/NCBI View Article : Google Scholar

73 

Coelho BP, Fernandes CFL, Boccacino JM, Souza MCDS, Melo-Escobar MI, Alves RN, Prado MB, Iglesia RP, Cangiano G, Mazzaro GR and Lopes MH: Multifaceted WNT signaling at the crossroads between epithelial-mesenchymal transition and autophagy in glioblastoma. Front Oncol. 10(597743)2020.PubMed/NCBI View Article : Google Scholar

74 

Alkailani MI, Aittaleb M and Tissir F: WNT signaling at the intersection between neurogenesis and brain tumorigenesis. Front Mol Neurosci. 15(1017568)2022.PubMed/NCBI View Article : Google Scholar

75 

Rajakulendran N, Rowland KJ, Selvadurai HJ, Ahmadi M, Park NI, Naumenko S, Dolma S, Ward RJ, So M, Lee L, et al: Wnt and Notch signaling govern self-renewal and differentiation in a subset of human glioblastoma stem cells. Genes Dev. 33:498–510. 2019.PubMed/NCBI View Article : Google Scholar

76 

Rim EY, Clevers H and Nusse R: The Wnt pathway: From signaling mechanisms to synthetic modulators. Annu Rev Biochem. 91:571–598. 2022.PubMed/NCBI View Article : Google Scholar

77 

Sareddy GR, Pratap UP, Viswanadhapalli S, Venkata PP, Nair BC, Krishnan SR, Zheng S, Gilbert AR, Brenner AJ, Brann DW and Vadlamudi RK: PELP1 promotes glioblastoma progression by enhancing Wnt/β-catenin signaling. Neurooncol Adv. 1(vdz042)2019.PubMed/NCBI View Article : Google Scholar

78 

Latour M, Her NG, Kesari S and Nurmemmedov E: WNT signaling as a therapeutic target for glioblastoma. Int J Mol Sci. 22(8428)2021.PubMed/NCBI View Article : Google Scholar

79 

Tang C, Guo J, Chen H, Yao CJ, Zhuang DX, Wang Y, Tang WJ, Ren G, Yao Y, Wu JS, et al: Gene mutation profiling of primary glioblastoma through multiple tumor biopsy guided by 1H-magnetic resonance spectroscopy. Int J Clin Exp Pathol. 8:5327–5335. 2015.PubMed/NCBI

80 

Morris LG, Ramaswami D and Chan TA: The FAT epidemic: A gene family frequently mutated across multiple human cancer types. Cell Cycle. 12:1011–1012. 2013.PubMed/NCBI View Article : Google Scholar

81 

Fargeas CA, Lorico A and Corbeil D: Commentary: Could we address the interplay between CD133, Wnt/β-catenin, and TERT signaling pathways as a potential target for glioblastoma therapy? Front Oncol. 11(712358)2021.PubMed/NCBI View Article : Google Scholar

82 

Shevchenko V, Arnotskaya N, Zaitsev S, Sharma A, Sharma HS, Bryukhovetskiy A, Pak O, Khotimchenko Y and Bryukhovetskiy I: Proteins of Wnt signaling pathway in cancer stem cells of human glioblastoma. Int Rev Neurobiol. 151:185–200. 2020.PubMed/NCBI View Article : Google Scholar

83 

Manoranjan B, Chokshi C, Venugopal C, Subapanditha M, Savage N, Tatari N, Provias JP, Murty NK, Moffat J, Doble BW and Singh SK: A CD133-AKT-Wnt signaling axis drives glioblastoma brain tumor-initiating cells. Oncogene. 39:1590–1599. 2020.PubMed/NCBI View Article : Google Scholar

84 

Jhanwar-Uniyal M, Wainwright JV, Mohan AL, Tobias ME, Murali R, Gandhi CD and Schmidt MH: Diverse signaling mechanisms of mTOR complexes: mTORC1 and mTORC2 in forming a formidable relationship. Adv Biol Regul. 72:51–62. 2019.PubMed/NCBI View Article : Google Scholar

85 

Shahcheraghi SH, Tchokonte-Nana V, Lotfi M, Lotfi M, Ghorbani A and Sadeghnia HR: Wnt/beta-catenin and PI3K/Akt/mTOR signaling pathways in glioblastoma: Two main targets for drug design: A review. Curr Pharm Des. 26:1729–1741. 2020.PubMed/NCBI View Article : Google Scholar

86 

Liu N, Guo XH, Liu JP and Cong YS: Role of telomerase in the tumor microenvironment. Clin Exp Pharmacol Physiol. 47:357–364. 2020.PubMed/NCBI View Article : Google Scholar

87 

Abad E, Graifer D and Lyakhovich A: DNA damage response and resistance of cancer stem cells. Сancer Lett. 474:106–117. 2020.PubMed/NCBI View Article : Google Scholar

