You are on page 1of 15

Seminars in Cancer Biology 88 (2023) 123–137

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Transcriptional factors targeting in cancer stem cells for tumor modulation


Archana Chaudhary a, Syed Shadab Raza b, Rizwanul Haque a, *
a
Department of Biotechnology, School of Earth Biological and Environmental Sciences, Central University of South Bihar, Gaya, Bihar, India
b
Laboratory for Stem Cell & Restorative Neurology, Era’s Lucknow Medical College and Hospital, Era University, Lucknow, India

A R T I C L E I N F O A B S T R A C T

Keywords: Cancer Stem Cells (CSCs) are now considered the primary "seeds" for the onset, development, metastasis, and
Cancer Stem Cells (CSC) recurrence of tumors. Despite therapeutic breakthroughs, cancer remains the leading cause of death worldwide.
Transcription factors (TFs) This is because the tumor microenvironment contains a key population of cells known as CSCs, which promote
Octamer-binding transcription factor 4 (OCT-4)
tumor aggression. CSCs are self-renewing cells that aid tumor recurrence by promoting tumor growth and
Sex determining region Y-box 2 (SOX-2)
persisting in patients after many traditional cancer treatments. According to reports, numerous transcription
factors (TF) play a key role in maintaining CSC pluripotency and its self-renewal property. The understanding of
the functions, structures, and interactional dynamics of these transcription factors with DNA has modified the
hypothesis, paving the way for novel transcription factor-targeted therapies. These TFs, which are crucial and are
required by cancer cells, play a vital function in the etiology of human cancer. Such CSC TFs will help with gene
expression profiling, which provides crucial data for predicting the prognosis of patients. To overcome anti-
cancer medication resistance and completely eradicate cancer, a potent therapy combining TFs-based CSC tar­
gets with traditional chemotherapy may be developed. In order to develop therapies that could eliminate CSCs,
we here concentrated on the effect of TFs and other components of signalling pathways on cancer stemness.

1. Introduction direct and convincing experimental evidence to support the idea that
some tumor cells have a small number of fast-growing and self-renewing
It was first proposed in 1994 that tiny groups of cancer cells called cell populations [8,10], as well as the implications of this finding for
Cancer Stem Cells (CSCs) are responsible for the formation of the bulk of therapy (Fig. 1). The role of TFs in tumor onset and growth has recently
tumor cells. These studies showed that while the bulk of malignant cells received a lot of attention, especially since the discovery of multiple
fail to engraft and cause disease, a tiny percentage of human leukemia stemness factors in human somatic cells that can cause cell reprogram­
cells could cause leukemia in SCID (severe combined immunodeficient) ming to resemble embryonic stem cells [11,12].
mice [1,2]. Further research on hematological malignancies led to the
hypothesis that CSCs, like normal stem cells, live at the apex of a hier­ 2. Role of TFs in CSCs
archical cluster of mature cells, are capable of self-renewal, and can
produce numerous progeny that are considered "mature." Critical in­ Transcription factors are key gene expression regulators that are
vestigations carried out, afterwards discovered CSC activity in a range of necessary for CSC self-renewal and pluripotency [8]. One critical goal is
malignancies [3]. As we all know, metastasis and reoccurrence to find master transcription factors that determine the identity of CSCs.
following successful primary tumor elimination are two important ob­ Years of cell transplantation research demonstrated that abnormal
stacles to cancer therapy [1,4]. As a result, cancer eradication necessi­ expression of transcription factors like OCT-4, NANOG, and SOX-2 iso­
tates the elimination of CSCs, which persist after treatments cause tumor forms and pseudogenes has a significant impact on cancer trans­
relapse or recurrence [5,6]. Since CSC can produce any sort of cell formation, metastasis, and carcinogenesis, though the precise
observed in the majority of the tumor, they can start the development of underlying mechanisms remain unknown [10,13]. Novel approaches
secondary tumors [7,8]. CSCs are cell lineages that exist within a that specifically target CSCs while causing no harm to normal cells are
growing tumor in various malignancies, such as intestinal adenomas, desperately needed to advance current cancer treatment regimens.
glioblastomas, and squamous skin tumors. These tumors have given There is a wealth of evidence that OCT-4, SOX-2, and NANOG over

* Corresponding author.
E-mail address: rhaque@cub.ac.in (R. Haque).

https://doi.org/10.1016/j.semcancer.2022.12.010
Received 13 July 2022; Received in revised form 29 December 2022; Accepted 30 December 2022
Available online 2 January 2023
1044-579X/© 2022 Published by Elsevier Ltd.
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

expression occurs in human malignancies, and is linked with tumor 25], but consistent changes in their gene expression, level of proteins,
development, metastasis, and distant recurrence after and post-translational modification states have not been observed in the
chemo-radiotherapy [10,13–15]. Over expression of OCT-4, SOX-2 and majority of malignancies [26]. Evidence recently suggested that Zeb1 (a
NANOG in tumor stem cells modifies signaling pathways to prevent zinc finger E-box), an essential transcriptional regulator in hematopoi­
apoptosis [10,16–18]. The amounts of OCT-4, NANOG, and SOX-2 esis that controls HSC renewal, multilineage differentiation fates, and
mRNA transcripts along with markers of stem cells found in cancer apoptosis, must be carefully coordinated in order to reduce leukemic
cells and in niches of CSC are usually greater than those found in potential in AML[27]. In addition to the therapeutic approach of CSC
non-tumor tissue [8,15,16]. A number of human malignancies, TFs, the limited cross reactivity of CSC surface markers with normal
including breast cancer, prostate cancer, and oral squamous carcinoma tissue cells makes them potential therapeutic targets for CSCs [29]. The
cells, have been shown to co-upregulate the expression of OCT-4, most frequently utilized markers for CSC research and cancer thera­
NANOG, and SOX2 [19]. It has been shown that OCT-4, the most sig­ peutic targets are nestin, CD133, and CD44. Colon CSCs, which are
nificant pluripotent factor, is up-regulated in a number of tumor cells, identified by the cell surface markers CD133, CD24, and CD44, are
including human oral squamous cell carcinoma, murine Lewis lung among the CSCs that have been shown to have a role in the growth of
carcinoma, bladder cancer, and seminoma cancer [10,19]. Transcrip­ tumors, treatment resistance, and the spread of the disease to other or­
tional factors linked to stemness can be specifically targeted to treat gans. Reports suggested that high levels of nestin, CD44/CD133, and
different types of tumor illnesses. These stem cell factors promote the OCT-4 expression were present in pancreatic CSCs, which made them
self-renewal properties of CSCs through interactions with various tran­ more resistant to gemcitabine than normal pancreatic cells. They are
scription factors, including Stat3, Zic3, and Hesx1, which can exhibit also strong anti-apoptotic but have limited proliferative abilities [29,
abnormal expression in a variety of human malignancies [20]. However, 30]. Therefore, the distinct population of pancreatic stem cells may be
very little research has been done on the regulating mechanism of crucial in the emergence of widespread metastases and multidrug
transcription factors and their role in mediating CSCs’ stemness. Recent resistance in pancreatic cancer. Researchers also showed that autophagy
studies showed that CSC transcription factors could keep CSCs stem-like and the maintenance of tumor cells with upregulated CD44 and down­
by encouraging the expression of genes relevant to stemness[10,20]. The regulated CD24, which are common features of breast CSCs, have a
key player in the reprogramming of breast CSCs is the glycolysis gate­ mechanistic link. Autophagy inhibition increased CD24 gene transcrip­
keeper PDK1, which also offers a potential treatment for breast cancer tional activation, enhancing the phenotype of CD44 (+)CD24(+) cells
[21]. Additionally, it has been shown that NFIs (the nuclear factor one), that resembles epithelial cells rather than the mesenchymal CD24
a transcription factor that is crucial for the growth of numerous (-/low) CD44(+) offspring. Treatment with chloroquine increased the
mammalian organ systems, regulate the accessibility of chromatin at proportion of CD24 (+) cells while concurrently decreasing the consti­
distal regulatory regions that facilitate the metastasis and growth of tutive upregulation of vimentin in breast cancer cell lines [31]. A range
CSCs, as well as its remodeling and epigenetic regulation of gene of malignancies, including lung, pancreatic, and prostate cancers, have
expression[22]. Apart from this, STAT3 also has a key role in controlling been discovered to have significant nestin expression [30,32]. It has
both healthy stem cells and tumor stem cells, which are physically and been shown that transformed cells from a range of human tumors ex­
functionally linked with p63, a member of the p53 protein family [23]. press nestin along with other CSC TFs, and it has been proven that this
expression is associated with the clinical development of particular tu­
3. Regulation of TFs in various cancer stem cells’ stemness mors [30]. Increased nestin expression indicates that converted cells are
young and invasive and is related to tumor grade in a variety of tumor
Most of the TFs are highly up-regulated in certain cancers such as forms [30]. Additionally, a correlation between nestin expression levels
glioblastomas, colonic adenomas, and squamous skin tumors [10,24, and the clinical course of the disease was discovered for melanoma,

Fig. 1. The theory of CSC resistance and its therapeutic implications are illustrated by contrasting CSC targeted therapies with conventional cancer therapies. CSCs
can survive the cell death caused by conventional chemotherapy because of their distinct drug-resistance characteristics and plasticity, allowing them to grow new
tumors. It is proposed that CSC targets and bulk tumor ablation be used in combination therapy to enhance cancer patients’ clinical outcomes.

124
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

ovarian carcinoma, breast cancer, gastrointestinal malignancies, 47]. OCT-4, also known as Pou5f1, is a transcription factor that regulates
gastrointestinal malignancies, germ cell tumors, and osteosarcoma the pluripotency of ESCs (embryonic stem cells) [25]. It induces cell
[33–39]. In these cancers, nestin expression is therefore thought to be a proliferation by regulating T-cell leukemia/lymphoma1 (TCL1) tran­
predictive marker; nevertheless, its prognostic usefulness needs to be scription and upregulating AKT1 kinase activity [48]. The
confirmed in large cohorts of patients. The common stem cell TFs OCT-4, OCT-4/Akt1/Tcl1 pathway is activated when OCT-4 is activated, and
NANOG, CD133, SOX-2, and nestin are expressed by CSCs in prostate tumor cells undergo apoptosis inhibition [47]. Different tumors activate
cancer patients [40]. Furthermore, CSCs were isolated from human­ or suppress different cancer-related pathways, but it appears that OCT4,
pancreatic cancer using sphere creation, and nestin, CD44, CD133, NANOG, and SOX-2 are always up-regulated to either activate or sup­
NANOG, and OCT-4 expression in these cells was discovered [30,41]. It press OCT-4/Tcl1/Akt1 pathways [47,48]. OCT-4 overexpression pro­
is interesting to note that breast CSCs also express nestin and OCT-4, motes carcinogenesis and suppresses cancer cell death in cervical cancer
exhibit higher tumorigenicity by developing mammospheres in an cells [24]. According to a chromatin immunoprecipitation study, OCT-4
invitro environment [42]. Compared to cells with limited nestin increases miR-125b production by binding directly to the miR-125b-1
expression, those with high nestin expression had a 100-fold higher promoter [49–52]. When the miR-125b response element was changed
tumorigenicity [42,43]. However, nestin was knocked down in breast or eliminated, the untranslated region reporter activity of wild-type
CSCs, resulting in cell cycle arrest, induced apoptosis, and suppression of BAK1 3′ was significantly suppressed in the Luciferase assay [47,52].
spontaneous EMT and cell migration [43]. Nestin’s molecular silencing OCT-4 post-translational modification alters its activity and calls for
interfered with the induction of catenin/Wnt, a crucial route for further research into its relationship to chemo resistance and prognosis.
signaling in breast CSCs [43]. In addition to this, younger age, higher Radiation- or chemotherapy enriched CSCs in various malignancies
grades of malignant tumors, lymph node metastases, and poorer sur­ showed phenotypic changes as well as an increase in stem cell marker
vivals have all been linked to nestin/OCT-4 positivity in cancer tissues expression such as OCT-4, SOX-2, NANOG, CD133, and ALDH, although
[42,43]. Nestin and other CSC markers have been shown to co-express the mechanisms of OCT-4 in promoting chemo resistance are not well
with other known CSCs TFs, such as SOX-2, NANOG, OCT-3/4, KLF4 understood [47]. In a number of CSCs, OCT-4 is implicated in tumor
and MYC in circulating breast cancer cells [44]. Using an in silico vasculogenesis. In the year 2020, Liu et al. demonstrated that liver
translational approach, researchers hypothesized that the amalgamation cancer stem cells (LCSCs) can undergo trans-differentiation into endo­
of these CSC markers has predictive value in a significant cohort of thelial cells in response to endothelial induction, that
colorectal cancer patients [28]. Life-threatening malignancies may be endothelium-specific functions and markers exist in vitro, and that
completely cured by such treatments that target TFs along with CSC LCSCs contribute to neo vasculogenesis in vivo [52]. Transcription
markers, which have significant potential for eliminating the CSC pop­ factors like OCT-4, SOX-2, and NANOG show tremendous clinical
ulation. So here, we will look at the role of various TFs in the growth of promise. They can be effective carcinogenesis markers and molecular
CSCs as well as the potential of TF networks as direct and indirect switches that regulate the fate of CSC cells during the cancer develop­
therapeutic targets in CSCs. ment [53,54]. CSCs exhibit a lot of OCT-4 during the occurrence and
growth of malignant tumors and its degree of expression is positively
4. Different types of transcription factors in CSCs connected with various malignancies.OCT-4 was found in both the
cytoplasm and nucleus of breast cancer cells, according to the findings
Researchers demonstrated that cancer is caused by a tumor cell [54,55]. The human OCT-4 gene can produce at least 3 transcripts,
subpopulation with stem cell characteristics, which were later dubbed referred to as pseudogenes (OCT-4A, OCT-4B, and OCT-4B1), and 4
CSCs [45]. Despite advancements in terms of diagnosis and therapy, the protein isoforms through alternative splicing and alternative translation
tumor disease prognosis remains deplorable, and the patients’ long-term initiation (OCT-4A, OCT-4B-190, OCT-4B-265, and OCT-4B-164) [53,
cure rate has plateaued within the last 30 years [46]. CSCs are respon­ 56]. OCT-4 has a slightly larger molecular weight in bladder tumor cells
sible for treatment failure, and it has been established that aggressive than in normal bladder cells, which may be the result of distinct post­
cancer cells exhibit high expressional levels of various transcription translational modifications of OCT-4 [57]. OCT-4, SOX-2, and NANOG
factors such as OCT-4, SOX, NANOG, and KLF-4, among many others are deregulated in signaling pathways, although their distinct behaviors
that have been designated as distinct markers [10,46]. Despite intensive in ESCs and CSCs may be influenced by the expression of their isoforms
attempts to discover CSCs, novel therapeutic strategies for CSC eradi­ and pseudogenes. Breast cancer specimens were found to express two or
cation remain elusive due to our limited understanding of transcrip­ more factors that induce stem cells, like c-MYC, KLF4, OCT-4, NANOG,
tional regulation in CSC biology. It will be crucial to comprehend how and SOX-2[36,37,57,58]. Therefore, CSC-inducing factors in CSCs could
the TFs interact to promote normal cellular growth rather than malig­ be one of the crucial areas for cancer research in the future. OCT-4 was
nant outcomes in order to produce more efficient selective ligands that found to be highly activated in CD24/CD44 (+) tumor cells, suggesting
can combat CSCs in the future with fewer adverse effects. Additional that it could be a biomarker for cancer metastasis, progression, and
investigation into the features of such transcription factors and the CSC progression [58]. Breast cancer stem-like cells may express more CD44,
regulation mechanism could aid in the identification of a potential CXC chemokine receptor-4 (CXCR4), and OPN (osteopontin) while
molecular target for therapeutic intervention in a variety of malig­ expressing less CD24 [58]. In patients with surgically removed rectal
nancies. We illustrate how OCT-4, NANOG, and SOX-2 are regulated in cancer (RC) getting adjuvant therapy, the expression of OCT-4, NANOG,
various tumors and how they are usually up-regulated to either inhibit and SOX-2 was investigated for their associations with
or activate the cancer-related process. clinico-pathological traits and overall survival. It was discovered that
OCT-4 expression in tumor tissue can serve as a standalone prognostic
5. Octamer-binding transcription factor 4 (Oct4) marker and is related to poor differentiation, tumor size, and its stage
[59]. The Raf-kinase antagonist protein, which inhibits metastasis and
The wealth of data demonstrating the role of CSCs in the context of serves as a chemo-immuno-sensitizer to medication- or
chemo resistance has fueled interest in OCT-4. The homeo domain immune-mediated apoptosis, is inversely linked with the expression of
transcription factor Octamer-binding transcription factor 4 (OCT-4) has OCT-4,suppress SOX-2 expression by inhibiting the route for
long been recognized as a master controller of pluripotency and self- mitogen-activated protein kinase and induces NANOG by preventing the
renewal properties [47]. According to a large body of research, OCT-4 kinase controlled by extracellular signals. Raf-kinase inhibitor proteins
has also been found in tumor cells and tissues, as well as in a discrete prevent OCT-4 and SOX-2 from forming the heterodimer that is required
subset of undefined tumor-initiating cells (TICs), which are important to bind to the promoter of NANOG and trigger the transcription of
for tumor development, metastasis, and treatment-resistant cancer[10, NANOG. These reports demonstrated a direct link between the

