IMR Press / FBL / Volume 28 / Issue 12 / DOI: 10.31083/j.fbl2812362
Open Access Review
Cancer Stem Cell Biomarkers in the Nervous System
Show Less
1 College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 100044 Beijing, China
2 Chinese Research Academy of Environmental Sciences, 100012 Beijing, China
*Correspondence: xuhanli@bjtu.edu.cn (Hanli Xu); fan.jingpu@craes.org.cn (Jingpu Fan)
Front. Biosci. (Landmark Ed) 2023, 28(12), 362; https://doi.org/10.31083/j.fbl2812362
Submitted: 29 June 2023 | Revised: 11 October 2023 | Accepted: 25 October 2023 | Published: 29 December 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Cancer stem cells (CSCs) have been increasingly recognized in recent years. CSCs from human neural tumors are one of the root causes of metastatic tumor progression, therapeutic resistance and recurrence. However, there is a lack of comprehensive literature that systematically consolidates the biomarkers specific to CSCs in neurological cancers. Therefore, this review provides a comprehensive summary of cancer stem cell (CSC) biomarkers for neurological tumors such as glioma, meningioma, medulloblastoma and neurofibroma. It also points out the possible functions of these biomarkers in diagnosis, treatment and prognosis, providing a broader perspective. First, we quantitatively screened key words such as CSCs, biomarkers, and expression by bibliometric analysis and clarified the intrinsic connections between the key words. Then, we describe the CSC biomarkers of major neurological tumors and their pathway mechanisms, and provide an in-depth analysis of the commonalities and differences with the biomarkers of non-CSCs. In addition, many studies have shown that antipsychotic drugs can inhibit tumor growth and reduce the expression of CSC biomarkers, which facilitates targeted therapy against tumors in the nervous system. Therefore, this study will focus on the biomarkers of CSCs in the nervous system, hoping to provide guidance for future in-depth exploration and monitoring of neurological tumors for clinical applications.

Keywords
cancer stem cell
neurological tumors
biomarker
nervous system
1. Introduction

Cancer is still one of the major causes of death worldwide, with cancer stem cells (CSCs) being a particularly complex component [1, 2]. In the WHO Classification of Tumors of the Central Nervous System 2021, the tumors covered include glioma, meningioma, medulloblastoma and pituitary tumors [3]. However, tumors related to the peripheral nervous system include neurofibromas and schwannomas [4]. The incidence of each tumor in the nervous system varies widely depending on the histology. Incidence rates were highest for neurofibroma (1/3000), meningioma (7.44 per 100,000), glioblastomas (3.19 per 100,000) and medulloblastoma (0.71 per 100,000) [5, 6]. As one of the major tumor types, glioma is highly malignant and has a poor prognosis even after the application of comprehensive treatment. Recent studies have shown that glioblastoma has surpassed pancreatic cancer and liver cancer as the most difficult to treat tumors [7]. Therefore, it is of great interest to investigate CSCs in the nervous system.

Stem cells are able to differentiate into any cell of an organism and have the ability of self-renewal [8]. Among them, CSCs are a small population of stem cells, and are one of the underlying causes of highly metastatic, recurrence-prone and untreatable cancers. Not only can they give rise to all cell types present in the tumor tissue from which they originate through cell differentiation, they can also drive tumorigenesis and cause recurrence [9, 10, 11]. CSC biomarkers can indicate the malignancy of tumors and respond to the development of tumors. Certain molecules are specifically expressed in CSCs or cancer stem cell-like cells [9]. These natural growth molecules can be detected in body fluids or tissues, thus acting as biomarkers to identify CSC and monitor treatment effectiveness, and used to isolate cell populations with CSC characteristics [12]. However, because CSC biomarkers are not universally expressed across tumor types, one marker cannot be used for all tumors [13]. The diagnosis, treatment and prognosis of the same tumor may also involve different biomarkers [12]. Therefore, classification studies on CSC biomarkers of different tumors are of great interest for monitoring and treating tumors.

A large number of biomarkers have been identified in the nervous system. However, there are relatively fewer CSC biomarkers. Moreover, most studies have concentrated on a single CSC biomarker rather than multiple biomarkers that may be more reliable for CSC identification. Furthermore, given the intrinsic connection between the nervous system and mental illness, there may be a potential function of drugs applied to mental illness on neurological CSCs [14]. To address the above three shortcomings, this research aims to review CSC biomarkers for their diagnostic and prognostic potential in several neurological tumors, including glioma, medulloblastoma, meningioma, and neurofibromas.

2. Bibliometric Analysis

An authorship-based bibliometric map showed that neurological CSC biomarker research tends to be in “small teams” (Fig. 1A). Meanwhile, each team formed a self-completeness academic community, and the mutual communication between each community was much less than that within the community. It can often be self-sufficient and has less interaction with the outside world. However, neurological CSC marker research involves a complicated process, which is difficult for an isolated group to generate systematic data. Team collaboration to achieve an aggregation effect on CSC biomarker research will be highly appreciated.

Fig. 1.

Content of bibliometric analysis. (A) Authorship-based bibliometric map of the studies. (B) Co-occurrences of the keywords based on the full counting method. The strength of the links was normalized by the association strength method and the weight of keywords refers to the total link strength. Only the links with a minimum strength of three are displayed.

The word frequency analysis on neurological CSC (Fig. 1B) suggests that most of the literature focused on hotspot information about stem cells, such as expression, glioma, biomarker, and prognosis. Expression clustered with the words gene, proliferation, tumor, and overexpression, predicts that overexpression of biomarkers such as genes can inhibit tumor cell metastasis and proliferation. It also induces apoptosis and reduces the stemness of CSCs. The high association of CSCs with words such as recognition, invasion, and epithelial-mesenchymal transition (EMT) suggests that CSC formation is related to EMT and that identification and detection of CSCs by flow cytometry methods are also important. EMT is a key process in CSC formation. In this process, epithelial cells acquire the characteristics of mesenchymal cells, with enhanced cell invasion and migration ability, and allow the cells to acquire properties such as self-renewal. The co-occurrence of the terms glioma and glioblastoma, CD133, and growth indicates that most of the research on glioma, one of the major tumor species, has focused on glioblastoma. It also predicts a correlation between the formation of the biomarker CD133 and the growth of the tumor. A careful reading of the literature also confirms that glycosylation of CD133 maintains the self-renewal of CSCs and tumor growth and promotes the ability of glioma stem cells (GSCs) to resist chemotherapy. In addition, the words prognosis, meningioma, DNA methylation, and survival time were clustered into one category, indicating that the study of prognostic markers mainly focused on meningioma and that the expression level of prognostic markers correlated with postoperative survival time. Further studies showed that the differences in DNA methylation among meningioma patients led to different clinical prognostic characteristics. The expression levels of prognostic markers were significantly correlated with postoperative survival time. If the expression of markers was positively correlated with prognosis, increased markers indicated a good prognosis and a longer postoperative survival time for the patients. In conclusion, the primary subjects for most of the literature are humans versus animals. By constructing xenograft models derived from cell lines with biomarkers, the investigators found that they were more malignant and aggressive, exhibiting a strong stemness phenotype.