88 

Fan D, Yue Q, Chen J, Wang C, Yu R, Jin Z, Yin S, Wang Q, Chen L, Liao X, et al: Reprogramming the immunosuppressive microenvironment of IDH1 wild-type glioblastoma by blocking Wnt signaling between microglia and cancer cells. Oncoimmunology. 10(1932061)2021.PubMed/NCBI View Article : Google Scholar

89 

Hao J, Han X, Huang H, Yu X, Fang J, Zhao J, Prayson RA, Bao S and Yu JS: Sema3C signaling is an alternative activator of the canonical WNT pathway in glioblastoma. Nat Commun. 14(2262)2023.PubMed/NCBI View Article : Google Scholar

90 

Montemurro N, Pahwa B, Tayal A, Shukla A, De Jesus Encarnacion M, Ramirez I, Nurmukhametov R, Chavda V and De Carlo A: Macrophages in recurrent glioblastoma as a prognostic factor in the synergistic system of the tumor microenvironment. Neurol Int. 15:595–608. 2023.PubMed/NCBI View Article : Google Scholar

91 

Chan MK, Chung JY, Tang PC, Chan AS, Ho JY, Lin TP, Chen J, Leung KT, To KF, Lan HY and Tang PM: TGF-β signaling networks in the tumor microenvironment. Cancer Lett. 550(215925)2022.PubMed/NCBI View Article : Google Scholar

92 

Yun EJ, Kim S, Hsieh JT and Baek ST: Wnt/β-catenin signaling pathway induces autophagy-mediated temozolomide-resistance in human glioblastoma. Cell Death Dis. 11(771)2020.PubMed/NCBI View Article : Google Scholar

93 

Matias D, Predes D, Niemeyer Filho P, Lopes MC, Abreu JG, Lima FRS and Moura Neto V: Microglia-glioblastoma interactions: New role for Wnt signaling. Biochim Biophys Acta Rev Cancer. 1868:333–340. 2017.PubMed/NCBI View Article : Google Scholar

94 

Tao W, Chu C, Zhou W, Huang Z, Zhai K, Fang X, Huang Q, Zhang A, Wang X, Yu X, et al: Dual role of WISP1 in maintaining glioma stem cells and tumor-supportive macrophages in glioblastoma. Nat Commun. 11(3015)2020.PubMed/NCBI View Article : Google Scholar

95 

Bayik D and Lathia JD: Cancer stem cell-immune cell crosstalk in tumor progression. Nat Rev Cancer. 21:526–536. 2021.PubMed/NCBI View Article : Google Scholar

96 

Omuro A: Immune-checkpoint inhibitors for glioblastoma: What have we learned? Arq Neuropsiquiatr. 80 (5 Suppl 1):S266–S269. 2022.PubMed/NCBI View Article : Google Scholar

97 

Verdugo E, Puerto I and Medina MÁ: An update on the molecular biology of glioblastoma, with clinical implications and progress in its treatment. Cancer Commun (Lond). 42:1083–1111. 2022.PubMed/NCBI View Article : Google Scholar

98 

Sautter L, Hofheinz R, Tuettenberg J, Grimm M, Vajkoczy P, Groden C, Schmieder K, Hochhaus A, Wenz F and Giordano FA: Open-label phase II evaluation of imatinib in primary inoperable or incompletely resected and recurrent glioblastoma. Oncology. 98:16–22. 2020.PubMed/NCBI View Article : Google Scholar

99 

Kim JY, Jo Y, Oh HK and Kim EH: Sorafenib increases tumor treating fields-induced cell death in glioblastoma by inhibiting STAT3. Am J Cancer Res. 10:3475–3486. 2020.PubMed/NCBI

100 

Alamón C, Dávila B, García MF, Sánchez C, Kovacs M, Trias E, Barbeito L, Gabay M, Zeineh N, Gavish M, et al: Sunitinib-containing carborane pharmacophore with the ability to inhibit tyrosine kinases receptors FLT3, KIT and PDGFR-β, exhibits powerful in vivo anti-glioblastoma activity. Cancers (Basel). 12(3423)2020.PubMed/NCBI View Article : Google Scholar

101 

Reardon DA, Desjardins A, Vredenburgh JJ, O'Rourke DM, Tran DD, Fink KL, Nabors LB, Li G, Bota DA, Lukas RV, et al: Rindopepimut with bevacizumab for patients with relapsed EGFRvIII-expressing glioblastoma (ReACT): Results of a double-blind randomized phase II trial. Clin Cancer Res. 26:1586–1594. 2020.PubMed/NCBI View Article : Google Scholar

102 

Padovan M, Eoli M, Pellerino A, Rizzato S, Caserta C, Simonelli M, Michiara M, Caccese M, Anghileri E, Cerretti G, et al: Depatuxizumab mafodotin (Depatux-M) plus temozolomide in recurrent glioblastoma patients: Real-world experience from a multicenter study of italian association of neuro-oncology (AINO). Cancers (Basel). 13(2773)2021.PubMed/NCBI View Article : Google Scholar

