Next Article in Journal
Prognostic Factors Research in Breast Cancer Patients: New Paths
Next Article in Special Issue
Celastrol and Resveratrol Modulate SIRT Genes Expression and Exert Anticancer Activity in Colon Cancer Cells and Cancer Stem-like Cells
Previous Article in Journal
Understanding Aberrant Signaling to Elude Therapy Escape Mechanisms in Myeloproliferative Neoplasms
Previous Article in Special Issue
Low Levels of TRIM28-Interacting KRAB-ZNF Genes Associate with Cancer Stemness and Predict Poor Prognosis of Kidney Renal Clear Cell Carcinoma Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Intrinsic and Extrinsic Factors Impacting Cancer Stemness and Tumor Progression

1
Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
2
School of Veterinary Medicine and Science (SVMS) and Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(4), 970; https://doi.org/10.3390/cancers14040970
Submission received: 24 December 2021 / Revised: 3 February 2022 / Accepted: 8 February 2022 / Published: 15 February 2022

Abstract

:

Simple Summary

Presently, the study of cancer stem cells is important because these cells increase the cancer complexity, confer tumors the ability to grow, resist treatment, and survive in adverse conditions. One of the properties that these cells have is stemness. Cancer stemness is modulated by the tumor microenvironment, which influences cancer stem cell function and survival. This review includes information about cancer stem cells and their regulation by extrinsic and intrinsic factors. Pluripotency factors and signaling pathways, which regulate and modulate cancer stemness are summarized in this review. In addition, it provides an overview of the models that allow the study of cancer stem cells for the development of new targeted therapies.

Abstract

Tumor heterogeneity represents an important limitation to the development of effective cancer therapies. The presence of cancer stem cells (CSCs) and their differentiation hierarchies contribute to cancer complexity and confer tumors the ability to grow, resist treatment, survive unfavorable conditions, and invade neighboring and distant tissues. A large body of research is currently focusing on understanding the properties of CSCs, including their cellular and molecular origin, as well as their biological behavior in different tumor types. In turn, this knowledge informs strategies for targeting these tumor initiating cells and related cancer stemness. Cancer stemness is modulated by the tumor microenvironment, which influences CSC function and survival. Several advanced in vitro models are currently being developed to study cancer stemness in order to advance new knowledge of the key molecular pathways involved in CSC self-renewal and dormancy, as well as to mimic the complexity of patients’ tumors in pre-clinical drug testing. In this review, we discuss CSCs and the modulation of cancer stemness by the tumor microenvironment, stemness factors and signaling pathways. In addition, we introduce current models that allow the study of CSCs for the development of new targeted therapies.

Graphical Abstract

1. Introduction

Cancer stem cells (CSCs) are a population of cells present in malignant tumors that share many similarities with normal stem cells or progenitor cells. The common characteristics of these cells include the ability to self-renew and differentiate into multiple lineages, leading to activation of tumor growth and heterogeneity. Mutations that occur in the stem cell pool may contribute to the process of oncogenesis [1]. In addition, there is a link between aging and an increase in the incidence of cancer. There is an accumulation of damage that can lead to the inactivation of tumor suppressor genes and the activation of oncogenes with aging [2]. The origin of CSCs remains unclear, as well as whether they originate from normal stem cells or non-stem cells through re-acquisition of stem cell traits (stemness) through a change in differentiation [3,4,5] (Figure 1). CSCs are generally a rare population of cells, usually amounting to 0.01–2% of the total tumor mass. A notable exception to this evidence is represented by malignant melanoma which comprises a high proportion of CSCs [6]. CSCs share many characteristics with their normal counterparts, including expression of surface markers and regulation by common signaling pathways [7,8,9]. Most CSC surface markers (Table 1) are expressed by embryonic or adult stem cells and are rarely expressed by normal differentiated tissue cells [10]. The expression of surface markers is used to identify and isolate CSCs by fluorescence-activated cell sorting (FACS) and magnetic-activated cell sorting (MACS) [11,12]. Additionally, for the isolation and enrichment of CSCs, the following methods are used: intracellular enzyme activity, promoter-driven fluorescent protein expression, suspension/adherent culture, immunoselection, etc. [13]. However, CSC markers can be characterized not only by the expression of surface markers, but also by intrinsic and extrinsic factors which include transcription factors, microRNAs (for example, miR-21, miR-210, miR-34a, and miR-16) [14], signaling molecules [15,16,17], and extracellular matrix (ECM) [18].
All of these factors regulate CSC function, under the influence of the tumor microenvironment (TME). Differently from normal stem cells, whose proliferation is finely regulated by interactions with their physiological niche, CSCs demonstrate abnormal regulation of self-renewal that enables their expansion either by symmetrical or asymmetrical cell division [7]. Differences between the niche surrounding normal cells and CSCs are at the basis of CSCs ability to transition into different cell states, to remain dormant for long periods of time, and to metastasize to different sites [39]. The niche is a specialized microenvironment that regulates normal stem cell function through cell-to-cell interaction and paracrine signaling. Escape of normal stem cells from control of proliferation and apoptosis provided by the niche is associated with malignant transformation and the formation of a “CSC niche” which induces the recruitment of cells of the TME (mesenchymal stem cells (MSC), immune cells, endothelial cells, and tumor associated fibroblasts (TAF)), secreting growth factors and cytokines essential for sustaining CSC self-renewal [40,41]. By maintaining differentiation capacity, CSC can then lead to the formation of heterogeneous tumors characterized by the growth of phenotypically different subclones [42] of transit-amplifying cells which accumulate and accelerate tumor growth [3]. These heterogeneous cell populations demonstrate high plasticity potential [43,44] and high resistance to stressful factors present in the TME, such as low oxygen or lack of nutrients [39,45].
The CSC niche therefore supports the basic properties of CSCs, preserves their phenotypic plasticity, protects them from the immune system, and contributes to their metastatic potential [39]. Key to the CSC plasticity and metastatic potential is the process of epithelial–mesenchymal transition (EMT) [46,47]. EMT can be induced by the cells of the TME secreting cytokine (e.g., TGFβ and interleukins) to causes changes in cytoskeleton organization, loss of apical-basal polarity, expression of E-cadherin, and acquisition of mesenchymal features and motility, as well as remodeling of the extracellular matrix (ECM) [48,49]. Through these changes, EMT induces a CSC phenotype which enables tumor metastasis and confers resistance to therapy [50,51,52]. It should be emphasized that metastasis is associated with circulating tumor cells that originate from the primary tumor, but these cells have properties of stemness and EMT, which contribute to their penetration and circulation in the blood and increased metastatic ability [53].
Indeed, CSCs are inherently resistant to radiation and cytotoxic drugs, and therefore responsible for minimal residual disease and cancer recurrence [54]. Currently, the study of cancer stemness poses a number of questions, which include: how different are CSCs from normal stem cells; are CSC properties changing during the course of disease; are there differences in stemness between different cancer subtypes; how CSC knowledge can be used to advance precision medicine [55].
In this review, we discuss cancer stemness, CSCs, and their regulation by extrinsic and intrinsic factors. Stemness can be affected by both physical and chemical factors. The main attention of this article is focused on chemical factors, molecular pathways of CSC interaction, and their microenvironment. A deeper understanding of the interactions between CSCs and the microenvironment, including the mechanisms responsible for switching cancer cells from non-CSC to CSC status, is essential for the discovery of effective new therapies. In addition, knowledge about extrinsic and intrinsic factors and their influence on stem formation can provide targeted therapy as well as prevent cancer recurrence and metastasis.

2. Pluripotency Factors as Intrinsic Factors Regulating Cancer Stemness

CSC function is determined by a dysregulation of stemness-related signaling pathways. A reduced level of tumor differentiation and increased self-renewal are a characteristic of stemness. Transcription factors which are master regulators of self-renewal and pluripotency in embryonic stem cells (ESCs) have been demonstrated to play a key role in the regulation of stemness in cancer [56]. These transcription factors include the octamer-binding transcription factor 4 (OCT4), the sex-determining region Y-box 2 (SOX2), the homeobox transcription factor NANOG, the Kruppel-like factor 4 (KLF4), and the proto-oncogene C-MYC [57,58,59].
Expression of these factors can reprogram somatic cells into induced cancer stem cells and promote cell plasticity allowing cancer cells to adapt, survive, grow, and resist therapies. This effect has been demonstrated by a recent study showing acquisition of stemness after induced expression of OCT4, SOX2, and NANOG and high expression of pluripotency genes in advanced prostate, bladder, and renal cancers which was correlated with aggressive disease and drug resistance [60]. In addition, pluripotency factors have been shown to mediate cell plasticity in the TME and of enhance ECM production leading to metastasis [61]. Expression of pluripotency factors also regulate the expression of EMT mediators SNAI1 and SNAI2 [62].
Ectopic expression of OCT4 induced a block of differentiation and dysplasia in epithelial tissues [63,64]. Expression of OCT4 has been found in several cancer types and it contributes to the self-renewal and chemoresistance of CSCs [65,66]. Indeed, OCT4 induces the expression of the drug transporter ABCG2, which is highly expressed in CSCs and responsible for drug resistance [67]. Moreover, a relationship between OCT4 translation and metastasis of colorectal cancer to the liver have been demonstrated [68]. Similarly, it has been shown that OCT4 expression in lung cancer cells promotes the polarization of M2 type macrophages due the macrophage colony-stimulating factor (M-CSF) secretion, which leads to increase in tumor growth and metastasis [69].
SOX2 expression is also associated with cancer stemness [70,71]. Expression of this transcription factor is increased in cells and tumor tissue of patients with triple-negative breast cancer (TNBC). Importantly, inhibition of SOX2 suppresses proliferation and invasion of breast cancer cells, inducing cell apoptosis in vitro and inhibiting tumor growth and metastasis in vivo [72]. SOX2 knockout in a mouse model of osteosarcoma also induces a sharp decrease in frequency and occurrence of tumors [73]. In addition, SOX2 and CD133 co-expression can be associated with poor outcome in colon, stomach, and ovarian cancers, as well as melanoma and advanced cancers with bone metastases [74].
NANOG is also involved in maintaining embryonic stem cell self-renewal and cancer stemness [75,76,77]. It has been shown that an increase in the number of oral cancer stem-like cells is associated with increase expression of NANOG and increase malignancy [78]. The expression of this transcription factor increases with the degree of dysplasia and is an early predictor of cancer risk in patients with oral cavity malignant diseases [79]. Mutation in the tumor suppressor SPOP and negative regulator of NANOG also leads to increased stemness of prostate cancer and a negative prognosis in prostate cancer [80]. Dehghan Harati et al. have shown that the expression of NANOG is associated with the increased activity of ALDH and radioresistance, as well as with repair of double-strand DNA breaks [81].
Together with other pluripotency genes, KLF4 plays an important role in the regulation of cell growth, proliferation, and differentiation [82]. In embryonic stem cells, KLF4 activates the expression of telomerase reverse transcriptase (TERT) and contributes to the maintenance of self-renewal [83]. In cancer, KLF4 can act either as oncogene by inhibiting apoptosis or tumor suppressor by inducing p21-dependent cell cycle arrest. For instance, KLF4 is highly expressed in a subset of human melanomas and ectopic KLF4 expression enhances melanoma cell growth by decreasing apoptosis [84]. It has also been shown that KLF4 expression is associated with stemness of osteosarcoma [85]. However, KLF4 can also function as tumor suppressor and its knockdown can promote migration and invasion of non-small-cell lung carcinoma (NSCLC) [86].
Similarly, enhanced expression of KLF4 by lentiviral transduction increased sensitivity of ovarian cancer cells to the chemotherapeutic drugs paclitaxel and cisplatin [87].
Finally, C-MYC coordinates various biological processes in stem cells, such as cell cycle, cell metabolism, self-renewal, differentiation, and apoptosis [88]. Mutations in MYC genes have been found in many tumors and C-MYC is upregulated and acts as an oncogene in more than 50% of human cancers [89]. The expression of C-MYC correlates with the level of differentiation in cancer, as expression of C-MYC induces de-differentiation and acquisition of CSC properties, including glutamine metabolic addiction, dormancy and therapeutic resistance [90]. Dysregulation of MYC usually plays an important role in maintaining the number of invasive CSCs. For example, increased expression of MYC is associated with glioblastoma CSC-induced cell proliferation and invasion, and apoptosis inhibition [91].