3. Functions and Pathways Involved in Stem Cell Biomarkers of Tumor Species
3.1 Characteristics of the Tumor Species

Glioma is the most common primary intracranial tumor, accounting for nearly 80% of malignant brain tumors, with a high mortality and malignancy rate [15]. According to the WHO 2007 pathological criteria, gliomas can be classified into four grades according to their malignancy: grade I is Fibrillary astrocytoma, grade II is Diffuse astrocytoma, grade III is Anaplastic astrocytoma, and grade IV is Glioblastoma multiform (GBM) [16]. The WHO 2016 and 2021 pathology criteria are refined according to molecular typing and age stage. Among them, grade I and II gliomas are called low-grade malignant astrocytoma, grade III and IV gliomas are malignant glioma cells, and grade III gliomas are prone to progress to grade IV GBM [3, 17, 18]. As a more aggressive tumor, GBM is the most malignant, with a recurrence rate of 100% and an average survival time of less than 12 months in most cases [19, 20]. Medulloblastoma (MB) is one of the most common malignant brain tumors in children, accounting for approximately 10% of all tumor deaths in young infants [21]. After standard treatment, approximately 1/3 of MB patients experience recurrence, and the probability of long-term survival after recurrence is low [22]. Current genetically-defined categories include WNT-activated, SHH-activated TP53 wildtype, SHH-activated TP53-mutant, and non-WNT/non-SHH [23]. Meningioma (MG) is a common intracranial and intraspinal neoplasm accounting for 25–30% of all primary neurological tumors, and its postoperative recurrence rate often ranges from 13% to 40% [24, 25]. The WHO classification system describes 15 different MG subtypes, 9 of which are allotted WHO grade I, 3 WHO grade II, and 1 WHO grade III. The risk of recurrence is relatively low for WHO grade I meningiomas, but WHO grade III meningiomas are characterized by rapid growth, early recurrence, and a risk of systemic metastasis [26]. There are three types of neurofibromas: neurofibromatosis type 1 (NF1), accounting for 96% of all cases; neurofibromatosis type 2 (NF2), accounting for 3%; and schwannomatosis, accounting for <1% [27]. NF1 is one of the most common human monogenic diseases, with a prevalence of 1 in 3000, and consists mainly of Schwann cells [28] (Table 1).

Table 1.Overview of cancer stem cell (CSC) biomarkers for neurological tumors.
Tumors of the nervous system Morbidity Recurrence rate Gene expression Protein expression Pathway(s)
Glioma 6.02/100,000 100% (in Glioblastoma) NADH, CD133, KLF4, SOX2, ALDH1, NANOG, MYC, ABCG2, HOXB5, BRAP Wnt/β-linked protein, TGFβ/PI3K/AKT/mTOR, STAT3, NF-κB
Medulloblastoma 0.71/100,000 1/3 Sox2, Klf4, Nestin, Bmi1 CD133, CD15, Nestin, CD24, SOX2 PI3K/AKT, MAPK
Meningioma 7.44/100,000 13% to 40% NANOG, SOX2
Neurofibroma 1/3000 PROM1, NKX2-2 CD68, OCT3/4, NANOG, SOX2 PI3K/AKT/GSK3β, Wnt

Notes: Proteins are in normal font, genes are in italics.

3.2 CSC Biomarkers
3.2.1 Glioma

Biomarkers for glioma CSC diagnosis include NADH (Table 2, Ref. [24, 27, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41]). Yuan et al. [29] found that glioma cells demonstrate stem cell-like properties characteristic of GSCs at NADHhigh autofluorescence, so the autofluorescence of NADH can be an appropriate marker to sort GSCs. Biomarkers of glioma CSC treatment are CD133, ALDH1, KLF4, SOX2, NANOG, MYC, ABCG2, Nestin and Msi-1 [30, 31, 32, 33]. Tian et al. [32] demonstrated that the BET inhibitor ZBC260 inhibits tumor growth and GSC-like properties through the Wnt/β-linked protein pathway. The main biomarkers of stem cells such as aldehyde dehydrogenase 1 (ALDH1), KLF4, SOX2, NANOG, MYC and ABCG2 are significantly reduced with increasing levels of ZBC260 to achieve therapeutic effects. Moreover, Wei et al. [33] demonstrated that low expression of MAN1A1 results in the formation of a high-mannose type N-glycan on CD133 in GSCs. This interaction with DNMT1 facilitates GSC self-renewal and tumorigenesis, suggesting a potential therapeutic approach. Several studies have identified key molecular targets for inhibiting GSC-like traits. These findings suggest promising avenues for developing therapies to combat GSC-like traits in glioma. The expression levels of CD133, IL6, TGF-β, BRAP, HOXB5 and GAS5 are prognostic biomarkers in glioma patients [31, 34, 35, 36]. Liu et al. [31] found a positive correlation between the expression of CD133, a CSC biomarker for glioma, and WHO tumor grade. Conversely, CD133 expression is negatively correlated with prognosis. Its elevation in glioma tissues predicts poor prognosis in glioma patients, while regulatory T cells increase IL6 expression by secreting TGF-β to activate NF-κB signaling, and IL6 then contributes to GSC stemness by activating the STAT3 signaling pathway. Wang et al. [34] discovered that the expression of BRAP is linked to favorable outcomes in individuals with glioma. BRAP hinders the proliferation and migration of glioma cells, as well as the self-renewal of GSCs, by targeting the TGF-β/PI3K/AKT/mTOR signaling pathway. These findings suggest that BRAP could serve as a prognostic marker for glioma patients. Wu et al. [35] showed that GAS5 overexpression reduces cell viability, inhibits the migration of GBM cells, and impairs the stemness of GSCs through miR-let-125e and miR-6a. Moreover, GAS5 expression levels are significantly correlated with postoperative survival time and reduced in high-grade glioma tissues and cells, making it a prognostic biomarker. Zhao et al. [36] found that HOXB5 is overexpressed in gliomas, which correlates with poor patient survival and regulates the proliferation of GSCs. Functionally, they confirmed that HOXB5 promoted the proliferation of GSCs by activating IL6-mediated JAK2/STAT3 signaling (Fig. 2).