103 

Lassman AB, Pugh SL, Wang TJC, Aldape K, Gan HK, Preusser M, Vogelbaum MA, Sulman EP, Won M, Zhang P, et al: Depatuxizumab mafodotin in EGFR-amplified newly diagnosed glioblastoma: A phase III randomized clinical trial. Neuro Oncol. 25:339–350. 2023.PubMed/NCBI View Article : Google Scholar

104 

Cloughesy T, Finocchiaro G, Belda-Iniesta C, Recht L, Brandes AA, Pineda E, Mikkelsen T, Chinot OL, Balana C, Macdonald DR, et al: Randomized, double-blind, placebo-controlled, multicenter phase II study of onartuzumab plus bevacizumab versus placebo plus bevacizumab in patients with recurrent glioblastoma: efficacy, safety, and hepatocyte growth factor and O6-methylguanine-DNA methyltransferase biomarker analyses. J Clin Oncol. 35:343–351. 2017.PubMed/NCBI View Article : Google Scholar

105 

Cloughesy TF, Drappatz J, de Groot J, Prados MD, Reardon DA, Schiff D, Chamberlain M, Mikkelsen T, Desjardins A, Ping J, et al: Phase II study of cabozantinib in patients with progressive glioblastoma: Subset analysis of patients with prior antiangiogenic therapy. Neuro Oncol. 20:259–267. 2018.PubMed/NCBI View Article : Google Scholar

106 

Zhang T, Xin Q and Kang JM: Bevacizumab for recurrent glioblastoma: A systematic review and meta-analysis. Eur Rev Med Pharmacol Sci. 25:6480–6491. 2021.PubMed/NCBI View Article : Google Scholar

107 

Wen PY, Rodon JA, Mason W, Beck JT, DeGroot J, Donnet V, Mills D, El-Hashimy M and Rosenthal M: Phase I, open-label, multicentre study of buparlisib in combination with temozolomide or with concomitant radiation therapy and temozolomide in patients with newly diagnosed glioblastoma. ESMO Open. 5(e000673)2020.PubMed/NCBI View Article : Google Scholar

108 

Rosenthal M, Clement PM, Campone M, Gil-Gil MJ, DeGroot J, Chinot O, Idbaih A, Gan H, Raizer J, Wen PY, et al: Buparlisib plus carboplatin or lomustine in patients with recurrent glioblastoma: A phase Ib/II, open-label, multicentre, randomised study. ESMO Open. 5(e000672)2020.PubMed/NCBI View Article : Google Scholar

109 

Kaley TJ, Panageas KS, Pentsova EI, Mellinghoff IK, Nolan C, Gavrilovic I, DeAngelis LM, Abrey LE, Holland EC, Omuro A, et al: Phase I clinical trial of temsirolimus and perifosine for recurrent glioblastoma. Ann Clin Transl Neurol. 7:429–436. 2020.PubMed/NCBI View Article : Google Scholar

110 

Schiff D, Jaeckle KA, Anderson SK, Galanis E, Giannini C, Buckner JC, Stella P, Flynn PJ, Erickson BJ, Schwerkoske JF, et al: Phase 1/2 trial of temsirolimus and sorafenib in the treatment of patients with recurrent glioblastoma: North central cancer treatment group study/alliance N0572. Cancer. 124:1455–1463. 2018.PubMed/NCBI View Article : Google Scholar

111 

Lin Z, Xu H, Yang R, Li Z, Zheng H, Zhang Z, Peng J, Zhang X, Qi S, Liu Y and Huang G: Effective treatment of a BRAF V600E-mutant epithelioid glioblastoma patient by vemurafenib: a case report. Anticancer Drugs. 33:100–104. 2022.PubMed/NCBI View Article : Google Scholar

112 

Herrera-Rios D, Li G, Khan D, Tsiampali J, Nickel AC, Aretz P, Hewera M, Suwala AK, Jiang T, Steiger HJ, et al: A computational guided, functional validation of a novel therapeutic antibody proposes Notch signaling as a clinical relevant and druggable target in glioma. Sci Rep. 10(16218)2020.PubMed/NCBI View Article : Google Scholar

113 

Bogdahn U, Hau P, Stockhammer G, Venkataramana NK, Mahapatra AK, Suri A, Balasubramaniam A, Nair S, Oliushine V, Parfenov V, et al: Targeted therapy for high-grade glioma with the TGF-β2 inhibitor trabedersen: Results of a randomized and controlled phase IIb study. Neuro Oncol. 13:132–142. 2011.PubMed/NCBI View Article : Google Scholar