125
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

expression of all three TFs and the expression of the Raf-kinase inhibitor active alkaline phosphatase (AP) in significant amounts, with signifi­
protein, suggesting that this protein’s level of expression may be regu­ cantly greater proportions of CD44(+). The genes PRKAA1 (which codes
lating the phenotype of CSCs [60]. OCT-4 expression, like most stem cell for the catalytic AMPK 1 (AMP-activated protein kinase1 subunit),
factors, appears to be important in cancer, with increased as well as DDIT4/REDD1 (a stress response gene that acts as a negative regulator
decreased expression interfering with various cancer-related processes. of mTOR(Target of Rapamycin), and DEPTOR (a naturally occurring
endogenous inhibitor of mTOR activity) were noticeably down regu­
6. Sex determining region Y-box 2 (SOX-2) lated in three separate SOX2-overexpressing CSC-like cell lines.
Insulin-receptor gene expression in MCF-7/Rep cells was differently
SOX-2 is a crucial transcription factor in CSCs that regulates em­ upregulated. Immunoblotting tests demonstrated over expression of
bryonic stem cell pluripotency as well as other developmental processes p70S6K and enhanced phosphorylation of mTOR in
[49]. SOX-2 expression has been linked to breast, colorectal, gastric, and SOX-2-overexpressing CSC-like cell types, which was consistent with the
lung cancers, as well as a link between cancer and stemness[10,61]. downregulation of AMPK expression. Through these reports, researchers
SOX-2 functions in CSCs through a number of signaling channels, and showed that transcriptional suppression of mTOR repressors is an
there is evidence that it opposes the Wnt pathway, which promotes intrinsic process occurring during the acquisition of CSC-like properties
differentiation and may suppress SOX-2 expression [62,63]. In prostate by differentiated populations of luminal-like breast cancer cells. They
cancer stem cells (PCSCs), stimulation of Epidermal Growth Factor Re­ did this by using an in vitro model of the de novo generation of CSC-like
ceptor (EGFR) signaling pathways causes a substantial increase in SOX-2 states through the nuclear reprogramming of an established breast
mRNA along with protein levels, promoting CSC self-renewal [47,64]. cancer cell line. These reports might open up a new avenue for learning
Further investigation has revealed that the CSC SOX-2 expression status how to control the AMPK/mTOR pathway to avoid the emergence of
is at the root of many clinical types of tumors, implying that SOX-2 CSCs [71]. Studies have also reported that through the stimulation of the
serves a crucial function in the development of cancer through CSC Rap1/P110 β cascade, aberrant AKT activation supports CSC charac­
amplification [64]. SOX-2 is classified as a proto-oncogene, which teristics in hepatocellular carcinoma (HCC). By increasing SOX-9
means that amplification, mutation, and overexpression of the SOX-2 expression and improving its protein stability, CD73, a traditional sur­
gene can result in a variety of malignant diseases with metastasis [10, face marker of multipotent mesenchymal stem cells, contributes signif­
61]. SOX-2 amplification is linked to enhanced proliferation, metastasis, icantly to the maintenance of CSC characteristics. Eradicating CSCs and
tumor burden, and a poor prognosis [65]. In lung cancer, SOX-2 defi­ reversing Lenvatinib resistance in HCC may be accomplished by tar­
ciency reduces CSC tumorigenicity and controls the expression of a geting CD73 [72]. Studies also suggest that overexpressionof SOX-2may
number of cancer-regulating genes, including, c-MYC, NOTCH1, WNT1, be closely linked to the malignant transformation of ovarian tumors, as
and WNT2 [66]. Furthermore, downregulation of SOX-2 results in cell evidenced by the fact that the level of SOX-2 expression in human
cycle arrest, which ultimately leads to a reduced proliferation rate in ovarian tumors was directly related to the severity of the cancer [73]. As
glioma, lung and breast cancer cells [61,63]. Another recent study a result, SOX-2 promotes tumorigenesis and self-renewal in CSCs while
showed that SOX-2 is expressed in early breast cancers but OCT-3/4 and suppressing differentiation. Therefore, these innovative updates related
NANOG are not, indicating that a variety of variables at the level of to SOX-2 TF might aid in the future creation of effective cancer therapy.
translation and protein synthesis influence gene expression [66]. SOX-2
and DNA (cytosine-5) Methyltransferase 1 (DNMT1) predicted a poor 7. NANOG
prognosis for breast cancer. According to reports, the DNMT1/FOXO3a
(Forkhead box protein O3)/Forkhead box protein M1 (FOXM1)/SOX-2 NANOG is normally silent in normal somatic cells and is only
pathway has a significant role in controlling the characteristics of Breast expressed in germline fibroblasts, ESCs, and in a few cancer cells;
Cancer Stem Cells(BCSCs), providing prospective therapeutic targets for however, altered expression has been observed in lung cancer, breast
breast cancer [67]. SOX-2 overexpression caused mammosphere for­ cancer, brain cancer, cervical cancer, colon cancer, and gastric cancer
mation in xenograft tumor models, whereas SOX-2 knockdown inhibited [10,25]. Cell cycle arrest and apoptosis induced by NANOG knockdown
mammosphere formation and prolonged tumor formation in breast diminish gastric cancer cell growth and metastatic capacity [74].
cancer cell [68]. SOX-2 expression was induced following sphere for­ NANOG was discovered to have anti-tumorigenic properties in glio­
mation by activation of the distal enhancer of the SOX-2 promoter, the blastoma, prostate, and breast cancer cell lines [10,25,75]. Reports
same element that controls SOX-2 transcription in pluripotent stem cells. suggest that NANOG promotes chemo-resistance by increasing
By situating the tumor-initiating event in any cell along the axis of epithelial-mesenchymal transition and cell migration [76,77]. Similarly,
mammary differentiation, researchers can infer tumor heterogeneity in vivo, NANOG overexpression stimulates tumorigenicity and colony
and highlight SOX-2 reactivation as a key early stage in the development formation in colorectal CSCs [78,79]. Patients with gastric cancer who
of breast cancers [68]. Down regulated SOX-2 expression significantly have a high NANOG level are associated with a decreased 5-year sur­
reduced lung cancer cell metastasis and tumor growth [61,68]. CSCs are vival rate. NANOG expression is high in primary tumors such as HCC,
developed and maintained through the SOX-2 signaling pathway, which and it has been linked to disease progression (stage III/IV of tumor node
is active in CSCs produced from lung cancer cells [66]. It is still unclear metastasis (TNM)) [79,80]. In a study of prostatic cell lines, xenografts,
how the SOX-2 signaling pathway and the genes OCT-4 and NANOG and primary tumors, NANOG short hairpin RNA was discovered to
function in the growth and maintenance of CSCs. Previous reports sug­ inhibit the growth of initial cancer cells in the prostate (PCA) spheres,
gest that SOX-2 signaling is involved in the development of CSCs and tumorigenesis, and clonal growth [76,80,81].
that its deregulation can effectively suppress the metastasis and growth According to a Kaplan-Meier analysis of 43 pancreatic cancer tissue
of non-small cell lung carcinoma cells [61]. This TF may control onco­ microarray analyses, a high level of NANOG and OCT-4 expression was
gene expression in CSCs to aid in the progression of human lung cancer associated with a poor prognosis and a lower chance of survival for the
[66]. SOX-2 binds to specific DNA sequences and acts as transcription patient [82]. NANOG is a pro-tumorigenic factor that can be used as a
factors to either repress or activate target gene expression using their biomarker in the clinic to diagnose cancer and predict the prognosis and
highly conserved high mobility group [69,70]. In MCF-7/Rep (rapa­ efficacy of anticancer therapies. Researchers demonstrated that when
mycin resistant breast cancer) cells, the exogenous SOX-2 trans gene was human breast CSCs were tested using real-time PCR, they showed higher
silenced in tandem but the endogenous SOX-2 stemness gene was expression of NANOG, SOX-2, OCT-3/4,CD34, and Nestin [44]. Based on
distinctly reactivated according to the transcriptional analyses of OCT-4, the results of qPCR in breast cancer, they revealed that several CSC
SOX-2, KLF-4, and c-MYC transcription factors. Compared to MCF-7 transcriptional factors’ expression levels increased in accordance with
parental cells, some MCF-7/Rep cells, but not all of them, developed the disease’s stage. The gene expression levels, with the exception of

126
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

OCT-3/4, were increased in accordance with the stage. Less variation mitogen-activated protein kinase pathway [101]. KLF4 expression was
was seen between stages II-III, although between stages I and II and III found to be positively connected with CD9/CD81 expression and
and IV, it was higher [44,68]. Gynecologic tumorigenesis was found to negatively related to downstream MAPKinase/JNK(c-Jun N-terminal
be significantly impacted by NANOG overexpression [83]. According to Kinase) signaling, implying that future therapies should target the
immune histochemical analysis, NANOG andSOX-2 expression were KLF4-CD9/CD81-Jnk pathway [91].
mentioned as potential candidates for involvement in the initial stages of
breast cancer [68]. Scientists also revealed that NANOG expression was 9. SALL1 (Spalt like transcription factor 1)
linked to resistance to hormonal therapy and chemotherapy as well as a
worse prognosis [84]. Breast cancer metastasis and proliferation were The SALL family of zinc finger domains includes the cysteine-
linked to 70 genes, including NANOG, and it was illustrated that the histidine-containing proteins SALL-1, SALL-2, SALL-3,and SALL-4.
expression of these genes was connected to prognosis [85]. In light of SALL-1 is downregulated in malignancies like breast cancer, various
this, it appears that NANOG may function as a prognostic indicator both leukemias, and glioblastoma cells, which demonstrate its functions as a
at the protein and gene levels [86,87]. NANOG removal may also be a tumor suppressor [102,103]. Sall1 overexpression also suppresses the
therapeutic goal because it is thought to prevent CSC self-renewal and Wnt/-catenin signalling pathway by affecting the oncogene c-MYC and
identify the source of cancer recurrence and metastasis. Wnt downstream targets Cyclin D1 (Ccnd1) [10,104]. SALL-1 causes the
mesenchymal to epithelial transition by decreasing the mesenchymal
8. Kruppel-like factor 4 (KLF4) markers N-cadherin and vimentin while increasing the epithelial marker
E-cadherin [104]. Depending on the tissue, its genetic environment, the
Kruppel-like factor 4 (KLF4), associated with three zinc finger do­ control of epigenetic processes, and the particular SALL isoforms
mains on chromosome 9q31, is needed for the generation of cells with involved, SALL proteins can either act as oncogenes or tumor suppres­
induced pluripotency (iPSCs) [87]. KLF4 expression is lower in sors, or as proteins with a dual function [102]. Because isoforms differ in
cancerous tissues than in healthy ones, particularly in NSCLC (non-small structure, expression, and cellular localization, they may be in charge of
cell lung cancer), indicating tumor-suppressing property [88]. KLF4 has some of the various functions of several SALL proteins in cancer [102,
also been discovered to be a tumor suppressor in cancers of the digestive 105]. For the purpose of preserving the pluripotency of embryonic stem
system, where it has been connected to growth restriction by blocking cells, it has been proposed that direct communication with the central
the G1/S cell cycle [85,87,89]. KLF4 overexpression has been shown to master regulators SOX-2, OCT-4, and SALL4 is essential [76]. Recent
promote the growth of CSCs in osteosarcoma and act as a studies have shown that SALL4 regulates several AT-rich genes that
tumor-inducing gene in nasopharyngeal carcinoma [46]. Studies reveal encourage neuronal cell differentiation while maintaining the pluripo­
that KLF4 promotes the development of breast tumors, and 70% of tent state in ESCs [105]. Leukemia, testicular, breast, colon, ovarian, and
breast tumor cells have this TF upregulated [10,46]. In various tumor liver malignancies have all been found to express SALL4 [106–108].
cell lines, KLF4 has both oncogenic and tumor-suppressive properties. SALL4 expression has been investigated as a predictor for poor prognosis
KLF4 and the estrogen receptor (ER) directly interact with active p53, in hepatocellular carcinoma, gliomas, and myelodysplastic syndromes
and KLF4 regulates the expression of oncogenes connected to estrogen, [109].
showing that lowering KLF4 in breast cancer cells may boost the pro­
liferative response to estrogen [84]. Bladder cancer cells (T24) depleted 10. MYC
of METTL3 (m6A axis, a core “writer” protein in bladder cancer) were
used to show that KLF4 is a downstream target of METTL3(RNA meth­ In stem cells, MYC regulates a wide range of both protein-coding and
yltransferaseMETTL3) [90]. According to Nagata et al.a higher breast non-coding genes as well as various biological functions like cell dif­
cancer recurrence rate was linked to lower KLF4 expression [84]. Akaogi ferentiation, metabolism, and self-renewal [25]. A well-known tran­
et al. in 2009 reported that gene expression in breast cancer cells showed scription factor called MYC promotes somatic cell proliferation
reduced Klf4 expression as compared to healthy mammary gland cells effectively, and this property, along with the finding that the majority of
[92]. In breast cancer samples, it was possible to find the expression of human CSCs exhibit its deregulated expression, suggests that MYC might
several factors that induce stem cells, including KLF4,c-MYC, NANOG, make a potent therapeutic target. Human cancer cells frequently exhibit
OCT-4, and SOX-2. In contrast, KLF4 may have an inhibitory effect on deregulation of the oncogenic transcription factor c-MYC due to protein
the spread and proliferation of this malignancy [92]. Recent research stabilization, amplification, mutation, or over expression [110]. C-MYC
showed that KLF4 knockdown in MCF-7 cells accelerated the prolifera­ is a key transcriptional regulator in both malignancies and stem cells.
tion of these cells in the presence of oestrogen [91,92]. Scientists One of the most extensively researched oncogenes, C-MYC over­
investigated the relationship between KLF4 and ERalpha (Estrogen re­ expression, has been demonstrated to result in cancer in mouse models.
ceptor alpha) and discovered that KLF4 binds to ERalpha’s DNA-binding MYC is frequently deregulated in various human cancers and is among
region [93]. KLF4 prevents ERalpha from binding to promoter regions the most efficient oncogenes for determining the phenotype of cell
that contain oestrogen response elements, which lowers the transcrip­ transformation both in vitro and in vivo, and it helps in invasiveness as
tion of ERalpha target genes [92]. According to past findings, p53 well as in survival of CSCs [111]. Additionally, deactivating MYC in HCC
transcriptionally activates KLF4 following DNA damage [93]. It was also (Hepatocellular carcinoma) leads to a process by which stem cells
demonstrated that p53 activation increased KLF4 expression, which develop into biliary duct cells and hepatocytes, resulting in the forma­
decreased ERalpha’s transcriptional activity. p53, ERalpha, and KLF4are tion of bile duct anatomy, which may be connected to the loss of feto­
connected by a novel molecular network, as p53 and ERalpha both protein, a tumor marker, and elevated levels of the hepatocyte markers
contribute to cell proliferation and apoptosis [92,93]. Myeloma, leuke­ cytokeratin 8, cytokeratin 19, as well as liver stem cell markers [14,25].
mia, prostate, breast, brain, head and neck, oral, and testicular cancers Glioblastoma multiform stem cells also contain MYC, which promotes
have been reported to express KLF4 [94]. Additionally, the expression of invasion and cell proliferation while inhibiting apoptosis [112]. The
KLF4 can serve as a prognostic indicator for head and neck squamous number of copies of the MYC gene was also found to be higher in human
cell carcinoma and colon cancer[95–97]. Reports also suggest, nuclear and mouse prostate CSCs [25]. Reports demonstrate that when MYC is
localization of KLF4 has been linked to a worse prognosis in oral and deactivated in hepatocellular carcinoma, tumor cells begin to differen­
nasopharyngeal malignancies [98] as well as an aggressive phenotype in tiate and die in large numbers, but certain cells exhibit stem cell char­
early-stage breast cancer [99,100]. Raf-kinase inhibitor protein prevents acteristics and resume their ability to proliferate when MYC is activated
KLF4 from directly interacting with Raf-kinase inhibitor protein via [113,114]. The liver tumors in a mouse model regressed after MYC was
controlling the KLF4/OCT-4/SOX-2 complex through the inactivated, and the tumor dormancy continued for more than 8 months