3. Signaling Pathways Modulate Cancer Stemness

Several signaling pathways that are known mediators of juxtacrine (cell–cell) and paracrine extracellular signaling in the local TME have been identified to be key extrinsic players in the regulation of cancer stemness. These include Wnt, Notch, Hedgehog (Hh), Janus kinase/signal transducers and activators of transcription (JAK/STAT), and phosphatidylinositol 3-kinase/serine/threonine-protein kinase/mammalian target of the rapamycin (PI3K/AKT/mTOR) (reviewed in detail in Yang et al.) [56]. Moreover, some of these pathways also participate in epithelial-to-mesenchymal and mesenchymal-to-epithelial (MET) transitions, thus regulating cell identity and plasticity [92].
In addition, there are a large number of studies related to other signaling pathways involved in cancer progression, self-renewal, and metastasis of CSCs [56,93]. For example, recent developments to target and inhibit NF-κB in the ovarian cancer or disruption of the NF-κB/IL-8 signaling in breast cancer can potential targeted therapy for CSCs [94,95,96]. Signaling regulation can be complex in different types of tumors, with cross-interaction of pathways participating in the regulation of CSCs [54,97].

3.1. Wnt Signaling

The activation of the Wnt pathway is common in cancer and can be caused by mutations in Wnt signaling components [98,99,100], as well as in downstream targets. Indeed, aberrant activation of Wnt mediators such as APC, β-catenin, Axin, Wnt1, and others are found in many cancers. For instance, thyroid receptor-interacting protein 6 (TRIP6) is an adapter protein that belongs to Lim proteins Zixin family and plays an important role in regulating the function of CSCs in breast cancer through regulation of Wnt/β-Catenin signaling [101]. Similarly, B-cell lymphoma/leukemia 11A (BCL11A) contributes to formation and invasion of tumor cells, stem cell self-renewal and activation of signalling by Wnt/β-Catenin and the EMT pathway. In addition, BCL11A is associated with lung metastasis and increase stemness of breast cancer cells [102].
Interestingly, glioblastoma cells expressing high levels of Wnt demonstrated expression of OCT-4, SOX2, NANOG, NESTIN, and CD133, thus suggesting a role of Wnt signaling in the maintenance of glioma CSCs [103].

3.2. Notch Signaling

The Notch pathway is also important for CSC function, and it is activated in tumors surviving and adapting to their microenvironment. Activation of the Notch pathway contributes to self-renewal, metastasis, and suppression of apoptosis. For example, the aberrant transmission of Notch signals (Notch1 and Notch4) contributes to self-renewal and metastasis of breast CSCs [104]. High levels of Notch1, Notch3, JAG1, JAG2, and the target HES-1 are found in pancreatic and breast cancers [105,106]. Notch signaling is activated under hypoxic conditions in breast cancer mediating chemoresistance and CSC expansion, which can be reversed by treatment with Notch inhibitors [107]. In addition, suppression of Notch1 via miR-34a can lead to an increase in breast cancer cell chemosensitivity to paclitaxel with a reduction in CSC proliferation and expression of the stemness marker ALDH1 [108]. Glioma stem cells are also regulated by activation of Notch1 and they show increased expression of the CSC genes OCT4 and CD133 under hypoxia [109].

3.3. Hedgehog Signaling

Together with Wnt and Notch signaling, the Hedgehog pathway is involved in embryonic development and organogenesis, including the nervous system, and organs such as lung, heart, and bowel [110]. Abnormal activation of the Hedgehog signaling pathway can be detected in CSCs [111,112]. For instance, it contributes to self-renewal, proliferation, and tumorigenicity of lung adenocarcinoma stem cells [113]. Through activation of the PTCH1 receptor and downstream effector Gli-1, Hedgehog signaling stimulates the transcription of the target genes OCT4, SOX2, NANOG, and C-MYC [114]. Zhu et al. showed that SHH, PTCH1, and Gli-1 are activated by TSPAN8 expression in breast CSCs leading to increased expression of NANOG, OCT4, and ALDHA1 genes, as well as increased stem cell self-renewal and cell survival after treatment with adriamycin and paclitaxel [115]. Similarly, Hedgehog signaling stimulates self-renewal of glioma CSCs as they overexpress SHH, PTCH11, and GLI1 [116]. The Hedgehog pathway has also been shown to be important for pancreatic CSCs, as inhibition of the ligand SHH by inhibition of sialidase-2 (Neu2) and desialylation leads to a decrease in stemness [117].

3.4. JAK/STAT Signaling

The JAK/STAT pathway promotes survival, self-renewal, hematopoiesis, and neurogenesis of ESCs [118]. This pathway is also activated in CSCs [119]. Among the different subtypes of STAT proteins, activation of STAT3 plays an important role in CSC function by regulating oncogenic signaling pathways. STAT3 is constitutively activated in many different cancers, including pancreatic breast, prostate, ovarian liver, colorectal, and bone cancers, as well as leukemia and melanoma. In addition, STAT3 activation is associated with the generation of glioblastoma stem cells and the metastatic potential of colon CSCs [90]. As well as STAT3, it has been shown that suppression of STAT1 reduces the formation of lung A549 tumor spheres which was maintained by the suppression of factors associated with stemness, such as SOX2, OCT4, and NANOG [120].

3.5. AKT/mTOR Signaling

The PI3K/AKT/mTOR signaling pathway is important for cell proliferation and survival, and abnormal activation of PI3K/mTOR signals is commonly found in cancer [121,122]. The activation of this pathway also increases the migration, invasion, and resistance of the CSCs [123]. The transmission of PI3K/AKT signals is part of the main molecular stemness program both in mouse and human pluripotent stem cells. The oncogenic version of PIK3CAH1047R in cancer causes constitutive activation of the PI3K pathway and is associated with increased stemness in a dose-dependent manner, as shown in mouse models of breast, lung, and colorectal cancers [124]. Activation of the PI3KCA is also associated with induction of EMT and stem cell plasticity through multiple signals, including TGFβ [49].

4. Influence of the Microenvironment on CSC

Stem cells cannot survive outside their niche environment or in the absence of specific pluripotency factors and signaling pathways that support stem cell function [125]. Importantly, these factors can facilitate the emergence of stem cells from more differentiated cells, as these retain the ability to dedifferentiate and return to a more primitive developmental state [126].
The plasticity demonstrated by cancer cells is key in cancer as extrinsic factors can promote the acquisition of stemness by reprogramming cancer cells into CSCs. These factors include cytokine and growth factors secreted cells of the TME (mesenchymal stem cells (MSCs), macrophages, tumor-associated fibroblasts (TAFs)), as well as extracellular vesicles (EVs), and hypoxia [127]. In epithelial tissues, the activation of EMT has been linked to the formation of both normal cells and CSCs [128]. Fundamental to the process of gastrulation during embryo development, EMT is activated in the adult during wound healing and in cancer [129].
EMT is a reversible process with cells changing phenotypes from epithelial to mesenchymal and then back to epithelial through MET. These highly dynamic processes are regulated by paracrine signaling, most notably TGF-β, Wnt, and others involved in maintaining stem cell function, as described above. These pathways then induce expression of factors triggering EMT, including transcription factors of the TWIST, SNAIL, and ZEB families, splicing factors and microRNAs (e.g., miR34, miR200) which drive the loss of expression of adhesion molecules such as E-cadherin (encoded by the CDH1 gene), as well as the acquisition of mesenchymal markers, such as Vimentin [130].
Phenotypic plasticity linked to EMT has important implications for CSCs and their cellular origin in different tumor types. For instance, both epithelial and mesenchymal cells in the human breast can adopt a CSC phenotype and co-exist in tumor. Indeed, epithelial CSCs are proliferative and express ALDH, whereas mesenchymal CSCs are mostly quiescent and display a CD44hi/CD24- profile [131]. This dynamic equilibrium is regulated by the TME and the resulting heterogeneity is at the basis of the existence of different disease molecular and pathological subtypes in most solid tumors [42].
Factors associated with inflammation, such as tumor necrosis factor (TNF), interleukin-6 (IL-6), and IL-1β, can activate EMT [132]. For instance, IL-6 serum levels are high in osteosarcoma patients and the cytokine stimulates osteosarcoma stemness as measured in a self-renewal spheroid assay [133]. It was also found that IL-1β can increase the formation of colon cancer spheres, which show an up-regulation of stemness factor genes and increased drug resistance [134]. Finally, tumor necrosis factor (TNF)-α promotes HPV-associated oral carcinogenesis by increasing stemness [135].
These signaling pathways are also involved in the communication between cancer-associated fibroblasts (CAFs) present in the tissue stroma and cancer cells. Indeed, CAFs can activate signaling promoting cancer stemness through activation of Wnt and Notch signaling. CSCs, in turn, can influence CAFs through activation of signals involved in cancer progression, including the Hedgehog pathway [136]. Inter-related signaling pathways also link hypoxia with EMT. Indeed, hypoxia can directly induce EMT via the activation of the hypoxia-inducible factor (HIF)-1α through cross-talk with TGFβ and Wnt/β catenin pathways. In addition, hypoxia can also induce EMT via HIF-independent pathways which include AMPK, PIK/AKT, MAPK, NF-kB, and Notch signalling [137].
Other non-cellular components of the TME can modulate CSCs, including ECM and EVs. Among ECM molecules, tenascin-C is involved in the stimulation of self-renewal of CSCs. In breast cancer, it promotes stemness through upregulation of the CSC marker LRG5 [138] and it is also associated with poor prognosis in glioblastoma and represents a candidate CSC markers in this cancer type [139]. In addition, the ECM provides a physical barrier to CSCs from cytotoxic drugs and may promote EMT, self-renewal, expression of CSC markers, and drug resistance. ECM properties such as stiffness and porosity affect various CSC functions. The rigidity of the ECM is involved in the regulation of self-renewal and differentiation of stem cells [140,141]. Tumor ECM is usually more rigid than normal tissue ECM due to overexpression of collagens, proteoglycans, and ECM-modifying enzymes (lysyl oxidases) [142].
Finally, EVs isolated from tumor and stromal cells are involved in various stages of tumor progression such as proliferation, angiogenesis, metastasis, and drug resistance [143]. Tumor cells secrete a heterogeneous set of EVs, which differ in size, biogenesis, and molecular composition, which include cytoplasmic proteins, proteins interacting with lipid rafts, DNA, and RNA [144]. Communication through EVs is important for the maintenance of CSCs. For instance, Evs released by glioblastoma stem cells promote self-renewal and angiogenesis through endothelial tube formation [145]. Similarly, exosomes derived from TAFs promote the formation of colorectal cancer spheres by activating Wnt signaling and ultimately increasing the number of CSCs [146]. Gonzalez et al. also showed that stem/progenitor-enriched mammospheres from primary mammary epithelial cells can secrete extracellular vesicles that are capable of altering the expression levels of genes involved in EMT and stem cell markers [147].