114 

Hadizadeh M, AminJafari A, Parvizpour S and Ghasemi S: Novel targets to overcome antiangiogenesis therapy resistance in glioblastoma multiforme: Systems biology approach and suggestion of therapy by galunisertib. Cell Biol Int. 46:1649–1660. 2022.PubMed/NCBI View Article : Google Scholar

115 

Wick A, Desjardins A, Suarez C, Forsyth P, Gueorguieva I, Burkholder T, Cleverly AL, Estrem ST, Wang S, Lahn MM, et al: Phase 1b/2a study of galunisertib, a small molecule inhibitor of transforming growth factor-beta receptor I, in combination with standard temozolomide-based radiochemotherapy in patients with newly diagnosed malignant glioma. Invest New Drugs. 38:1570–1579. 2020.PubMed/NCBI View Article : Google Scholar

116 

Hsu SY, Lee SC, Liu HC, Peng SF, Chueh FS, Lu TJ, Lee HT and Chou YC: Phenethyl isothiocyanate suppresses the proinflammatory cytokines in human glioblastoma cells through the PI3K/Akt/NF-κB signaling pathway in vitro. Oxid Med Cell Longev. 2022(2108289)2022.PubMed/NCBI View Article : Google Scholar

117 

Li S, He Y, Chen K, Sun J, Zhang L, He Y, Yu H and Li Q: RSL3 drives ferroptosis through NF-κB pathway activation and GPX4 depletion in glioblastoma. Oxid Med Cell Longev. 2021(2915019)2021.PubMed/NCBI View Article : Google Scholar

118 

Volmar MNM, Cheng J, Alenezi H, Richter S, Haug A, Hassan Z, Goldberg M, Li Y, Hou M, Herold-Mende C, et al: Cannabidiol converts NF-κB into a tumor suppressor in glioblastoma with defined antioxidative properties. Neuro Oncol. 23:1898–1910. 2021.PubMed/NCBI View Article : Google Scholar

119 

Navone SE, Guarnaccia L, Cordiglieri C, Crisà FM, Caroli M, Locatelli M, Schisano L, Rampini P, Miozzo M, La Verde N, et al: Aspirin affects tumor angiogenesis and sensitizes human glioblastoma endothelial cells to temozolomide, bevacizumab, and sunitinib, impairing vascular endothelial growth factor-related signaling. World Neurosurg. 120:e380–e391. 2018.PubMed/NCBI View Article : Google Scholar

120 

Kast RE: Adding high-dose celecoxib to increase effectiveness of standard glioblastoma chemoirradiation. Ann Pharm Fr. 79:481–488. 2021.PubMed/NCBI View Article : Google Scholar

121 

Yin D, Jin G, He H, Zhou W, Fan Z, Gong C, Zhao J and Xiong H: Celecoxib reverses the glioblastoma chemo-resistance to temozolomide through mitochondrial metabolism. Aging (Albany NY). 13:21268–21282. 2021.PubMed/NCBI View Article : Google Scholar

122 

Pantovic A, Bosnjak M, Arsikin K, Kosic M, Mandic M, Ristic B, Tosic J, Grujicic D, Isakovic A, Micic N, et al: In vitro antiglioma action of indomethacin is mediated via AMP-activated protein kinase/mTOR complex 1 signalling pathway. Int J Biochem Cell Biol. 83:84–96. 2017.PubMed/NCBI View Article : Google Scholar

123 

Allani SK, Weissbach H and Lopez Toledano MA: Sulindac induces differentiation of glioblastoma stem cells making them more sensitive to oxidative stress. Neoplasma. 65:376–388. 2018.PubMed/NCBI View Article : Google Scholar

124 

Qiu J, Shi Z and Jiang J: Cyclooxygenase-2 in glioblastoma multiforme. Drug Discov Today. 22:148–156. 2017.PubMed/NCBI View Article : Google Scholar

125 

Li H, Jiao S, Li X, Banu H, Hamal S and Wang X: Therapeutic effects of antibiotic drug tigecycline against cervical squamous cell carcinoma by inhibiting Wnt/β-catenin signaling. Biochem Biophys Res Commun. 467:14–20. 2015.PubMed/NCBI View Article : Google Scholar

126 

Wieland A, Trageser D, Gogolok S, Reinartz R, Höfer H, Keller M, Leinhaas A, Schelle R, Normann S, Klaas L, et al: Anticancer effects of niclosamide in human glioblastoma. Clin Cancer Res. 19:4124–4136. 2013.PubMed/NCBI View Article : Google Scholar

127 

Li H, Liu S, Jin R, Xu H, Li Y, Chen Y and Zhao G: Pyrvinium pamoate regulates MGMT expression through suppressing the Wnt/β-catenin signaling pathway to enhance the glioblastoma sensitivity to temozolomide. Cell Death Discov. 7(288)2021.PubMed/NCBI View Article : Google Scholar