Table 2.CSC biomarkers for neurological tumors.
Diagnostic biomarker Pathway(s) Prognostic biomarker Pathway(s) Treatment biomarker Pathway(s) References
Glioma NADH (+) CD133, IL6, TGF-β (-) TGF-β–NF-κB CD133 (+) Wnt/β-linked protein [29, 30, 31, 32, 33, 34, 35, 36]
GAS5 (-) –IL6–STAT3 ALDH1, KLF4, SOX2, NANOG,
HOXB5 (-) IL-6/IL-6R/STAT3 MYC, ABCG2 (+)
BRAP (+) TGFβ/PI3K/AKT/mTOR
Medulloblastoma CD133, CD15, Nestin, CD24, SOX2 (+) CD133, CD15 (+) PI3K/AKT, MAPK [37, 38, 39, 40]
Sox2, Klf4, Nestin, Bmi1 (+) PI3K/AKT
Meningioma NANOG (+) SOX2 (-) [24, 41]
Neurofibroma PROM1, NKX2-2 (+) Wnt CD68, NANOG, OCT3/4, SOX2 (+) PI3K/AKT/GSK3β [27, 41]

Notes: tumor growth, prognostic effect and marker content were positively correlated as (+); Proteins are in normal font, genes are in italics.

Fig. 2.

Mechanisms of CSC biomarkers in glioma. GSC, glioma stem cell. Created with BioRender.com.

3.2.2 Medulloblastoma

Medulloblastoma (MB) comprises a subpopulation of CSCs, and the key properties of CSCs depend on phosphatidylinositol 3 kinase (PI3K). Thus, activation of the PI3K/AKT pathway contributes to the development of MB [37]. Biomarkers for the diagnosis of MB CSC include CD133, Nestin, SOX2, CD24 and CD15 [38, 39]. Dietl et al. [39] demonstrated that MB3W1 cells, which exhibit high ALDH activity, also express high levels of CD133 and CD15, suggesting that they possess characteristics commonly associated with CSCs. Biomarkers for the treatment of MB CSC include CD133, CD15, OCT4, NANOG, Sox2, Klf4, Nestin and Bmi1 [37, 40]. Eckerdt et al. [37] mentioned that PIK3CA expression correlates positively with the expression of several stem cell/pluripotency markers such as Sox2, Klf4, Nestin and Bmi1. Moreover, they observed a significant increase in Nestin expression, a CSC biomarker, and phosphorylation of AKT, which contributes to tumorigenesis and mediates survival and drug resistance in brain CSCs. Gong et al. [40]discovered that increased expression of the neuropilin-1 in cancer cells promotes the development of CSC features. However, MR438, when used in conjunction withthe neuropilin-1, effectively suppresses the activation of PI3K/AKT and MAPK pathways, reducing the expression of CSC biomarkers like CD133 and CD15, thus limiting the progression of MB (Fig. 3).

Fig. 3.

Mechanisms of CSC biomarkers in medulloblastoma. Created with BioRender.com.

3.2.3 Meningioma

The CSC Biomarkers for the diagnosis of intracranial meningioma (MG) include KLF4, MYC, CD133, SOX2, OCT4, NANOG, NESTIN and VIMENTIN [24]. The CSC biomarker of prognosis for intracranial MG is SOX2. Clinical studies by Di Bonaventura et al. [41] have shown a close correlation between SOX2 expression and MG grading, with prognostic implications for both overall survival and recurrence-free survival. Therefore, the assessment of SOX2 can be a valuable tool in guiding adjuvant treatment for MG patients.

3.2.4 Neurofibroma

Biomarkers for the diagnosis of neurofibroma CSC are PROM1 and NKX2-2. A recent study by Luscan et al. [28] found that PROM1 and NKX2-2 are overexpressed in malignant peripheral nerve sheath tumors (MPNSTs), suggesting a higher proportion of CSCs in this group. Additionally, the study demonstrated that activation of the Wnt signaling pathway can promote Schwann-mesenchymal transition, leading to the generation of CSCs during NF1 tumorigenesis. The biomarkers of neurofibroma CSC treatment are CD68, OCT3/4, NANOG and SOX2. Jia et al. [42] demonstrated that inhibiting PI3K/AKT signaling and GSK3β, as well as macrophage-induced stem cell biomarkers, effectively suppressed the formation of spheres induced by CM. It offers a potential therapeutic approach for treating neurofibromas (Fig. 4).

Fig. 4.

Mechanisms of CSC biomarkers in neurofibroma. SMT, Schwann-mesenchymal transition. Created with BioRender.com.

4. Differences and Correlations between CSC Biomarkers and Non-CSC Biomarkers

Non-CSC diagnostic biomarkers in glioma include APRP protein. Ali et al. [43] reported that glioma patients with higher tumor grades had increased levels of positive APRP fibrinogen and reduced levels of negative APRP biomarker albumin. In glioma, non-CSC therapeutic biomarkers such as PD-L1, as well as methyladenine DNA modifications, can serve as potential targets for therapy [44, 45]. Xie et al. [45] found that N6-methyladenine DNA modification was enriched in human glioblastoma. Reducing this modification, which can inhibit tumor growth, is an effective therapeutic strategy. The expression levels of VASH1, TP73 mRNA, HOXA5 and FMR1NB protein are prognostic biomarkers for glioma patients [46, 47, 48, 49]. Aili et al. [46] showed that high VASH1 expression is associated with poor prognosis and that VASH1 affects tumor cell invasion and migration by influencing microtubule formation and immune infiltration in the tumor microenvironment. Ding et al. [49] found that HOXA5 was a prognostic biomarker, and its overexpression could promote glioma progression by affecting glioma cell proliferation.

The non-CSC diagnostic biomarkers for MB are CADH1, FGFR4 and FIBB and other urinary proteins. Hao et al. [50] found that urinary proteins such as CADH1, FGFR4 and FIBB can be used to differentiate between MB patients and healthy individuals. The biomarkers of non-CSC prognosis in MB include miR-137, PBK and UBE2G1 [51, 52, 53]. Ji et al. [53] demonstrated that upregulated miR-137 inhibited cell proliferation, migration, invasion and cell cycle progression, and induced apoptosis by targeting KDM1A. Thus, it was concluded that miR-137 downregulation was significantly associated with poor prognosis of MB.

The non-CSC diagnostic biomarkers for intracranial MG are miR-497 and VIMENTIN [54, 55]. Abdelrahman et al. [54] found that serum miR-497 is a valid and easily measured biomarker for the diagnosis and classification of MG in clinical studies. Kim et al. [55] showed that all primary cell lines derived from both benign and atypical MG express high levels of the MG biomarker VIMENTIN, which is strongly associated with malignancy. The DVL1 protein is a biomarkers of non-CSC treatment in intracranial meningioma. Bukovac et al. [56] showed that higher nuclear expression of DVL1 protein and higher expression of active β-linked protein significantly correlated with higher MG grade. Therefore, DVL1 protein can be used as a biomarker for MG development and Wnt signaling pathway activation. Moreover, biomarkers associated with the prognosis of non-CSC in intracranial MG include total DNA methylation and GSKIP protein [57, 58]. Barciszewska et al. [57] concluded that since 5-methylcytosine levels in peripheral blood can reflect graded levels in tumor tissue, changes in 5-methylcytosine can be used as a biomarker of DNA damage and tumor malignancy in intracranial MG. Cheng et al. [58] found that high GSKIP protein expression was associated with poor prognosis of MG. Moreover, the GSKIP protein is bound to protein kinase A and glycogen synthase kinase 3β to regulate different biological processes and malignant tumorigenesis through the Wnt pathway.