127
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

[114]. The regulatory network of ovarian CSCs that suppresses genes reversion of transformed cancer cells’ totipotency or pluripotency [10,
linked to adhesion and the cell cycle includes MYC. These TFs-based 138,139]. YB-1 deletion may cause CSCs to differentiate, which would
therapies treat chemo resistance and recurrence of various cancers, reduce their capacity to form spheres and downregulate their stemness
including ovarian cancer and liver cancer, taking into account the genes. In contrast to published findings, YB-I rescue in YB-1 mutant CSCs
therapeutic importance of a quiescent tumor enriched with CSCs [114]. did not reprogramme the differentiated CSCs into stem cells [140,141].
Studies have shown that up to 70% of malignant cancers exhibit C-MYC It has been observed that the simultaneous presence of YB-1 and the
over expression, including leukemia and lymphoma as well as tumors of other four (SOX-2, OLIG1,POU3F2, and OCT-4) or five (SALL2,SOX-2,
the brain, pancreatic, breast, colon, renal, head, neck, prostate, testis, Bmi-1, POU3F2, and OCT-4) transcription factors prevented the con­
and salivary gland[94,115,116]. In early HCC and uterine cervix cancer, version of YB-1-deleted melanoma stem cells or breast CSCs into CSCs
C-MYC expression has also been linked to a poor prognosis [107,117]. [138]. Therefore, YB-1 is necessary for the transformation of differen­
This information suggests that MYC, in combination with other factors, tiated cancer cells into totipotent or pluripotent states [138]. For better
promotes tumorigenesis in conjunction with other factors. understanding, a pictorial representation of various regulatory mecha­
nisms associated with these TFs in CSCs for the modulation of various
11. STAT3 (Signal transducer and activator of transcription 3) tumors is shown in Fig. 2, and their comprehensive role is summarized in
Table 1.
STAT3 is widely expressed in gastric and colorectal CSCs, and it plays
a significant role in controlling a number of physiological activities, 15. Biomedical perspective of CSCs
including inflammation, proliferation, and invasion in CSCs [118–121].
High levels of STAT3 expression have been linked to the stemness and 15.1. Targeting transcription factors for therapeutic anti-tumor strategies
invasive features of gastric CSCs [120,121]. Numerous studies suggest in CSCs
that STAT3 activation is a marker of poor prognosis with regard to its
prognostic value [122–124]. One of the main oncogenic proteins Cancer therapies now focus on the TFs and signalling pathways that
involved in proliferation, angiogenesis, invasion, and resistance to control the maintenance and survival of CSCs. The OCT-4, SOX-2,
chemotherapy and radiation [125]. Additionally, its inhibition reduces NANOG, KLF4, MYC, Wnt, Notch, and Hhsignalling pathways, as well as
the development of tumors, making it a desirable therapeutic target for the TGF-, JAK-STAT, PI3K, and NF-kβ signalling pathways, are the pri­
CRC(colorectal cancer) [119]. mary TFs and signalling pathways at the moment. When tumors are
developing in CSCs, these TFs and pathways frequently interact. Early
12. GLI1 (GLI family zinc finger 1) clinical trials for OCT-4, SOX-2, NANOG, Notch, and Hh pathway in­
hibitors have seen significant advancements; however, it has proven
The Sonic Hedgehog (SHH) pathway member GLI1 appears to be challenging to target the Wnt pathway [25,142]. Fig. 3 depicts the
crucial for the maintenance of cancer cells with stem-like characteristics various therapeutic implications of targeting CSC transcription factors in
in various cancers like gastric cancer (GC) and colorectal cancer (CRC) cancer therapy. Combination therapies may be the focus of future trials.
[126,127]. Its expression in GC is much higher in tissues where the This field, however, is still in its early stages, and more research is
cancer has spread, and it is favorably linked with a more aggressive needed to produce some useful results.
tumor phenotype [126]. Additionally, it has been noted that GLI1 over
expression fosters a CSC phenotype by promoting cell motility, prolif­ 15.1.1. Available therapeutic strategies for targeting the TF
eration, and resistance to treatment [128]. GLI1 is also crucial for CSC The clinical potential of various transcription factors in CSCs is
traits associated with aggression and the metastatic spread of CRC cells, promising. These TFs function as crucial carcinogenesis markers [143]
which reduce survival. and as molecular switches that regulate the fate of CSC cells during the
course of cancer development [144]. A number of pluripotent tran­
13. SOX-9 (SRY transcription factor 9) scription factors, such as OCT-4, SOX-2, NANOG, KLF4, and MYC, as
well as some intracellular signalling pathways, like Wnt, NF-B, Notch,
Developmental processes like chondrogenesis, neurogenesis, and Hh, JAK-STAT, PI3K/AKT/mTOR, TGF/Smad, and PPAR, are crucial
neural crest development are regulated by SOX-9 [129]. Furthermore, it regulators of CSCs. Other regulatory extracellular factors, including
is required for the maintenance of stem cells and the determination of vascular niches, hypoxia, TAM, CAF, and exosomes, play a vital role in
cell fate in a variety of organs, including the digestive system, during CSC management. So to combat CSCs, researchers have also created
embryonic development and adulthood [130,131]. Its significance in GC medications, vaccines, antibodies, and CAR-T (chimeric antigen recep­
is still unclear; some studies support a link between reduced survival and tor T cell) cells that target these TFs, pathways, and exosomes [25,144].
high SOX-9 expression, while others link a poor prognosis to low SOX-9 Importantly, numerous CSC clinical trials have been conducted, and
expression [132,133]. SOX-9 has also been linked to CRC due to its they point to a bright future for cancer treatment [25]. A special class of
oncogenic and tumor suppressor activities [134,135]. Previous research therapeutic targets, including blockage of differentiation and cell death
suggests that this TF may be associated with a poor prognosis and may gene expression profiles, which are defining characteristics of malig­
regulate the CSC pool, which is the primary mechanism by which tumor nancies, is represented by mutated or deregulated transcription factors.
proliferation and progression are influenced [136,137]. Numerous direct mechanisms, such as chromosomal translocations,
gene amplification or deletion, point mutations, and altered expression,
14. YB-1 as well as indirect mechanisms involving non-coding DNA mutations
that affect transcription factor binding, result in altered transcription
According to previous studies, the transcription factor YB-1 is mainly factor activity in a variety of cancer types and can aid in the treatment of
linked to the stemness of stem cells in breast cancer and melanoma and CSCs [145]. Inhibition of transcription factor-cofactor protein-protein
is necessary for keeping CSCs stem-like and turning tumor cells back into interactions, inhibition of transcription factor-DNA binding, modulation
CSCs [138]. It is confirmed that YB-1 is involved in the effective pres­ of transcription factor activity by altering levels of ubiquitylation and
ervation of CSC properties [116]. According to prior research, YB-1 is subsequent proteasome degradation, as well as inhibition of transcrip­
required for both the preservation of CSC stemness and the trans­ tion factor expression regulators, are just a few methods that have been
formation of CSCs from tumor cells [10,138]. The expression of GLP-1, demonstrated to target transcription factor activity in preclinical and, in
GINS1, p21, Notch2, and FZD-1 is increased by YB-1, which also sustains some cases, clinical studies. These breakthroughs in drug research have
CSCs’ stemness, inhibits apoptosis, and is needed for the activation and a great chance of producing special compounds that will probably have

128
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

Fig. 2. A diagram illustrating how different CSC TFs are modulated in different cancer types. In order to promote stemness and tumor-propagating potential, these
transcription factors differentially regulate a network of regulatory mechanisms, which in turn drive the development of various types of cancer cells. The stemness of
CSCs is regulated by a number of molecular mechanisms that are brought about by these transcription factors.

an impact on how cancer is treated in the future [146]. It is possible to works as a mobilizer of hematopoietic stem cells in NHL and patients with
suppress tumors by limiting the expression of CSC TFs or by sabotaging multiple myeloma [152]. AMD3100 treatment for refractory or relapsed
the altered molecular pathways in CSCs [147]. Finally, the improved AML (acute myeloid leukemia) resulted in complete remission in 46% of
efficacy of clinical trials combining CSC TF modulators with chemo­ patients, regardless of white count recovery, in phase I/II research [153,
therapy has already suggested that the functional redundancy of CSC 154]. Musashi-1 (MSI1) and Musashi-2 (MSI2) are two RNA-binding
TFs may be overcome by combining TF modulators with or without proteins (RBPs) that are increasingly being recognized as regulators of
conventional cancer treatment. The use of cysteine-reactive inhibitors, numerous vital biological processes important for the development,
modulation of auto-inhibition, proteolysis targeting chimaeras (PRO­ progression, and resistance to cancer treatment. These proteins support
TACs), targeting intrinsically disordered regions of transcription factors, the populations of CSCs and control metastasis, invasion, and the emer­
and combining transcription factor inhibitors with kinase inhibitors to gence of more aggressive cancer traits, such as drug resistance. Despite
prevent the emergence of resistance are other recent developments in the fact that RBPs are challenging therapeutic targets, early attempts to
the field of targeting transcription factors [148]. produce specific inhibitors for MSI are encouraging, and strategies based
Over the last decade, cytotoxic T-lymphocyte-associated antigen 4 on RNA interference to inhibit these proteins have shown encouraging
(CTLA4), programmed death 1 (PD-1), and programmed death receptor results in preclinical trials against CSCs[155]. Transmembrane 4 L-six
ligand1 (PD-L1) have become more widely recognized [149,150]. In family member 1 (TM4SF1) has been shown to alter SOX-2 expression in a
patients suffering from advanced cancer, blocking TFshas produced sig­ Wnt/-catenin-dependent manner Furthermore, they reported that
nificant rates of response and demonstrated exceptional clinical out­ TM4SF1 knockdown reduced the binding of c-MYC tothe SOX-2 gene
comes [142,151]. CSC-specific surface biomarkers are targeted by promoter by suppressing c-MYC production, preventing metastasis and
monoclonal antibodies (mAbs), a new cancer therapy technology. To tumor growth in a xenograft mouse model [156]. CSC TFs, including
improve accessibility and affordability of radioimmunotherapy for re­ OCT-4, NANOG, and SOX-2, have been shown to be inhibited by the
fractory or recurring non-lymphoma Hodgkin’s (NHL), a phase II clinical vitamin B precursor fursultiamine TTFD (thiamine tetrahydrofurfuryl
trial for Rituximab radioiodine replacement was carried out, in which 35 disulfide), reducing their expression in esophageal carcinoma spheroids
patients were involved, and showed a good response rate of 71% and a and mouse xenografts [157]. Recently, it has been demonstrated that
total remission rate of 54%, with just 2 occurrences of grade IV hema­ circular RNA like Qki5-circZKSCAN1-FMRP-CCAR1targets theWnt­
tologic toxicity [25]. Chemokine receptors such as CXCR4 and SDF-1 signaling axis and could be a key therapeutic target for the treatment of
have been discovered in the vast majority of cancer cells, particularly CSCs in hepatocellular carcinoma(HCC). Results from RNA immune
CSCs. Plerixafor (AMD3100) is the most well-studied CXCR4 drug, and it precipitation-sequencing (RIP-seq) and bioinformatics analysis revealed

129
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

Table 1
A comprehensive summery of various TFs in the CSC population in different cancer types.The expression of TFs and an overview of different regulatory mechanisms
modulated by CSCs TFs in various tumors.
S. TFs Tumor types Expressional Phenotypes associated Molecular mechanisms Ref.
No analysis

1 OCT-4 Colorectal cancer Overexpression Progression of the cell cycle & Oct4/Tcl1/Akt1 pathway is activated. [47]
Apoptosis inhibition.
Cervical cancer Overexpression CSC-like phenotype. Upregulation of Akt1 pathway. [45]
Liver cancer Upregulation Increase neovascularization. Upregulation of epithelial-to- [50]
mesenchymal transition (EMT).
prostate cancer Upregulation Uncontrolled cell proliferation Downregulation of Ca2+channels, and [19,52]
CSC-like phenotype. upregulation of EMT, WNT/β,CATENIN,
and EGFR/PI3K/AKT pathways.
Squamous Cell Carcinoma of the Head and Upregulation Aggressive phenotype, inhibit Activation of Akt/mTORC1, and STAT3. [82]
Neck apoptosis.
Breast cancer Overexpression Increased tumor cell proliferation. Upregulation of WNT/ β-CATENIN, EMT [179,
and AMPK/mTOR pathways. 180]
Renal, Testis, Seminoma, Brain, Lung, Upregulation Self-renewal of stem cells and Activation of MAP4K4-Survivin, [181]
Bladder, Ovarian, and Leukemia preservation of pluripotency. downregulation of EGFR/Src/Akt and
BMP4 pathways.
2 SOX2 Gastric cancer Overexpression High proliferative and metastatic Upregulation of AKT signaling [159,
capacity, apoptosis and cell cycle 192,193]
arrest.
Squamous cell carcinoma Downregulation Decreased proliferation by cell Unknown [66]
cycle arrest.
Non-small cell lung cancer Overexpression Progression of the cell cycle & Activation of MAP kinase pathway and [182,
Apoptosis inhibition. down regulation of EGFR/Src/Akt 189]
pathways.
Breast cancer Overexpression Cell cycle progression, Apoptosis Upregulation of Wnt, EMT signaling and [183]
bypassing undifferentiated stat. downregulation of AMP kinase as well as
mTOR pathway
Colorectal Cancer Overexpression Proliferation, tumor burden, and Activation of BMP, MET,WNT/ [182,
high metastasis contribute to a poor β-CATENIN and downregulation of 185]
prognosis. mTOR
Neuroblastoma cells Overexpression Proliferation, tumor burden, Unknown [184]
metastasis, and a poor prognosis
Liver, Testis, and Prostate cancer. Upregulation Maintenance of pluripotency and Upregulation of EMT, WNT/ [73,178,
stem cell self-renewal. β-CATENIN,EGFR, PI3K and AKT 185,192,
pathways 193]
3 NANOG Hepatocellular carcinoma Elevated Cells invaded and induced EMT Activation of Akt/mTORC1, STAT3 [186]
expression with the high Vimentin/low E- pathway
Cadherin signature.
Breast cancer Elevated Anti-apoptotic proteins are Modulation of Hedgehog (Hh) & Notch [75]
expression upregulated and miR-21 levels are signaling pathways
increased.
Lung cancer Over expression Migration, colony formation, and Activation of MAP4K4 and AKT [61]
cycle progression. pathway.
Glioblastoma Over expression Proliferation, tumor burden, Modulation of Akt pathways. [187,
metastasis, and a poor prognosis. 188]
Head and Neck Squamous Cell Carcinoma Overexpression Aggressive phenotype, inhibit Unknown [24,77,
apoptosis. 187]
4 KLF-4 Colorectal Cancer Down regulation DNA synthesis has increased. Activation of the p38MAPK pathway [46]
Breast cancer Overexpression CSC-like phenotype of uncontrolled Activation of Wnt, Notch, TGF-β,and [87]
cell proliferation. Hedgehog pathways.
Nasopharyngeal carcinoma. Upregulation Increased tumor cell proliferation. Unknown [188]
Head and neck squamous cell carcinoma Upregulation Self-renewal of stem cells and Activation of MAP kinase pathway [190,
preservation of pluripotency. 191]
5 C-MYC Leukemia, Lymphoma, Myeloma, Brain, Overexpression Stem cell self-renewal. Modulation of Wnt-signaling pathways. [108,
Breast, Colon, Head and Neck, Pancreas, 115,116,
Prostate, Renal, Salivary-gland, testis 177]

that these circular RNAs exercised their inhibitory action by competi­ Wnt/-catenin and PI3K/AKT signalling pathways, both of which are
tively binding FMRP (Fragile X Messenger Ribonucleoprotein 1). This important regulators of normal intestinal growth and over activation
prevented FMRP from binding to the mRNA for cell cycle and apoptosis [161–164]. Additionally, it was shown that CD44 knockdown changed
regulator 1 (CCAR1), which in turn reduced Wnt’s signaling ability to breast CSCs into non-BCSCs with a drastic reduction in their
activate transcription. Additionally, it was shown that the RNA-splicing cancer-causing potential and also changed the cell cycle and expressional
protein Quaking 5 was downregulated in HCC tissues and was respon­ profile of steaminess-related genes and TFsso that they were more com­
sible for the decline in circZKSCAN1 [158]. Napabucasin is an orally parable to those seen in non-BCSCs. Therefore, knockdown of CD44 is a
administered naturally occurring drug that induces apoptosis in CSCs and successful tactic for reducing BCSC stemness, which causes stemness loss
inhibits CSC self-renewal in gastrointestinal malignancies by targeting and increases susceptibility to chemotherapy or radiation [165]. Re­
the transcription factors NANOG, STAT3, and -catenin signalling path­ searchers in recent years have also suggested that autophagy may be an
ways in CSCs [159,160]. Ina Phase I/II clinical trial with important element in the suppression of mTOR-mediated CSCs. Putative
FOLFOX-bevacizumab or FOLFOX, patients reported that the soy-derived esophageal CSCs may be removed with the help of mTOR pathway
compound genistein was well tolerated [116]. Similarly, genistein may stimulation. Additionally, the mTOR inhibitor Torin-1 promoted auto­
improve efficacy in the treatment of colorectal CSCs by targeting the phagy activity, while the mTOR stimulator MHY1485 inhibited

130
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

Fig. 3. Targeting transcription factors in CSCs


has therapeutic implications for cancer therapy.
A novel approach to cancer chemoprevention
and therapy may involve targeting CSCs TFs.
CSCs exhibit a variety of responses to survive
during and after therapy, such as EMT, self-
renewal, tumor environment, epigenetic,
quiescence modification, and targeting TFs that
modulate these pathways, which act as a game
changer in cancer therapeutics. TFs are known
to act on CSCs and the signalling pathways that
are involved in CSC self-renewal.

autophagy. In cells treated with the autophagy inhibitor hydroxy­ 16. Challenges for targeting TFs in cancer therapeutics
chloroquine, torin-1-mediated CSC induction was drastically decreased.
Esophageal cancer patient-derived organoids revealed a distinct Until now, such CSC-based therapies have only been performed in
CD44 + /CD24-CSC population, and MHY1485 also decreased this pop­ the laboratory, but more research is being done to improve CSC
ulation [166]. Thioridazine-loaded nanoparticles (Thz-HPPMc) could reprogramming and transcription factor-based therapies so that they
greatly enhance the cancer-suppressing impact, while hyaluronic acid may be used safely, precisely, and successfully in clinical settings.
(HA)-coated nanoparticles (HPMMc)could remain in the tumor site of Numerous methods, such as exosome-directed CSC reprogramming,
BALB/c nude mice for a longer time. These recent findings showed how small molecule, microRNA, and transcription factor-mediated cancer
effective the multifunctional nanoparticle technology was at getting rid stem cell reprogramming, have made great strides and shown great
of CSCs[167]. Additionally, after cisplatin treatment, a unique group of innovation, making them more adaptable and practical in both pre-
likely target genes associated with the Hedgehog pathway and angio­ clinical and clinical settings. Although clinical therapies on humans
genesis genes that are crucial for controlling chemo-resistance and remain technically and morally difficult, recent research suggests that a
self-renewability in triple negative breast cancer were only deregulated number of non-coding RNAs (ncRNAs), including long non-coding RNAs
in CSC-like cells. Researchers discovered that cisplatin is less harmful to (lncRNAs) and microRNAs (miRNAs), may regulate the transcription
CSC-like cells than to parental cells through analyses of cellular and gene factors and downstream signalling pathways that CSCs activate to con­
expression. Understanding the role of deregulated potential target genes trol their growth and replication [25]. Making links between these
produced by cisplatin in CSCs may help identify therapeutic targets for findings may help in the creation of innovative treatments that can
cisplatin-resistant breast cancer [168]. The combination of CDDP target CSCs with specificity and lower cancer recurrence rates [174].
(cis-diamminedichloroplatinum) and a carbon ion laser offers a greater Although numerous clinical studies on these CSCs have been attempted
ability to inhibit TN (triple negative) breast CSCs with heightened up to this point, many obstacles still need to be overcome before CSCs
apoptosis and severe, permanent DNA damage, as demonstrated by Sai can be effectively eliminated. Because of the vital role of CSCs in tumor
et al. in the year 2015. However, compared to X-ray paired with CDDP or formation and poor patient outcomes, approaches to CSC-specific
carbon ion beam alone, the combination of carbon ion beam and CDDP treatment have emerged as a significant new strategy for prospective
dramatically reduced colony and spheroid formation and slowed cell cancer therapeutics. However, targeting the CSC microenvironment for
cycle progression (sub-G1 arrest). After combining CDDP with either an therapeutic targets remains a challenge now.
X-ray or a carbon ion beam, the proportion of CSCs was higher and more
enriched [169,170]. Additionally, in their next report, they demonstrated 16.1. Potential solutions and future perspective
that, compared to an individual carbon ion or X-rays paired with gem­
citabine, a carbon ion beam has a higher potential to destroy pancreatic In order to develop treatments that may eliminate CSCs, we
CSCs through irreversible clustered DNA damage. Further, studies have comprehensively explore the effect of TFs and other components of
shown that the Chinese medicine beta-element can reduce the expression signaling pathways on cancer stemness in this paper, which will be able
of the genes for the cancer resistance proteins BCRP and P-glycoprotein to effectively detect and destroy these life-threatening CSCs. CSCs will
(P-gp), as well as the growth of BCSCs and the rate at which their spheres therefore be crucial as potential therapeutic targets in developing
form in the doxorubicin-resistant breast cancer cell line MCF-7/ADM cancer-controlling medications and improving patient clinical out­
[171]. Recently, scientists also discovered that the growth of breast comes. The discovery of innovative treatments for the complete elimi­
cancer has been found to be inhibited by phytoestrogens, and more nation of CSCs will improve treatment modalities, and this will require
recently, it has been revealed that they particularly inhibit breast CSCs. unlocking the biology of CSCs in carcinogenesis and metastasis. This
Among the natural compounds investigated, phytoestrogens, especially review helps in comprehending how harmful CSCs are and why man­
genistein, S-equol, naringenin, resveratrol, and pterostilbene,have aging them is difficult. This paper covers a number of subjects that are
distinguished themselves as significant players with the capacity to the subject of ongoing research. We focused on important transcrip­
inhibit BCSCs through a variety of pathways [172]. In breast cancer cell tional regulators and signaling channels that are known to regulate CSC
lines of non-hereditary origin, olaparib, a recognized inhibitor of poly behavior and interact in a variety of intricate ways. The transcriptional
ADP ribose, an enzyme primarily involved in DNA repair, exhibits networks that control CSCs are still not fully understood, and some
anti-CSC action, possibly through activation of p-ERK [173]. findings are controversial. In order to further understand and charac­
terize these regulatory processes, particularly through network analysis,
novel therapeutic targets may be found.

131
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

Small populations of CSCs create cellular heterogeneity, which is pluripotency of CSC transcription factors are getting more and more
linked to a long cell life span, relative quiescence, and treatment resis­ complex. Both OCT-4 and SOX-2 exhibit real oncogenic behavior in
tance. This leads to cancer that is hard to treat, or "refractory." Due to some cancers. Contrarily, CSCs appear to be physiologically quite
their ability to self-renew and resistance to drugs, CSCs allow cancer diverse within individual tumors as well as between various tumor
cells to grow and change even after they have been treated with anti­ types. Even though pluripotency transcription factors are definitely
cancer drugs. There is not a single marker that can be used to recognize linked to cancer stemness, it does not look like they cover the whole
all CSCs. In addition, cancer cells become more plastic during metastasis range of this stemness. Some CSCs preserve their stemness through
in different types of malignancy. It is critical to comprehend the dif­ biological processes that are entirely dissimilar from pluripotency
ferences between CSCs and normal stem cells. The above-mentioned TFs stemness, occasionally even by activating signaling pathways that
in this review will definitely help us to separate, recognize, and examine encourage pluripotent stem cell development. Unexpected side effects
these populations within heterogeneous tumors. Examining these are regularly experienced when pluripotency transcription factors are
characteristic TFs will aid in gene expression profiling, which offers forced to express in cancer cells, including partial epigenetic normali­
essential information that enables patient prognosis prediction. Clari­ zation, phenotypic reversion, and loss of the potential to initiate tumors.
fying CSC characteristics could help in the creation of cancer therapies The recurring tumorigenicity loss observed in induced pluripotent can­
that take into account the tumor microenvironment and molecular cer cells may be due to the substantial dose dependence of reprogram­
subtypes. ming pluripotency factors in addition to the extremely different
signaling contexts that are active in pluripotent and CSCs, respectively.
17. Discussion Finally, during pluripotency (cancer) reprogramming, numerous
signaling molecules have opposing cell-autonomous and
The cancer disease continuously develops over time and has been non-cell-autonomous effects. Therefore, the idea of CSC heterogeneity
threatening human life for over a decade. As a result of the development provides the greatest justification for the impact of pluripotency tran­
of numerous cancer treatment options, including chemotherapy, radia­ scription factors in cancer [178]. However, it is still unclear what
tion, and surgery, the incidence rate of cancer in women has largely transcriptional processes control the formation of CSCs that are tolerant
remained stable while slightly declining in men over the past ten years to harsh therapies. The in vivo therapeutic promise of TFs-based CSC
(2006–2015), and the death rate from cancer has also decreased therapy will ultimately be confirmed in the future through testing on
(2007–2016) [14,25]. Conventional cancer therapy is only effective for mouse models for efficiency, safety, and specificity, as well as other
a small subset of malignant tumors due to metastasis, heterogeneity, animal models. The in vivo dose and distribution, as well as the insta­
recurrence, resistance to radio/chemotherapy, and avoidance of bility of CSCs and potential off-target consequences, provide challenges
immunological surveillance. The existence of CSCs could be the cause of for CSCs’ complete inhibition. CSC reprogramming obstacles are
all of these failures. By arresting in the G0 phase and forming new tu­ currently preventing the promising strategy from being translated to
mors, CSCs can cause multidrug resistance, cancer relapse, and radiation therapeutic applications. However, based on the constantly expanding
resistance [9139]. As a result, CSCs may be considered the most prom­ horizon in CSC biology, they appear to be solvable. The significance of
ising cancer treatment targets. With a better understanding of CSC transcription factors in CSC biology, as well as their role in the devel­
characteristics, tumor biology research has entered a new era. Under­ opment of various cancers and drug resistance, is summarized in this
standing CSC biological properties is therefore crucial for tumor diag­ article. In order to better understand how transcriptional regulation of
nosis and treatment. Since their discovery in leukemia in 1994, TF different cancers affects the growth and development of a tumor as well
targets for cancer modulation have been thought to be a promising as its cell resistance to therapy, we looked at all of the major tran­
approach for cancer therapy [25]. There are transcriptional factors at scriptional regulators. Our findings potentially lay the groundwork for
play in a variety of human disorders, including cancer, where they make novel therapies for the removal of metastatic cancer and CSCs, and they
up about 20% of all oncogenes currently known [175,176]. Finding the also contribute to a deeper understanding of the characteristics of CSCs
processes underlying drug resistance is a key objective in cancer and suggest novel ways to use targeted therapy in cancer patients. There
research, and new studies have linked CSCs to intrinsic resistance have been many clinical trials on these CSCs, suggesting that the future
models. The pathological self-renewal features of CSCs may be influ­ of cancer therapy is promising. However, before CSCs can be effectively
enced by overexpression of stem cell-specific transcription factors, while eliminated, a number of challenges must be surmounted.
surface molecules mediate intercellular interactions and their sur­
roundings. Other stemness-related indicators and mechanisms might 18. Conclusion and future prospects
encourage the growth, development, and metastasis of cancer cells.
Understanding the stemness-related characteristics of cancers will not It is now obvious that the ability of CSCs to self-renew and metas­
only shed new light on the development of effective therapeutic stra­ tasize cannot be defeated by conventional chemotherapy. CSCs have
tegies that specifically target these stemness-related characteristics but been recognized and isolated using a combination of transcription fac­
will also contribute to our understanding of the underlying molecular tors and their functional characteristics. Despite this development, there
pathways in the pathogenesis of cancer.SOX-2, OCT-4, NANOG, MYC, is still a lack of precise CSC TFs to target this diverse cancer etiology.
andKLF4 are just a few of the pluripotent transcription factors that This must be solved to create therapeutic approaches with more speci­
control the biological activity of CSCs. Numerous extracellular factors, ficity and fewer adverse effects. In various pre-clinical models, targeting
such as hypoxia, the vascular niche, tumor-associated macrophages, CSC-associated oncogenes and signaling pathways, which were dis­
exosomes, the extracellular matrix, cancer-associated mesenchymal cussed above with small molecule inhibitors or therapeutic targets, led
stem cells, and cancer-associated fibroblasts, have all been implicated in to decreased CSC functionality and numbers, as well as tumor remission.
the development of cancer. Several intracellular signalling pathways, CSCs become resistant to traditional chemotherapeutics. However,
including Notch, Wnt, JAK-STAT, NF-kβ, Hedgehog, AKT/mammalian desensitizing CSCs to those same medications involves targeting various
target of rapamycin, and phosphoinositis, are also altered in cancer [25]. TFs. The application of therapies, including regulation of CSC-specific
It makes sense that certain embryonic transcription factors would be TFs, alone and along with traditional therapy will result in promising
reactivated or re-expressed in CSCs [177]. As a result, these transcrip­ and enhanced CSC targeting effects in various tumor types, as shown in
tion factors are crucial in controlling CSC development. Numerous nu­ Fig. 4 graphical abstract. Anti-CSC strategies may be able to specifically
clear signaling channels and transcription factors control how different promote differentiation and prevent proliferation. Targeting CSCs by
cell types develop tumors. Although these CSC TFs are typically well altering these TFs and the tumor microenvironment has produced pos­
established as tumor-promoting factors, the clinical implications of the itive outcomes. TFs alone might not be effective enough to yield clinical

132
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

Fig. 4. Role of transcription factors in the therapeutic strategies of CSCs. CSCs is mainly targeted by different transcription factors that regulate various signaling
pathways, surface markers and gene regulations. These TFs also induces various inhibitors of ATP binding cassette transporters, immune evasions, and tumor
microenvironment which ultimately leads to the induction of CSCs apoptosis and thus inhibits tumor proliferation and self-renewal properties.

results. Consequently, combination therapy using both conventional and [10] A. Liu, X. Yu, S. Liu, Pluripotency transcription factors and cancer stem cells:
small genes make a big difference, Chin. J. Cancer 32 (2013) 483–487, https://
anti-CSC drugs may be relevant in the future to enhance the efficacy of
doi.org/10.5732/cjc.012.10282.
treatment. [11] M.L. Wang, S.H. Chiou, C.W. Wu, Targeting cancer stem cells: emerging role of
Nanog transcription factor, OncoTargets Ther. 6 (2013) 1207–1220, https://doi.
org/10.2147/OTT.S38114.
Acknowledgments [12] A. Marusyk, V. Almendro, K. Polyak, Intra-tumour heterogeneity: a looking glass
for cancer, Nat. Rev. Cancer 12 (2012) 323–334, https://doi.org/10.1038/
The work was supported by grant No. EMR/2017/004171 from DST- nrc3261.
[13] W.A. Da Silveira, P. Palma, R.D. Sicchieri, R. Villacis, Mandarano, T. Oliveira, et
SERB (Science and Engineering Research Board) India. The first author al., Transcription factor networks derived from breast cancer stem cells control
thanks the Council for Scientific and Industrial Research (CSIR-UGC) for the immune response in the basal subtype, Sci. Rep. 7 (2017) 2851, https://doi.
providing fellowship. org/10.1038/s41598-017-02761-6.
[14] P. Valent, D. Bonnet, R. De Maria, T. Lapidot, M. Copland, J.V. Melo, et al.,
Cancer stem cell definitions and terminology: the devil is in the details, Nat. Rev.
Cancer 12 (2012) 767–775, https://doi.org/10.1038/nrc3368.
Competing interests [15] M. Lambert, S. Jambon, S. Depauw, M.H. David-Cordonnier, Targeting
transcription factors for cancer treatment, Molecules 23 (6) (2018) 1479, https://
doi.org/10.3390/molecules23061479.
The authors declare no competing interest. [16] A.S. Bhagwat, C.R. Vakoc, Targeting transcription factors in cancer, Trends
Cancer 1 (1) (2015) 53–65, https://doi.org/10.1016/j.trecan.2015.07.001.
[17] M. Didiasova, L. Schaefer, M. Wygrecka, Targeting GLI transcription factors in
Authors contribution cancer, Molecules 23 (5) (2018) 1003, https://doi.org/10.3390/
molecules23051003.
[18] J.E. Carpentino, M.J. Hynes, H.D. Appelman, T. Zheng, D.A. Steindler, E.W. Scott,
Rizwanul Haque contributed to the study’s conception and design. et al., Aldehyde dehydrogenase-expressing colon stem cells contribute to
Formal analysis and investigation were carried out by Archana tumorigenesis in the transition from colitis to cancer, Cancer Res. 69 (20) (2009)
8208–8215, https://doi.org/10.1158/0008-5472.CAN-09-1132.
Chaudhary. Archana Chaudhary drafted the manuscript. RizwanulHa­ [19] F. BaghaiNaini, P. Aminishakib, A. Abdollahi, M. Hodjat, H. Mohammadpour,
que and Syed Shadab Raza reviewed and edited the previous version of N. KardouniKhoozestani, et al., Relative expression of OCT4, SOX2 and NANOG
the manuscript. All authors read and approved the final manuscript. in oral squamous cell carcinoma versus adjacent non- tumor tissue, Asian Pac. J.
Cancer Prev. 20 (6) (2019) 1649–1654, https://doi.org/10.31557/
APJCP.2019.20.6.1649.
References [20] F. Yang, P. Cui, Y. Lu, X. Zhang, Requirement of the transcription factor YB-1 for
maintaining the stemness of cancer stem cells and reverting differentiated cancer
[1] Nancy A. Speck, A pernicious cycle affecting premalignant stem cells, N. Engl. J. cells into cancer stem cells, Stem Cell Res. Ther. 10 (1) (2019) 233, https://doi.
Med. 386 (2022) 596–598, https://doi.org/10.1056/NEJMcibr2117528. org/10.1186/s13287-019-1360-4.
[2] Z. Yu, T.G. Pestell, M.P. Lisanti, R.G. Pestell, Cancer stem cells, Int. J. Biochem. [21] F. Peng, J.H. Wang, W.J. Fan, Y.T. Meng, M.M. Li, T.T. Li, et al., Glycolysis
Cell Biol. 44 (12) (2012) 2144–2151, https://doi.org/10.1016/j. gatekeeper PDK1 reprograms breast cancer stem cells under hypoxia, Oncogene
biocel.2012.08.022. 37 (8) (2018) 1062–1074, https://doi.org/10.1038/onc.2017.368.
[3] J.L. Guan, A.K. Simon, M. Prescott, J.A. Menendez, F. Liu, F. Wang, C. Wang, [22] M. Fane, L. Harris, A.G. Smith, M. Piper, Nuclear factor one transcription factors
E. Wolvetang, A. Vazquez-Martin, J. Zhang, Autophagy in stem cells, Autophagy as epigenetic regulators in cancer, Int. J. Cancer 140 (12) (2017) 2634–2641,
9 (6) (2013) 830–849, https://doi.org/10.4161/auto.24132. https://doi.org/10.1002/ijc.30603.
[4] A.I. Riggio, K.E. Varley, A.L. Welm, The lingering mysteries of metastatic [23] M. Galoczova, P. Coates, B. Vojtesek, STAT3, stem cells, cancer stem cells and
recurrence in breast cancer, Br. J. Cancer 124 (2021) 13–26, https://doi.org/ p63, Cell. Mol. Biol. Lett. 23 (2018) 12, https://doi.org/10.1186/s11658-018-
10.1038/s41416-020-01161-4. 0078-0.
[5] T.M. Sissung, D.F. William, Stem cell clinics: risk of proliferation, Lancet Oncol. [24] J. Chen, Y. Li, T.S. Yu, R.M. McKay, D.K. Burns, S.G. Kernie, et al., A restricted cell
21 (2020) 205–206, https://doi.org/10.1016/S1470-2045(19)30787-9. population propagates glioblastoma growth after chemotherapy, Nature 488
[6] L. Phi, I.N. Sari, Y.G. Yang, S.H. Lee, N. Jun, K.S. Kim, et al., Cancer Stem Cells (7412) (2012) 522–526, https://doi.org/10.1038/nature11287.
(CSCs) in drug resistance and their therapeutic implications in cancer treatment, [25] L. Yang, P. Shi, G. Zhao, J. Xu, W. Peng, J. Zhang, et al., Targeting cancer stem
Stem Cells Int. (2018) 5416923, https://doi.org/10.1155/2018/5416923. cell pathways for cancer therapy, Signal Transduct. Target. Ther. 5 (1) (2020) 8,
[7] I. Baccelli, A. Trumpp, The evolving concept of cancer and metastasis stem cells, https://doi.org/10.1038/s41392-020-0110-5.
J. Cell Biol. 198 (2012) 281–293, https://doi.org/10.1083/jcb.201202014. [26] J. Maddox, A. Shakya, S. South, D. Shelton, J.N. Andersen, S. Chidester, et al.,
[8] Z. Yu, T.G. Pestell, M.P. Lisanti, R.G. Pestell, Cancer stem cells, Int. J. Biochem. Transcription factor Oct1 is a somatic and cancer stem cell determinant, PLoS
Cell Biol. 44 (2012) 2144–2151, https://doi.org/10.1016/j.biocel.2012.08.022. Genet 8 (11) (2012), e1003048, https://doi.org/10.1371/journal.pgen.1003048.
[9] D.R. Pattabiraman, R.A. Weinberg, Tackling the cancer stem cells - what [27] A. Almotiri, H. Alzahrani, J.B. Menendez-Gonzalez, A. Abdelfattah, B. Alotaibi,
challenges do they pose? Nat. Rev. Drug Discov. 13 (2014) 497–512, https://doi. L. Saleh, A. Greene, et al., Zeb1 modulates hematopoietic stem cell fates required
org/10.1038/nrd4253.

133
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

for suppressing acute myeloid leukemia, J. Clin. Investig. 131 (1) (2021), [51] W. Li, Y. Zhou, X. Zhang, Y. Yang, S. Dan, T. Su, et al., Dual inhibiting OCT4 and
e129115, https://doi.org/10.1172/JCI129115. AKT potently suppresses the propagation of human cancer cells, Sci. Rep. 7
[28] J.L. Huang, M. Oshi, I. Endo, K. Takabe, Clinical relevance of stem cell surface (2017) 46246, https://doi.org/10.1038/srep46246.
markers CD133, CD24, and CD44 in colorectal cancer, Am. J. Cancer Res. 11 (10) [52] H.L. Liu, H.T. Tang, H.L. Yang, T.T. Deng, Y.P. Xu, S.Q. Xu, et al., Oct4 regulates
(2021) 5141–5154. the transition of cancer stem-like cells to tumor endothelial-like cells in human
[29] D. Wang, H. Zhu, Y. Zhu, Y. Liu, H. Shen, R. Yin, et al., CD133(+)/CD44(+)/Oct4 liver cancer, Front. Cell Dev. Biol. 8 (2020), 563316, https://doi.org/10.3389/
(+)/Nestin(+) stem-like cells isolated from Panc-1 cell line may contribute to fcell.2020.563316.
multi-resistance and metastasis of pancreatic cancer, Acta Histochem. 115 (4) [53] I. Ben-Porath, M.W. Thomson, V.J. Carey, R. Ge, G.W. Bell, A. Regev, et al., An
(2013) 349–356, https://doi.org/10.1016/j.acthis.2012.09.007. embryonic stem cell-like gene expression signature in poorly differentiated
[30] J. Neradil, R. Veselska, Nestin as a marker of cancer stem cells, Cancer Sci. 106 aggressive human tumors, Nat. Genet. 40 (5) (2008) 499–507, https://doi.org/
(7) (2015) 803–811, https://doi.org/10.1111/cas.12691. 10.1038/ng.127.
[31] S. Cufí, A. Vazquez-Martin, C. Oliveras-Ferraros, B. Martin-Castillo, L. Vellon, J. [54] X. Wang, J. Dai, Concise review: isoforms of OCT4 contribute to the confusing
A. Menendez, Autophagy positively regulates the CD44(+) CD24(-/low) breast diversity in stem cell biology, Stem Cells 28 (5) (2010) 885–893, https://doi.org/
cancer stem-like phenotype, Cell Cycle 10 (22) (2011) 3871–3885, https://doi. 10.1002/stem.419.
org/10.4161/cc.10.22.17976. [55] C.G. Liu, Y. Lu, B.B. Wang, Y.J. Zhang, R.S. Zhang, Y. Lu, et al., Clinical
[32] K. Narita, Y. Matsuda, M. Seike, Z. Naito, A. Gemma, T. Ishiwata, Nestin regulates implications of stem cell gene Oct-4 expression in breast cancer, Ann. Surg. 253
proliferation, migration, invasion and stemness of lung adenocarcinoma, Int. J. (6) (2011) 1165–1171, https://doi.org/10.1097/SLA.0b013e318214c54e.
Oncol. 44 (4) (2014) 1118–1130, https://doi.org/10.3892/ijo.2014.2278. [56] J. Takeda, S. Seino, G.I. Bell, Human Oct3 gene family: cDNA sequences,
[33] P.R. Pandey, J. Saidou, K. Watabe, Role of myoepithelial cells in breast tumor alternative splicing, gene organization, chromosomal location, and expression at
progression, Front. Biosci. 15 (1) (2010) 226–236, https://doi.org/10.2741/ low levels in adult tissues, Nucleicacidsresearch 20 (17) (1992) 4613–4620,
3617. https://doi.org/10.1093/nar/20.17.4613.
[34] Q.Z. He, X.Z. Luo, Q. Zhou, K. Wang, S.X. Li, Y. Li, et al., Expression of nestin in [57] A.G. Schepers, H.J. Snippert, D.E. Stange, M. van den Born, J.H. van Es, M. van de
ovarian serous cancer and its clinicopathologic significance, Eur. Rev. Med. Weterin, et al., Lineage tracing reveals Lgr5+ stem cell activity in mouse
Pharmacol. Sci. 17 (21) (2013) 2896–2901. intestinal adenomas, Science 337 (6095) (2012) 730–735, https://doi.org/
[35] J.R. Parfitt, C.A. McLean, M.G. Joseph, C.J. Streutker, S. Al-Haddad, D.K. Driman, 10.1126/sci.
Granular cell tumours of the gastrointestinal tract: expression of nestin and [58] L.J. Ling, S. Wang, X.A. Liu, E.C. Shen, Q. Ding, C. Lu, et al., A novel mouse model
clinicopathological evaluation of 11 patients, Histopathology 48 (4) (2006) of human breast cancer stem-like cells with high CD44+CD24-/lower phenotype
424–430, https://doi.org/10.1111/j.1365-2559.2006.02352.x. metastasis to human bone, Chin. Med. J. 121 (20) (2008) 1980–1986.
[36] K. Sakurada, M. Saino, W. Mouri, A. Sato, C. Kitanaka, T. Kayama, Nestin [59] L. You, X. Guo, Y. Huang, Correlation of cancer stem-cell markers OCT4, SOX2,
expression in central nervous system germ cell tumors, Neurosurg. Rev. 31 (2) and NANOG with clinicopathological features and prognosis in operative patients
(2008) 173–177, https://doi.org/10.1007/s10143-007-0115-3. with rectal cancer, Yonsei Med. J. 59 (1) (2018) 35–42, https://doi.org/10.3349/
[37] I. Zambo, M. Hermanova, D. AdamkovaKrakorova, P. Mudry, K. Zitterbart, ymj.2018.59.1.35.
M. Kyr, et al., Nestin expression in high-grade osteosarcomas and its clinical [60] S. Lee, S. Wottrich, B. Bonavida, Crosstalks between Raf-kinase inhibitor protein
significance, Oncol. Rep. 27 (5) (2012) 1592–1598, https://doi.org/10.3892/ and cancer stem cell transcription factors (Oct4, KLF4, Sox2, Nanog), Tumour
or.2012.1687. Biol.: J. Int. Soc. Oncodev. Biol. Med. 39 (4) (2017), https://doi.org/10.1177/
[38] A. Nambirajan, M.C. Sharma, R.K. Gupta, V. Suri, M. Singh, C. Sarkar, Study of 1010428317692253.
stem cell marker nestin and its correlation with vascular endothelial growth [61] R. Xiang, D. Liao, T. Cheng, H. Zhou, Q. Shi, T.S. Chuang, et al., Downregulation
factor and microvascular density in ependymomas, Neuropathol. Appl. of transcription factor SOX2 in cancer stem cells suppresses growth and
Neurobiol. 40 (6) (2014) 714–725, https://doi.org/10.1111/nan.12097. metastasis of lung cancer, Br. J. Cancer 104 (9) (2011) 1410–1417, https://doi.
[39] M.N. Sabet, A. Rakhshan, E. Erfani, Z. Madjd, Co-expression of putative cancer org/10.1038/bjc.2011.94.
stem cell markers, CD133 and Nestin, in skin tumors, Asian Pac. J. Cancer Prev.: [62] S. Liu, Y. Cong, D. Wang, Y. Sun, L. Deng, Y. Liu, et al., Breast cancer stem cells
APJCP 15 (19) (2014) 8161–8169, https://doi.org/10.7314/ transition between epithelial and mesenchymal states reflective of their normal
apjcp.2014.15.19.8161. counterparts, Stem Cell Rep. 2 (1) (2013) 78–91, https://doi.org/10.1016/j.
[40] T. Liu, F. Xu, X. Du, D. Lai, T. Liu, Y. Zhao, et al., Establishment and stemcr.2013.11.009.
characterization of multi-drug resistant, prostate carcinoma-initiating stem-like [63] Z.H. Ren, C.P. Zhang, T. Ji, Expression of SOX2 in oral squamous cell carcinoma
cells from human prostate cancer cell lines 22RV1, Mol. Cell. Biochem. 340(1-2) ( and the association with lymph node metastasis, Oncol. Lett. 11 (3) (2016)
(2010) 265–273, https://doi.org/10.1007/s11010-010-0426-5. 1973–1979, https://doi.org/10.3892/ol.2016.4207.
[41] C.J. Shi, J. Gao, M. Wang, X. Wang, R. Tian, F. Zhu, et al., CD133 (+) gallbladder [64] J. He, J. Shi, K. Zhang, J. Xue, J. Li, J. Yang, et al., Sox2 inhibits Wnt-β-catenin
carcinoma cells exhibit self-renewal ability and tumorigenicity, World J. signaling and metastatic potency of cisplatin-resistant lung adenocarcinoma cells,
Gastroenterol. 17 (24) (2011) 2965–2971, https://doi.org/10.3748/wjg.v17. Mol. Med. Rep. 15 (4) (2017) 1693–1701, https://doi.org/10.3892/
i24.2965. mmr.2017.6170.
[42] C. Liu, X. Cao, Y. Zhang, H. Xu, R. Zhang, Y. Wu, Co-expression of Oct-4 and [65] S. Zhang, X. Xiong, Y. Sun, Functional characterization of SOX2 as an anticancer
Nestin in human breast cancers, Mol. Biol. Rep. 39 (5) (2012) 5875–5881, target, Signal Transduct. Target. Ther. 5 (1) (2020) 135, https://doi.org/
https://doi.org/10.1007/s11033-011-1398-6. 10.1038/s41392-020-00242-3.
[43] Z. Zhao, P. Lu, H. Zhang, H. Xu, N. Gao, M. Li, et al., Nestin positively regulates [66] S. Chen, Y. Xu, Y. Chen, X. Li, W. Mou, L. Wang, et al., SOX2 gene regulates the
the Wnt/β-catenin pathway and the proliferation, survival and invasiveness of transcriptional network of oncogenes and affects tumorigenesis of human lung
breast cancer stem cells, Breast Cancer Res.: BCR 16 (4) (2014) 408, https://doi. cancer cells, PLoS One 7 (5) (2012), e36326, https://doi.org/10.1371/journal.
org/10.1186/s13058-014-0408-8. pone.0036326.
[44] P. Apostolou, M. Toloudi, M. Chatziioannou, E. Ioannou, I. Papasotiriou, Cancer [67] H. Liu, Y. Song, H. Qiu, Y. Liu, K. Luo, Y. Yi, et al., Downregulation of FOXO3a by
stem cells stemness transcription factors expression correlates with breast cancer DNMT1 promotes breast cancer stem cell properties and tumorigenesis, Cell
disease stage, Curr. Stem Cell Res. Ther. 7 (6) (2012) 415–419, https://doi.org/ Death Differ. 27 (3) (2020) 966–983, https://doi.org/10.1038/s41418-019-0389-
10.2174/157488812804484639. 3.
[45] Y.D. Wang, N. Cai, X.L. Wu, H.Z. Cao, L.L. Xie, P.S. Zheng, OCT4 promotes [68] O. Leis, A. Eguiara, E. Lopez-Arribillaga, M.J. Alberdi, S. Hernandez-Garcia,
tumorigenesis and inhibits apoptosis of cervical cancer cells by miR-125b/BAK1 K. Elorriaga, et al., Sox2 expression in breast tumours and activation in breast
pathway, Cell Death Dis. 4 (8) (2013), e760, https://doi.org/10.1038/ cancer stem cells, Oncogene 31 (11) (2012) 1354–1365, https://doi.org/
cddis.2013.272. 10.1038/onc.2011.338.
[46] X.T. Qi, Y.L. Li, Y.Q. Zhang, T. Xu, B. Lu, L. Fang, et al., KLF4 functions as an [69] Y. Kamachi, M. Uchikawa, H. Kondoh, Pairing SOX off: with partners in the
oncogene in promoting cancer stem cell-like characteristics in osteosarcoma cells, regulation of embryonic development, Trends Genet. 16 (4) (2000) 182–187,
ActapharmacologicaSinica 40 (4) (2019) 546–555, https://doi.org/10.1038/ https://doi.org/10.1016/s0168-9525(99)01955-1.
s41401-018-0050-6. [70] M. Wilson, P. Koopman, Matching SOX: partner proteins and co-factors of the
[47] Y.J. Wang, M. Herlyn, The emerging roles of Oct4 in tumor-initiating cells, Am. J. SOX family of transcriptional regulators, Curr. Opin. Genet. Dev. 12 (4) (2002)
Physiol., Cell Physiol. 309 (11) (2015) C709–C718, https://doi.org/10.1152/ 441–446, https://doi.org/10.1016/s0959-437x(02)00323-4.
ajpcell.00212.2015. [71] B. Corominas-Faja, S. Cufí, C. Oliveras-Ferraros, E. Cuyàs, E. López-Bonet,
[48] Y. Pekarsky, A. Koval, C. Hallas, R. Bichi, M. Tresini, S. Malstrom, et al., Tcl1 R. Lupu, et al., Nuclear reprogramming of luminal-like breast cancer cells
enhances Akt kinase activity and mediates its nuclear translocation, Proc. Natl. generates Sox2-overexpressing cancer stem-like cellular states harboring
Acad. Sci. USA 97 (7) (2000) 3028–3033, https://doi.org/10.1073/ transcriptional activation of the mTOR pathway, Cell Cycle 12 (18) (2013)
pnas.97.7.3028. 3109–3124, https://doi.org/10.4161/cc.26173.
[49] J. Wang, H. Zeng, H. Li, J. Zhang, S. Wang, Roles of sex-determining region Y-box [72] X.L. Ma, B. Hu, W.G. Tang, S.H. Xie, N. Ren, L. Guo, et al., CD73 sustained cancer-
2 in cell pluripotency and tumor-related signaling pathways, Mol. Clin. Oncol. 3 stem-cell traits by promoting SOX9 expression and stability in hepatocellular
(6) (2015) 1203–1207, https://doi.org/10.3892/mco.2015.639. carcinoma, J. Hematol. Oncol. 13 (1) (2020) 11, https://doi.org/10.1186/
[50] X.Q. Wang, W.M. Ongkeko, L. Chen, Z.F. Yang, P. Lu, K.K. Chen, et al., Octamer 4 s13045-020-0845-z.
(Oct4) mediates chemotherapeutic drug resistance in liver cancer cells through a [73] F. Ye, Y. Li, Y. Hu, C. Zhou, Y. Hu, H. Chen, Expression of Sox2 in human ovarian
potential Oct4-AKT-ATP-binding cassette G2 pathway, Hepatology 52 (2) (2010) epithelial carcinoma, J. Cancer Res. Clin. Oncol. 137 (1) (2011) 131–137, https://
528–539, https://doi.org/10.1002/hep.23692. doi.org/10.1007/s00432-010-0867-y.

134
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

[74] N. Gawlik-Rzemieniewska, I. Bednarek, The role of NANOG transcriptional factor and neck squamous cell carcinoma, Cancer Sci. 102 (4) (2011) 895–902, https://
in the development of malignant phenotype of cancer cells, Cancer Biol. Ther. 17 doi.org/10.1111/j.1349-7006.2011.01859.x.
(1) (2016) 1–10, https://doi.org/10.1080/15384047.2015.1121348. [98] C.J. Chen, L.S. Hsu, S.H. Lin, M.K. Chen, H.K. Wang, J.D. Hsu, et al., Loss of
[75] C. Scheel, E.N. Eaton, S.H. Li, C.L. Chaffer, F. Reinhardt, K.J. Kah, et al., Paracrine nuclear expression of Krüppel-like factor 4 is associated with poor prognosis in
and autocrine signals induce and maintain mesenchymal and stem cell states in patients with oral cancer, Hum. Pathol. 43 (7) (2012) 1119–1125, https://doi.
the breast, Cell 145 (6) (2011) 926–940, https://doi.org/10.1016/j. org/10.1016/j.humpath.2011.09.003.
cell.2011.04.029. [99] N.V. Patel, A.M. Ghaleb, M.O. Nandan, V.W. Yang, Expression of the tumor
[76] C.R. Jeter, B. Liu, X. Liu, X. Chen, C. Liu, T. Calhoun-Davis, et al., NANOG suppressor Krüppel-like factor 4 as a prognostic predictor for colon cancer,
promotes cancer stem cell characteristics and prostate cancer resistance to Cancer Epidemiol., Biomark. Prev. 19 (10) (2010) 2631–2638, https://doi.org/
androgen deprivation, Oncogene 30 (36) (2011) 3833–3845, https://doi.org/ 10.1158/1055-9965.EPI-10-0677.
10.1038/onc.2011.114. [100] A.Y. Pandya, L.I. Talley, A.R. Frost, T.J. Fitzgerald, V. Trivedi, M. Chakravarthy,
[77] C. Huang, C. Yoon, X.H. Zhou, Y.C. Zhou, W.W. Zhou, H. Liu, et al., ERK1/2- et al., Nuclear localization of KLF4 is associated with an aggressive phenotype in
Nanog signaling pathway enhances CD44(+) cancer stem-like cell phenotypes early-stage breast cancer, Clin. Cancer Res. 10 (8) (2004) 2709–2719, https://doi.
and epithelial-to-mesenchymal transition in head and neck squamous cell org/10.1158/1078-0432.ccr-03-0484.
carcinomas, Cell Death Dis. 11 (4) (2020) 266, https://doi.org/10.1038/s41419- [101] S. Lee, S. Wottrich, B. Bonavida, Crosstalks between Raf-kinase inhibitor protein
020-2448-6. and cancer stem cell transcription factors (Oct4, KLF4, Sox2, Nanog), Tumour
[78] L.E. IvSantaliz-Ruiz, X. Xie, M. Old, T.N. Teknos, Q. Pan, Emerging role of nanog Biol. 39 (4) (2017), https://doi.org/10.1177/1010428317692253.
in tumorigenesis and cancer stem cells, Int. J. Cancer 135 (12) (2014) [102] C. Álvarez, A. Quiroz, D. Benítez-Riquelme, E. Riffo, A.F. Castro, R. Pincheira,
2741–2748, https://doi.org/10.1002/ijc.28690. SALL Proteins; Common and Antagonistic Roles in Cancer, Cancers 13 (24)
[79] Z. Hosseini-Khah, M.R. Babaei, M. Tehrani, M. Cucchiarini, H. Madry, A. Ajami, (2021) 6292, https://doi.org/10.3390/cancers13246292.
et al., SOX2 and Bcl-2 as a novel prognostic value in hepatocellular carcinoma [103] D. Chi, W. Zhang, Y. Jia, D. Cong, S. Hu, Spalt-Like transcription factor 1 (SALL1)
progression, Curr. Oncol. 28 (4) (2021) 3015–3029, https://doi.org/10.3390/ gene expression inhibits cell proliferation and cell migration of human glioma
curroncol28040264. cells through the Wnt/β-Catenin signaling pathway, Med. Sci. Monit. Basic Res.
[80] C.R. Jeter, M. Badeaux, G. Choy, D. Chandra, L. Patrawala, C. Liu, et al., 25 (2019) 128–138, https://doi.org/10.12659/MSMBR.915067.
Functional evidence that the self-renewal gene NANOG regulates human tumor [104] A. Wells, C. Yates, C.R. Shepard, E-cadherin as an indicator of mesenchymal to
development, Stem Cells 27 (5) (2009) 993–1005, https://doi.org/10.1002/ epithelial reverting transitions during the metastatic seeding of disseminated
stem.29. carcinomas, Clin. Exp. Metastas-.-. 25 (6) (2008) 621–628, https://doi.org/
[81] C.R. Jeter, T. Yang, J. Wang, H.P. Chao, D.G. Tang, Concise review: NANOG in 10.1007/s10585-008-9167-1.
cancer stem cells and tumor development: an update and outstanding questions, [105] J. Yang, C. Gao, L. Chai, Y. Ma, A novel SALL4/OCT4 transcriptional feedback
Stem Cells 33 (8) (2015) 2381–2390, https://doi.org/10.1002/stem.2007. network for pluripotency of embryonic stem cells, PLoS One 5 (5) (2010),
[82] M.X. Zhang, W. Gan, C.Y. Jing, S.S. Zheng, Y. Yi, J. Zhang, et al., High expression e10766, https://doi.org/10.1371/journal.pone.0010766.
of Oct4 and Nanog predict poor prognosis in intrahepatic cholangiocarcinoma [106] Q. Chen, J. Qian, J. Lin, J. Yang, Y. Li, C.Z. Wang, et al., Expression of SALL4 gene
patients after curative resection, J. Cancer 10 (5) (2019) 1313–1324, https://doi. in patients with acute and chronic myeloid leukemia, Zhongguoshiyanxue ye
org/10.7150/jca.28349. xuezazhi 21 (2) (2010) 315–319, https://doi.org/10.7534/j.issn.1009-
[83] V.C. Mak, M.K. Siu, O.G. Wong, K.K. Chan, H.Y. Ngan, A.N. Cheung, 2137.2013.02.011.
Dysregulatedstemness-related genes in gynecological malignancies, Histol. [107] G. Riou, M. Barrois, M.G. Lê, M. George, V. Le Doussal, C. Haie, C-myc proto-
Histopathol. 27 (9) (2012) 1121–1130, https://doi.org/10.14670/HH-27.1121. oncogene expression and prognosis in early carcinoma of the uterine cervix,
[84] T. Nagata, Y. Shimada, S. Sekine, R. Hori, K. Matsui, T. Okumura, et al., Lancet 1 (8536) (1987) 761–763, https://doi.org/10.1016/s0140-6736(87)
Prognostic significance of NANOG and KLF4 for breast cancer, Breast Cancer 21 92795-4.
(1) (2014) 96–101, https://doi.org/10.1007/s12282-012-0357-y. [108] F. Wang, Y. Guo, Q. Chen, Z. Yang, N. Ning, Y. Zhang, et al., Stem cell factor
[85] A. Naderi, A.E. Teschendorff, N.L. Barbosa-Morais, S.E. Pinder, A.R. Green, D. SALL4, a potential prognostic marker for myelodysplastic syndromes, J. Hematol.
G. Powe, et al., A gene-expression signature to predict survival in breast cancer Oncol. 6 (1) (2013) 73, https://doi.org/10.1186/1756-8722-6-73.
across independent data sets, Oncogene 26 (10) (2007) 1507–1516, https://doi. [109] K.J. Yong, L. Chai, D.G. Tenen, Oncofetal gene SALL4 in aggressive hepatocellular
org/10.1038/sj.onc.1209920. carcinoma, N. Engl. J. Med. 369 (12) (2013) 1171–1172, https://doi.org/
[86] M. Pitrone, G. Pizzolanti, A. Coppola, L. Tomasello, S. Martorana, G. Pantuso, et 10.1056/NEJMc1308785.
al., Knockdown of NANOG reduces cell proliferation and induces G0/G1 cell cycle [110] L. Soucek, G.I. Evan, The ups and downs of Myc biology, Curr. Opin. Genet. Dev.
arrest in human adipose stem cells, Int. J. Mol. Sci. 20 (10) (2019) 2580, https:// 20 (1) (2010) 91–95, https://doi.org/10.1016/j.gde.2009.11.001.
doi.org/10.3390/ijms20102580. [111] D.M. Miller, S.D. Thomas, A. Islam, D. Muench, K. Sedoris, c-Myc and cancer
[87] S.K. Ray, The transcription regulator Krüppel-Like Factor 4 and its dual roles of metabolism, Clin. Cancer Res. 18 (20) (2012) 5546–5553, https://doi.org/
oncogene in glioblastoma and tumor suppressor in neuroblastoma, Forum 10.1158/1078-0432.CCR-12-0977.
Immunopathol. Dis. Ther. 7 (1–2) (2016) 127–139, https://doi.org/10.1615/ [112] G.J. Yoshida, Emerging roles of Myc in stem cell biology and novel tumor
ForumImmunDisTher.2016017227. therapies, J. Exp. Clin. Cancer Res.: CR 37 (1) (2018) 173, https://doi.org/
[88] A. Taracha-Wisniewska, G. Kotarba, S. Dworkin, T. Wilanowski, Recent 10.1186/s13046-018-0835-y.
discoveries on the involvement of Krüppel-Like factor 4 in the most common [113] C.M. Shachaf, A.M. Kopelman, C. Arvanitis, A. Karlsson, S. Beer, S. Mandl, et al.,
cancer types, Int. J. Mol. Sci. 21 (22) (2020) 8843, https://doi.org/10.3390/ MYC inactivation uncovers pluripotent differentiation and tumour dormancy in
ijms21228843. hepatocellular cancer, Nature 431 (7012) (2004) 1112–1117, https://doi.org/
[89] J. Cui, M. Shi, M. Quan, K. Xie, Regulation of EMT by KLF4 in gastrointestinal 10.1038/nature03043.
cancer, Curr. Cancer Drug Targets 13 (9) (2013) 986–995, https://doi.org/ [114] H. Park, S. Hwang, J.Y. Jeong, S.G. Jun, M.C. Choi, W.D. Joo, et al., Integrative
10.2174/15680096113136660104. analysis of transcription factors and microRNAs in ovarian cancer cell spheroids,
[90] H. Xie, J. Li, Y. Ying, H. Yan, K. Jin, X. Ma, et al., METTL3/YTHDF2 m6A axis J. Ovarian Res. 13 (1) (2020) 16, https://doi.org/10.1186/s13048-020-00618-7.
promotes tumorigenesis by degrading SETD7 and KLF4 mRNAs in bladder cancer, [115] J. Kim, S.H. Orkin, Embryonic stem cell-specific signatures in cancer: insights into
J. Cell. Mol. Med. 24 (7) (2020) 4092–4104, https://doi.org/10.1111/ genomic regulatory networks and implications for medicine, Genome Med. 3 (11)
jcmm.15063. (2011) 75, https://doi.org/10.1186/gm291.
[91] Y. Li, S. Yu, L. Li, J. Chen, M. Quan, Q. Li, et al., KLF4-mediated upregulation of [116] C.V. Dang, MYC on the path to cancer, Cell 149 (1) (2012) 22–35, https://doi.
CD9 and CD81 suppresses hepatocellular carcinoma development via JNK org/10.1016/j.cell.2012.03.003.
signaling, Cell Death Dis. 11 (4) (2020) 299, https://doi.org/10.1038/s41419- [117] N. Colombo, S. Carinelli, A. Colombo, C. Marini, D. Rollo, C. Sessa, et al., Cervical
020-2479-z. cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up,
[92] K. Akaogi, Y. Nakajima, I. Ito, S. Kawasaki, S.H. Oie, A. Murayama, KLF4 Ann. Oncol. 23 (2012) 27–32, https://doi.org/10.1093/annonc/mds268.
suppresses estrogen-dependent breast cancer growth by inhibiting the [118] G. Yu, J. Wang, Y. Chen, X. Wang, J. Pan, G. Li, Z. Jia, Q. Li, J.C. Yao, K. Xie,
transcriptional activity of ERalpha, Oncogene 28 (32) (2009) 2894–2902, Overexpression of phosphorylated mammalian target of rapamycin predicts
https://doi.org/10.1038/onc.2009.151. lymph node metastasis and prognosis of chinese patients with gastric cancer, Clin.
[93] W. Liu, M.M. Ip, M.B. Podgorsak, G.M. Das, Disruption of estrogen receptor alpha- Cancer Res.: Off. J. Am. Assoc. Cancer Res. 15 (5) (2009) 1821–1829, https://doi.
p53 interaction in breast tumors: a novel mechanism underlying the anti-tumor org/10.1158/1078-0432.CCR-08-2138.
effect of radiation therapy, Breast Cancer Res. Treat. 115 (1) (2009) 43–50, [119] L. Lin, A. Liu, Z. Peng, H.J. Lin, P.K. Li, C. Li, J. Lin, STAT3 is necessary for
https://doi.org/10.1007/s10549-008-0044-z. proliferation and survival in colon cancer-initiating cells, Cancer Res. 71 (23)
[94] M. Schoenhals, A. Kassambara, J. De Vos, D. Hose, J. Moreaux, B. Klein, (2011) 7226–7237, https://doi.org/10.1158/0008-5472.CAN-10-4660.
Embryonic stem cell markers expression in cancers, Biochem. Biophys. Res. [120] M. Hajimoradi, Z. Mohammad Hassan, M. Ebrahimi, M. Soleimani, M. Bakhshi,
Commun. 383 (2) (2009) 157–162, https://doi.org/10.1016/j.bbrc.2009.02.156. J. Firouzi, F.S. Samani, STAT3 is overactivated in gastric cancer stem-like cells,
[95] J. Matsuoka, M. Yashiro, K. Sakurai, N. Kubo, H. Tanaka, K. Muguruma, et al., Cell J. 17 (4) (2016) 617–628, https://doi.org/10.22074/cellj.2016.3834.
Role of the stemness factors sox2, oct3/4, and nanog in gastric carcinoma, J. Surg. [121] M.B. Sonbol, D.H. Ahn, T. Bekaii-Saab, Therapeutic targeting strategies of cancer
Res. 174 (1) (2012) 130–135, https://doi.org/10.1016/j.jss.2010.11.903. stem cells in gastrointestinal malignancies, Biomedicines 7 (1) (2019) 17, https://
[96] J. Ingruber, D. Savic, T.B. Steinbichler, S. Sprung, F. Fleischer, R. Glueckert, et al., doi.org/10.3390/biomedicines7010017.
KLF4, Slug and EMT in head and neck squamous cell carcinoma, Cells 10 (3) [122] D.Y. Kim, S.T. Cha, D.H. Ahn, H.Y. Kang, C.I. Kwon, K.H. Ko, S.G. Hwang, P.
(2021) 539, https://doi.org/10.3390/cells10030539. W. Park, K.S. Rim, S.P. Hong, STAT3 expression in gastric cancer indicates a poor
[97] S.K. Tai, M.H. Yang, S.Y. Chang, Y.C. Chang, W.Y. Li, T.L. Tsai, et al., Persistent prognosis, J. Gastroenterol. Hepatol. 24 (4) (2009) 646–651, https://doi.org/
Krüppel-like factor 4 expression predicts progression and poor prognosis of head 10.1111/j.1440-1746.2008.05671.x.

135
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

[123] J.Y. Deng, D. Sun, X.Y. Liu, Y. Pan, H. Liang, STAT-3 correlates with lymph node [145] J.H. Bushweller, Targeting transcription factors in cancer - from undruggable to
metastasis and cell survival in gastric cancer, World J. Gastroenterol. 16 (42) reality, Nat. Rev. Cancer 19 (11) (2019) 611–624, https://doi.org/10.1038/
(2010) 5380–5387, https://doi.org/10.3748/wjg.v16.i42.5380. s41568-019-0196-7.
[124] K. Ji, M. Zhang, Q. Chu, Y. Gan, H. Ren, L. Zhang, L. Wang, X. Li, W. Wang, The [146] L. Cheng, Establishing a germ cell origin for metastatic tumors using OCT4
role of p-STAT3 as a prognostic and clinicopathological marker in colorectal immunohistochemistry, Cancer 101 (9) (2004) 2006–2010, https://doi.org/
cancer: a systematic review and meta-analysis, PLoS One 11 (8) (2016), 10.1002/cncr.20566.
e0160125, https://doi.org/10.1371/journal.pone.0160125. [147] G. Gu, J. Yuan, M. Wills, S. Kasper, Prostate cancer cells with stem cell
[125] Q. Lin, R. Lai, L.R. Chirieac, C. Li, V.A. Thomazy, I. Grammatikakis, G. characteristics reconstitute the original human tumor in vivo, Cancer Res. 67 (10)
Z. Rassidakis, W. Zhang, Y. Fujio, K. Kunisada, S.R. Hamilton, H.M. Amin, (2007) 4807–4815, https://doi.org/10.1158/0008-5472.CAN-06-4608.
Constitutive activation of JAK3/STAT3 in colon carcinoma tumors and cell lines: [148] H.M. Meng, P. Zheng, X.Y. Wang, C. Liu, H.M. Sui, S.J. Wu, J. Zhou, Y.Q. Ding,
inhibition of JAK3/STAT3 signaling induces apoptosis and cell cycle arrest of J. Li, Over-expression of Nanog predicts tumor progression and poor prognosis in
colon carcinoma cells, Am. J. Pathol. 167 (4) (2005) 969–980, https://doi.org/ colorectal cancer, Cancer Biol. Ther. 9 (4) (2010) 295–302, https://doi.org/
10.1016/S0002-9440(10)61187-X. 10.4161/cbt.9.4.10666.
[126] Y. Zhang, Z. Jin, H. Zhou, X. Ou, Y. Xu, H. Li, C. Liu, B. Li, Suppression of prostate [149] J. Delyon, C. Mateus, D. Lefeuvre, E. Lanoy, L. Zitvogel, N. Chaput, et al.,
cancer progression by cancer cell stemness inhibitor napabucasin, Cancer Med. 5 Experience in daily practice with ipilimumab for the treatment of patients with
(6) (2016) 1251–1258, https://doi.org/10.1002/cam4.675. metastatic melanoma: an early increase in lymphocyte and eosinophil counts is
[127] Z. Yang, C. Zhang, W. Qi, Y. Cui, Y. Xuan, GLI1 promotes cancer stemness through associated with improved survival, Ann. Oncol. 24 (6) (2013) 1697–1703,
intracellular signaling pathway PI3K/Akt/NFκB in colorectal adenocarcinoma, https://doi.org/10.1093/annonc/mdt027.
Exp. Cell Res. 373 (1–2) (2018) 145–154, https://doi.org/10.1016/j. [150] S.L. Topalian, F.S. Hodi, J.R. Brahmer, S.N. Gettinger, D.C. Smith, D.
yexcr.2018.10.006. F. McDermott, et al., Safety, activity, and immune correlates of anti-PD-1
[128] Y. Yao, D. Zhou, D. Shi, H. Zhang, S. Zhan, X. Shao, K. Sun, L. Sun, G. Wu, K. Tian, antibody in cancer, N. Engl. J. Med. 366 (26) (2012) 2443–2454, https://doi.org/
X. Zhu, S. He, GLI1 overexpression promotes gastric cancer cell proliferation and 10.1056/NEJMoa1200690.
migration and induces drug resistance by combining with the AKT-mTOR [151] T. Hoey, W.C. Yen, F. Axelrod, J. Basi, L. Donigian, S. Dylla, et al., DLL4 blockade
pathway, Biomed. Pharmacother. 111 (2019) 993–1004, https://doi.org/ inhibits tumor growth and reduces tumor-initiating cell frequency, Cell Stem Cell
10.1016/j.biopha.2019.01.018. 5 (2) (2009) 168–177, https://doi.org/10.1016/j.stem.2009.05.019.
[129] A. Jo, S. Denduluri, B. Zhang, Z. Wang, L. Yin, Z. Yan, R. Kang, L.L. Shi, J. Mok, M. [152] A. Cashen, S. Lopez, F. Gao, G. Calandra, R. MacFarland, K. Badel, et al., A phase
J. Lee, R.C. Haydon, The versatile functions of Sox9 in development stem cells, II study of plerixafor (AMD3100) plus G-CSF for autologous hematopoietic
and human diseases, Genes&Dis. 1 (2) (2014) 149–161, https://doi.org/10.1016/ progenitor cell mobilization in patients with Hodgkin lymphoma, Biol. Blood
j.gendis.2014.09.004. Marrow Transplant. 14 (11) (2008) 1253–1261, https://doi.org/10.1016/j.
[130] P. Bastide, C. Darido, J. Pannequin, R. Kist, S. Robine, C. Marty-Double, F. Bibeau, bbmt.2008.08.011.
G. Scherer, D. Joubert, F. Hollande, P. Blache, P. Jay, Sox9 regulates cell [153] G.L. Uy, M.P. Rettig, I.H. Motabi, K. McFarland, K.M. Trinkaus, L.M. Hladnik, et
proliferation and is required for Paneth cell differentiation in the intestinal al., A phase 1/2 study of chemosensitization with the CXCR4 antagonist plerixafor
epithelium, J. Cell Biol. 178 (4) (2007) 635–648, https://doi.org/10.1083/ in relapsed or refractory acute myeloid leukemia, Blood 119 (17) (2012)
jcb.200704152. 3917–3924, https://doi.org/10.1182/blood-2011-10-383406.
[131] M. Huch, H. Clevers, Sox9 marks adult organ progenitors, Nat. Genet. 43 (1) [154] S.A. Mani, W. Guo, M.J. Liao, E.N. Eaton, A. Ayyanan, A.Y. Zhou, et al., The
(2011) 9–10, https://doi.org/10.1038/ng0111-9. epithelial-mesenchymal transition generates cells with properties of stem cells,
[132] M. Sun, H. Uozaki, R. Hino, A. Kunita, A. Shinozaki, T. Ushiku, T. Hibiya, Cell 133 (4) (2008) 704–715, https://doi.org/10.1016/j.cell.2008.03.027.
K. Takeshita, M. Isogai, K. Takada, M. Fukayama, SOX9 expression and its [155] A.E. Kudinov, J. Karanicolas, E.A. Golemis, Y. Boumber, Musashi, RNA-Binding
methylation status in gastric cancer, VirchowsArchiv: Int. J. Pathol. 460 (3) Proteins as cancer drivers and novel therapeutic targets, Clin. Cancer Res. 23 (9)
(2012) 271–279, https://doi.org/10.1007/s00428-012-1201-7. (2017) 2143–2153, https://doi.org/10.1158/1078-0432.CCR-16-2728.
[133] J.C. Santos, E. Carrasco-Garcia, M. Garcia-Puga, P. Aldaz, M. Montes, [156] Q. Tang, J. Chen, Z. Di, W. Yuan, Z. Zhou, Z. Liu, TM4SF1 promotes EMT and
M. Fernandez-Reyes, C.C. de Oliveira, C.H. Lawrie, M.J. Araúzo-Bravo, M. cancer stemness via the Wnt/β-catenin/SOX2 pathway in colorectal cancer,
L. Ribeiro, A. Matheu, SOX9 elevation acts with canonical WNT signaling to drive J. Exp. Clin. Cancer Res. 39 (1) (2020) 232, https://doi.org/10.1186/s13046-
gastric cancer progression, Cancer Res. 76 (22) (2016) 6735–6746, https://doi. 020-01690-z.
org/10.1158/0008-5472.CAN-16-1120. [157] C.H. Lin, H.Y. Li, Y.P. Liu, P.F. Kuo, W.C. Wang, F.C. Lin, High-CLDN4 ESCC cells
[134] C. Darido, M. Buchert, J. Pannequin, P. Bastide, H. Zalzali, T. Mantamadiotis, J. harbor stem-like properties and indicate for poor concurrent chemoradiation
F. Bourgaux, V. Garambois, P. Jay, P. Blache, D. Joubert, F. Hollande, Defective therapy response in esophageal squamous cell carcinoma, Ther. Adv. Med. Oncol.
claudin-7 regulation by Tcf-4 and Sox-9 disrupts the polarity and increases the 11 (2019), https://doi.org/10.1177/1758835919875324.
tumorigenicity of colorectal cancer cells, Cancer Res. 68 (11) (2008) 4258–4268, [158] Y.J. Zhu, B. Zheng, G.J. Luo, X.K. Ma, X.Y. Lu, X.M. Lin, Circular RNAs negatively
https://doi.org/10.1158/0008-5472.CAN-07-5805. regulate cancer stem cells by physically binding FMRP against CCAR1 complex in
[135] C. Prévostel, P. Blache, The dose-dependent effect of SOX9 and its incidence in hepatocellular carcinoma, Theranostics 9 (12) (2019) 3526–3540, https://doi.
colorectal cancer, Eur. J. Cancer 86 (2017) 150–157, https://doi.org/10.1016/j. org/10.7150/thno.32796.
ejca.2017.08.037 (Oxf., Engl.: 1990). [159] T. Bekaii-Saab, B. El-Rayes, Identifying and targeting cancer stem cells in the
[136] M.L. Espersen, J. Olsen, D. Linnemann, E. Høgdall, J.T. Troelsen, Clinical treatment of gastric cancer, Cancer 123 (2017) 1303–1312, https://doi.org/
implications of intestinal stem cell markers in colorectal cancer, Clin. Colorectal 10.1002/cncr.30538.
Cancer 14 (2) (2015) 63–71, https://doi.org/10.1016/j.clcc.2014.12.004. [160] J.C. Bendell, J.M. Hubbard, B.H. O’Neil, D.J. Jonker, A. Starodub, J.D. Peyton, et
[137] B.M. Javier, R. Yaeger, L. Wang, F. Sanchez-Vega, A. Zehir, S. Middha, al., Phase 1b/II study of cancer stemness inhibitor napabucasin (BBI-608) in
J. Sadowska, E. Vakiani, J. Shia, D. Klimstra, M. Ladanyi, C.A. Iacobuzio- combination with FOLFIRI +/- bevacizumab (bev) in metastatic colorectal cancer
Donahue, J.F. Hechtman, Recurrent, truncating SOX9 mutations are associated (mCRC) patients (pts), J. Clin. Oncol. 35 (2017), https://doi.org/10.1200/
with SOX9 overexpression, KRAS mutation, and TP53 wild type status in JCO.2017.35.15_suppl.3529, 3529–3529.
colorectal carcinoma, Oncotarget 7 (32) (2016) 50875–50882, https://doi.org/ [161] F. Loupakis, C. Cremolini, G. Masi, S. Lonardi, V. Zagonel, L. Salvatore, et al.,
10.18632/oncotarget.9682. Initial therapy with FOLFOXIRI and bevacizumab for metastatic colorectal
[138] F. Yang, P. Cui, Y. Lu, X. Zhang, Requirement of the transcription factor YB-1 for cancer, N. Engl. J. Med. 371 (17) (2014) 1609–1618, https://doi.org/10.1056/
maintaining the stemness of cancer stem cells and reverting differentiated cancer NEJMoa1403108.
cells into cancer stem cells, Stem Cell Res. Ther. 10 (1) (2019) 233, https://doi. [162] S. Pintova, S. Dharmupari, E. Moshier, N. Zubizarreta, C. Ang, R.F. Holcombe,
org/10.1186/s13287-019-1360-4. Genistein combined with FOLFOX or FOLFOX–Bevacizumab for the treatment of
[139] S. Vinogradov, X. Wei, Cancer stem cells and drug resistance: the potential of metastatic colorectal cancer: phase I/II pilot study, Cancer Chemother.
nanomedicine, Nanomed. (Lond., Engl. ) 7 (4) (2012) 597–615, https://doi.org/ Pharmacol. 84 (2019) 591–598, https://doi.org/10.1007/s00280-019-03886-3.
10.2217/nnm.12.22. [163] Y. Zhang, H. Chen, Genistein attenuates WNT signaling by up-regulating sFRP2 in
[140] M.L. Suvà, E. Rheinbay, S.M. Gillespie, A.P. Patel, H. Wakimoto, S.D. Rabkin, et a human colon cancer cell line, Exp. Biol. Med. 236 (6) (2011) 714–722, https://
al., Reconstructing and reprogramming the tumor-propagating potential of doi.org/10.1258/ebm.2011.010347.
glioblastoma stem-like cells, Cell 157 (3) (2014) 580–594, https://doi.org/ [164] H. Wang, Q. Li, H. Chen, Genistein affects histone modifications on Dickkopf-
10.1016/j.cell.2014.02.030. related protein 1 (DKK1) gene in SW480 human colon cancer cell line, PloSone 7
[141] K. Takahashi, S. Yamanaka, Induction of pluripotent stem cells from mouse (7) (2012), e40955, https://doi.org/10.1371/journal.pone.0040955.
embryonic and adult fibroblast cultures by defined factors, Cell 126 (4) (2006) [165] P.V. Pham, N.L. Phan, N.T. Nguyen, N.H. Truong, T.T. Duong, D.V. Le, et al.,
663–676, https://doi.org/10.1016/j.cell.2006.07.024. Differentiation of breast cancer stem cells by knockdown of CD44: promising
[142] H.R. Sun, S. Wang, S.C. Yan, Y. Zhang, P.J. Nelson, H.L. Jia, et al., Therapeutic differentiation therapy, J. Transl. Med. 9 (2011) 209, https://doi.org/10.1186/
strategies targeting cancer stem cells and their microenvironment, Front. Oncol. 9 1479-5876-9-209.
(2019) 1104, https://doi.org/10.3389/fonc.2019.01104. [166] L. Du, D. Wang, P.W. Nagle, A. Groen, H. Zhang, C.T. Muijs, et al., Role of mTOR
[143] I. Ben-Porath, M.W. Thomson, V.J. Carey, R. Ge, G.W. Bell, A. Regev, R. through autophagy in esophageal cancer stemness, Cancers 14 (7) (2022) 1806,
A. Weinberg, An embryonic stem cell-like gene expression signature in poorly https://doi.org/10.3390/cancers14071806.
differentiated aggressive human tumors, Nat. Genet. 40 (5) (2008) 499–507, [167] W. Li, X. Zhang, Y. Nan, L. Jia, J. Sun, L. Zhang, et al., Hyaluronidase and pH
https://doi.org/10.1038/ng.127. Dual-Responsive nanoparticles for targeted breast cancer stem cells, Front. Oncol.
[144] X. Wang, J. Dai, Concise review: isoforms of OCT4 contribute to the confusing 11 (2021), 760423, https://doi.org/10.3389/fonc.2021.760423.
diversity in stem cell biology, Stem Cells 28 (5) (2010) 885–893, https://doi.org/ [168] M.Z. Koh, W.Y. Ho, S.K. Yeap, N.M. Ali, L. Boo, N.B. Alitheen, Regulation of
10.1002/stem.419. cellular and cancer stem cell-related putative gene expression of parental and

136
A. Chaudhary et al. Seminars in Cancer Biology 88 (2023) 123–137

CD44+CD24- Sorted MDA-MB-231 Cells by cisplatin, Pharmaceuticals 14 (5) and loss of tumorigenicity, Stem Cells 27 (1) (2009) 40–48, https://doi.org/
(2021) 391, https://doi.org/10.3390/ph14050391. 10.1634/stemcells.2008-0493.
[169] S. Sai, G. Vares, E.H. Kim, K. Karasawa, B. Wang, M. Nenoi, et al., Carbon ion [183] F. Jiang, Q. Qiu, A. Khanna, N.W. Todd, J. Deepak, L. Xing, et al., Aldehyde
beam combined with cisplatin effectively disrupts triple negative breast cancer dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer, Mol.
stem-like cells in vitro, Mol. Cancer 14 (2015) 166, https://doi.org/10.1186/ Cancer Res. 7 (3) (2009) 330–338, https://doi.org/10.1158/1541-7786.MCR-08-
s12943-015-0429-7. 0393.
[170] S. Sai, T. Wakai, G. Vares, S. Yamada, T. Kamijo, T. Kamada, Combination of [184] L. Vermeulen, E. De Sousa, F. Melo, M. van der Heijden, K. Cameron, J.H. de
carbon ion beam and gemcitabine causes irreparable DNA damage and death of Jong, T. Borovski, et al., Wnt activity defines colon cancer stem cells and is
radioresistant pancreatic cancer stem-like cells in vitro and in vivo, Oncotarget 6 regulated by the microenvironment, Nat. Cell Biol. 12 (5) (2010) 468–476,
(8) (2015) 5517–5535, https://doi.org/10.18632/oncotarget.3584. https://doi.org/10.1038/ncb2048.
[171] Y. Dong, L. Li, L. Wang, T. Zhou, J.W. Liu, Y.J. Gao, Preliminary study of the [185] C. Li, Y. Yan, W. Ji, L. Bao, H. Qian, L. Chen, et al., OCT4 positively regulates
effects of β-elemene on MCF-7/ADM breast cancer stem cells, Genet. Mol. Res. 14 Survivin expression to promote cancer cell proliferation and leads to poor
(1) (2015) 2347–2355, https://doi.org/10.4238/2015.March.27.20. prognosis in esophageal squamous cell carcinoma, PloS One 7 (11) (2012),
[172] M. Nguyen, C. Osipo, Targeting breast cancer stem cells using naturally occurring e49693, https://doi.org/10.1371/journal.pone.0049693.
phytoestrogens, Int. J. Mol. Sci. 23 (12) (2022) 6813, https://doi.org/10.3390/ [186] J.P. Thiery, Epithelial-mesenchymal transitions in tumor progression, Nat. Rev.
ijms23126813. Cancer 2 (6) (2002) 442–454, https://doi.org/10.1038/nrc822.
[173] T. Shimo, J. Kurebayashi, N. Kanomata, T. Yamashita, Y. Kozuka, T. Moriya, et [187] Y. Du, C. Ma, Z. Wang, Z. Liu, H. Liu, T. Wang, et al., Nanog, a novel prognostic
al., Antitumor and anticancer stem cell activity of a poly ADP-ribose polymerase marker for lung cancer, Surg. Oncol. 22 (4) (2013) 224–229, https://doi.org/
inhibitor olaparib in breast cancer cells, Breast Cancer 21 (1) (2014) 75–85, 10.1016/j.suronc.2013.08.001.
https://doi.org/10.1007/s12282-012-0356-z. [188] L.Y. Bourguignon, C.C. Spevak, G. Wong, W. Xia, E. Gilad, Hyaluronan-CD44
[174] H. Yan, P. Bu, Non-coding RNAs in cancer stem cells, Cancer Lett. 421 (2018) interaction with protein kinase C (epsilon) promotes oncogenic signaling by the
121–126, https://doi.org/10.1016/j.canlet.2018.01.027. stem cell marker Nanog and the Production of microRNA-21, leading to down-
[175] S. Pece, D. Tosoni, S. Confalonieri, G. Mazzarol, M. Vecchi, S. Ronzoni, et al., regulation of the tumor suppressor protein PDCD4, anti-apoptosis, and
Biological and molecular heterogeneity of breast cancers correlates with their chemotherapy resistance in breast tumor cells, J. Biol. Chem. 284 (39) (2009)
cancer stem cell content, Cell 140 (1) (2010) 62–73, https://doi.org/10.1016/j. 26533–26546, https://doi.org/10.1074/jbc.M109.027466.
cell.2009.12.007. [189] Y. Inoue, S. Matsuura, N. Kurabe, T. Kahyo, H. Mori, A. Kawase, et al.,
[176] Z. Islam, A.M. Ali, A. Naik, M. Eldaw, J. Decock, P.R. Kolatkar, Transcription Clinicopathological and survival analysis of japanese patients with resected non-
factors: the fulcrum between cell development and carcinogenesis, Front. Oncol. small-cell lung cancer harboring NKX2-1, SETDB1, MET, HER2, SOX2, FGFR1, or
11 (2021), 681377, https://doi.org/10.3389/fonc.2021.681377. PIK3CA gene amplification, J. Thorac. Oncol. 10 (11) (2015) 1590–1600, https://
[177] H. Zhang, Z.Z. Wang, Mechanisms that mediate stem cell self-renewal and doi.org/10.1097/JTO.0000000000000685.
differentiation, J. Cell. Biochem. 103 (3) (2008) 709–718, https://doi.org/ [190] D.L. Pham, V. Scheble, P. Bareiss, A. Fischer, C. Beschorner, A. Adam, et al., SOX2
10.1002/jcb.21460. expression and prognostic significance in ovarian carcinoma, Int. J. Gynecol.
[178] J. Hatina, M. Kripnerová, Z. Houdek, M. Pešta, F. Tichánek, Pluripotency Pathol. 32 (4) (2013) 358–367, https://doi.org/10.1097/
stemness and cancer: more questions than answers, Adv. Exp. Med. Biol. 1376 PGP.0b013e31826a642b.
(2022) 77–100, https://doi.org/10.1007/5584_2021_663. [191] Z. Liu, H. Yang, W. Luo, Q. Jiang, C. Mai, Y. Chen, et al., Loss of cytoplasmic KLF4
[179] W. Zhao, X. Ji, F. Zhang, L. Li, L. Ma, Embryonic stem cell markers, Molecules 17 expression is correlated with the progression and poor prognosis of
(6) (2012) 6196–6236, https://doi.org/10.3390/molecules17066196. nasopharyngeal carcinoma, Histopathology 63 (3) (2013) 362–370, https://doi.
[180] K. Xu, Z. Zhu, F. Zeng, Expression and significance of Oct4 in bladder cancer, org/10.1111/his.12176.
J. Huazhong Univ. Sci. Technol. 27 (6) (2007) 675–677, https://doi.org/ [192] K. Weina, J. Utikal, SOX2 and cancer: current research and its implications in the
10.1007/s11596-007-0614-z. clinic, Clin. Transl. Med. 3 (2014) 19, https://doi.org/10.1186/2001-1326-3-19.
[181] C.O. Rodini, D.E. Suzuki, N. Saba-Silva, A. Cappellano, J.E. de Souza, [193] X.T. For klf-4-ref- Qi, Y.L. Li, Y.Q. Zhang, T. Xu, B. Lu, L. Fang, J.Q. Gao, L.S. Yu,
S. Cavalheiro, et al., Expression analysis of stem cell-related genes reveal OCT4 as D.F. Zhu, B. Yang, Q.J. He, M.D. Ying, KLF4 functions as an oncogene in
a predictor of poor clinical outcome in medulloblastoma, J. Neuro-Oncol. 106 (1) promoting cancer stem cell-like characteristics in osteosarcoma cells,
(2012) 71–79, https://doi.org/10.1007/s11060-011-0647-9. ActapharmacologicaSinica 40 (4) (2019) 546–555, https://doi.org/10.1038/
[182] R.M. Gangemi, F. Griffero, D. Marubbi, M. Perera, M.C. Capra, P. Malatesta, et al., s41401-018-0050-6.
SOX2 silencing in glioblastoma tumor-initiating cells causes stop of proliferation

137

You might also like