5. Models of Cancer Stemness and TME Interaction

Culture models that allow cancer cells to re-acquire or enrich for stem cell characteristics have been developed to study CSCs. These culture models are aimed at preserving the biological characteristics of primary tumors, thus exhibiting the original tumor architecture and metabolic activity [148]. They also provide more accurate data on the effects of therapeutic agents and processes such as EMT and MET [149].
Among the 3D culture model systems which allow CSC self-renewal are spheroids and organoids cultured in suspension, scaffolds, matrix, and hydrogel cultures.

5.1. Spheroids

Spheroids are 3D tumor models that can be obtained by aggregation or spontaneous self-assembly of cancer cells. Aggregation can be caused by forced cell-to-cell contact by various methods such as hanging drop, liquid application/cell suspension culture, microwell culture (96-well round bottom plates), or microfluidics (i.e., gel encapsulation) [150]. Tumorsphere cultures are mainly generated by single cancer cell suspension in serum-free media. Under these conditions, only cells with self-renewal characteristics allow the formation of cell aggregates or spheroids that can proliferate and be serially passaged [151,152]. Spheroid cultures have been established from many different cancer types, including breast, prostate, bone, skin, brain, and colon tumors. Biomaterials can be used to support 3D CSC models, as they provide a physical structure to create a niche environment which is important to better sustain CSC function. Porous scaffolds are most commonly made of synthetic (polyglycolic and polylactic acids) or natural (collagen, alginate) polymers and hyaluronic acid. These have been used to support CSC proliferation in prostate and breast cancer, hepatocellular carcinoma, and glioblastoma. Hydrogels (natural and synthetic) are also widely used for 3D models of CSCs and they can be coupled with the use of extracellular matrix [153].

5.2. Organoids

Differently from spheroids, organoids are 3D cultures of stem cells that enable the differentiation and self-organization of tissue-like structures in vitro. These cultures are supported by embedding of cells in matrix and growth factors that normally support the survival and growth or stem cells in their natural niche. Organoids can be derived from cells isolated from primary tumors and many studies have demonstrated their ability to maintain tumor heterogeneity and pathological characteristics, even after multiple passaging. Organoids have been already established from a range of tumors, including colon, breast, pancreatic, liver, and prostate cancer. For example, 3D epithelial organoids can be established clonally from LRG5-positive intestinal stem cells and colon cancer cells [154].
Another application of organoids in CSC research is the use of iPSC technology and gene editing. Indeed, organoids can be generated through differentiation of gene-edited normal iPSC to carrying cancer driver mutations as well as iPSC derived from tumor cells isolated from cancer patients. The cancer iPSC-derived organoids have been shown to contain CSC that establish heterogeneous tumoroid structures that recapitulate primary tumors [155].

5.3. TME Interaction

Although the described models are powerful tools to maintain and enrich CSCs ex vivo, they face the challenge of mimicking the intra-tumor heterogeneity of primary tumors due to the modulation of CSC by their microenvironment. The bidirectional crosstalk between CSC and TME maintains cancer stemness. Niche cells of the TME communicate with CSC inducing self-renewal and tumor progression. Physical characteristics of the CSC niche, such as hypoxia and interaction with ECM, also affect CSC function [156]. These features can be modeled in vitro to some extent and current research efforts are directed to create model systems that closely resemble patient’s cancer complexity. Histomorphological analysis shows that the spheroid model is divided into several zones, like a primary tumor. The outer marginal zone is characterized by preferentially proliferating cells, the middle zone by dormant cells, and the central part by a necrotic core due to lack of oxygen and nutrients. The central part of spheroids is characterized by the accumulation of lactate due to the significant consumption of glucose in the proliferating layer of cells whilst the distribution of glucose occurs evenly in all areas of spheroids [157]. Glycolytic metabolism and hypoxia are critical for maintaining stemness of CSCs. A hypoxic microenvironment can also activate CSC signalling. For instance, breast CSCs in mammospheres activate Wnt and Notch signalling in hypoxia [107]. This is mirrored by glioblastoma and colorectal CSC activating Notch, Hedgehog, and Wnt pathways [158]. ECM proteins play an important role in the modulation of stemness in CSC models. In a 3D model of colon cancer using afibrin gel, CSC properties were enriched with the activation of NANOG [133]. Similarly, collagen type I (Col-I) oligomer 3D matrices induce stemness and EMT in pancreatic cells [159].
Interactions with cells of the TME can be effectively modelled to study modulation of CSCs. Breast cancer cells can adopt a proliferative and EMT phenotype when cultured in 3D spheroids in the presence of mesenchymal stem cells (MSCs) [160]. The effect of MSCs on tumor cells has also been studied by creating hybrids with gastric cancer cells using polyethylene glycol (PEG) in vitro. These hybrids show a change in EMT with down-regulation of E-cadherin and up-regulation of vimentin, N-cadherin, α-smooth muscle actin (α-SMA), and fibroblast activation protein (FAP). Hybrids increase the expression of the stemness factors OCT4, NANOG, SOX2, and LIN28, upregulate CD44 and CD133 as well as promote gastric xenograft tumor growth in vivo [161].
Therefore, the co-culture of cancer cell in 3D with cells of the TME can model the complexity of patient’s tumors which, combined with the use of microfluidic devises and 3D bioprinting, can be used to investigate many aspects of tumor stemness.
As the eradication of CSCs is clinically important, the study of CSCs in a TME context can provide invaluable clues in the fight against cancer, such as the discovery of new biomarkers for early detection of cancer and effective therapies [162,163].

6. Conclusions and Future Perspectives

The survival rate of cancer patients has increased significantly over the past decades, thanks to the development of anticancer therapy which includes interdisciplinary care, improved chemotherapy drugs, and targeted biological agents. However, despite these advances, the problems of cancer relapse and treatment of advanced-stage tumors remains unresolved. Currently, there is no therapy that would effectively treat patients with advanced metastatic disease. The main factors that complicate the treatment of patients are cellular heterogeneity, as well as the structural and molecular complexity of the tumors. Intratumor heterogeneity arises from genetic mutations, interactions with the microenvironment, and the presence of tumor stem cells. Activation of signaling pathways that promote self-renewal and chemoresistance allow CSCs not only to successfully survive unfavorable conditions but also to self-renew with the production of resistant clones. Therefore, targeting cancer stemness is an increasingly important therapeutic approach to halt tumor progression, recurrence, and resistance to therapy.
To this end, there is some evidence that some compounds have a direct effect on CSCs. For instance, salinomycin, a potassium ionophore, can selectively kill breast cancer stem cells and induce epithelial differentiation of breast tumor cells, promoting effective tumor removal [164]. In addition, epigallocatechin gallate reduces the stem and oncogenicity of human lung cancer cells by inhibiting AXL receptors [165].
Although promising, data of effective CSC therapies are limited and therefore the development of CSC-specific therapies requires better knowledge of the relationship between CSCs and their microenvironment. A deeper understanding of the intrinsic and extrinsic factors involved in shaping this interaction is likely to be essential to overcome the barrier of CSC plasticity. Although therapeutic strategies aimed at inhibiting the signaling pathways of CSC are available (Table 2), it is necessary to continue researching new strategies that can target factors inducing cancer stemness, including TME (EMT, hypoxia, CAFs, vesicles), metabolism, and chemoresistance [166].
However, a significant obstacle to the development of effective therapies that can take into account cancer complexity is the discrepancy between model systems and patient’s tumors. Modelling of the TME is especially complex and therefore the use of clinically relevant and close to patient models can address the probe of the high attrition rate in cancer drug development. In addition, the combination of different models can add to their predictive value. For instance, the use of CSCs to create cultures of non-adherent spheres, three-dimensional tumor organoids, and patient-derived xenografts have shown that treatment with Wnt and Notch inhibitors can block the proliferation and self-renewal of CSCs [11].
Thus, the use of advanced and complex model systems will expand the possibilities of CSC therapy, which will stimulate the development of precision medicine.

Author Contributions

Conceptualization, A.P., Z.G., V.S. and A.R.; writing—original draft preparation, A.P., Z.G., C.A. and V.S.; writing—review and editing, V.S. and A.R.; supervision, V.S. and A.R. All authors have read and agreed to the published version of the manuscript.

Funding

This paper has been supported by the Russian Science Foundation grant 21-74-10021.