128 

Liu Y, Fang S, Sun Q and Liu B: Anthelmintic drug ivermectin inhibits angiogenesis, growth and survival of glioblastoma through inducing mitochondrial dysfunction and oxidative stress. Biochem Biophys Res Commun. 480:415–421. 2016.PubMed/NCBI View Article : Google Scholar

129 

Hajikhani B, Nasiri MJ, Hosseini SS, Khalili F, Karimi-Yazdi M, Hematian A, Nojookambari NY, Goudarzi M, Dadashi M and Mirsaeidi M: Clofazimine susceptibility testing of Mycobacterium avium complex and Mycobacterium abscessus: A meta-analysis study. J Glob Antimicrob Resist. 26:188–193. 2021.PubMed/NCBI View Article : Google Scholar

130 

Park L, Wallace CE, Vasile G and Buckley C: A case of lepromatous leprosy with erythema nodosum leprosum. Cureus. 15(e33846)2023.PubMed/NCBI View Article : Google Scholar

131 

Ahmed K, Koval A, Xu J, Bodmer A and Katanaev VL: Towards the first targeted therapy for triple-negative breast cancer: Repositioning of clofazimine as a chemotherapy-compatible selective Wnt pathway inhibitor. Cancer Lett. 449:45–55. 2019.PubMed/NCBI View Article : Google Scholar

132 

Xu J, Koval A and Katanaev VL: Beyond TNBC: Repositioning of clofazimine against a broad range of Wnt-dependent cancers. Front Oncol. 10(602817)2020.PubMed/NCBI View Article : Google Scholar

133 

Keswani RK, Tian C, Peryea T, Girish G, Wang X and Rosania GR: Repositioning clofazimine as a macrophage-targeting photoacoustic contrast agent. Sci Rep. 6(23528)2016.PubMed/NCBI View Article : Google Scholar

134 

Trexel J, Yoon GS, Keswani RK, McHugh C, Yeomans L, Vitvitsky V, Banerjee R, Sud S, Sun Y, Rosania GR and Stringer KA: Macrophage-mediated clofazimine sequestration is accompanied by a shift in host energy metabolism. J Pharm Sci. 106:1162–1174. 2017.PubMed/NCBI View Article : Google Scholar

135 

Yoon GS, Sud S, Keswani RK, Baik J, Standiford TJ, Stringer KA and Rosania GR: Phagocytosed clofazimine biocrystals can modulate innate immune signaling by inhibiting TNFα and boosting IL-1RA secretion. Mol Pharm. 12:2517–2527. 2015.PubMed/NCBI View Article : Google Scholar

136 

Geribaldi-Doldán N, Fernández-Ponce C, Quiroz RN, Sánchez-Gomar I, Escorcia LG, Velásquez EP and Quiroz EN: The role of microglia in glioblastoma. Front Oncol. 10(603495)2021.PubMed/NCBI View Article : Google Scholar

137 

Castellani G, Croese T, Peralta Ramos JM and Schwartz M: Transforming the understanding of brain immunity. Science. 380(eabo7649)2023.PubMed/NCBI View Article : Google Scholar

138 

Rustenhoven J and Kipnis J: Brain borders at the central stage of neuroimmunology. Nature. 612:417–429. 2022.PubMed/NCBI View Article : Google Scholar : De Leo A, Ugolini A and Veglia F: Myeloid cells in glioblastoma microenvironment. Cells 10: 18, 2020.

139 

Lewellis SW and Knaut H: Attractive guidance: How the chemokine SDF1/CXCL12 guides different cells to different locations. Semin Cell Dev Biol. 23:333–340. 2012.PubMed/NCBI View Article : Google Scholar

140 

Giordano FA, Link B, Glas M, Herrlinger U, Wenz F, Umansky V, Brown JM and Herskind C: Targeting the post-irradiation tumor microenvironment in glioblastoma via inhibition of CXCL12. Cancers (Basel). 11(272)2019.PubMed/NCBI View Article : Google Scholar

141 

Wang S, Chen C, Li J, Xu X, Chen W and Li F: The CXCL12/CXCR4 axis confers temozolomide resistance to human glioblastoma cells via up-regulation of FOXM1. J Neurol Sci. 414(116837)2020.PubMed/NCBI View Article : Google Scholar

142 

Dolina JS, Van Braeckel-Budimir N, Thomas GD and Salek-Ardakani S: CD8+ T cell exhaustion in cancer. Front Immunol. 12(715234)2021.PubMed/NCBI View Article : Google Scholar

143 

Belk JA, Daniel B and Satpathy AT: Epigenetic regulation of T cell exhaustion. Nat Immunol. 23:848–860. 2022.PubMed/NCBI View Article : Google Scholar