For diagnostic biomarkers of neurofibromas, a study identified the downregulation of miR-204 as a novel diagnostic biomarker and potential therapeutic target for patients with NF1 with MPNSTs.16 [59]. Biomarkers for non-CSC treatment in neurofibromas include PD-L1 [60]. Farschtschi et al. [60] found that PD-L1 is upregulated in MPNST patients in a study and could be a potential biomarker for malignant transformation in NF1 patients.

Overall, there is some overlap between CSC and non-CSC type biomarkers in the nervous system, but these biomarkers may play different roles in different pathways. Ki67, for instance, serves as a biomarker for non-stem cell therapy in intracranial MG and as a diagnostic biomarker for stem cells in MB [38, 61]. De Carvalho et al. [61] found that Ki67 observed in recurrent tumors of MG was higher than in primary tumors and correlated with poor prognosis. Moreover, elevated Ki67 indicates larger recurrent tumors and more severe peritumor edema. These findings indicate that Ki67 plays multiple roles in diverse tumor types and holds potential as both a diagnostic indicator and a prognostic factor. However, HOXA5 is a prognostic biomarker for both glioma cells and GSCs, both of which are associated with poor patient survival and regulate the proliferation of GSCs [36, 49].

Vimentin serves as a diagnostic biomarker for both CSCs and non-CSCs in intracranial MG [55, 62]. In non-CSCs, strong expression of Vimentin is significantly associated with malignancy [55]. However, Vimentin is a CSC biomarker that regulates several signaling pathways, such as MAPK, PI3K/Akt, and Wnt/β-catenin. This protein affects the proliferation, differentiation, and invasiveness of CSCs, contributing to the recurrence of rare and aggressive MG variants (Table 3, Ref. [43, 44, 45, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60]).

Table 3.Non-CSC biomarkers for neurological tumors.
Diagnostic biomarker Prognostic biomarker Pathway(s) Treatment biomarker References
Glioma APRP (+) PD-L1 (-) Methyladenizne [43, 44, 45, 47, 48, 49]
VASH1 (-) DNA modifications (-)
TP73 mRNA (-)
HOXB5 (+)
Medulloblastoma CADH1, FGFR4, FIBB (+) miR-137 (-) [50, 51, 52, 53]
PBK (-)
UBE2G1 (+)
Meningioma miR-497 (+) DVL1 (+) Wnt [54, 55, 56, 57, 58]
VIMENTIN (+) GSKIP (-) Wnt
DNA methylation (+)
Neurofibroma miR-204 (+) PD-L1 (+) [59, 60]

Notes: tumor growth, prognostic effect and marker content were positively correlated as (+); Proteins are in normal font, genes are in italics.

5. Therapeutic Targets of CSCs

Because neurological tumors such as gliomas are resistant to conventional treatment and prone to recurrence, research to explore their precise molecular mechanisms and reliable therapeutic targets has received widespread attention [63]. Baisiwala demonstrated that overexpression of LNX1 activates the Notch1 signaling pathway and increases the population of GSCs [20]. However, knocking down LNX1 modulates Notch1 activity, leading to downregulation of the Notch1. CSCs are significantly reduced after TMZ treatment, thereby improving patient survival. These results suggest that LNX1-regulated Notch1 signaling contributes to treatment resistance and is a possible therapeutic target. Moreover, Lu et al. [64] discovered that knockdown of EH Domain Containing 1 (EHD1) reduced CSC-like traits in glioma cells. EHD1 binding to CD44 enhances CD44 stability, whereas CD44 overexpression reduces the inhibitory effect of EHD1 knockdown on GSC-like traits. Thus, EHD1 is positively correlated with the expression of CSC biomarkers in glioma tissues and has the potential to be an effective target for glioma progression. Mazor et al. [65] showed that lncRNA TP73-AS1 is associated with the aggressiveness of glioblastoma, and its high expression is associated with poor patient prognosis. Meanwhile, TP73-AS1 downregulation resensitizes GBM cancer stem cells to TMZ treatment and may serve as a promising therapeutic target.

Eckerdt et al. [37, 66] also found that signaling in the PI3K-AKT-mTOR axis was blocked by dual inhibition of PI3Kα and mTOR. It also enhanced the inhibition of tumor growth in MB in vivo, contributing to the development of more effective therapeutic strategies. Wang et al. [67] showed that miR-222-3p is also a therapeutic target. It not only inhibits cell viability, but also suppresses MB stem cell-like cell formation via the Notch2/c-myc pathway. Filipponi et al. [68] discovered that OCT4 induces the BMP4 signaling pathway, which promotes both stem cell pluripotency and MB tumorigenesis. Conversely, inhibiting BMP4-ALK2/3 signaling reduces tumor-initiating cell (TIC) ability and CSC biomarker expression, suggesting potential implications for MB treatment.

Additionally, published research suggests that taking antipsychotic medication may reduce the risk of cancer development in patients. Many antipsychotic drugs can also treat tumors in the nervous system and inhibit tumor growth [14]. SOX2-OT, an evolutionarily conserved long noncoding RNA, contains SOX2 in its intronic region, which can regulate SOX2 expression [69]. SOX2-OT have been linked to several health conditions, including psychiatric disorders, cancer and diabetes complications. SOX2 is a proven biomarker for CSCs in many nervous system tumors. Therefore, CSCs of the nervous system are associated with the occurrence of mental illness. Penflulidol is an FDA-approved antipsychotic drug specifically for schizophrenia that has potential anticancer effects against glioblastoma. Ranjan et al. [70] discovered that penfluridol inhibits the activation of Akt by blocking its phosphorylation at Ser473, thereby reducing Akt-mediated GLI1 expression. Furthermore, it directly regulates CSC biomarkers such as OCT4, Nanog and Sox2, decreasing CSC characteristics and inhibiting tumor growth. Dong et al. [71] found that the traditional antipsychotic drug fluspirilene can be utilized as a targeted treatment for GSCs. It works by inhibiting the STAT3-related signaling pathway, thereby reducing the proliferation of GSCs and their stemness, ultimately treating GBM. In addition, Cheng et al. [72] screened the antipsychotic drug thioridazine by public gene expression data. It was shown to have potent anti-GBM and anti-GSCs properties, inhibiting GBM tumorigenesis and inducing autophagy in vivo. And pimozide, which is used clinically for the treatment of schizophrenia, inhibits USP1 in GBM cells, resulting in decreased levels of ID1, Sox2, as well as inhibiting stem cell maintenance [73].