Acknowledgments

This paper has been supported by the Kazan Federal University Strategic Academic Leadership Program (PRIORITY-2030).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Feinberg, A.P.; Ohlsson, R.; Henikoff, S. The epigenetic progenitor origin of human cancer. Nat. Rev. Genet. 2006, 7, 21–33. [Google Scholar] [CrossRef] [PubMed]
  2. Elkashty, O.A.; Hariharan, A.; Tran, S.D. Aging, stem cells and cancer updated. Aging 2021, 13, 20854–20855. [Google Scholar] [CrossRef] [PubMed]
  3. Sell, S. On the Stem Cell Origin of Cancer. Am. J. Pathol. 2010, 176, 2584–2594. [Google Scholar] [CrossRef] [PubMed]
  4. Beck, B.; Blanpain, C. Unravelling cancer stem cell potential. Nat. Rev. Cancer 2013, 13, 727–738. [Google Scholar] [CrossRef] [PubMed]
  5. Albini, A.; Bruno, A.; Gallo, C.; Pajardi, G.; Noonan, D.M.; Dallaglio, K. Cancer stem cells and the tumor microenvironment: Interplay in tumor heterogeneity. Connect Tissue Res. 2015, 56, 414–425. [Google Scholar] [CrossRef] [Green Version]
  6. Girouard, S.D.; Murphy, G.F. Melanoma stem cells: Not rare, but well done. Lab. Investig. 2011, 91, 647–664. [Google Scholar] [CrossRef] [Green Version]
  7. Reya, T.; Morrison, S.J.; Clarke, M.F.; Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 2001, 414, 105–111. [Google Scholar] [CrossRef] [Green Version]
  8. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef]
  9. Capp, J.P. Cancer Stem Cells: From Historical Roots to a New Perspective. J. Oncol. 2019, 2019, 5189232 . [Google Scholar] [CrossRef]
  10. Kim, W.T.; Ryu, C.J. Cancer stem cell surface markers on normal stem cells. BMB Rep. 2017, 50, 285–298. [Google Scholar] [CrossRef] [Green Version]
  11. Fendler, A.; Bauer, D.; Busch, J.; Jung, K.; Wulf-Goldenberg, A.; Kunz, S.; Song, K.; Myszczyszyn, A.; Elezkurtaj, S.; Erguen, B.; et al. Inhibiting WNT and NOTCH in renal cancer stem cells and the implications for human patients. Nat. Commun. 2020, 11, 929. [Google Scholar] [CrossRef] [Green Version]
  12. Dou, J.; Gu, N. Emerging strategies for the identification and targeting of cancer stem cells. Tumor Biol. 2010, 31, 243–253. [Google Scholar] [CrossRef]
  13. Duan, J.J.; Qiu, W.; Xu, S.L.; Wang, B.; Ye, X.Z.; Ping, Y.F.; Zhang, X.; Bian, X.W.; Yu, S.C. Strategies for isolating and enriching cancer stem cells: Well begun is half done. Stem Cells Dev. 2013, 22, 2221–2239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Goossens, S.; Vandamme, N.; Van Vlierberghe, P.; Berx, G. EMT transcription factors in cancer development re-evaluated: Beyond EMT and MET. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 584–591. [Google Scholar] [CrossRef] [PubMed]
  15. Mercurio, A.M. VEGF/Neuropilin Signaling in Cancer Stem Cells. Int. J. Mol. Sci. 2019, 20, 490. [Google Scholar] [CrossRef] [Green Version]
  16. Pattabiraman, D.R.; Weinberg, R.A. Tackling the cancer stem cells—What challenges do they pose? Nat. Rev. Drug Discov. 2014, 13, 497–512. [Google Scholar] [CrossRef] [Green Version]
  17. Farace, C.; Pisano, A.; Grinan-Lison, C.; Solinas, G.; Jimenez, G.; Serra, M.; Carrillo, E.; Scognamillo, F.; Attene, F.; Montella, A.; et al. Deregulation of cancer-stem-cell-associated miRNAs in tissues and sera of colorectal cancer patients. Oncotarget 2020, 11, 116–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Nallanthighal, S.; Heiserman, J.P.; Cheon, D.J. The Role of the Extracellular Matrix in Cancer Stemness. Front. Cell Dev. Biol. 2019, 7, 86. [Google Scholar] [CrossRef] [PubMed]
  19. Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. [Google Scholar] [CrossRef] [PubMed]
  20. Jijiwa, M.; Demir, H.; Gupta, S.; Leung, C.; Joshi, K.; Orozco, N.; Huang, T.; Yildiz, V.O.; Shibahara, I.; de Jesus, J.A.; et al. CD44v6 Regulates Growth of Brain Tumor Stem Cells Partially through the AKT-Mediated Pathway. PLoS ONE 2011, 6, e24217. [Google Scholar] [CrossRef]
  21. Matsui, W.; Huff, C.A.; Wang, Q.J.; Malehorn, M.T.; Barber, J.; Tanhehco, Y.; Smith, B.D.; Civin, C.I.; Jones, R.J. Characterization of clonogenic multiple myeloma cells. Blood 2004, 103, 2332–2336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Singh, S.K.; Hawkins, C.; Clarke, I.D.; Squire, J.A.; Bayani, J.; Hide, T.; Henkelman, R.M.; Cusimano, M.D.; Dirks, P.B. Identification of human brain tumour initiating cells. Nature 2004, 432, 396–401. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, S.; Balch, C.; Chan, M.W.; Lai, H.C.; Matei, D.; Schilder, J.M.; Yan, P.S.; Huang, T.H.M.; Nephew, K.P. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008, 68, 4311–4320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Szotek, P.P.; Pieretti-Vanmarcke, R.; Masiakos, P.T.; Dinulescut, D.M.; Connolly, D.; Foster, R.; Dombkowski, D.; Preffer, F.; MacLaughlin, D.T.; Donahoe, P.K. Ovarian cancer side population defines cells with stem cell-like characteristics and Mullerian Inhibiting Substance responsiveness. Proc. Natl. Acad. Sci. USA 2006, 103, 11154–11159. [Google Scholar] [CrossRef] [Green Version]
  25. Sims-Mourtada, J.; Izzo, J.G.; Ajani, J.; Chao, K.S.C. Sonic Hedgehog promotes multiple drug resistance by regulation of drug transport. Oncogene 2007, 26, 5674–5679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Collins, A.T.; Berry, P.A.; Hyde, C.; Stower, M.J.; Maitland, N.J. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 2005, 65, 10946–10951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Vermeulen, L.; Melo, F.; van der Heijden, M.; Cameron, K.; de Jong, J.H.; Borovski, T.; Tuynman, J.B.; Todaro, M.; Merz, C.; Rodermond, H.; et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat. Cell Biol. 2010, 12, 468–476. [Google Scholar] [CrossRef]
  28. Katoh, Y.; Katoh, M. Comparative genomics on PROM1 gene encoding stem cell marker CD133. Int. J. Mol. Med. 2007, 19, 967–970. [Google Scholar] [CrossRef]
  29. Van Der Flier, L.G.; Sabates-Bellver, J.; Oving, I.; Haegebarth, A.; De Palo, M.; Anti, M.; Van Gijn, M.E.; Suijkerbuijk, S.; Van De Wetering, M.; Marra, G.; et al. The intestinal Wnt/TCF signature. Gastroenterology 2007, 132, 628–632. [Google Scholar] [CrossRef]
  30. Dalerba, P.; Dylla, S.J.; Park, I.K.; Liu, R.; Wang, X.H.; Cho, R.W.; Hoey, T.; Gurney, A.; Huang, E.H.; Simeone, D.M.; et al. Phenotypic characterization of human colorectal cancer stem cells. Proc. Natl. Acad. Sci. USA 2007, 104, 10158–10163. [Google Scholar] [CrossRef] [Green Version]
  31. Lonardo, E.; Hermann, P.C.; Mueller, M.T.; Huber, S.; Balic, A.; Miranda-Lorenzo, I.; Zagorac, S.; Alcala, S.; Roderiguez-Arabaolaza, I.; Ramirez, J.C.; et al. Nodal/Activin Signaling Drives Self-Renewal and Tumorigenicity of Pancreatic Cancer Stem Cells and Provides a Target for Combined Drug Therapy. Cell Stem Cell 2011, 9, 433–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Li, C.W.; Lee, C.J.; Simeone, D.M. Identification of Human Pancreatic Cancer Stem Cells. In Cancer Stem Cells: Methods Protoc; Yu, J.S., Ed.; Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2009; Volume 568, pp. 161–173. [Google Scholar]
  33. Yang, Z.F.; Ho, D.W.; Ng, M.N.; Lau, C.K.; Yu, W.C.; Ngai, P.; Chu, P.W.K.; Lam, C.T.; Poon, R.T.P.; Fan, S.T. Significance of CD90(+) cancer stem cells in human liver cancer. Cancer Cell 2008, 13, 153–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Eramo, A.; Lotti, F.; Sette, G.; Pilozzi, E.; Biffoni, M.; Di Virgilio, A.; Conticello, C.; Ruco, L.; Peschle, C.; De Maria, R. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008, 15, 504–514. [Google Scholar] [CrossRef] [PubMed]
  35. Mei, Y.; Cai, D.Y.; Dai, X.F. Modulating cancer stemness provides luminal a breast cancer cells with HER2 positive-like features. J. Cancer 2020, 11, 1162–1169. [Google Scholar] [CrossRef] [Green Version]
  36. Liu, S.L.; Dontu, G.; Mantle, I.D.; Patel, S.; Ahn, N.S.; Jackson, K.W.; Suri, P.; Wicha, M.S. Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res. 2006, 66, 6063–6071. [Google Scholar] [CrossRef] [Green Version]
  37. Liu, S.L.; Ginestier, C.; Ou, S.J.; Clouthier, S.G.; Patel, S.H.; Monville, F.; Korkaya, H.; Heath, A.; Dutcher, J.; Kleer, C.G.; et al. Breast Cancer Stem Cells Are Regulated by Mesenchymal Stem Cells through Cytokine Networks. Cancer Res. 2011, 71, 614–624. [Google Scholar] [CrossRef] [Green Version]
  38. Bocci, F.; Gearhart-Serna, L.; Boareto, M.; Ribeiro, M.; Ben-Jacob, E.; Devi, G.R.; Levine, H.; Onuchic, J.N.; Jolly, M.K. Toward understanding cancer stem cell heterogeneity in the tumor microenvironment. Proc. Natl. Acad. Sci. USA 2019, 116, 148–157. [Google Scholar] [CrossRef] [Green Version]
  39. Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [Green Version]
  40. Viatour, P. Bridges between Cell Cycle Regulation and Self-Renewal Maintenance. Genes Cancer 2012, 3, 670–677. [Google Scholar] [CrossRef] [Green Version]
  41. Simons, B.D.; Clevers, H. Strategies for homeostatic stem cell self-renewal in adult tissues. Cell 2011, 145, 851–862. [Google Scholar] [CrossRef] [Green Version]