144 

Yang T, Kong Z and Ma W: PD-1/PD-L1 immune checkpoint inhibitors in glioblastoma: Clinical studies, challenges and potential. Hum Vaccin Immunother. 17:546–553. 2021.PubMed/NCBI View Article : Google Scholar

145 

Arrieta VA, Dmello C, McGrail DJ, Brat DJ, Lee-Chang C, Heimberger AB, Chand D, Stupp R and Sonabend AM: Immune checkpoint blockade in glioblastoma: From tumor heterogeneity to personalized treatment. J Clin Invest. 133(e163447)2023.PubMed/NCBI View Article : Google Scholar

146 

Caramanna I, de Kort JM, Brandes AA, Taal W, Platten M, Idbaih A, Frenel JS, Wick W, Preetha CJ, Bendszus M, et al: Corticosteroids use and neurocognitive functioning in patients with recurrent glioblastoma: Evidence from European organization for research and treatment of cancer (EORTC) trial 26101. Neurooncol Pract. 9:310–316. 2022.PubMed/NCBI View Article : Google Scholar

147 

Pitter KL, Tamagno I, Alikhanyan K, Hosni-Ahmed A, Pattwell SS, Donnola S, Dai C, Ozawa T, Chang M, Chan TA, et al: Corticosteroids compromise survival in glioblastoma. Brain. 139:1458–1471. 2016.PubMed/NCBI View Article : Google Scholar

148 

Klement RJ and Champ CE: Corticosteroids compromise survival in glioblastoma in part through their elevation of blood glucose levels. Brain. 140(e16)2017.PubMed/NCBI View Article : Google Scholar

149 

Yovino S, Kleinberg L, Grossman SA, Narayanan M and Ford E: The etiology of treatment-related lymphopenia in patients with malignant gliomas: Modeling radiation dose to circulating lymphocytes explains clinical observations and suggests methods of modifying the impact of radiation on immune cells. Cancer Invest. 31:140–144. 2013.PubMed/NCBI View Article : Google Scholar

150 

Kut C and Kleinberg L: Radiotherapy, lymphopenia and improving the outcome for glioblastoma: A narrative review. Chin Clin Oncol. 12(4)2023.PubMed/NCBI View Article : Google Scholar

151 

Saito T, Sugiyama K, Hama S, Yamasaki F, Takayasu T, Nosaka R, Muragaki Y, Kawamata T and Kurisu K: Prognostic importance of temozolomide-induced neutropenia in glioblastoma, IDH-wildtype patients. Neurosurg Rev. 41:621–628. 2018.PubMed/NCBI View Article : Google Scholar

152 

Aboody KS, Brown A, Rainov NG, Bower KA, Liu S, Yang W, Small JE, Herrlinger U, Ourednik V, Black PM, et al: Neural stem cells display extensive tropism for pathology in adult brain: Evidence from intracranial gliomas. Proc Natl Acad Sci USA. 97:12846–12851. 2000.PubMed/NCBI View Article : Google Scholar

153 

Snyder EY, Park KI, Flax JD, Liu S, Rosario CM, Yandava BD and Aurora S: Potential of neural ‘stem-like’ cells for gene therapy and repair of the degenerating central nervous system. Adv Neurol. 72:121–132. 1997.PubMed/NCBI

154 

Bryukhovetskiy IS, Mischenko PV, Tolok EV, Zaitcev SV, Khotimchenko YS and Bryukhovetskiy AS: Directional migration of adult hematopoeitic progenitors to C6 glioma in vitro. Oncol Lett. 9:1839–1844. 2015.PubMed/NCBI View Article : Google Scholar

155 

Hass R, von der Ohe J and Dittmar T: Hybrid formation and fusion of cancer cells in vitro and in vivo. Cancers (Basel). 13(4496)2021.PubMed/NCBI View Article : Google Scholar

156 

Goldenberg DM: Horizontal transmission of malignancy by cell-cell fusion. Expert Opin Biol Ther. 12 (Suppl 1):S133–S139. 2012.PubMed/NCBI View Article : Google Scholar

157 

Sun Z, Wang L, Zhou Y, Dong L, Ma W, Lv L, Zhang J and Wang X: Glioblastoma stem cell-derived exosomes enhance stemness and tumorigenicity of glioma cells by transferring Notch1 protein. Cell Mol Neurobiol. 40:767–784. 2020.PubMed/NCBI View Article : Google Scholar

158 

Khan F, Pang L, Dunterman M, Lesniak MS, Heimberger AB and Chen P: Macrophages and microglia in glioblastoma: Heterogeneity, plasticity, and therapy. J Clin Invest. 133(e163446)2023.PubMed/NCBI View Article : Google Scholar

159 

Wang G, Zhong K, Wang Z, Zhang Z, Tang X, Tong A and Zhou L: Tumor-associated microglia and macrophages in glioblastoma: From basic insights to therapeutic opportunities. Front Immunol. 13(964898)2022.PubMed/NCBI View Article : Google Scholar

160 

Bryukhovetskiy IS, Mischenko PV, Tolok EV, Zaitcev SV, Khotimchenko YS and Bryukhovetskiy AS: Directional migration of adult hematopoeitic progenitors to C6 glioma in vitro. Oncol Lett. 9:1839–1844. 2015.PubMed/NCBI View Article : Google Scholar

161 

Lee-Six H, Øbro NF, Shepherd MS, Grossmann S, Dawson K, Belmonte M, Osborne RJ, Huntly BJP, Martincorena I, Anderson E, et al: Population dynamics of normal human blood inferred from somatic mutations. Nature. 561:473–478. 2018.PubMed/NCBI View Article : Google Scholar

162 

Jaiswal S: Clonal hematopoiesis and nonhematologic disorders. Blood. 136:1606–1614. 2020.PubMed/NCBI View Article : Google Scholar

163 

Kast RE, Hill QA, Wion D, Mellstedt H, Focosi D, Karpel-Massler G, Heiland T and Halatsch ME: Glioblastoma-synthesized G-CSF and GM-CSF contribute to growth and immunosuppression: Potential therapeutic benefit from dapsone, fenofibrate, and ribavirin. Tumour Biol. 39(1010428317699797)2017.PubMed/NCBI View Article : Google Scholar

164 

Bryukhovetskiy I, Manzhulo I, Mischenko P, Milkina E, Dyuizen I, Bryukhovetskiy A and Khotimchenko Y: Cancer stem cells and microglia in the processes of glioblastoma multiforme invasive growth. Oncol Lett. 12:1721–1728. 2016.PubMed/NCBI View Article : Google Scholar

165 

Zaitsev S, Sharma HS, Sharma A, Manzhulo I, Polevshchikov A, Kudriavtsev I, Khotimchenko Y, Pak O, Bryukhovetskiy A and Bryukhovetskiy I: Pro-inflammatory modification of cancer cells microsurroundings increases the survival rates for rats with low differentiated malignant glioma of brain. Int Rev Neurobiol. 151:253–279. 2020.PubMed/NCBI View Article : Google Scholar

166 

Choe JH, Watchmaker PB, Simic MS, Gilbert RD, Li AW, Krasnow NA, Downey KM, Yu W, Carrera DA, Celli A, et al: SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med. 13(eabe7378)2021.PubMed/NCBI View Article : Google Scholar

167 

Meister H, Look T, Roth P, Pascolo S, Sahin U, Lee S, Hale BD, Snijder B, Regli L, Ravi VM, et al: Multifunctional mRNA-based CAR T cells display promising antitumor activity against glioblastoma. Clin Cancer Res. 28:4747–4756. 2022.PubMed/NCBI View Article : Google Scholar

168 

Maggs L, Cattaneo G, Dal AE, Moghaddam AS and Ferrone S: CAR T cell-based immunotherapy for the treatment of glioblastoma. Front Neurosci. 15(662064)2021.PubMed/NCBI View Article : Google Scholar

169 

Wang D, Starr R, Chang WC, Aguilar B, Alizadeh D, Wright SL, Yang X, Brito A, Sarkissian A, Ostberg JR, et al: Chlorotoxin-directed CAR T cells for specific and effective targeting of glioblastoma. Sci Transl Med. 12(eaaw2672)2020.PubMed/NCBI View Article : Google Scholar

170 

Agliardi G, Liuzzi AR, Hotblack A, De Feo D, Núñez N, Stowe CL, Friebel E, Nannini F, Rindlisbacher L, Roberts TA, et al: Intratumoral IL-12 delivery empowers CAR-T cell immunotherapy in a pre-clinical model of glioblastoma. Nat Commun. 12(444)2021.PubMed/NCBI View Article : Google Scholar

171 

Brown CE, Rodriguez A, Palmer J, Ostberg JR, Naranjo A, Wagner JR, Aguilar B, Starr R, Weng L, Synold TW, et al: Off-the-shelf, steroid-resistant, IL13Rα2-specific CAR T cells for treatment of glioblastoma. Neuro Oncol. 24:1318–1330. 2022.PubMed/NCBI View Article : Google Scholar

172 

Wang G, Zhang Z, Zhong K, Wang Z, Yang N, Tang X, Li H, Lu Q, Wu Z, Yuan B, et al: CXCL11-armed oncolytic adenoviruses enhance CAR-T cell therapeutic efficacy and reprogram tumor microenvironment in glioblastoma. Mol Ther. 31:134–153. 2023.PubMed/NCBI View Article : Google Scholar

173 

Ghouzlani A, Kandoussi S, Tall M, Reddy KP, Rafii S and Badou A: Immune checkpoint inhibitors in human glioma microenvironment. Front Immunol. 12(679425)2021.PubMed/NCBI View Article : Google Scholar