Apart from their medical applications, antipsychotic drugs have demonstrated additional pharmacological properties. They can regulate cell signaling pathways and participate in various intracellular functions, ultimately weakening the characteristics of CSCs and inhibiting cancer cell proliferation [74].

6. Detection of Neurological CSCs

The proportion of CSCs in tumor tissue is very low, typically only 0.01–2% of the total tumor mass. CSCs and normal stem cells also share similar biomarkers and signaling pathways [75]. Therefore, it is important to identify and detect CSCs in the nervous system. Many recent studies have focused on the pathological characteristics and detection methods of neurological CSCs.

CSCs can be identified by specific surface markers. For example, CD133 can be used to identify stem cells in normal and cancerous tissues and has been widely used as a marker to identify CSCs [76]. The assessment of the expression rate of relevant genes and protein markers in CSC reflects their density in the patient’s tumor tissue [77]. However, as with non-CSC, most identification of molecular biomarkers for CSC is based on antibody recognition of specific proteins. Cell surface markers are usually detected using immunohistochemistry, western blot (WB), and flow cytometry (FCM) methods. For immunohistochemistry staining and WB, the avidin-biotin-peroxidase complex method can be used, where the expression level of biomarkers is assessed by the percentage of positively stained cells and the intensity of staining [34, 78]. FCM involves incubating cells with a fluorescent dye The expression of biomarkers can also be determined using a cell sorter by further incubation with labeled secondary antibodies [79].

Second, Hematoxylin and eosin is the most essential pathological staining technique when examing tissues and cells under the microscope. Sometimes, abdominal tumors are embedded in paraffin and then sectioned for Hematoxylin and eosinstaining [37]. Moreover, microstructural characterization was performed with a high-resolution scanning electron microscope [80].

Detection tools for CSCs include fluorescence-activated cell sorter (FACS), magnetic-activated cell sorter (MACS), sphere formation assays, and techniques such as immunohistochemistry, FCM and WB for cell surface markers. For CSC characterization, FACS and MACS provide data for identifying individual CSCs in cell populations [81, 82]. FACS selection is a strategy for isolating CSCs based on specific markers expressed by CSCs using flow cytometry. By labeling cells with fluorescent or magnetic bead-coupled antibodies, the CSC population can be separated from tumor cells by FACS or MACS. In practice, FACS selection is purer, more accurate and superior to MACS [83].

Alternatively, the colony-forming ability of CSCs was used for isolation and identification [75]. Sphere formation assays is an in vitro method that is commonly used to identify CSCs and study their properties [82]. In vitro formation of floating clone (from one cell) colonies, called different “glioma spheres” or errant “neurospheres”, is commonly used as an indicator of potential stem cells. These colonies themselves can generate new colonies and can give rise to a variety of cell types within the tumor [84].

7. Discussions

This review has established the credibility and repeatability of the research through strict literature screening standards in the early stage. However, most of the literature is one-sided in describing biomarkers of a single tumor and is not well-informed in terms of diagnosis, treatment and prognosis. In contrast, this study comprehensively summarizes the sets of multiple CSC biomarkers, including neurological tumors such as glioma, MB, MG and neurofibroma, as a more accurate and reliable method to monitor the effects. The commonalities and differences in their functional pathways were also analyzed, providing a broader perspective. The bibliometric analysis we completed also remedied the shortcomings of other articles by obtaining more comprehensive information about CSC biomarkers in the nervous system. In addition, since most tumor recurrence and metastasis are caused by the promotion of CSCs, research addresses CSCs from the root cause to provide guidance for clinical application of CSC biomarkers in oncology.

However, CSCs with self-renewal and pluripotency capabilities are plastic and their plasticity is affected by the microenvironment [85, 86]. Recent studies have revealed that infiltrating macrophages can regulate the PI3K/AKT/GSK3β signaling pathway to induce stem cell-like characteristics and promote the growth of neurofibroma [42], so the tumor microenvironment plays an important role in regulating cell stemness. In addition, due to the heterogeneity of CSCs and the different specific signaling pathways they activate, a single CSC biomarker is incompetent to discriminate between CSC and non-CSC. The blockage of a single CSC-associated pathway may not effectively block the occurrence of cancers [85].

To date, the impact of CSC on mental illness in the nervous system has not been confirmed. Previous studies have reported that antipsychotic drugs might be able to treat CSC through pathways such as STAT3 and Akt, inhibiting the proliferation and spread of cancer cells [70, 71]. However, the effects of these drugs on different types of tumors and CSCs may vary. Meanwhile, current research is relatively limited and not only does not directly point out that neurological CSC accelerates the degree of tumor progression but also does not provide a specific assessment of the change in the degree of progression. Therefore, further research on this issue is also necessary to address these concerns.

8. Conclusions

This review provides a comprehensive summary of CSC biomarkers for neurological tumors such as glioma, meningioma, medulloblastoma and neurofibroma. In glioma, the CSC diagnostic biomarkers include NADH. And biomarkers of CSC treatment are CD133, ALDH1, KLF4, SOX2, NANOG, MYC, ABCG2, Nestin and Msi-1. The expression levels of CD133, IL6, TGF-β, BRAP, HOXB5 and GAS5 are prognostic biomarkers in glioma patients. Also, the CSC diagnostic biomarkers of MB include CD133, Nestin, SOX2, CD24 and CD15. Biomarkers for the treatment of CSC include CD133, CD15, OCT4, NANOG, Sox2, Klf4, Nestin and Bmi1. Biomarkers for the diagnosis of intracranial MG CSC include KLF4, MYC, CD133, SOX2, OCT4, NANOG, NESTIN and VIMENTIN. And the biomarker of prognosis for intracranial MG CSC is SOX2. In neurofibroma, the CSC diagnostic biomarkers include PROM1 and NKX2-2. The biomarkers of neurofibroma CSC treatment are CD68, OCT3/4, NANOG and SOX2. In addition, many antipsychotic drugs can also treat tumors in the nervous system and inhibit tumor growth. Therefore, this study hopes to provide guidance for future in-depth exploration and monitoring of tumors for clinical applications.

Abbreviations

CSC, cancer stem cell; EMT, epithelial-mesenchymal transition; GSC, glioma stem cell; GBM, glioblastoma multiform; MB, medulloblastoma; MG, meningioma; NF1, neurofibromatosis type 1; NF2, neurofibromatosis type 2; ALDH1, dehydrogenase 1; PI3K, phosphatidylinositol 3 kinase; MPNSTs, nerve sheath tumors; FACS, fluorescence-activated cell sorter; MACS, magnetic-activated cell sorter; FCM, flow cytometry; WB, western blot; EHD1, EH Domain Containing 1.

Author Contributions

HX, JF and XL conceived and designed the work. XL, JH, YK, XC, QY and LZ collected and summarized the literature. XL, YK, HX and JF interpreted the content of the review, generated tables and figures and wrote the original manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We thank our laboratory members for technical help and insightful discussions, as well as the assistance from Biorender software in creating illustrations.

Funding

This research was funded in part by the National Natural Science Foundation of China (No. 42307366), the Fundamental Research Funds for the Central Universities (2021RC207) and the Beijing Natural Science Foundation (7214242).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Wu C, Li M, Meng H, Liu Y, Niu W, Zhou Y, et al. Analysis of status and countermeasures of cancer incidence and mortality in China. Science China. Life Sciences. 2019; 62: 640–647.
[2]
Kang Y, Zhai X, Lu S, Vuletic I, Wang L, Zhou K, et al. A Hybrid Imaging Platform(CT/PET/FMI) for Evaluating Tumor Necrosis and Apoptosis in Real-Time. Frontiers in Oncology. 2022; 12: 772392.
[3]
Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro-Oncology. 2021; 23: 1231–1251.
[4]
Ly KI, Blakeley JO. The Diagnosis and Management of Neurofibromatosis Type 1. The Medical Clinics of North America. 2019; 103: 1035–1054.
[5]
Ostrom QT, Gittleman H, Farah P, Ondracek A, Chen Y, Wolinsky Y, et al. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006–2010. Neuro-Oncology. 2013; 15: ii1–ii56.
[6]
Wang ZC, Li HB, Wei CJ, Wang W, Li QF. Community-boosted neurofibromatosis research in China. The Lancet. Neurology. 2022; 21: 773–774.
[7]
Chen F, Wendl MC, Wyczalkowski MA, Bailey MH, Li Y, Ding L. Moving pan-cancer studies from basic research toward the clinic. Nature Cancer. 2021; 2: 879–890.
[8]
Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Research & Therapy. 2019; 10: 68.
[9]
Corrò C, Moch H. Biomarker discovery for renal cancer stem cells. The Journal of Pathology. Clinical Research. 2018; 4: 3–18.
[10]
Hu X, Cong Y, Luo HH, Wu S, Zhao LE, Liu Q, et al. Cancer Stem Cells Therapeutic Target Database: The First Comprehensive Database for Therapeutic Targets of Cancer Stem Cells. Stem Cells Translational Medicine. 2017; 6: 331–334.
[11]
Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, et al. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Frontiers in Immunology. 2020; 11: 1280.
[12]
Sun JH, Luo Q, Liu LL, Song GB. Liver cancer stem cell markers: Progression and therapeutic implications. World Journal of Gastroenterology. 2016; 22: 3547–3557.
[13]
Prasetyanti PR, Medema JP. Intra-tumor heterogeneity from a cancer stem cell perspective. Molecular Cancer. 2017; 16: 41.
[14]
Dalton SO, Johansen C, Poulsen AH, Nørgaard M, Sørensen HT, McLaughlin JK, et al. Cancer risk among users of neuroleptic medication: a population-based cohort study. British Journal of Cancer. 2006; 95: 934–939.
[15]
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA: a Cancer Journal for Clinicians. 2020; 70: 7–30.
[16]
Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathologica. 2007; 114: 97–109.
[17]
Komori T. The 2016 WHO Classification of Tumours of the Central Nervous System: The Major Points of Revision. Neurologia Medico-Chirurgica. 2017; 57: 301–311.
[18]
Weller M, Wick W, Aldape K, Brada M, Berger M, Pfister SM, et al. Glioma. Nature Reviews. Disease Primers. 2015; 1: 15017.
[19]
Lin Y, Zhou J, Xu J, Zhao K, Liu X, Wang G, et al. Effects of combined radiosurgery and temozolomide therapy on epidermal growth factor receptor and variant III in glioblastoma multiforme. Oncology Letters. 2018; 15: 5751–5759.
[20]
Baisiwala S, Hall RR, 3rd, Saathoff MR, M Shireman J, Park C, Budhiraja S, et al. LNX1 Modulates Notch1 Signaling to Promote Expansion of the Glioma Stem Cell Population during Temozolomide Therapy in Glioblastoma. Cancers. 2020; 12: 3505.
[21]
Xu Z, Murad N, Malawsky D, Tao R, Rivero-Hinojosa S, Holdhof D, et al. OLIG2 Is a Determinant for the Relapse of MYC-Amplified Medulloblastoma. Clinical Cancer Research. 2022; 28: 4278–4291.
[22]
Richardson S, Hill RM, Kui C, Lindsey JC, Grabovksa Y, Keeling C, et al. Emergence and maintenance of actionable genetic drivers at medulloblastoma relapse. Neuro-Oncology. 2022; 24: 153–165.
[23]
Cotter JA, Hawkins C. Medulloblastoma: WHO 2021 and Beyond. Pediatric and Developmental Pathology. 2022; 25: 23–33.
[24]
Shivapathasundram G, Wickremesekera AC, Tan ST, Itinteang T. Tumour stem cells in meningioma: A review. Journal of Clinical Neuroscience. 2018; 47: 66–71.
[25]
Zhao L, Zhao W, Hou Y, Wen C, Wang J, Wu P, et al. An Overview of Managements in Meningiomas. Frontiers in Oncology. 2020; 10: 1523.
[26]
Goldbrunner R, Stavrinou P, Jenkinson MD, Sahm F, Mawrin C, Weber DC, et al. EANO guideline on the diagnosis and management of meningiomas. Neuro-Oncology. 2021; 23: 1821–1834.
[27]
Tamura R. Current Understanding of Neurofibromatosis Type 1, 2, and Schwannomatosis. International Journal of Molecular Sciences. 2021; 22: 5850.
[28]
Luscan A, Shackleford G, Masliah-Planchon J, Laurendeau I, Ortonne N, Varin J, et al. The activation of the WNT signaling pathway is a Hallmark in neurofibromatosis type 1 tumorigenesis. Clinical Cancer Research. 2014; 20: 358–371.
[29]
Yuan Y, Yan Z, Miao J, Cai R, Zhang M, Wang Y, et al. Autofluorescence of NADH is a new biomarker for sorting and characterizing cancer stem cells in human glioma. Stem Cell Research & Therapy. 2019; 10: 330.
[30]
Chu IR, Pan RL, Yang CS. Differentiation of Tumorigenic C6 Glioma Cells Induced by Enhanced IL-6 Signaling. Medicina. 2020; 56: 625.
[31]
Liu S, Zhang C, Wang B, Zhang H, Qin G, Li C, et al. Regulatory T cells promote glioma cell stemness through TGF-β-NF-κB-IL6-STAT3 signaling. Cancer Immunology, Immunotherapy. 2021; 70: 2601–2616.
[32]
Tian T, Guo T, Zhen W, Zou J, Li F. BET degrader inhibits tumor progression and stem-like cell growth via Wnt/β-catenin signaling repression in glioma cells. Cell Death & Disease. 2020; 11: 900.
[33]
Wei Y, Chen Q, Huang S, Liu Y, Li Y, Xing Y, et al. The Interaction between DNMT1 and High-Mannose CD133 Maintains the Slow-Cycling State and Tumorigenic Potential of Glioma Stem Cell. Advanced Science. 2022; 9: e2202216.
[34]
Wang B, Cao C, Liu X, He X, Zhuang H, Wang D, et al. BRCA1-associated protein inhibits glioma cell proliferation and migration and glioma stem cell self-renewal via the TGF-β/PI3K/AKT/mTOR signalling pathway. Cellular Oncology. 2020; 43: 223–235.
[35]
Wu S, Ren K, Zhao J, Li J, Jia B, Wu X, et al. LncRNA GAS5 represses stemness and malignancy of gliomas via elevating the SPACA6-miR-125a/let-7e Axis. Frontiers in Oncology. 2022; 12: 803652.
[36]
Zhao J, Jiang Y, Zhang H, Zhou J, Chen L, Li H, et al. The SRSF1/circATP5B/miR-185-5p/HOXB5 feedback loop regulates the proliferation of glioma stem cells via the IL6-mediated JAK2/STAT3 signaling pathway. Journal of Experimental & Clinical Cancer Research. 2021; 40: 134.
[37]
Eckerdt F, Bell JB, Beauchamp EM, Clymer J, Blyth GT, Kosciuczuk EM, et al. Potent Antineoplastic Effects of Combined PI3Kα-MNK Inhibition in Medulloblastoma. Molecular Cancer Research. 2019; 17: 1305–1315.
[38]
Casciati A, Tanori M, Manczak R, Saada S, Tanno B, Giardullo P, et al. Human Medulloblastoma Cell Lines: Investigating on Cancer Stem Cell-Like Phenotype. Cancers. 2020; 12: 226.
[39]
Dietl S, Schwinn S, Dietl S, Riedel S, Deinlein F, Rutkowski S, et al. MB3W1 is an orthotopic xenograft model for anaplastic medulloblastoma displaying cancer stem cell- and Group 3-properties. BMC Cancer. 2016; 16: 115.
[40]
Gong C, Valduga J, Chateau A, Richard M, Pellegrini-Moïse N, Barberi-Heyob M, et al. Stimulation of medulloblastoma stem cells differentiation by a peptidomimetic targeting neuropilin-1. NEURO ONCOLOGY. 2018; 20: i131–i132.
[41]
Di Bonaventura R, Martini M, Cenci T, Caccavella VM, Barresi V, Gessi M, et al. Dissecting Stemness in Aggressive Intracranial Meningiomas: Prognostic Role of SOX2 Expression. International Journal of Molecular Sciences. 2022; 23: 11690.
[42]
Jia J, Zhang H, Zhang H, Liu W, Shu M. Infiltrating Macrophages Induced Stem-cell-like Features Through PI3K/AKT/GSK3β Signaling to Promote Neurofibroma Growth. Archives of Medical Research. 2020; 51: 124–134.
[43]
Ali H, Harting R, de Vries R, Ali M, Wurdinger T, Best MG. Blood-Based Biomarkers for Glioma in the Context of Gliomagenesis: A Systematic Review. Frontiers in Oncology. 2021; 11: 665235.
[44]
Chen RQ, Liu F, Qiu XY, Chen XQ. The Prognostic and Therapeutic Value of PD-L1 in Glioma. Frontiers in Pharmacology. 2019; 9: 1503.
[45]
Xie Q, Wu TP, Gimple RC, Li Z, Prager BC, Wu Q, et al. N6-methyladenine DNA Modification in Glioblastoma. Cell. 2018; 175: 1228–1243.e20.
[46]
Aili Y, Maimaitiming N, Maimaiti A, Liu W, Qin H, Ji W, et al. Identification of VASH1 as a Potential Prognostic Biomarker of Lower-Grade Glioma by Quantitative Proteomics and Experimental Verification. Journal of Oncology. 2022; 2022: 2621969.
[47]
Bi SQ, Peng Y, Wei ZD, Yao SZ, Luo B, Ge YY, et al. FMR1NB Involved in Glioma Tumorigenesis Is a Promising Target for Prognosis and Therapy. Current Medical Science. 2022; 42: 803–816.
[48]
Chen Y, Wang Y, He Q, Wang W, Zhang T, Wang Z, et al. Integrative analysis of TP73 profile prognostic significance in WHO grade II/III glioma. Cancer Medicine. 2021; 10: 4644–4657.
[49]
Ding F, Chen P, Bie P, Piao W, Cheng Q. HOXA5 Is Recognized as a Prognostic-Related Biomarker and Promotes Glioma Progression Through Affecting Cell Cycle. Frontiers in Oncology. 2021; 11: 633430.
[50]
Hao X, Guo Z, Sun H, Liu X, Zhang Y, Zhang L, et al. Urinary protein biomarkers for pediatric medulloblastoma. Journal of Proteomics. 2020; 225: 103832.
[51]
Deng Y, Wen H, Yang H, Zhu Z, Huang Q, Bi Y, et al. Identification of PBK as a hub gene and potential therapeutic target for medulloblastoma. Oncology Reports. 2022; 48: 125.
[52]
Du BS, Yuan L, Sun LG, Zhang ZY. WGCNA screening of prognostic markers in medulloblastoma. Zhonghua Yi Xue Za Zhi. 2020; 100: 460–464. (In Chinese)
[53]
Ji W, Zhe X, Li L, Cheng Y, Zhao X, Liang P, et al. Prognostic Value of miR-137 in Children with Medulloblastoma and its Regulatory Effect on Tumor Progression. Neuromolecular Medicine. 2022; 24: 215–223.
[54]
Abdelrahman A, Negroni C, Sahm F, Adams CL, Urbanic-Purkart T, Khalil M, et al. miR-497 and 219 in blood aid meningioma classification. Journal of Neuro-Oncology. 2022; 160: 137–147.
[55]
Kim E, Kim M, So K, Park YS, Woo CG, Hyun SH. Characterization and comparison of genomic profiles between primary cancer cell lines and parent atypical meningioma tumors. Cancer Cell International. 2020; 20: 345.
[56]
Bukovac A, Dragičević K, Kafka A, Orešković D, Cesarec-Augustinović S, Pećina-Šlaus N. Decoding the Role of DVL1 in Intracranial Meningioma. International Journal of Molecular Sciences. 2021; 22: 11996.
[57]
Barciszewska AM. Total DNA methylation as a biomarker of DNA damage and tumor malignancy in intracranial meningiomas. BMC Cancer. 2020; 20: 509.
[58]
Cheng YW, Chen YY, Lin CJ, Lieu AS, Tsai HP, Kwan AL. High expression of GSKIP is associated with poor prognosis in meningioma. Medicine. 2022; 101: e32209.
[59]
Hanemann CO, Blakeley JO, Nunes FP, Robertson K, Stemmer-Rachamimov A, Mautner V, et al. Current status and recommendations for biomarkers and biobanking in neurofibromatosis. Neurology. 2016; 87: S40–S48.
[60]
Farschtschi S, Kluwe L, Park SJ, Oh SJ, Mah N, Mautner VF, et al. Upregulated immuno-modulator PD-L1 in malignant peripheral nerve sheath tumors provides a potential biomarker and a therapeutic target. Cancer Immunology, Immunotherapy. 2020; 69: 1307–1313.
[61]
de Carvalho GTC, da Silva-Martins WC, de Magalhães KCSF, Nunes CB, Soares AN, Tafuri LSDA, et al. Recurrence/Regrowth in Grade I Meningioma: How to Predict? Frontiers in Oncology. 2020; 10: 1144.
[62]
Rath P, Miller DC, Litofsky NS, Anthony DC, Feng Q, Franklin C, et al. Isolation and characterization of a population of stem-like progenitor cells from an atypical meningioma. Experimental and Molecular Pathology. 2011; 90: 179–188.
[63]
Peng Z, Liu C, Wu M. New insights into long noncoding RNAs and their roles in glioma. Molecular Cancer. 2018; 17: 61.
[64]
Lu Y, Wang W, Tan S. EHD1 promotes the cancer stem cell (CSC)-like traits of glioma cells via interacting with CD44 and suppressing CD44 degradation. Environmental Toxicology. 2022; 37: 2259–2268.
[65]
Mazor G, Levin L, Picard D, Ahmadov U, Carén H, Borkhardt A, et al. The lncRNA TP73-AS1 is linked to aggressiveness in glioblastoma and promotes temozolomide resistance in glioblastoma cancer stem cells. Cell Death & Disease. 2019; 10: 246.
[66]
Eckerdt F, Clymer J, Bell JB, Beauchamp EM, Blyth GT, Goldman S, et al. Pharmacological mTOR targeting enhances the antineoplastic effects of selective PI3Kα inhibition in medulloblastoma. Scientific Reports. 2019; 9: 12822.
[67]
Wang H, Wang Y, Wang Y. MiR-222-3p inhibits formation of medulloblastoma stem-like cells by targeting Notch2/c-myc signaling pathway. Immunopharmacology and Immunotoxicology. 2022; 44: 548–555.
[68]
Filipponi D, Pagnuzzi-Boncompagni M, Pagès G. Inhibiting ALK2/ALK3 Signaling to Differentiate and Chemo-Sensitize Medulloblastoma. Cancers. 2022; 14: 2095.
[69]
Li PY, Wang P, Gao SG, Dong DY. Long Noncoding RNA SOX2-OT: Regulations, Functions, and Roles on Mental Illnesses, Cancers, and Diabetic Complications. BioMed Research International. 2020; 2020: 2901589.
[70]
Ranjan A, Srivastava SK. Penfluridol suppresses glioblastoma tumor growth by Akt-mediated inhibition of GLI1. Oncotarget. 2017; 8: 32960–32976.
[71]
Dong Y, Furuta T, Sabit H, Kitabayashi T, Jiapaer S, Kobayashi M, et al. Identification of antipsychotic drug fluspirilene as a potential anti-glioma stem cell drug. Oncotarget. 2017; 8: 111728–111741.
[72]
Cheng HW, Liang YH, Kuo YL, Chuu CP, Lin CY, Lee MH, et al. Identification of thioridazine, an antipsychotic drug, as an antiglioblastoma and anticancer stem cell agent using public gene expression data. Cell Death & Disease. 2015; 6: e1753.
[73]
Shaw V, Srivastava S, Srivastava SK. Repurposing antipsychotics of the diphenylbutylpiperidine class for cancer therapy. Seminars in Cancer Biology. 2021; 68: 75–83.
[74]
Hendouei N, Saghafi F, Shadfar F, Hosseinimehr SJ. Molecular mechanisms of anti-psychotic drugs for improvement of cancer treatment. European Journal of Pharmacology. 2019; 856: 172402.
[75]
Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduction and Targeted Therapy. 2020; 5: 8.
[76]
Barzegar Behrooz A, Syahir A, Ahmad S. CD133: beyond a cancer stem cell biomarker. Journal of Drug Targeting. 2019; 27: 257–269.
[77]
Marquardt S, Solanki M, Spitschak A, Vera J, Pützer BM. Emerging functional markers for cancer stem cell-based therapies: Understanding signaling networks for targeting metastasis. Seminars in Cancer Biology. 2018; 53: 90–109.
[78]
Qian M, Chen Z, Wang S, Guo X, Zhang Z, Qiu W, et al. PLEKHG5 is a novel prognostic biomarker in glioma patients. International Journal of Clinical Oncology. 2019; 24: 1350–1358.
[79]
Shi Y, Ping YF, Zhou W, He ZC, Chen C, Bian BSJ, et al. Tumour-associated macrophages secrete pleiotrophin to promote PTPRZ1 signalling in glioblastoma stem cells for tumour growth. Nature Communications. 2017; 8: 15080.
[80]
Paolillo M, Colombo R, Serra M, Belvisi L, Papetti A, Ciusani E, et al. Stem-Like Cancer Cells in a Dynamic 3D Culture System: A Model to Study Metastatic Cell Adhesion and Anti-Cancer Drugs. Cells. 2019; 8: 1434.
[81]
Sharifzad F, Ghavami S, Verdi J, Mardpour S, Mollapour Sisakht M, Azizi Z, et al. Glioblastoma cancer stem cell biology: Potential theranostic targets. Drug Resistance Updates. 2019; 42: 35–45.
[82]
Bahmad HF, Cheaito K, Chalhoub RM, Hadadeh O, Monzer A, Ballout F, et al. Sphere-Formation Assay: Three-Dimensional in vitro Culturing of Prostate Cancer Stem/Progenitor Sphere-Forming Cells. Frontiers in Oncology. 2018; 8: 347.
[83]
Luo C, Ding Z, Tu Y, Tan J, Luo Q, Song G. Biomaterial-based platforms for cancer stem cell enrichment and study. Cancer Biology & Medicine. 2021; 18: 458–469. (online ahead of print)
[84]
Ludwig K, Kornblum HI. Molecular markers in glioma. Journal of Neuro-Oncology. 2017; 134: 505–512.
[85]
Eun K, Ham SW, Kim H. Cancer stem cell heterogeneity: origin and new perspectives on CSC targeting. BMB Reports. 2017; 50: 117–125.
[86]
Yin S, Guo Y, Wen X, Zeng H, Chen G. Increased expression of PD-L1 in endometrial cancer stem-like cells is regulated by hypoxia. Frontiers in Bioscience-Landmark. 2022;27:23.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top