  42. Brooks, M.D.; Burness, M.L.; Wicha, M.S. Therapeutic Implications of Cellular Heterogeneity and Plasticity in Breast Cancer. Cell Stem Cell 2015, 17, 260–271. [Google Scholar] [CrossRef] [Green Version]
  43. Cabrera, M.C.; Hollingsworth, R.E.; Hurt, E.M. Cancer stem cell plasticity and tumor hierarchy. World J. Stem Cells 2015, 7, 27–36. [Google Scholar] [CrossRef]
  44. Chaffer, C.L.; Brueckmann, I.; Scheel, C.; Kaestli, A.J.; Wiggins, P.A.; Rodrigues, L.O.; Brooks, M.; Reinhardt, F.; Su, Y.; Polyak, K.; et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc. Natl. Acad. Sci. USA 2011, 108, 7950–7955. [Google Scholar] [CrossRef] [Green Version]
  45. Borovski, T.; Melo, F.D.E.; Vermeulen, L.; Medema, J.P. Cancer Stem Cell Niche: The Place to Be. Cancer Res. 2011, 71, 634–639. [Google Scholar] [CrossRef] [Green Version]
  46. Jing, Y.; Han, Z.; Zhang, S.; Liu, Y.; Wei, L. Epithelial-Mesenchymal Transition in tumor microenvironment. Cell Biosci. 2011, 1, 29. [Google Scholar] [CrossRef] [Green Version]
  47. Mittal, V. Epithelial Mesenchymal Transition in Tumor Metastasis. Annu. Rev. Pathol. 2018, 13, 395–412. [Google Scholar] [CrossRef]
  48. Abell, A.N.; Johnson, G.L. Implications of Mesenchymal Cells in Cancer Stem Cell Populations: Relevance to EMT. Curr. Pathobiol. Rep. 2014, 2, 21–26. [Google Scholar] [CrossRef] [Green Version]
  49. Shibue, T.; Weinberg, R.A. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 611–629. [Google Scholar] [CrossRef] [Green Version]
  50. Mego, M.; Reuben, J.; Mani, S. Epithelial-Mesenchymal Transition (EMT) and Cancer Stem Cells (CSCs): The Traveling Metastasis; Cancer Drug Discovery and Development; Humana Press: Totowa, NJ, USA, 2017; p. 14. [Google Scholar]
  51. Tanabe, S.; Quader, S.; Cabral, H.; Ono, R. Interplay of EMT and CSC in Cancer and the Potential Therapeutic Strategies. Front. Pharm. 2020, 11, 904. [Google Scholar] [CrossRef]
  52. Aponte, P.M.; Caicedo, A. Stemness in Cancer: Stem Cells, Cancer Stem Cells, and Their Microenvironment. Stem Cells Int. 2017, 2017, 5619472. [Google Scholar] [CrossRef]
  53. Lin, D.; Shen, L.; Luo, M.; Zhang, K.; Li, J.; Yang, Q.; Zhu, F.; Zhou, D.; Zheng, S.; Chen, Y.; et al. Circulating tumor cells: Biology and clinical significance. Signal Transduct Target 2021, 6, 404. [Google Scholar] [CrossRef]
  54. Visvader, J.E.; Lindeman, G.J. Cancer stem cells: Current status and evolving complexities. Cell Stem Cell 2012, 10, 717–728. [Google Scholar] [CrossRef] [Green Version]
  55. Laplane, L.; Solary, E. Towards a classification of stem cells. Elife 2019, 8, e46563. [Google Scholar] [CrossRef]
  56. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target 2020, 5, 8. [Google Scholar] [CrossRef] [Green Version]
  57. Muller, M.; Hermann, P.C.; Liebau, S.; Weidgang, C.; Seufferlein, T.; Kleger, A.; Perkhofer, L. The role of pluripotency factors to drive stemness in gastrointestinal cancer. Stem Cell Res. 2016, 16, 349–357. [Google Scholar] [CrossRef] [Green Version]
  58. Zhu, Y.; Huang, S.; Chen, S.; Chen, J.; Wang, Z.; Wang, Y.; Zheng, H. SOX2 promotes chemoresistance, cancer stem cells properties, and epithelial-mesenchymal transition by beta-catenin and Beclin1/autophagy signaling in colorectal cancer. Cell Death Dis. 2021, 12, 449. [Google Scholar] [CrossRef]
  59. Yasumizu, Y.; Rajabi, H.; Jin, C.; Hata, T.; Pitroda, S.; Long, M.D.; Hagiwara, M.; Li, W.; Hu, Q.; Liu, S.; et al. MUC1-C regulates lineage plasticity driving progression to neuroendocrine prostate cancer. Nat. Commun. 2020, 11, 338. [Google Scholar] [CrossRef] [Green Version]
  60. Hepburn, A.C.; Steele, R.E.; Veeratterapillay, R.; Wilson, L.; Kounatidou, E.E.; Barnard, A.; Berry, P.; Cassidy, J.R.; Moad, M.; El-Sherif, A.; et al. The induction of core pluripotency master regulators in cancers defines poor clinical outcomes and treatment resistance. Oncogene 2019, 38, 4412–4424. [Google Scholar] [CrossRef]
  61. Huang, J.; Zhang, L.; Wan, D.; Zhou, L.; Zheng, S.; Lin, S.; Qiao, Y. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target 2021, 6, 153. [Google Scholar] [CrossRef]
  62. Wang, D.; Lu, P.; Zhang, H.; Luo, M.; Zhang, X.; Wei, X.; Gao, J.; Zhao, Z.; Liu, C. Oct-4 and Nanog promote the epithelial-mesenchymal transition of breast cancer stem cells and are associated with poor prognosis in breast cancer patients. Oncotarget 2014, 5, 10803–10815. [Google Scholar] [CrossRef] [Green Version]
  63. Ohnishi, K.; Semi, K.; Yamamoto, T.; Shimizu, M.; Tanaka, A.; Mitsunaga, K.; Okita, K.; Osafune, K.; Arioka, Y.; Maeda, T.; et al. Premature Termination of Reprogramming In Vivo Leads to Cancer Development through Altered Epigenetic Regulation. Cell 2014, 156, 663–677. [Google Scholar] [CrossRef] [Green Version]
  64. Hochedlinger, K.; Yamada, Y.; Beard, C.; Jaenisch, R. Ectopic expression of Oct-4 blocks progenitor-cell differentiation and causes dysplasia in epithelial tissues. Cell 2005, 121, 465–477. [Google Scholar] [CrossRef] [Green Version]
  65. Murakami, S.; Ninomiya, W.; Sakamoto, E.; Shibata, T.; Akiyama, H.; Tashiro, F. SRY and OCT4 Are Required for the Acquisition of Cancer Stem Cell-Like Properties and Are Potential Differentiation Therapy Targets. Stem Cells 2015, 33, 2652–2663. [Google Scholar] [CrossRef]
  66. Du, Z.; Jia, D.; Liu, S.; Wang, F.; Li, G.; Zhang, Y.; Cao, X.; Ling, E.A.; Hao, A. Oct4 is expressed in human gliomas and promotes colony formation in glioma cells. Glia 2009, 57, 724–733. [Google Scholar] [CrossRef]
  67. Wang, X.Q.; Ongkeko, W.M.; Chen, L.; Yang, Z.F.; Lu, P.; Chen, K.K.; Lopez, J.P.; Poon, R.T.P.; Fan, S.T. Octamer 4 (Oct4) Mediates Chemotherapeutic Drug Resistance in Liver Cancer Cells Through a Potential Oct4-AKT-ATP-Binding Cassette G2 Pathway. Hepatology 2010, 52, 528–539. [Google Scholar] [CrossRef]
  68. Fujino, S.; Miyoshi, N. Oct4 Gene Expression in Primary Colorectal Cancer Promotes Liver Metastasis. Stem Cells Int. 2019, 2019, 7896524. [Google Scholar] [CrossRef] [Green Version]
  69. Lu, C.S.; Shiau, A.L.; Su, B.H.; Hsu, T.S.; Wang, C.T.; Su, Y.C.; Tsai, M.S.; Feng, Y.H.; Tseng, Y.L.; Yen, Y.T.; et al. Oct4 promotes M2 macrophage polarization through upregulation of macrophage colony-stimulating factor in lung cancer. J. Hematol. Oncol. 2020, 13, 62. [Google Scholar] [CrossRef]
  70. Hagey, D.W.; Klum, S.; Kurtsdotter, I.; Zaouter, C.; Topcic, D.; Andersson, O.; Bergsland, M.; Muhr, J. SOX2 regulates common and specific stem cell features in the CNS and endoderm derived organs. PLoS Genet. 2018, 14, e1007224. [Google Scholar] [CrossRef] [Green Version]
  71. Schaefer, T.; Lengerke, C. SOX2 protein biochemistry in stemness, reprogramming, and cancer: The PI3K/AKT/SOX2 axis and beyond. Oncogene 2020, 39, 278–292. [Google Scholar] [CrossRef] [Green Version]
  72. Liu, P.; Tang, H.; Song, C.; Wang, J.; Chen, B.; Huang, X.; Pei, X.; Liu, L. SOX2 Promotes Cell Proliferation and Metastasis in Triple Negative Breast Cancer. Front. Pharm. 2018, 9, 942. [Google Scholar] [CrossRef] [Green Version]
  73. Maurizi, G.; Verma, N.; Gadi, A.; Mansukhani, A.; Basilico, C. Sox2 is required for tumor development and cancer cell proliferation in osteosarcoma. Oncogene 2018, 37, 4626–4632. [Google Scholar] [CrossRef] [PubMed]
  74. Han, S.; Huang, T.; Wu, X.; Wang, X.; Liu, S.; Yang, W.; Shi, Q.; Li, H.; Hou, F. Prognostic Value of CD133 and SOX2 in Advanced Cancer. J. Oncol. 2019, 2019, 3905817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Heurtier, V.; Owens, N.; Gonzalez, I.; Mueller, F.; Proux, C.; Mornico, D.; Clerc, P.; Dubois, A.; Navarro, P. The molecular logic of Nanog-induced self-renewal in mouse embryonic stem cells. Nat. Commun. 2019, 10, 1109. [Google Scholar] [CrossRef] [Green Version]
  76. Chiou, S.H.; Wang, M.L.; Chou, Y.T.; Chen, C.J.; Hong, C.F.; Hsieh, W.J.; Chang, H.T.; Chen, Y.S.; Lin, T.W.; Hsu, H.S.; et al. Coexpression of Oct4 and Nanog enhances malignancy in lung adenocarcinoma by inducing cancer stem cell-like properties and epithelial-mesenchymal transdifferentiation. Cancer Res. 2010, 70, 10433–10444. [Google Scholar] [CrossRef] [Green Version]
  77. Lin, T.; Ding, Y.Q.; Li, J.M. Overexpression of Nanog protein is associated with poor prognosis in gastric adenocarcinoma. Med. Oncol. 2012, 29, 878–885. [Google Scholar] [CrossRef]
  78. Chiou, S.H.; Yu, C.C.; Huang, C.Y.; Lin, S.C.; Liu, C.J.; Tsai, T.H.; Chou, S.H.; Chien, C.S.; Ku, H.H.; Lo, J.F. Positive correlations of Oct-4 and Nanog in oral cancer stem-like cells and high-grade oral squamous cell carcinoma. Clin. Cancer Res. 2008, 14, 4085–4095. [Google Scholar] [CrossRef] [Green Version]
  79. de Vicente, J.C.; Rodriguez-Santamarta, T.; Rodrigo, J.P.; Allonca, E.; Vallina, A.; Singhania, A.; Donate-Perez Del Molino, P.; Garcia-Pedrero, J.M. The Emerging Role of NANOG as an Early Cancer Risk Biomarker in Patients with Oral Potentially Malignant Disorders. J. Clin. Med. 2019, 8, 1376. [Google Scholar] [CrossRef] [Green Version]
  80. Wang, X.; Jin, J.; Wan, F.; Zhao, L.; Chu, H.; Chen, C.; Liao, G.; Liu, J.; Yu, Y.; Teng, H.; et al. AMPK Promotes SPOP-Mediated NANOG Degradation to Regulate Prostate Cancer Cell Stemness. Dev. Cell 2019, 48, 345–360.e7. [Google Scholar] [CrossRef] [Green Version]