174 

Gatto L, Franceschi E, Di Nunno V, Maggio I, Lodi R and Brandes AA: Engineered CAR-T and novel CAR-based therapies to fight the immune evasion of glioblastoma: Gutta cavat lapidem. Expert Rev Anticancer Ther. 21:1333–1353. 2021.PubMed/NCBI View Article : Google Scholar

175 

Bryukhovetskiy I: Cell-based immunotherapy of glioblastoma multiforme. Oncol Lett. 23(133)2022.PubMed/NCBI View Article : Google Scholar

176 

Baharom F, Ramirez-Valdez RA, Khalilnezhad A, Khalilnezhad S, Dillon M, Hermans D, Fussell S, Tobin KKS, Dutertre CA, Lynn GM, et al: Systemic vaccination induces CD8+ T cells and remodels the tumor microenvironment. Cell. 185:4317–4332.e15. 2022.PubMed/NCBI View Article : Google Scholar

177 

Liau LM, Ashkan K, Brem S, Campian JL, Trusheim JE, Iwamoto FM, Tran DD, Ansstas G, Cobbs CS, Heth JA, et al: Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma: A phase 3 prospective externally controlled cohort trial. JAMA Oncol. 9:112–121. 2023.PubMed/NCBI View Article : Google Scholar

178 

Keskin DB, Anandappa AJ, Sun J, Tirosh I, Mathewson ND, Li S, Oliveira G, Giobbie-Hurder A, Felt K, Gjini E, et al: Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 565:234–239. 2019.PubMed/NCBI View Article : Google Scholar

179 

Chen KS, Reinshagen C, Van Schaik TA, Rossignoli F, Borges P, Mendonca NC, Abdi R, Simon B, Reardon DA, Wakimoto H and Shah K: Bifunctional cancer cell-based vaccine concomitantly drives direct tumor killing and antitumor immunity. Sci Transl Med. 15(eabo4778)2023.PubMed/NCBI View Article : Google Scholar

180 

Wang J, Weiss T, Neidert MC, Toussaint NC, Naghavian R, Sellés Moreno C, Foege M, Tomas Ojer P, Medici G, Jelcic I, et al: Vaccination with designed neopeptides induces intratumoral, cross-reactive CD4+ T-cell responses in glioblastoma. Clin Cancer Res. 28:5368–5382. 2022.PubMed/NCBI View Article : Google Scholar

181 

Yu TW, Chueh HY, Tsai CC, Lin CT and Qiu JT: Novel GM-CSF-based vaccines: One small step in GM-CSF gene optimization, one giant leap for human vaccines. Hum Vaccin Immunother. 12:3020–3028. 2016.PubMed/NCBI View Article : Google Scholar

182 

Li L, Zhou J, Dong X, Liao Q, Zhou D and Zhou Y: Dendritic cell vaccines for glioblastoma fail to complete clinical translation: Bottlenecks and potential countermeasures. Int Immunopharmacol. 109(108929)2022.PubMed/NCBI View Article : Google Scholar

183 

Liau LM, Ashkan K, Tran DD, Campian JL, Trusheim JE, Cobbs CS, Heth JA, Salacz M, Taylor S, D'Andre SD, et al: First results on survival from a large phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J Transl Med. 16(142)2018.PubMed/NCBI View Article : Google Scholar

184 

Zhu P, Li SY, Ding J, Fei Z, Sun SN, Zheng ZH, Wei D, Jiang J, Miao JL, Li SZ, et al: Combination immunotherapy of glioblastoma with dendritic cell cancer vaccines, anti-PD-1 and poly I:C. J Pharm Anal. 13:616–624. 2023.PubMed/NCBI View Article : Google Scholar

185 

Medikonda R, Dunn G, Rahman M, Fecci P and Lim M: A review of glioblastoma immunotherapy. J Neurooncol. 151:41–53. 2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

February-2024
Volume 20 Issue 2

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kosianova А, Pak O and Bryukhovetskiy I: Regulation of cancer stem cells and immunotherapy of glioblastoma (Review). Biomed Rep 20: 24, 2024
APA
Kosianova, А., Pak, O., & Bryukhovetskiy, I. (2024). Regulation of cancer stem cells and immunotherapy of glioblastoma (Review). Biomedical Reports, 20, 24. https://doi.org/10.3892/br.2023.1712
MLA
Kosianova, А., Pak, O., Bryukhovetskiy, I."Regulation of cancer stem cells and immunotherapy of glioblastoma (Review)". Biomedical Reports 20.2 (2024): 24.
Chicago
Kosianova, А., Pak, O., Bryukhovetskiy, I."Regulation of cancer stem cells and immunotherapy of glioblastoma (Review)". Biomedical Reports 20, no. 2 (2024): 24. https://doi.org/10.3892/br.2023.1712