  81. Dehghan Harati, M.; Rodemann, H.P.; Toulany, M. Nanog Signaling Mediates Radioresistance in ALDH-Positive Breast Cancer Cells. Int J Mol Sci 2019, 20, 1151. [Google Scholar] [CrossRef] [Green Version]
  82. Ghaleb, A.M.; Yang, V.W. Kruppel-like factor 4 (KLF4): What we currently know. Gene 2017, 611, 27–37. [Google Scholar] [CrossRef]
  83. Hsieh, M.H.; Chen, Y.T.; Chen, Y.T.; Lee, Y.H.; Lu, J.; Chien, C.L.; Chen, H.F.; Ho, H.N.; Yu, C.J.; Wang, Z.Q.; et al. PARP1 controls KLF4-mediated telomerase expression in stem cells and cancer cells. Nucleic Acids Res. 2017, 45, 10492–10503. [Google Scholar] [CrossRef] [PubMed]
  84. Riverso, M.; Montagnani, V.; Stecca, B. KLF4 is regulated by RAS/RAF/MEK/ERK signaling through E2F1 and promotes melanoma cell growth. Oncogene 2017, 36, 3322–3333. [Google Scholar] [CrossRef]
  85. Qi, X.T.; Li, Y.L.; Zhang, Y.Q.; Xu, T.; Lu, B.; Fang, L.; Gao, J.Q.; Yu, L.S.; Zhu, D.F.; Yang, B.; et al. KLF4 functions as an oncogene in promoting cancer stem cell-like characteristics in osteosarcoma cells. Acta Pharm. Sin. 2019, 40, 546–555. [Google Scholar] [CrossRef] [PubMed]
  86. Ding, X.; Zhong, T.; Jiang, L.; Huang, J.; Xia, Y.; Hu, R. miR-25 enhances cell migration and invasion in non-small-cell lung cancer cells via ERK signaling pathway by inhibiting KLF4. Mol. Med. Rep. 2018, 17, 7005–7016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Wang, B.; Shen, A.; Ouyang, X.; Zhao, G.; Du, Z.; Huo, W.; Zhang, T.; Wang, Y.; Yang, C.; Dong, P.; et al. KLF4 expression enhances the efficacy of chemotherapy drugs in ovarian cancer cells. Biochem. Biophys. Res. Commun. 2017, 484, 486–492. [Google Scholar] [CrossRef] [PubMed]
  88. Dang, C.V. MYC on the path to cancer. Cell 2012, 149, 22–35. [Google Scholar] [CrossRef] [Green Version]
  89. Beroukhim, R.; Mermel, C.H.; Porter, D.; Wei, G.; Raychaudhuri, S.; Donovan, J.; Barretina, J.; Boehm, J.S.; Dobson, J.; Urashima, M.; et al. The landscape of somatic copy-number alteration across human cancers. Nature 2010, 463, 899–905. [Google Scholar] [CrossRef]
  90. Yoshida, G.J. Emerging roles of Myc in stem cell biology and novel tumor therapies. J. Exp. Clin. Cancer Res. 2018, 37, 1–20. [Google Scholar]
  91. Galardi, S.; Savino, M.; Scagnoli, F.; Pellegatta, S.; Pisati, F.; Zambelli, F.; Illi, B.; Annibali, D.; Beji, S.; Orecchini, E.; et al. Resetting cancer stem cell regulatory nodes upon MYC inhibition. EMBO Rep. 2016, 17, 1872–1889. [Google Scholar] [CrossRef] [Green Version]
  92. Fabregat, I.; Malfettone, A.; Soukupova, J. New Insights into the Crossroads between EMT and Stemness in the Context of Cancer. J. Clin. Med. 2016, 5, 37. [Google Scholar] [CrossRef] [Green Version]
  93. Sever, R.; Brugge, J.S. Signal transduction in cancer. Cold Spring Harb. Perspect. Med. 2015, 5, a006098. [Google Scholar] [CrossRef] [Green Version]
  94. Harrington, B.S.; Annunziata, C.M. NF-kappaB Signaling in Ovarian Cancer. Cancers 2019, 11, 1182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Choi, H.S.; Kim, J.H.; Kim, S.L.; Lee, D.S. Disruption of the NF-kappaB/IL-8 Signaling Axis by Sulconazole Inhibits Human Breast Cancer Stem Cell Formation. Cells 2019, 8, 1007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Rinkenbaugh, A.L.; Baldwin, A.S. The NF-kappaB Pathway and Cancer Stem Cells. Cells 2016, 5, 16. [Google Scholar] [CrossRef] [PubMed]
  97. Katoh, M. Networking of WNT, FGF, Notch, BMP, and Hedgehog signaling pathways during carcinogenesis. Stem Cell Rev. 2007, 3, 30–38. [Google Scholar] [CrossRef] [PubMed]
  98. Clements, W.M.; Wang, J.; Sarnaik, A.; Kim, O.J.; MacDonald, J.; Fenoglio-Preiser, C.; Groden, J.; Lowy, A.M. beta-Catenin mutation is a frequent cause of Wnt pathway activation in gastric cancer. Cancer Res 2002, 62, 3503–3506. [Google Scholar]
  99. Abd El-Rehim, D.; Ali, M.M. Aberrant expression of beta-catenin in invasive ductal breast carcinomas. J. Egypt Natl. Canc. Inst. 2009, 21, 185–195. [Google Scholar]
  100. Kudo, J.; Nishiwaki, T.; Haruki, N.; Ishiguro, H.; Shibata, Y.; Terashita, Y.; Sugiura, H.; Shinoda, N.; Kimura, M.; Kuwabara, Y.; et al. Aberrant nuclear localization of beta-catenin without genetic alterations in beta-catenin or Axin genes in esophageal cancer. World J. Surg. Oncol. 2007, 5, 21. [Google Scholar] [CrossRef] [Green Version]
  101. Zhao, X.; Jiang, C.; Xu, R.; Liu, Q.; Liu, G.; Zhang, Y. TRIP6 enhances stemness property of breast cancer cells through activation of Wnt/beta-catenin. Cancer Cell Int. 2020, 20, 51. [Google Scholar] [CrossRef] [Green Version]
  102. Zhu, L.; Pan, R.; Zhou, D.; Ye, G.; Tan, W. BCL11A enhances stemness and promotes progression by activating Wnt/beta-catenin signaling in breast cancer. Cancer Manag. Res. 2019, 11, 2997–3007. [Google Scholar] [CrossRef] [Green Version]
  103. Liu, S.; Pong, U.K.; Zhang, J.T.; Tsang, L.L.; Huang, J.W.; Tu, S.P.; Jiang, X.H. R-spodin2 enhances canonical Wnt signaling to maintain the stemness of glioblastoma cells. Cancer Cell Int. 2018, 18, 156. [Google Scholar] [CrossRef] [PubMed]
  104. Stylianou, S.; Clarke, R.B.; Brennan, K. Aberrant activation of notch signaling in human breast cancer. Cancer Res. 2006, 66, 1517–1525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Abel, E.V.; Kim, E.J.; Wu, J.J.; Hynes, M.; Bednar, F.; Proctor, E.; Wang, L.D.; Dziubinski, M.L.; Simeone, D.M. The Notch Pathway Is Important in Maintaining the Cancer Stem Cell Population in Pancreatic Cancer. PLoS ONE 2014, 9, e91983. [Google Scholar] [CrossRef] [PubMed]
  106. Koury, J.; Zhong, L.; Hao, J.J. Targeting Signaling Pathways in Cancer Stem Cells for Cancer Treatment. Stem Cells Int. 2017, 2017, 2925869 . [Google Scholar] [CrossRef]
  107. Yan, Y.Y.; Liu, F.X.; Han, L.; Zhao, L.; Chen, J.J.; Olopade, O.I.; He, M.; Wei, M.J. HIF-2 alpha promotes conversion to a stem cell phenotype and induces chemoresistance in breast cancer cells by activating Wnt and Notch pathways. J. Exp. Clin. Cancer Res. 2018, 37, 256. [Google Scholar] [CrossRef] [Green Version]
  108. Kang, L.; Mao, J.; Tao, Y.; Song, B.; Ma, W.; Lu, Y.; Zhao, L.; Li, J.; Yang, B.; Li, L. MicroRNA-34a suppresses the breast cancer stem cell-like characteristics by downregulating Notch1 pathway. Cancer Sci. 2015, 106, 700–708. [Google Scholar] [CrossRef]
  109. Zeng, F.; Chen, H.; Zhang, Z.H.; Yao, T.; Wang, G.; Zeng, Q.X.; Duan, S.H.; Zhan, Y.Q. Regulating glioma stem cells by hypoxia through the Notch1 and Oct3/4 signaling pathway. Oncol. Lett. 2018, 16, 6315–6322. [Google Scholar] [CrossRef]
  110. Petrova, R.; Joyner, A.L. Roles for Hedgehog signaling in adult organ homeostasis and repair. Development 2014, 141, 3445–3457. [Google Scholar] [CrossRef] [Green Version]
  111. Amantini, C.; Morelli, M.B.; Nabissi, M.; Cardinali, C.; Santoni, M.; Gismondi, A.; Santoni, G. Capsaicin triggers autophagic cell survival which drives epithelial mesenchymal transition and chemoresistance in bladder cancer cells in an Hedgehog-dependent manner. Oncotarget 2016, 7, 50180–50194. [Google Scholar] [CrossRef] [Green Version]
  112. Villegas, V.E.; Rondon-Lagos, M.; Annaratone, L.; Castellano, I.; Grismaldo, A.; Sapino, A.; Zaphiropoulos, P.G. Tamoxifen Treatment of Breast Cancer Cells: Impact on Hedgehog/GLI1 Signaling. Int. J. Mol. Sci. 2016, 17, 308. [Google Scholar] [CrossRef] [Green Version]
  113. Po, A.; Silvano, M.; Miele, E.; Capalbo, C.; Eramo, A.; Salvati, V.; Todaro, M.; Besharat, Z.M.; Catanzaro, G.; Cucchi, D.; et al. Noncanonical GLI1 signaling promotes stemness features and in vivo growth in lung adenocarcinoma. Oncogene 2017, 36, 4641–4652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Borah, A.; Raveendran, S.; Rochani, A.; Maekawa, T.; Kumar, D.S. Targeting self-renewal pathways in cancer stem cells: Clinical implications for cancer therapy. Oncogenesis 2015, 4, e177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Zhu, R.; Gires, O.; Zhu, L.; Liu, J.; Li, J.; Yang, H.; Ju, G.; Huang, J.; Ge, W.; Chen, Y.; et al. TSPAN8 promotes cancer cell stemness via activation of sonic Hedgehog signaling. Nat. Commun. 2019, 10, 2863. [Google Scholar] [CrossRef] [PubMed]
  116. Clement, V.; Sanchez, P.; de Tribolet, N.; Radovanovic, I.; Altaba, A.R.I. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr. Biol. 2007, 17, 165–172. [Google Scholar] [CrossRef] [PubMed]
  117. Nath, S.; Mondal, S.; Butti, R.; Prasanna Gunasekaran, V.; Chatterjee, U.; Halder, A.; Kundu, G.C.; Mandal, C. Desialylation of Sonic-Hedgehog by Neu2 Inhibits Its Association with Patched1 Reducing Stemness-Like Properties in Pancreatic Cancer Sphere-forming Cells. Cells 2020, 9, 1512. [Google Scholar] [CrossRef] [PubMed]
  118. Chambers, I. The molecular basis of pluripotency in mouse embryonic stem cells. Cloning Stem Cells 2004, 6, 386–391. [Google Scholar] [CrossRef]
  119. Zhou, J.; Wulfkuhle, J.; Zhang, H.; Gu, P.; Yang, Y.; Deng, J.; Margolick, J.B.; Liotta, L.A.; Petricoin, E., 3rd; Zhang, Y. Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc. Natl. Acad. Sci. USA 2007, 104, 16158–16163. [Google Scholar] [CrossRef] [Green Version]
  120. Kaowinn, S.; Kaewpiboon, C.; Koh, S.S.; Kramer, O.H.; Chung, Y.H. STAT1HDAC4 signaling induces epithelialmesenchymal transition and sphere formation of cancer cells overexpressing the oncogene, CUG2. Oncol. Rep. 2018, 40, 2619–2627. [Google Scholar] [CrossRef]
  121. Dey, N.; De, P.; Leyland-Jones, B. PI3K-AKT-mTOR inhibitors in breast cancers: From tumor cell signaling to clinical trials. Pharm. 2017, 175, 91–106. [Google Scholar] [CrossRef]
  122. Tan, A.C. Targeting the PI3K/Akt/mTOR pathway in non-small cell lung cancer (NSCLC). Thorac. Cancer 2020, 11, 511–518. [Google Scholar] [CrossRef] [Green Version]
  123. Fitzgerald, T.L.; Lertpiriyapong, K.; Cocco, L.; Martelli, A.M.; Libra, M.; Candido, S.; Montalto, G.; Cervello, M.; Steelman, L.; Abrams, S.L.; et al. Roles of EGFR and KRAS and their downstream signaling pathways in pancreatic cancer and pancreatic cancer stem cells. Adv. Biol Regul 2015, 59, 65–81. [Google Scholar] [CrossRef] [PubMed]
  124. Madsen, R.R. PI3K in stemness regulation: From development to cancer. Biochem. Soc. Trans. 2020, 48, 301–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Ouspenskaia, T.; Matos, I.; Mertz, A.F.; Fiore, V.F.; Fuchs, E. WNT-SHH Antagonism Specifies and Expands Stem Cells prior to Niche Formation. Cell 2016, 164, 156–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Wahlster, L.; Daley, G.Q. Progress towards generation of human haematopoietic stem cells. Nat. Cell Biol. 2016, 18, 1111–1117. [Google Scholar] [CrossRef]
  127. Kitaeva, K.V.; Rutland, C.S.; Rizvanov, A.A.; Solovyeva, V.V. Cell Culture Based in vitro Test Systems for Anticancer Drug Screening. Front. Bioeng. Biotechnol. 2020, 8, 322. [Google Scholar] [CrossRef] [Green Version]
  128. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [Green Version]
  129. Nieto, M.A.; Huang, R.Y.J.; Jackson, R.A.; Thiery, J.P. EMT: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef] [Green Version]
  130. Lambert, A.W.; Weinberg, R.A. Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat. Rev. Cancer 2021, 21, 325–338. [Google Scholar] [CrossRef]
  131. Liu, Y.; Nenutil, R.; Appleyard, M.V.; Murray, K.; Boylan, M.; Thompson, A.M.; Coates, P.J. Lack of correlation of stem cell markers in breast cancer stem cells. Br. J. Cancer 2014, 110, 2063–2071. [Google Scholar] [CrossRef] [Green Version]
  132. Karin, M.; Clevers, H. Reparative inflammation takes charge of tissue regeneration. Nature 2016, 529, 307–315. [Google Scholar] [CrossRef]
  133. Zhang, M.; Xu, C.; Wang, H.Z.; Peng, Y.N.; Li, H.O.; Zhou, Y.J.; Liu, S.; Wang, F.; Liu, L.; Chang, Y.; et al. Soft fibrin matrix downregulates DAB2IP to promote Nanog-dependent growth of colon tumor-repopulating cells. Cell Death Dis 2019, 10, 151. [Google Scholar] [CrossRef] [Green Version]
  134. Li, Y.; Wang, L.; Pappan, L.; Galliher-Beckley, A.; Shi, J. IL-1beta promotes stemness and invasiveness of colon cancer cells through Zeb1 activation. Mol. Cancer 2012, 11, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Hong, H.S.; Akhavan, J.; Lee, S.H.; Kim, R.H.; Kang, M.K.; Park, N.H.; Shin, K.H. Proinflammatory cytokine TNFalpha promotes HPV-associated oral carcinogenesis by increasing cancer stemness. Int. J. Oral Sci. 2020, 12, 3. [Google Scholar] [CrossRef] [Green Version]
  136. Ciardiello, C.; Leone, A.; Budillon, A. The Crosstalk between Cancer Stem Cells and Microenvironment Is Critical for Solid Tumor Progression: The Significant Contribution of Extracellular Vesicles. Stem Cells Int. 2018, 2018, 6392198. [Google Scholar] [CrossRef] [PubMed]
  137. Tam, S.Y.; Wu, V.W.C.; Law, H.K.W. Hypoxia-Induced Epithelial-Mesenchymal Transition in Cancers: HIF-1 alpha and Beyond. Front. Oncol. 2020, 10, 486. [Google Scholar] [CrossRef] [PubMed]
  138. Ayob, A.Z.; Ramasamy, T.S. Cancer stem cells as key drivers of tumour progression. J. Biomed. Sci. 2018, 25, 20. [Google Scholar] [CrossRef]
  139. Brosicke, N.; Faissner, A. Role of tenascins in the ECM of gliomas. Cell Adhes. Migr. 2015, 9, 131–140. [Google Scholar] [CrossRef] [Green Version]
  140. Winer, J.P.; Janmey, P.A.; McCormick, M.E.; Funaki, M. Bone marrow-derived human mesenchymal stem cells become quiescent on soft substrates but remain responsive to chemical or mechanical stimuli. Tissue Eng. Part. A 2009, 15, 147–154. [Google Scholar] [CrossRef]
  141. You, Y.; Zheng, Q.; Dong, Y.; Xie, X.; Wang, Y.; Wu, S.; Zhang, L.; Wang, Y.; Xue, T.; Wang, Z.; et al. Matrix stiffness-mediated effects on stemness characteristics occurring in HCC cells. Oncotarget 2016, 7, 32221–32231. [Google Scholar] [CrossRef] [Green Version]
  142. Levental, K.R.; Yu, H.; Kass, L.; Lakins, J.N.; Egeblad, M.; Erler, J.T.; Fong, S.F.; Csiszar, K.; Giaccia, A.; Weninger, W.; et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 2009, 139, 891–906. [Google Scholar] [CrossRef] [Green Version]
  143. Chulpanova, D.S.; Kitaeva, K.V.; James, V.; Rizvanov, A.A.; Solovyeva, V.V. Therapeutic Prospects of extracellular vesicles in Cancer Treatment. Front. Immunol. 2018, 9, 1534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Shaimardanova, A.A.; Solovyeva, V.V.; Chulpanova, D.S.; James, V.; Kitaeva, K.V.; Rizvanov, A.A. Extracellular vesicles in the diagnosis and treatment of central nervous system diseases. Neural Regen. Res. 2020, 15, 586–596. [Google Scholar] [CrossRef] [PubMed]
  145. Bronisz, A.; Wang, Y.; Nowicki, M.O.; Peruzzi, P.; Ansari, K.I.; Ogawa, D.; Balaj, L.; De Rienzo, G.; Mineo, M.; Nakano, I.; et al. Extracellular Vesicles Modulate the Glioblastoma Microenvironment via a Tumor Suppression Signaling Network Directed by miR-1. Cancer Res. 2014, 74, 738–750. [Google Scholar] [CrossRef] [Green Version]
  146. Hu, Y.B.; Yan, C.; Mu, L.; Huang, K.Y.; Li, X.L.; Tao, D.D.; Wu, Y.Q.; Qin, J.C. Fibroblast-Derived Exosomes Contribute to Chemoresistance through Priming Cancer Stem Cells in Colorectal Cancer. PLoS ONE 2015, 10, e0125625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Gonzalez, E.; Piva, M.; Rodriguez-Suarez, E.; Gil, D.; Royo, F.; Elortza, F.; Falcon-Perez, J.M.; Vivanco, M. Human mammospheres secrete hormone-regulated active extracellular vesicles. PLoS ONE 2014, 9, e83955. [Google Scholar] [CrossRef]
  148. Gilazieva, Z.; Ponomarev, A.; Rutland, C.; Rizvanov, A.; Solovyeva, V. Promising Applications of Tumor Spheroids and Organoids for Personalized Medicine. Cancers 2020, 12, 2727. [Google Scholar] [CrossRef]
  149. Jensen, C.; Teng, Y. Is It Time to Start Transitioning From 2D to 3D Cell Culture? Front. Mol. Biosci. 2020, 7, 33. [Google Scholar] [CrossRef] [Green Version]
  150. Ishiguro, T.; Ohata, H.; Sato, A.; Yamawaki, K.; Enomoto, T.; Okamoto, K. Tumor-derived spheroids: Relevance to cancer stem cells and clinical applications. Cancer Sci. 2017, 108, 283–289. [Google Scholar] [CrossRef] [Green Version]
  151. Djordjevic, B.; Lange, C.S. Clonogenicity of mammalian cells in hybrid spheroids: A new assay method. Radiat. Env. Biophys. 1990, 29, 31–46. [Google Scholar] [CrossRef]
  152. Mikhail, A.S.; Eetezadi, S.; Allen, C. Multicellular tumor spheroids for evaluation of cytotoxicity and tumor growth inhibitory effects of nanomedicines in vitro: A comparison of docetaxel-loaded block copolymer micelles and Taxotere(R). PLoS ONE 2013, 8, e62630. [Google Scholar] [CrossRef]
  153. Zhang, C.Y.; Yang, Z.T.; Dong, D.L.; Jang, T.S.; Knowles, J.C.; Kim, H.W.; Jin, G.Z.; Xuan, Y.H. 3D culture technologies of cancer stem cells: Promising ex vivo tumor models. J. Tissue Eng. 2020, 11. [Google Scholar] [CrossRef] [PubMed]
  154. Drost, J.; Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 2018, 18, 407–418. [Google Scholar] [CrossRef] [PubMed]
  155. Papapetrou, E.P. Patient-derived induced pluripotent stem cells in cancer research and precision oncology. Nat. Med. 2016, 22, 1392–1401. [Google Scholar] [CrossRef] [PubMed]
  156. Morrison, S.J.; Spradling, A.C. Stem cells and niches: Mechanisms that promote stem cell maintenance throughout life. Cell 2008, 132, 598–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Hirschhaeuser, F.; Menne, H.; Dittfeld, C.; West, J.; Mueller-Klieser, W.; Kunz-Schughart, L.A. Multicellular tumor spheroids: An underestimated tool is catching up again. J. Biotechnol. 2010, 148, 3–15. [Google Scholar] [CrossRef]
  158. Kahlert, U.D.; Mooney, S.M.; Natsumeda, M.; Steiger, H.J.; Maciaczyk, J. Targeting cancer stem-like cells in glioblastoma and colorectal cancer through metabolic pathways. Int. J. Cancer 2017, 140, 10–22. [Google Scholar] [CrossRef]
  159. Puls, T.J.; Tan, X.H.; Whittington, C.F.; Voytik-Harbin, S.L. 3D collagen fibrillar microstructure guides pancreatic cancer cell phenotype and serves as a critical design parameter for phenotypic models of EMT. PLoS ONE 2017, 12, e0188870. [Google Scholar] [CrossRef] [Green Version]
  160. Pal, A.; Ashworth, J.C.; Collier, P.; Probert, C.; Jones, S.; Leza, E.P.; Meakin, M.L.; Ritchie, A.A.; Onion, D.; Clarke, P.A.; et al. A 3D Heterotypic Breast Cancer Model Demonstrates a Role for Mesenchymal Stem Cells in Driving a Proliferative and Invasive Phenotype. Cancers 2020, 12, 2290. [Google Scholar] [CrossRef]
  161. Xue, J.G.; Zhu, Y.; Sun, Z.X.; Ji, R.B.; Zhang, X.; Xu, W.R.; Yuan, X.; Zhang, B.; Yan, Y.M.; Yin, L.; et al. Tumorigenic hybrids between mesenchymal stem cells and gastric cancer cells enhanced cancer proliferation, migration and stemness. BMC Cancer 2015, 15, 793. [Google Scholar] [CrossRef] [Green Version]
  162. Nath, S.; Devi, G.R. Three-dimensional culture systems in cancer research: Focus on tumor spheroid model. Pharmacol. Ther. 2016, 163, 94–108. [Google Scholar] [CrossRef] [Green Version]
  163. Maliszewska-Olejniczak, K.; Brodaczewska, K.K.; Bielecka, Z.F.; Solarek, W.; Kornakiewicz, A.; Szczylik, C.; Porta, C.; Czarnecka, A.M. Development of extracellular matrix supported 3D culture of renal cancer cells and renal cancer stem cells. Cytotechnology 2019, 71, 149–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Gupta, P.B.; Onder, T.T.; Jiang, G.Z.; Tao, K.; Kuperwasser, C.; Weinberg, R.A.; Lander, E.S. Identification of Selective Inhibitors of Cancer Stem Cells by High-Throughput Screening. Cell 2009, 138, 645–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Namiki, K.; Wongsirisin, P.; Yokoyama, S.; Sato, M.; Rawangkan, A.; Sakai, R.; Iida, K.; Suganuma, M. (-)-Epigallocatechin gallate inhibits stemness and tumourigenicity stimulated by AXL receptor tyrosine kinase in human lung cancer cells. Sci Rep. 2020, 10, 2444. [Google Scholar] [CrossRef] [PubMed]
  166. Prasad, S.; Rarnachandran, S.; Gupta, N.; Kaushik, I.; Srivastava, S.K. Cancer cells stemness: A doorstep to targeted therapy. Biochim. Biophys. Acta-Mol. Basis Dis. 2020, 1866. [Google Scholar] [CrossRef]
  167. Konishi, J.; Kawaguchi, K.S.; Vo, H.; Haruki, N.; Gonzalez, A.; Carbone, D.P.; Dang, T.P. gamma-secretase inhibitor prevents Notch3 activation and reduces proliferation in human lung cancers. Cancer Res. 2007, 67, 8051–8057. [Google Scholar] [CrossRef] [Green Version]
  168. Moore, G.; Annett, S.; McClements, L.; Robson, T. Top Notch Targeting Strategies in Cancer: A Detailed Overview of Recent Insights and Current Perspectives. Cells 2020, 9, 1503. [Google Scholar] [CrossRef]
  169. Cook, N.; Basu, B.; Smith, D.M.; Gopinathan, A.; Evans, J.; Steward, W.P.; Palmer, D.; Propper, D.; Venugopal, B.; Hategan, M.; et al. A phase I trial of the gamma-secretase inhibitor MK-0752 in combination with gemcitabine in patients with pancreatic ductal adenocarcinoma. Br. J. Cancer 2018, 118, 793–801. [Google Scholar] [CrossRef]
  170. Herman, S.E.M.; Gordon, A.L.; Wagner, A.J.; Heerema, N.A.; Zhao, W.Q.; Flynn, J.M.; Jones, J.; Andritsos, L.; Puri, K.D.; Lannutti, B.J.; et al. Phosphatidylinositol 3-kinase-delta inhibitor CAL-101 shows promising preclinical activity in chronic lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular survival signals. Blood 2010, 116, 2078–2088. [Google Scholar] [CrossRef] [Green Version]
  171. Foster, P.; Yamaguchi, K.; Hsu, P.P.; Qian, F.; Du, X.N.; Wu, J.M.; Won, K.A.; Yu, P.W.; Jaeger, C.T.; Zhang, W.T.; et al. The Selective PI3K Inhibitor XL147 (SAR245408) Inhibits Tumor Growth and Survival and Potentiates the Activity of Chemotherapeutic Agents in Preclinical Tumor Models. Mol. Cancer Ther. 2015, 14, 931–940. [Google Scholar] [CrossRef] [Green Version]
  172. Eyler, C.E.; Foo, W.C.; Lafiura, K.M.; McLendon, R.E.; Hjelmeland, A.B.; Rich, J.N. Brain Cancer Stem Cells Display Preferential Sensitivity to Akt Inhibition. Stem Cells 2008, 26, 3027–3036. [Google Scholar] [CrossRef] [Green Version]
  173. Pal, S.K.; Reckamp, K.; Yu, H.; Figlin, R.A. Akt inhibitors in clinical development for the treatment of cancer. Expert Opin. Investig. Drugs 2010, 19, 1355–1366. [Google Scholar] [CrossRef] [Green Version]
  174. Tagawa, S.T.; Chatta, G.S.; Mazhari, R.; Benaim, E. Archexin, a novel AKT-1-specific inhibitor for the treatment of metastatic renal cancer: Preliminary phase I data. J. Clin. Oncol. 2016, 34. [Google Scholar] [CrossRef]
  175. Lindemann, R.K. Stroma-initiated hedgehog signaling takes center stage in B-cell lymphoma. Cancer Res. 2008, 68, 961–964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Jimeno, A.; Gordon, M.; Chugh, R.; Messersmith, W.; Mendelson, D.; Dupont, J.; Stagg, R.; Kapoun, A.M.; Xu, L.; Uttamsingh, S.; et al. A First-in-Human Phase I Study of the Anticancer Stem Cell Agent Ipafricept (OMP-54F28), a Decoy Receptor for Wnt Ligands, in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2017, 23, 7490–7497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Ko, A.H.; Chiorean, E.G.; Kwak, E.L.; Lenz, H.J.; Nadler, P.I.; Wood, D.L.; Fujimori, M.; Inada, T.; Kouji, H.; McWilliams, R.R. Final results of a phase Ib dose-escalation study of PRI-724, a CBP/beta-catenin modulator, plus gemcitabine (GEM) in patients with advanced pancreatic adenocarcinoma (APC) as second-line therapy after FOLFIRINOX or FOLFOX. J. Clin. Oncol. 2016, 34. [Google Scholar] [CrossRef]
  178. Cortes, J.E.; Faderl, S.; Pagel, J.; Jung, C.W.; Yoon, S.S.; Koh, Y.; Pardanani, A.D.; Hauptschein, R.S.; Lee, K.J.; Lee, J.H. Phase 1 study of CWP232291 in relapsed/refractory acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). J. Clin. Oncol. 2015, 33. [Google Scholar] [CrossRef]
  179. Jonker, D.J.; Stephenson, J.; Edenfield, W.J.; Supko, J.G.; Li, Y.Z.; Li, W.; Hitron, M.; Leggett, D.; Kerstein, D.; Li, C. A phase I extension study of BBI608, a first-in-class cancer stein cell (CSC) inhibitor, in patients with advanced solid tumors. J. Clin. Oncol. 2014, 32. [Google Scholar] [CrossRef]
  180. Laurie, S.A.; Jonker, D.J.; Edenfield, W.J.; Stephenson, J.; Keller, D.; Hitron, M.; Li, W.; Li, Y.Z.; Gada, K.; Gao, Y.; et al. A phase 1 dose-escalation study of BBI503, a first-in-class cancer stemness kinase inhibitor in adult patients with advanced solid tumors. J. Clin. Oncol. 2014, 32. [Google Scholar] [CrossRef]
Figure 1. CSCs and the modulation of cancer stemness by the tumor microenvironment, stemness factors, and signaling pathways. Impact of CSCs on tumor progression, recurrence, and resistance to therapy. Created with BioRender.com (accessed on 22 December 2021).
Figure 1. CSCs and the modulation of cancer stemness by the tumor microenvironment, stemness factors, and signaling pathways. Impact of CSCs on tumor progression, recurrence, and resistance to therapy. Created with BioRender.com (accessed on 22 December 2021).
Cancers 14 00970 g001
Table 1. CSCs markers in different types of tumors.
Table 1. CSCs markers in different types of tumors.
Cancer TypeCSCs MarkersReference
Blood tumorsCD34+ CD38 phenotype[19]
Brain tumorsCD133+, CD49f+, CD90+, epidermal growth factor receptor (EGFR)+, c-series ganglioside-specific antigen A2B5+, L1 cell adhesion molecule (L1CAM)+[20,21,22]
Ovary tumorsCD24+, aldehyde dehydrogenase (ALDH)+, CD44+/CD117+, epithelial cell adhesion molecule (EpCAM)+, CD133+[23,24]
Prostate tumorsEpCAM+, CD117+, α2β1 integrin+, ALDH+, CD44+, enhancer of zeste homolog (EZH)+, CXC chemokine receptor type 4 (CXCR4)+, E-cadherin+, CD133+[13,25,26]
Colon tumorsCD133+, CD44+, CD166+, CD24+, EpCAM+),[27,28,29,30]
Pancreatic tumorsCD133+, CD44+, CD24+, EpCAM+, tyrosine-protein kinase Met (cMet)+[31,32]
Liver tumorsCD44+, CD90+, CD206+, oval cell antigen 6 (OV-6)+), skin (CD20+, CD271+, ALDH+, CD133+[33]
Lung tumorsCD133+, ATP-binding cassette super-family G member 2 (ABCG2)high, CD166+, CD90+, CD87+, ALDH+, CD44+[34]
Breast tumorsALDH1+, CD24+, CD44+, CD90+, CD133+, α6-integrin+[35,36,37,38]
Table 2. Therapeutic agents targeting cancer stem cells.
Table 2. Therapeutic agents targeting cancer stem cells.
AgentNotesClinical TrialsReference
MRK-003,
MK-0752, R4733
Notch inhibitorsNCT00106145
NCT01154452
[167,168,169]
CAL-101,
XL-147
PI3K inhibitorsNCT01629615
NCT01613950
NCT00907205
[170,171]
KRX-0401,
RX-0201
AKT inhibitorsNCT00590954
NCT01028495
[172,173,174]
BMS-863923, IPI-926Hedgehog inhibitorsNCT01546038
NCT01130142
NCT01700049
[175]
OMP-54F28, PRI-724, CWP232291Wnt inhibitorsNCT01606579
NCT01351103
NCT02092363
[176,177,178]
BBI503,
BBI608
NANOG inhibitorsNCT02232633
NCT02315534
[179,180]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ponomarev, A.; Gilazieva, Z.; Solovyeva, V.; Allegrucci, C.; Rizvanov, A. Intrinsic and Extrinsic Factors Impacting Cancer Stemness and Tumor Progression. Cancers 2022, 14, 970. https://doi.org/10.3390/cancers14040970

AMA Style

Ponomarev A, Gilazieva Z, Solovyeva V, Allegrucci C, Rizvanov A. Intrinsic and Extrinsic Factors Impacting Cancer Stemness and Tumor Progression. Cancers. 2022; 14(4):970. https://doi.org/10.3390/cancers14040970

Chicago/Turabian Style

Ponomarev, Alexey, Zarema Gilazieva, Valeriya Solovyeva, Cinzia Allegrucci, and Albert Rizvanov. 2022. "Intrinsic and Extrinsic Factors Impacting Cancer Stemness and Tumor Progression" Cancers 14, no. 4: 970. https://doi.org/10.3390/cancers14040970

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop