You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Submitted Successfully!
Thank you for your contribution! You can also upload a video entry or images related to this topic. For video creation, please contact our Academic Video Service.
Version Summary Created by Modification Content Size Created at Operation
1 + 3958 word(s) 3958 2022-03-01 09:27:51 |
2 format correct Meta information modification 3958 2022-03-03 02:43:35 |

Video Upload Options

We provide professional Academic Video Service to translate complex research into visually appealing presentations. Would you like to try it?

Confirm

Are you sure to Delete?
Yes No
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Wilczyński, J. Cancer Stem Cells in Ovarian Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/20110 (accessed on 20 May 2025).
Wilczyński J. Cancer Stem Cells in Ovarian Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/20110. Accessed May 20, 2025.
Wilczyński, Jacek. "Cancer Stem Cells in Ovarian Cancer" Encyclopedia, https://encyclopedia.pub/entry/20110 (accessed May 20, 2025).
Wilczyński, J. (2022, March 02). Cancer Stem Cells in Ovarian Cancer. In Encyclopedia. https://encyclopedia.pub/entry/20110
Wilczyński, Jacek. "Cancer Stem Cells in Ovarian Cancer." Encyclopedia. Web. 02 March, 2022.
Cancer Stem Cells in Ovarian Cancer
Edit

Ovarian cancer is the most lethal neoplasm of the female genital organs. Despite indisputable progress in the treatment of ovarian cancer, the problems of chemo-resistance and recurrent disease are the main obstacles for successful therapy. One of the main reasons for this is the presence of a specific cell population of cancer stem cells.

cancer stem cells ovarian cancer stem cells

1. Introduction

Ovarian cancer (OC) is the most lethal tumor of the female genital tract due to aggressive behavior, late diagnosis and high recurrence potential. Most of the patients worldwide are admitted with advanced disease as the initial steps of cancer growth are usually clinically obscured. This is a reason why the 5-year survival in the whole patient population does not exceed 48% (data of American Cancer Society 2020. https://www.cancer.org/cancer/ovarian-cancer/detection-diagnosis-staging/survival-rates.html, accessed on 20 December 2021). Moreover, ovarian cancer shows chemoresistance to standard platinum-based chemotherapy especially in advanced and recurrent cases, the fact which further influences poor survival. Ovarian cancer disease includes a heterogenous group of neoplasia: among them, about 90% are epithelial (subtypes: mucinous, serous, endometrioid and clear cells), as suggested by several and recent morphological and ultrastructural studies [1]. Ovarian cancer is a heterogeneous disease which comprise malignant tumors of serous, mucinous, endometrial or clear cell histology. According to the differences of biological behavior and malignancy, OC has been divided into two types: type I tumors containing low-grade (LGOC) serous, mucinous and endometroid ovarian cancer with better prognosis and lower rate mortality, and type II highly malignant and rapidly progressing high-grade serous ovarian cancer (HGOC) with poor prognosis and mortality about 90% of all OC cases [2][3]. Genetic expression profiling studies support this clinical classification, as type I tumors are associated with relative genetic stability and mutations of PIK3CAPTENBRAFKRAS and ARID1A genes, while type II tumors possess high chromosomal instability, defective homologous recombination repair and are characterized mostly by TP53 mutations, but also by BRCA1BRCA2RB1 and CTNNB1 gene mutations [4][5][6][7][8][9]. Progenitor cells for type I OC are endometrial epithelial cells (for endometroid and clear cell tumors), tubal-peritoneal junction cells (for mucinous tumor) or fallopian epithelial cells and cortical inclusion cyst (CIC) epithelial cells (for LGOC), whereas for type II OC the progenitor cell originate from serous tubal intraepithelial cancer lesions (STIC) localized on tubal fimbriae. Early type I tumors frequently exist as so-called borderline tumors which do not show histologic signs of stromal invasion [1][2]. Recent gene profiling studies allowed for a proposal of a new classification based on both gene expression pattern and histological structure. According to this classification ovarian cancer could be divided into five subtypes: mesenchymal, immunoreactive, proliferative, differentiated and anti-mesenchymal. Mesenchymal and proliferative tumors comprise for 28% and 20% of OC, respectively. Mesenchymal subtype show desmoplasia and mesenchymal gene expression pattern, proliferative subtype show limited inflammatory infiltration and activation of signaling pathways for stemness. Both subtypes have an unfavorable prognosis. Otherwise, immunoreactive and anti-mesenchymal subtypes which comprise 21% and 12% of OC, have a better prognosis. The immunoreactive subtype is characterized by extensive T cell tumor infiltration and T-cell receptor and toll-like receptor signaling, while the anti-mesenchymal subtype shows a genotype which is opposite to the mesenchymal type. Differentiated subtype observed in 17% of OC tumors has gene pattern resembling serous borderline tumors and intermediate prognosis [10][11][12]. Extensive surgical debulking followed by platinum and taxane-based chemotherapy is a standard of care for invasive OC patients, however, extensive spread of tumor implants inside the peritoneal cavity, as well as a primary chemo-refractoriness or acquired chemoresistance of the tumor are responsible for unfavorable outcome. Recent studies suggest that a unique population of tumor cells called cancer stem cells (CSCs) are the most probable reason for cancer progression and therapy failure in OC.

2. Ovarian Cancer Stem Cells (OCSCs)—Markers

2.1. Cell Surface Markers

CSCs surface markers are not specific as they are also expressed on normal stem cells. Therefore, CSCs should also be identified by precisely defined behavior, such as spheroid formation or the reconstitution of tumors after transplantation to laboratory animals. Numerous markers have been suggested to identify CSCs, including OCSCs; however, their precise clinical significance is still unknown. Despite this, several surface cell markers identifying OCSCs isolated either from patient samples or experimental animals and cancer cell lines have been described (Table 1).
Table 1. Markers of OCSCs—function, correlation to clinicopathological features and their cell/tissue origin.
Marker Function Origin of Studied Cells Reference Association to Clinicopathological Features Cell/Tissue Origin Reference
CD44+ Increased tumorigenicity, sphere-formation, cells self-renewal Primary EOC tumors, cell cultures [13][14][15][16] Number of CD44+ cells higher in early stage EOC and correlated with shorter PFS
Expression correlated with high-grade, advanced (III/IV FIGO) EOC in younger (<60) patients
Higher number of CD44+ cells correlated with chemoresistance and shorter DFI
CD44+ correlated with Ki67 index, p53 positivity and tumor grade in HGSOC, mucinous and endometroid EOC
EOC-isolated cells
Recurrent EOC (88% HGSOC)
Primary and recurrent EOC (78% HGSOC)
EOC (HGSOC 62%) and BOT
[17][18][19][20]
CD44 v6+ Increased tumorigenicity, recapitulation of tumors Xenotransplantation model [21] Distant metastases more frequent and metastasis free survival shorter in CD44v6+—high group of patients
Increased number of CD44v6+ cells in primary tumors correlated with shorter OS
EOC FIGO I–III tumors
EOC FIGO III–IV tumors (71% HGSOC)
[22][21]
CD44+/MyD88+ Increased tumorigenicity, sphere-formation, resistance to apoptosis, chemoresistance Cell lines, ascites [23] Expression of MyD88 protein was an unfavorable prognostic factor for EOC patients Benign ovarian tumors, BOT and EOC (54% HGSOC) [24]
CD44+/CD117+ Increased tumorigenicity, sphere-formation, recapitulation of tumors, chemoresistance EOC tumors, xenograft models [13] CD44+CD117+ cell lines were less prone to paclitaxel-induced apoptosis EOC cell lines [23]
CD44+/CD24- Increased tumorigenicity, sphere-formation Cell lines [25] >25% CD44+/CD24- cells in ascites correlated with higher risk of recurrence and shorter PFS Ascites-isolated cells from advanced EOC [26]
CD44+/CD24+/ EpCAM+ Increased tumorigenicity, chemoresistance Cell lines, EOC-isolated cell lines, ascites [27][28] Ovarian cancer stem cells expressing EpCAM+ are less prone to chemotherapy and are a source of recurrent tumor after the treatment EOC I-IV FIGO stage (45% HGSOC, 14% clear cell, 17% endometroid, 12% mucinous) [27]
CD44+/CD166+ Increased tumorigenicity, sphere-formation Cell lines [29] Population of platinum-resistant cells is enriched in CD44+/CD166+ population EOC-isolated and standard cell lines [30]
CD44+ALDH1+ Increased tumorigenicity, chemoresistance Cell lines [31] >50% ALDH1+ cells correlated with shorter OS Advanced EOC (73% HGSOC) [31]
CD44+/CD133+/ALDH1A1+ Chemoresistance Cell lines, EOC-isolated cell lines [32] Expression of markers increased in recurrent compared to primary tumors Advanced primary and recurrent EOC [32]
CD133+ Increased tumorigenicity, enhanced vasculogenesis Cell lines, EOC tumors, xenograft models, ascites [33][34][35][36] Expression of CD133+ correlated with presence of HGSOC, higher FIGO stage, ascites, chemoresistance, shorter PFS and OS
No correlation with prognosis
Expression of CD133+ correlated with shorter PFS and OS
Expression of CD133+ correlated with shorter OS and platinum chemo-resistance
EOC (67% HGSOC)
EOC FIGO III–IV (72% HGSOC)
Advanced metastatic HGSOC
Advanced primary HGSOC
[37][38][39][40]
CD133+/ALDH1A+ Increased tumorigenicity, cells self-renewal, chemoresistance EOC tumors, cell lines, xenograft models [41][35] Expression of CD133+ correlated with III/IV FIGO stage, expression of CD133+/ALDH1A+ correlated with shorter PFS and OS HGSOC [42]
CD117+ Increased tumorigenicity, sphere-formation, recapitulation of tumors, chemoresistance EOC-isolated cell lines, xenograft model, ascites [43][44][45][46][47] Expression of CD117+ correlated with shorter PFS
40% of HGSOC were CD117+ and expression correlated with chemoresistance
Advanced metastatic HGSOC
HGSOC
[44][39]
CD24+ Increased tumorigenicity, stimulation of EMT Cell lines [48] Expression of CD24+ correlated with FIG stage and the presence of peritoneal and lymph node metastases 27% HGSOC
12% mucinous
18% clear-cell
18% endometaroid
23% others
[48]
BOT—borderline ovarian tumor; DFI—disease-free interval; EMT—epithelial-mesenchymal transition; EOC—epithelial ovarian cancer; FIGO—International Federation of Obstetrics and Gynecology; HGSOC—high-grade serous ovarian cancer; OS—overall survival; PFS—progression-free survival.

2.1.1. Glycoprotein CD44

Molecule CD44 is a cell-surface glycoprotein that is a receptor for the hyaluronic acid receptor. Its stimulation activates signaling pathways including epidermal growth factor receptor (EGFR)/Ras small GTPase protein (Ras)/extracellular signal-regulated kinase (ERK) and transcription factor homeobox protein NANOG-dependent signaling pathways, followed by cell proliferation, differentiation, and increased motility and stemness. Interaction between NANOG and STAT3 results in the up-regulation of multidrug resistance protein-1 (MDR1) and the effective efflux of chemotherapeutic drugs from OCSCs [49][13]. The population of CD44+ OC cells possess self-renewal, tumor initiating and sphere-forming capacities. Recurrent OC shows a higher expression of CD44-positive cells compared to primary tumors which is correlated with poor prognosis, although not all studies are consistent in this matter. CD44 exists in alternatively spliced variants which could be better correlated to the clinical history of OC. Between them CD44v6 was found in excess on OCSCs from distant metastases indicating metastasis-initiating activity via a hematogenous spread. In patients with FIGO stage I-III OC, distant metastasis-free survival was better in patients with CD44v6-low tumors. Silencing of the CD44-regulatory gene with siRNA resulted in tumor shrinkage in a mouse model of OC [13][14][15][16][50][51][52][22][21][53].

2.1.2. Receptor Tyrosine Kinase CD117

CD117 is receptor tyrosine kinase coded by c-kit proto-oncogene. It has an external ligand-binding domain specific for a stem cell factor (SCF) and a cytoplasmic domain with intrinsic tyrosine kinase activity. Binding CD117 to SCF causes the receptor proteins to dimerize, phosphorylation and activation. This activates several signaling pathways, mainly Ras/ERK, phosphoinositide-3-kinase (PI3K)/protein kinase-B (AKT), non-receptor tyrosine kinase Src and Janus kinase (JAK)/signal transducer and activator of transcription (STAT), responsible for regulating cell proliferation, differentiation, apoptosis and adhesion. CD117 identifies a population of sphere-forming non-adherent OC cells and a so-called “side population” of OC cells having the capacity to harbor a specific position on flow-cytometric panels due to the selective expression of ATP-binding cassette drug transporters (ABC membrane transporters) using Hoechst 33,342 dye-staining. The basis of the “side-population” assay is the ability of ABC transporters to provide a rapid efflux of lipophilic fluorescent dye, and they are identified using special gating strategies during the analysis of flow-cytometric plots. Since its first use for hematopoietic stem cell identification, the “side-population” assay has been successfully used for the identification of progenitor and stem cells in different tissues, including stem-like cancer cells displaying increased capacity of self-renewal, tumorigenicity and chemo-resistance [54]. The “side population” shows an increased expression of stemness genes. OC cells bearing both CD44 and CD117 markers are abundant in chemo-resistant OCSCs [52][53][55][56][43][44][45][57]. The presence of CD117+ OC cells are correlated with resistance to standard platinum-taxane-based chemotherapy and shorter recurrence intervals in treated OC patients, while the inhibition of CD117 results in the restitution of chemo-sensitivity. The level of CD117 expression correlates with both disease-free and overall survival. The expression of CD117 could be augmented by hypoxia and HIF-1α activation, and is followed by Wnt/β-catenin signaling [44][58][59]. SCF growth factor is expressed in high concentrations in the ascitic effusions collected from EOC patients. The soluble SCF form is produced by TAMs and fibroblasts (TAFs), whereas a minority of tumor cells only express the membrane-associated SCF form. C-kit is expressed by OCSCs and binds to both soluble and cell-associated SCF, as well as functionally responding to the ligand [60].

2.1.3. Prominin-1 CD133

Surface protein CD133 is also called prominin-1 and is a transmembrane glycoprotein activating the PI3K/Akt pathway. It is responsible for tumor maintenance, vascularization, and chemoresistance, and hence is considered to be an OCSC marker. It was found that CD133 mediates metastatic homing to peritoneal tissue in OC [61]. Endothelin receptor-A (ETRA) is expressed on CD133+ cells and the chemotherapy-induced inhibition of ETRA decreases the ability of CD133+ cells to form spheres [62]. The activity of nuclear factor-κ-light chain enhancer of activated B cells (NF-κB) and p38 MAPK pathways in response to IL-17 enhances the self-renewal capabilities of CD133+ cells. The expression of intracellular stemness markers octamer-binding transcription factor-4 (OCT4) and sex-determining region Y—box 2 (SOX2) is higher in CD133+ than CD133- cells [63]. Ovarian C-X-C chemokine receptor type-4 (CXCR4)+/CD133+ OC cells showed more effective platinum efflux and lower platinum sensitivity compared to CD133-negative cells [33]. The correlation between CD133 expression and advancement of the tumor (presence of HGOC, advanced clinical stage, presence of ascites, tumor non-responsive to chemotherapy), as well as patients’ survival has been pointed out [37]. Moreover, the population of CD133+ cells was more abundant in recurrent platinum-resistant tumors compared to primary OC tumors [64]. However, according to some studies, OCSCs indicate an inconsistent expression of the CD133 marker and CD133+ cells do not always have particular pro-tumorigenic properties. It is possible that OCSCs plasticity and some heterogeneity could account for this controversy. Another possibility is an alternating expression of the CD133 molecule in cytosolic and membrane compartments of OCSCs populations [65][66][67][34][68][41][35][69][70][71].

2.1.4. Heat-Stable Antigen CD24

CD24 is a transmembrane adhesion molecule also called a heat-stable antigen CD24. Through the activation of the STAT3 signaling pathway, it is able to stimulate cell adhesion and augment cell malignancy. The inhibition of the JAK2/STAT3 pathway in OC results in a loss of cancer stemness and reduced tumor growth [72]. The inhibition of the over-expressed JAK2/STAT3 pathway in patient-derived CD24+ OCSCs correlated with a better survival of OC patients [73]. The CD24 molecule may also regulate NANOG and OCT4 expression, thus stimulating tumor growth and resistance to chemotherapy. CD24-positive OC cells were isolated both from tissue specimens and from cellular cultures, and were shown to overcome anoikis and to form spheroid structures. CD24+ OC cells also displayed the up-regulation of genes regulating cellular stemness. The abundance of CD24+ cells was increased in metastatic peritoneal implants compared to the primary tumor, where it contributed to the attachment of OC cells to fibronectin and collagen of the peritoneal stroma. STAT3 signaling following CD24 stimulation is a well-established stimulator of stemness of OCSCs cells, while PI3K/AKT and mitogen-activated protein kinase (MAPK) signaling following CD24 stimulation could activate EMT. However, there are also scanty opinions that CD24 expression has no correlation with OCSCs’ self-renewal and chemoresistance [74][75][76][77][78][48][79][80][81]. Clinically, high CD24+ expression is a predictor of poor survival in OC patients [82].

2.1.5. MyD88 Protein

Myeloid differentiation primary response gene 88 (MyD88) is an adaptor protein for signals generated by toll-like receptor-type 4 (TLR-4). TLR-dependent signaling activates NF-κB which moderates the inflammatory pathway in tumor carcinogenesis. The TLR-4/MyD88 pathway has been modified in OC and is responsible for OC chemo-resistance. MyD88-positive ovarian cancer cells equate to OCSCs cells due to the resistance to pro-apoptotic signals and the ability to create a pro-inflammatory tumor microenvironment through leukocyte recruitment. MyD88-negative cells are more differentiated and less aggressive. The expression of MyD88 protein was found to be an unfavorable prognostic factor for OC patients [24][83].

2.1.6. Epithelial Cell Adhesion Molecule EpCAM

Epithelial cell adhesion molecule (EpCAM) is a type I transmembrane glycoprotein regulating intercellular adhesion, present on a subset of normal epithelia, as suggested by several recent studies [84]. EpCAM-positive OC cells have greater tumor-initiating potential compared to EpCAM-negative cells. The EpCAM/B-cell lymphoma-2 (Bcl-2) signaling pathway prevents platinum-dependent apoptosis of cancer cells resulting in chemo-resistance; therefore, EpCAM expression is increased in tumors of chemo-resistant patients and correlates with unfavorable outcome [27]. Similarly, the B-cell lymphoma extra-large (Bcl-xL) anti-apoptotic protein present in mitochondria was found to be over-expressed in recurrent chemo-resistant OC. The inhibition of Bcl-xL restored chemo-sensitivity of OC cells [85].

2.1.7. Multipositivity of Cell Surface Markers

Effective isolation of OCSCs usually demands the identification of two or more cell markers. Double positive CD44+/CD117+ cells are highly capable to recapitulate the original tumor after being transplanted into experimental animals, and are the main component of sphere-forming cells in ascites. This OCSC population also showed high mitochondrial concentrations of reactive oxygen species (ROS) suggesting that mitochondrial respiration is used to sustain OCSC’s viability in stress conditions and during starvation [86]. The level of CD44+CD24-OCSCs in OC patients has been suggested to have a prognostic value. In patients having more than 25% of CD44+CD24-OCSCs, a greater probability of recurrence and a shorter progression-free survival were observed. Similarly, primary tumors showed either a low or high expression of CD44+ALDH1+ OCSCs. Those exhibiting low expression had a better response to chemotherapy and longer progression-free survival [87][26][17][88]. Recurrent platinum-resistant ovarian tumors compared to primary tumors are enriched in the population of CD44+CD133+ALDH1A1+ OCSCs. The population of CD44+/E-cadherin-/CD34- inside ovarian tumors identify OCSCs cells with the ability to recapitulate the tumor and support neovascularization [89]. The population of CD44+CD166+ has stem cell characteristics through the increased capacity of forming spheres and the high activity of histone deacetylases regulating the OCSC’s phenotype [29]. CD133+/ALDH1+ cells have tumor initiating properties and induce neoangiogenesis. CD44+/MyD88+ cells are highly resistant against apoptosis and chemotherapeutic drugs. They can grow as adherent cells or are able to undergo EMT and form spheroid cell structures [90]. Similarly, CD44+/CD24+/EpCAM+ cells show OCSCs’ properties having increased migratory and invasive potential and chemo-resistance to platinum, taxane and doxorubicin [28]. Cells of CXCR4+CD133+ phenotype isolated from OC cell lines also have OCSC properties [67]. Neoadjuvant chemotherapy is a therapeutic option for patients in whom primary optimal cytoreductive surgery is unavailable due to extensive peritoneal carcinomatosis. However, studies have revealed that this form of management is associated with the enrichment of metastatic tumors in OCSCs defined as ALDH1+ cells showing chemo-resistance and correlated with bad prognosis [91][92]. Recent studies have shown that not only standard platinum-taxane-based chemotherapy contributes to the proliferation of OCSC population; targeted therapy with poly-(ADP) ribose polymerase (PARP) inhibitors which disturb tumor DNA repair systems also result in an enrichment of tumors with OCSCs followed by a restored ability to repair DNA [53].
The variegated opinions about OCSC markers could have several origins. The simplest explanation is that different OCSC populations are characterized by different markers. Another possibility is that due to ovarian cancer histological and genetic heterogeneity, the observed populations of OCSCs follow this heterogenic pattern. Moreover, OCSCs could differ between distinct localizations and in different stages of the disease. Finally, the marker diversity could result from OCSCs’ molecular and functional plasticity, where cells with different properties share stemness and tumor propagating abilities [93][94]. In this context, patients could have multiple populations of OCSCs which may vary between distinct tumors and patients. This possibility creates an unfavorable perspective for the success of therapy directed against OCSCs.

2.2. Intracellular and Functional Markers

2.2.1. Aldehyde Dehydrogenases ALDH

Aldehyde dehydrogenases (ALDH) are a group of enzymes converting aldehyde substrates to carboxylic acids via the oxidation process. The protective and detoxifying function of the ALDH1 subgroup is involved in the maintenance of cancer cells, especially OCSCs, against chemotherapeutics and radiation. In this subgroup, the most supporting role for the creation of the OCSC phenotype is assigned to ALDH1A1 and ALDH1A2 isotypes [95]. ALDH1-positive cell phenotype identifies the OCSC populations possessing self-renewal and stemness properties that are capable of sphere formation and restoring the tumor. ALDH1 exerts its function via the Wnt/β-catenin signaling pathway and ABC-transporters. ALDH1 activity is also regulated by NF-κB/transcription factor RelB non-canonical pathway [95]. Moreover, ALDH1 is engaged in the activation of OCSCs quiescence program by slowing down the proliferation of the cells and in the protection of DNA-repair programs that both contribute to ovarian cancer resistance. It was found that primary OC tumors contain less ALDH1+ cells compared to tumors pre-treated with neoadjuvant chemotherapy [91][32][96][97][98][99][100][101][102]. The observation that, in cultures of OC cells, the percentage of ALDH1+ cells was growing along with the dose escalation of platinum was a very alarming phenomenon, indicating great viability and endurance of this cell population [35]. Epidermal growth factor-like domain-6 (EGFL6), which functions as a stem cell regulatory factor activates an asymmetric division of ALDH1+ OC cells stimulating their proliferation [88][103][104]. In HGOC patients, tumors characterized by the higher percentage of double-positive ALDH+/epidermal growth factor receptor (EGFR) + cells, are associated with poor outcomes as compared with tumors that are either ALDH or EGFR negative [105]. In clear-cell carcinoma population of ALDH1high OCSCs, cells show markedly higher tumorigenic potential than ALDH1low cells. The expression of ALDH1high OCSCs is increased in advanced tumors and correlates with unfavorable prognosis and chemo-resistance [88][99][106].

2.2.2. Transcription Factors

A group of transcription factors have also been considered to be markers of OCSCs. These include NANOG, SOX2, forkhead-box protein M1 (FOXM1), and OCT4. NANOG is physiologically responsible for the maintenance of self-renewal and the pluripotency of embryonic stem cells (ESCs), the same role it plays in OC, where it additionally regulates EMT and chemo-resistance via the STAT3 signaling pathway. The expression of NANOG in OCSCs cells correlates with clinical stage and high grade, as well as resistance to standard chemotherapy [14]. Similar to NANOG, SOX2 is also responsible for the maintenance of self-renewal and the pluripotency of ESCs. The over-expression of SOX2 is related to the stemness of cells via the inhibition of the PI3K/AKT pathway, resulting in resistance to apoptosis. In OC, pathological SOX2-positive cells were identified in the epithelium of the tubal fimbriae of patients with HGOC tumors and patients with germline BRCA1/2 mutations [107][108]. OCT4 is involved in embryonic development and cellular pluripotency. Its function is to stabilize the higher-order structure of chromatin in the NANOG locus [109]. Up-regulation of OCT4 in OCSCs was correlated to tumor initiation and again, chemo-resistance. The cytoplasmic expression of OCT4 regulates EMT transformation and is a recognized predictor of adverse clinical outcomes in cancer. The co-expression of OCT4 with RNA-binding protein Lin28 was connected to advanced tumor growth and grade [110]. Increased levels of OCT4 were observed in OCSCs’ CD24-positive cells [75]. NANOG, OCT4 and SOX2 were over-expressed not only in tumor tissues but also in both ascites and spheres built from OCSCs cells [108][111][112]. FOXM1 is a member of the FOX family of transcription factors. It plays an important role in cell cycle control and progression, and in the maintenance of genomic stability. The over-expression of FOXM1 protein was observed in OCSCs exposed to elevated concentrations of lysophosphatidic acid (LPA) present in ascites fluid in OC patients. Increased FOXM1 levels were followed by the activation of wingless and Int-1 (Wnt)/β-catenin signaling and chemo-resistance. Alternatively, FOXM1 suppression resulted in the restitution of chemo-sensitivity and the loss of ability to spheroid formation in the peritoneal environment [113][114][115].
The cells characterized by OCSC phenotypes were identified both in ovarian surface epithelium (OSE) and fallopian tube epithelium (FTE) in mice, including ALDH1+, ALDH1A1+, ALDH1A2+, CD133+ and NANOG+ cells. These OCSCs markers were detected especially in the distal portion of the tube (fimbria) supporting the observation of HGOC origin [116][117].

3. Anti-OCSC Therapy

Cancer stem cells constitute a very attractive target for therapy. The effective elimination of this cell population would probably improve the treatment of advanced cancer and protect against recurrent lethal disease. Therefore, many different approaches to CSC-targeted management have been proposed and tested in vitro, in experimental settings and in clinical trials. CSCs are under extensive investigation in practically all known types of cancer. The most popular and tested targets for anti-CSC therapy are signaling pathways regulating the origin and function of CSCs, the surface and intracellular markers of CSCs, drugs changing the epigenetic regulation of CSCs’ function, and CSCs’ metabolism. Among them, there are inhibitors of Wnt (Ipafricept), Hedgehog (Vismodegib, Sonidegib), NOTCH (enoticumab, demcizumab, navicixizumab), MAPK (Salinomycin), and other signaling pathways (Metformin). Another group are drugs targeting CSC markers (Imatinib mesylate), epigenetic regulation by DNA-(cytosine-5)-methyltransferase-1 (DNMT1) (Decitabine, Guadecitabine, Azacitidine) or by histone deacetylase (HDAC) (Vorinostat, Belinostat, Etinostat). There are also some natural compounds being tested, such as curcumin, which has indicated anti-cancer activity in in vitro and animal studies. Another approach is to combine drugs or toxic agents with nanoparticles capable to transport them precisely into the tumor (examples are glucose-coated gold particles, paclitaxel albumin-bound nanoparticles, doxorubicin or mangostin encapsulated poly D/L lactide-co-glicolide acid—PLGA) [118]. Immunotherapy directed against tumor antigens with the use of chimeric antigen receptor T cells (CAR-T lymphocytes) has also been tested in many malignancies [119]. However, the immunosuppressive environment of solid tumors represents a barrier to this therapy’s success, due to low antigen expression on tumor cells [120]. A combination of several methods, for example, CAR-T cells and oncolytic viruses (Ovs), can allow the targeting of CSCs and the surrounding cancer niche. An OV-based strategy to overcome the mechanism of CAR-T cell evasion is to encode CAR-targeted TAAs in OVs to increase TAA expression more homogeneously across the tumor [121].

References

  1. Giusti, I.; Bianchi, S.; Nottola, S.A.; Macchiarelli, G.; Dolo, V. Clinical Electron Microscopy in the Study of Human Ovarian Tissues. Euromediterr. Biomed. J. 2019, 14, 145–151.
  2. Kurman, R.J.; Shih, I.-M. The Dualistic Model of Ovarian Carcinogenesis. Am. J. Pathol. 2016, 186, 733–747.
  3. Tothill, R.W.; Tinker, A.V.; George, J.; Brown, R.; Fox, S.B.; Lade, S.; Johnson, D.S.; Trivett, M.K.; Etemadmoghadam, D.; Locandro, B.; et al. Novel Molecular Subtypes of Serous and Endometrioid Ovarian Cancer Linked to Clinical Outcome. Clin. Cancer Res. 2008, 14, 5198–5208.
  4. Tsang, Y.T.; Deavers, M.T.; Sun, C.C.; Kwan, S.-Y.; Kuo, E.; Malpica, A.; Mok, S.C.; Gershenson, D.M.; Wong, K.-K. KRAS (but Not BRAF ) Mutations in Ovarian Serous Borderline Tumour Are Associated with Recurrent Low-Grade Serous Carcinoma. J. Pathol. 2013, 231, 449–456.
  5. Wu, R.; Hendrix-Lucas, N.; Kuick, R.; Zhai, Y.; Schwartz, D.R.; Akyol, A.; Hanash, S.; Misek, D.E.; Katabuchi, H.; Williams, B.O.; et al. Mouse Model of Human Ovarian Endometrioid Adenocarcinoma Based on Somatic Defects in the Wnt/β-Catenin and PI3K/Pten Signaling Pathways. Cancer Cell 2007, 11, 321–333.
  6. Saegusa, M.; Hashimura, M.; Yoshida, T.; Okayasu, I. β- Catenin Mutations and Aberrant Nuclear Expression during Endometrial Tumorigenesis. Br. J. Cancer 2001, 84, 209–217.
  7. Nakayama, K.; Nakayama, N.; Kurman, R.J.; Cope, L.; Pohl, G.; Samuels, Y.; Velculescu, V.E.; Wang, T.-L.; Shih, I.-M. Sequence Mutations and Amplification of PIK3CA and AKT2 Genes in Purified Ovarian Serous Neoplasms. Cancer Biol. Ther. 2006, 5, 779–785.
  8. Patch, A.-M.; Christie, E.L.; Etemadmoghadam, D.; Garsed, D.W.; George, J.; Fereday, S.; Nones, K.; Cowin, P.; Alsop, K.; Bailey, P.J.; et al. Whole–Genome Characterization of Chemoresistant Ovarian Cancer. Nature 2015, 521, 489–494.
  9. Vang, R.; Levine, D.A.; Soslow, R.A.; Zaloudek, C.; Shih, I.-M.; Kurman, R.J. Molecular Alterations of TP53 Are a Defining Feature of Ovarian High-Grade Serous Carcinoma. Int. J. Gynecol. Pathol. 2016, 35, 48–55.
  10. Integrated Genomic Analyses of Ovarian Carcinoma. Nature 2011, 474, 609–615.
  11. Konecny, G.E.; Wang, C.; Hamidi, H.; Winterhoff, B.; Kalli, K.R.; Dering, J.; Ginther, C.; Chen, H.-W.; Dowdy, S.; Cliby, W.; et al. Prognostic and Therapeutic Relevance of Molecular Subtypes in High-Grade Serous Ovarian Cancer. JNCI: J. Natl. Cancer Inst. 2014, 106, dju249.
  12. Irani, S. Emerging Insights into the Biology of Metastasis: A Review Article. Iran. J. Basic Med. Sci. 2019, 22, 833–847.
  13. 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.
  14. Bourguignon, L.Y.W.; Peyrollier, K.; Xia, W.; Gilad, E. Hyaluronan-CD44 Interaction Activates Stem Cell Marker Nanog, Stat-3-Mediated MDR1 Gene Expression, and Ankyrin-Regulated Multidrug Efflux in Breast and Ovarian Tumor Cells. J. Biol. Chem. 2008, 283, 17635–17651.
  15. Cheng, W.; Liu, T.; Wan, X.; Gao, Y.; Wang, H. MicroRNA-199a Targets CD44 to Suppress the Tumorigenicity and Multidrug Resistance of Ovarian Cancer-Initiating Cells. FEBS J. 2012, 279, 2047–2059.
  16. Chen, J.; Wang, J.; Zhang, Y.; Chen, D.; Yang, C.; Kai, C.; Wang, X.; Shi, F.; Dou, J. Observation of Ovarian Cancer Stem Cell Behavior and Investigation of Potential Mechanisms of Drug Resistance in Three-Dimensional Cell Culture. J. Biosci. Bioeng. 2014, 118, 214–222.
  17. Steffensen, K.D.; Alvero, A.B.; Yang, Y.; Waldstrøm, M.; Hui, P.; Holmberg, J.C.; Silasi, D.-A.; Jakobsen, A.; Rutherford, T.; Mor, G. Prevalence of Epithelial Ovarian Cancer Stem Cells Correlates with Recurrence in Early-Stage Ovarian Cancer. J. Oncol. 2011, 2011, 620523.
  18. Liu, M.; Mor, G.; Cheng, H.; Xiang, X.; Hui, P.; Rutherford, T.; Yin, G.; Rimm, D.L.; Holmberg, J.; Alvero, A.; et al. High Frequency of Putative Ovarian Cancer Stem Cells With CD44/CK19 Coexpression Is Associated With Decreased Progression-Free Intervals In Patients With Recurrent Epithelial Ovarian Cancer. Reprod. Sci. 2013, 20, 605–615.
  19. Zhang, J.; Chang, B.; Liu, J. CD44 Standard Form Expression Is Correlated with High-Grade and Advanced-Stage Ovarian Carcinoma but Not Prognosis. Hum. Pathol. 2013, 44, 1882–1889.
  20. Kar, K.; Ghosh, S.; Roy, A.K. A Study of CD44 Positive Cancer Cells in Epithelial Ovarian Cancer and Their Correlation with P53 And Ki67. J. Lab. Physicians 2021, 13, 050–057.
  21. Tjhay, F.; Motohara, T.; Tayama, S.; Narantuya, D.; Fujimoto, K.; Guo, J.; Sakaguchi, I.; Honda, R.; Tashiro, H.; Katabuchi, H. CD 44 Variant 6 Is Correlated with Peritoneal Dissemination and Poor Prognosis in Patients with Advanced Epithelial Ovarian Cancer. Cancer Sci. 2015, 106, 1421–1428.
  22. Motohara, T.; Fujimoto, K.; Tayama, S.; Narantuya, D.; Sakaguchi, I.; Tashiro, H.; Katabuchi, H. CD44 Variant 6 as a Predictive Biomarker for Distant Metastasis in Patients With Epithelial Ovarian Cancer. Obstet. Gynecol. 2016, 127, 1003–1011.
  23. Chen, M.; Su, J.; Feng, C.; Liu, Y.; Zhao, L.; Tian, Y. Chemokine CCL20 Promotes the Paclitaxel Resistance of CD44 + CD117 + Cells via the Notch1 Signaling Pathway in Ovarian Cancer. Mol. Med. Rep. 2021, 24, 635.
  24. d’Adhemar, C.J.; Spillane, C.D.; Gallagher, M.F.; Bates, M.; Costello, K.M.; Barry-O’Crowley, J.; Haley, K.; Kernan, N.; Murphy, C.; Smyth, P.C.; et al. The MyD88+ Phenotype Is an Adverse Prognostic Factor in Epithelial Ovarian Cancer. PLoS ONE 2014, 9, e100816.
  25. Shi, M.F.; Jiao, J.; Lu, W.G.; Ye, F.; Ma, D.; Dong, Q.G.; Xie, X. Identification of Cancer Stem Cell-like Cells from Human Epithelial Ovarian Carcinoma Cell Line. Cell. Mol. Life Sci. 2010, 67, 3915–3925.
  26. Meng, E.; Long, B.; Sullivan, P.; McClellan, S.; Finan, M.A.; Reed, E.; Shevde, L.; Rocconi, R.P. CD44+/CD24− Ovarian Cancer Cells Demonstrate Cancer Stem Cell Properties and Correlate to Survival. Clin. Exp. Metastasis 2012, 29, 939–948.
  27. Motohara, T.; Masuko, S.; Ishimoto, T.; Yae, T.; Onishi, N.; Muraguchi, T.; Hirao, A.; Matsuzaki, Y.; Tashiro, H.; Katabuchi, H.; et al. Transient Depletion of P53 Followed by Transduction of C-Myc and K-Ras Converts Ovarian Stem-like Cells into Tumor-Initiating Cells. Carcinogenesis 2011, 32, 1597–1606.
  28. Meirelles, K.; Benedict, L.A.; Dombkowski, D.; Pepin, D.; Preffer, F.I.; Teixeira, J.; Tanwar, P.S.; Young, R.H.; MacLaughlin, D.T.; Donahoe, P.K.; et al. Human Ovarian Cancer Stem/Progenitor Cells Are Stimulated by Doxorubicin but Inhibited by Mullerian Inhibiting Substance. Proc. Natl. Acad. Sci. USA 2012, 109, 2358–2363.
  29. Witt, A.E.; Lee, C.-W.; Lee, T.I.; Azzam, D.J.; Wang, B.; Caslini, C.; Petrocca, F.; Grosso, J.; Jones, M.; Cohick, E.B.; et al. Identification of a Cancer Stem Cell-Specific Function for the Histone Deacetylases, HDAC1 and HDAC7, in Breast and Ovarian Cancer. Oncogene 2017, 36, 1707–1720.
  30. Wu, Y.; Wang, T.; Xia, L.; Zhang, M. LncRNA WDFY3-AS2 Promotes Cisplatin Resistance and the Cancer Stem Cell in Ovarian Cancer by Regulating Hsa-MiR-139-5p/SDC4 Axis. Cancer Cell Int. 2021, 21, 284.
  31. Wang, Y.-C.; Yo, Y.-T.; Lee, H.-Y.; Liao, Y.-P.; Chao, T.-K.; Su, P.-H.; Lai, H.-C. ALDH1-Bright Epithelial Ovarian Cancer Cells Are Associated with CD44 Expression, Drug Resistance, and Poor Clinical Outcome. Am. J. Pathol. 2012, 180, 1159–1169.
  32. Steg, A.D.; Bevis, K.S.; Katre, A.A.; Ziebarth, A.; Dobbin, Z.C.; Alvarez, R.D.; Zhang, K.; Conner, M.; Landen, C.N. Stem Cell Pathways Contribute to Clinical Chemoresistance in Ovarian Cancer. Clin. Cancer Res. 2012, 18, 869–881.
  33. Baba, T.; Convery, P.A.; Matsumura, N.; Whitaker, R.S.; Kondoh, E.; Perry, T.; Huang, Z.; Bentley, R.C.; Mori, S.; Fujii, S.; et al. Epigenetic Regulation of CD133 and Tumorigenicity of CD133+ Ovarian Cancer Cells. Oncogene 2009, 28, 209–218.
  34. Curley, M.D.; Therrien, V.A.; Cummings, C.L.; Sergent, P.A.; Koulouris, C.R.; Friel, A.M.; Roberts, D.J.; Seiden, M.V.; Scadden, D.T.; Rueda, B.R.; et al. CD133 Expression Defines a Tumor Initiating Cell Population in Primary Human Ovarian Cancer. Stem Cells 2009, 27, 2875–2883.
  35. Silva, I.A.; Bai, S.; McLean, K.; Yang, K.; Griffith, K.; Thomas, D.; Ginestier, C.; Johnston, C.; Kueck, A.; Reynolds, R.K.; et al. Aldehyde Dehydrogenase in Combination with CD133 Defines Angiogenic Ovarian Cancer Stem Cells That Portend Poor Patient Survival. Cancer Res. 2011, 71, 3991–4001.
  36. Kusumbe, A.P.; Mali, A.M.; Bapat, S.A. CD133-Expressing Stem Cells Associated with Ovarian Metastases Establish an Endothelial Hierarchy and Contribute to Tumor Vasculature. Stem Cells 2009, 27, 498–508.
  37. Zhang, J.; Guo, X.; Chang, D.Y.; Rosen, D.G.; Mercado-Uribe, I.; Liu, J. CD133 Expression Associated with Poor Prognosis in Ovarian Cancer. Mod. Pathol. 2012, 25, 456–464.
  38. Ferrandina, G.; Martinelli, E.; Petrillo, M.; Prisco, M.G.; Zannoni, G.; Sioletic, S.; Scambia, G. CD133 Antigen Expression in Ovarian Cancer. BMC Cancer 2009, 9, 221.
  39. Stemberger-Papić, S.; Vrdoljak-Mozetic, D.; Ostojić, D.V.; Rubesa-Mihaljević, R.; Krigtofić, I.; Brncić-Fisher, A.; Kragević, M.; Eminović, S. Expression of CD133 and CD117 in 64 Serous Ovarian Cancer Cases. Coll. Antropol. 2015, 39, 745–753.
  40. Qin, Q.; Sun, Y.; Fei, M.; Zhang, J.; Jia, Y.; Gu, M.; Xia, R.; Chen, S.; Deng, A. Expression of Putative Stem Marker Nestin and CD133 in Advanced Serous Ovarian Cancer. Neoplasma 2012, 59, 310–316.
  41. Kryczek, I.; Liu, S.; Roh, M.; Vatan, L.; Szeliga, W.; Wei, S.; Banerjee, M.; Mao, Y.; Kotarski, J.; Wicha, M.S.; et al. Expression of Aldehyde Dehydrogenase and CD133 Defines Ovarian Cancer Stem Cells. Int. J. Cancer 2012, 130, 29–39.
  42. Ruscito, I.; Cacsire Castillo-Tong, D.; Vergote, I.; Ignat, I.; Stanske, M.; Vanderstichele, A.; Ganapathi, R.N.; Glajzer, J.; Kulbe, H.; Trillsch, F.; et al. Exploring the Clonal Evolution of CD133/Aldehyde-Dehydrogenase-1 (ALDH1)-Positive Cancer Stem-like Cells from Primary to Recurrent High-Grade Serous Ovarian Cancer (HGSOC). A Study of the Ovarian Cancer Therapy–Innovative Models Prolong Survival (OCTIPS) Consortium. Eur. J. Cancer 2017, 79, 214–225.
  43. He, Q.; Luo, X.; Wang, K.; Zhou, Q.; Ao, H.; Yang, Y.; Li, S.; Li, Y.; Zhu, H.; Duan, T. Isolation and Characterization of Cancer Stem Cells from High-Grade Serous Ovarian Carcinomas. Cell. Physiol. Biochem. 2014, 33, 173–184.
  44. Luo, L.; Zeng, J.; Liang, B.; Zhao, Z.; Sun, L.; Cao, D.; Yang, J.; Shen, K. Ovarian Cancer Cells with the CD117 Phenotype Are Highly Tumorigenic and Are Related to Chemotherapy Outcome. Exp. Mol. Pathol. 2011, 91, 596–602.
  45. Szotek, P.P.; Pieretti-Vanmarcke, R.; Masiakos, P.T.; Dinulescu, 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.
  46. Hu, L.; McArthur, C.; Jaffe, R.B. Ovarian Cancer Stem-like Side-Population Cells Are Tumourigenic and Chemoresistant. Br. J. Cancer 2010, 102, 1276–1283.
  47. Kobayashi, Y.; Seino, K.; Hosonuma, S.; Ohara, T.; Itamochi, H.; Isonishi, S.; Kita, T.; Wada, H.; Kojo, S.; Kiguchi, K. Side Population Is Increased in Paclitaxel-Resistant Ovarian Cancer Cell Lines Regardless of Resistance to Cisplatin. Gynecol. Oncol. 2011, 121, 390–394.
  48. Nakamura, K.; Terai, Y.; Tanabe, A.; Ono, Y.J.; Hayashi, M.; Maeda, K.; Fujiwara, S.; Ashihara, K.; Nakamura, M.; Tanaka, Y.; et al. CD24 Expression Is a Marker for Predicting Clinical Outcome and Regulates the Epithelial-Mesenchymal Transition in Ovarian Cancer via Both the Akt and ERK Pathways. Oncol. Rep. 2017, 37, 3189–3200.
  49. Muñoz-Galván, S.; Carnero, A. Targeting Cancer Stem Cells to Overcome Therapy Resistance in Ovarian Cancer. Cells 2020, 9, 1402.
  50. Du, Y.-R.; Chen, Y.; Gao, Y.; Niu, X.-L.; Li, Y.-J.; Deng, W.-M. Effects and Mechanisms of Anti-CD44 Monoclonal Antibody A3D8 on Proliferation and Apoptosis of Sphere-Forming Cells With Stemness From Human Ovarian Cancer. Int. J. Gynecol. Cancer 2013, 23, 1367–1375.
  51. Grass, G.D.; Tolliver, L.B.; Bratoeva, M.; Toole, B.P. CD147, CD44, and the Epidermal Growth Factor Receptor (EGFR) Signaling Pathway Cooperate to Regulate Breast Epithelial Cell Invasiveness. J. Biol. Chem. 2013, 288, 26089–26104.
  52. Lupia, M.; Cavallaro, U. Ovarian Cancer Stem Cells: Still an Elusive Entity? Mol. Cancer 2017, 16, 64.
  53. Zong, X.; Nephew, K.P. Ovarian Cancer Stem Cells: Role in Metastasis and Opportunity for Therapeutic Targeting. Cancers 2019, 11, 934.
  54. Golebiewska, A.; Brons, N.H.C.; Bjerkvig, R.; Niclou, S.P. Critical Appraisal of the Side Population Assay in Stem Cell and Cancer Stem Cell Research. Cell Stem Cell 2011, 8, 136–147.
  55. Abbaspour Babaei, M.; Kamalidehghan, B.; Saleem, M.; Zaman Huri, H.; Ahmadipour, F. Receptor Tyrosine Kinase (c-Kit) Inhibitors: A Potential Therapeutic Target in Cancer Cells. Drug Des. Dev. Ther. 2016, 10, 2443–2459.
  56. Chau, W.K.; Ip, C.K.; Mak, A.S.C.; Lai, H.-C.; Wong, A.S.T. C-Kit Mediates Chemoresistance and Tumor-Initiating Capacity of Ovarian Cancer Cells through Activation of Wnt/β-Catenin–ATP-Binding Cassette G2 Signaling. Oncogene 2013, 32, 2767–2781.
  57. Yan, H.C.; Fang, L.S.; Xu, J.; Qiu, Y.Y.; Lin, X.M.; Huang, H.X.; Han, Q.Y. The Identification of the Biological Characteristics of Human Ovarian Cancer Stem Cells. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 3497–3503.
  58. Raspollini, M.R.; Amunni, G.; Villanucci, A.; Baroni, G.; Taddei, A.; Taddei, G.L. C-KIT Expression and Correlation with Chemotherapy Resistance in Ovarian Carcinoma: An Immunocytochemical Study. Ann. Oncol. 2004, 15, 594–597.
  59. Li, S.-S.; Ma, J.; Wong, A.S.T. Chemoresistance in Ovarian Cancer: Exploiting Cancer Stem Cell Metabolism. J. Gynecol. Oncol. 2018, 29, e32.
  60. Mazzoldi, E.L.; Pavan, S.; Pilotto, G.; Leone, K.; Pagotto, A.; Frezzini, S.; Nicoletto, M.O.; Amadori, A.; Pastò, A. A Juxtacrine/Paracrine Loop between C-Kit and Stem Cell Factor Promotes Cancer Stem Cell Survival in Epithelial Ovarian Cancer. Cell Death Dis. 2019, 10, 412.
  61. Roy, L.; Bobbs, A.; Sattler, R.; Kurkewich, J.L.; Dausinas, P.B.; Nallathamby, P.; Cowden Dahl, K.D. CD133 Promotes Adhesion to the Ovarian Cancer Metastatic Niche. Cancer Growth Metastasis 2018, 11, 117906441876788.
  62. Klemba, A.; Purzycka-Olewiecka, J.K.; Wcisło, G.; Czarnecka, A.M.; Lewicki, S.; Lesyng, B.; Szczylik, C.; Kieda, C. Surface Markers of Cancer Stem-like Cells of Ovarian Cancer and Their Clinical Relevance. Współczesna Onkol. 2018, 2018, 48–55.
  63. Xiang, T.; Long, H.; He, L.; Han, X.; Lin, K.; Liang, Z.; Zhuo, W.; Xie, R.; Zhu, B. Interleukin-17 Produced by Tumor Microenvironment Promotes Self-Renewal of CD133+ Cancer Stem-like Cells in Ovarian Cancer. Oncogene 2015, 34, 165–176.
  64. Saha, S.; Parte, S.; Roy, P.; Kakar, S.S. Ovarian cancer stem cells: Characterization and role in tumorigenesis. Adv. Exp. Med. Biol. 2021, 1330, 151–169.
  65. Brescia, P.; Ortensi, B.; Fornasari, L.; Levi, D.; Broggi, G.; Pelicci, G. CD133 Is Essential for Glioblastoma Stem Cell Maintenance. Stem Cells 2013, 31, 857–869.
  66. Choi, Y.-J.; Ingram, P.N.; Yang, K.; Coffman, L.; Iyengar, M.; Bai, S.; Thomas, D.G.; Yoon, E.; Buckanovich, R.J. Identifying an Ovarian Cancer Cell Hierarchy Regulated by Bone Morphogenetic Protein 2. Proc. Natl. Acad. Sci. USA 2015, 112, E6882–E6888.
  67. Cioffi, M.; D’Alterio, C.; Camerlingo, R.; Tirino, V.; Consales, C.; Riccio, A.; Ieranò, C.; Cecere, S.C.; Losito, N.S.; Greggi, S.; et al. Identification of a Distinct Population of CD133+CXCR4+ Cancer Stem Cells in Ovarian Cancer. Sci. Rep. 2015, 5, 10357.
  68. Jang, J.-W.; Song, Y.; Kim, S.-H.; Kim, J.; Kim, K.M.; Choi, E.K.; Kim, J.; Seo, H.R. CD133 Confers Cancer Stem-like Cell Properties by Stabilizing EGFR-AKT Signaling in Hepatocellular Carcinoma. Cancer Lett. 2017, 389, 1–10.
  69. Wei, Y.; Jiang, Y.; Zou, F.; Liu, Y.; Wang, S.; Xu, N.; Xu, W.; Cui, C.; Xing, Y.; Liu, Y.; et al. Activation of PI3K/Akt Pathway by CD133-P85 Interaction Promotes Tumorigenic Capacity of Glioma Stem Cells. Proc. Natl. Acad. Sci. USA 2013, 110, 6829–6834.
  70. Yu, C.-C.; Hu, F.-W.; Yu, C.-H.; Chou, M.-Y. Targeting CD133 in the Enhancement of Chemosensitivity in Oral Squamous Cell Carcinoma-Derived Side Population Cancer Stem Cells. Head Neck 2016, 38, E231–E238.
  71. ZHU, Y.; YU, J.; WANG, S.; LU, R.; WU, J.; JIANG, B. Overexpression of CD133 Enhances Chemoresistance to 5-Fluorouracil by Activating the PI3K/Akt/P70S6K Pathway in Gastric Cancer Cells. Oncol. Rep. 2014, 32, 2437–2444.
  72. Abubaker, K.; Luwor, R.B.; Zhu, H.; McNally, O.; Quinn, M.A.; Burns, C.J.; Thompson, E.W.; Findlay, J.K.; Ahmed, N. Inhibition of the JAK2/STAT3 Pathway in Ovarian Cancer Results in the Loss of Cancer Stem Cell-like Characteristics and a Reduced Tumor Burden. BMC Cancer 2014, 14, 317.
  73. Jain, S.; Annett, S.L.; Morgan, M.P.; Robson, T. The Cancer Stem Cell Niche in Ovarian Cancer and Its Impact on Immune Surveillance. Int. J. Mol. Sci. 2021, 22, 4091.
  74. Davidson, B.; Holth, A.; Hellesylt, E.; Tan, T.Z.; Huang, R.Y.-J.; Tropé, C.; Nesland, J.M.; Thiery, J.P. The Clinical Role of Epithelial-Mesenchymal Transition and Stem Cell Markers in Advanced-Stage Ovarian Serous Carcinoma Effusions. Hum. Pathol. 2015, 46, 1–8.
  75. Davidson, B. CD24 Is Highly Useful in Differentiating High-Grade Serous Carcinoma from Benign and Malignant Mesothelial Cells. Hum. Pathol. 2016, 58, 123–127.
  76. Gao, M.-Q.; Choi, Y.-P.; Kang, S.; Youn, J.H.; Cho, N.-H. CD24+ Cells from Hierarchically Organized Ovarian Cancer Are Enriched in Cancer Stem Cells. Oncogene 2010, 29, 2672–2680.
  77. Jaggupilli, A.; Elkord, E. Significance of CD44 and CD24 as Cancer Stem Cell Markers: An Enduring Ambiguity. Clin. Dev. Immunol. 2012, 2012, 708036.
  78. Li, Y.; Sun, X.; Wang, H. Role of CD24 in Anoikis Resistance of Ovarian Cancer Cells. J. Huazhong Univ. Sci. Technol. 2015, 35, 390–396.
  79. Shen, Y.-A.; Wang, C.-Y.; Chuang, H.-Y.; Hwang, J.J.-J.; Chi, W.-H.; Shu, C.-H.; Ho, C.-Y.; Li, W.-Y.; Chen, Y.-J. CD44 and CD24 Coordinate the Reprogramming of Nasopharyngeal Carcinoma Cells towards a Cancer Stem Cell Phenotype through STAT3 Activation. Oncotarget 2016, 7, 58351–58366.
  80. Wang, Y.-C.; Wang, J.-L.; Kong, X.; Sun, T.-T.; Chen, H.-Y.; Hong, J.; Fang, J.-Y. CD24 Mediates Gastric Carcinogenesis and Promotes Gastric Cancer Progression via STAT3 Activation. Apoptosis 2014, 19, 643–656.
  81. Wang, X.; Zhang, Y.; Zhao, Y.; Liang, Y.; Xiang, C.; Zhou, H.; Zhang, H.; Zhang, Q.; Qing, H.; Jiang, B.; et al. CD24 Promoted Cancer Cell Angiogenesis via Hsp90-Mediated STAT3/VEGF Signaling Pathway in Colorectal Cancer. Oncotarget 2016, 7, 55663–55676.
  82. Kristiansen, G.; Denkert, C.; Schlüns, K.; Dahl, E.; Pilarsky, C.; Hauptmann, S. CD24 Is Expressed in Ovarian Cancer and Is a New Independent Prognostic Marker of Patient Survival. Am. J. Pathol. 2002, 161, 1215–1221.
  83. Keyvani, V.; Farshchian, M.; Esmaeili, S.-A.; Yari, H.; Moghbeli, M.; Nezhad, S.-R.K.; Abbaszadegan, M.R. Ovarian Cancer Stem Cells and Targeted Therapy. J. Ovarian Res. 2019, 12, 120.
  84. FU, J.; SHANG, Y.; QIAN, Z.; HOU, J.; YAN, F.; LIU, G.; DEHUA, L.; TIAN, X. Chimeric Antigen Receptor-T (CAR-T) Cells Targeting Epithelial Cell Adhesion Molecule (EpCAM) Can Inhibit Tumor Growth in Ovarian Cancer Mouse Model. J. Vet. Med. Sci. 2021, 83, 241–247.
  85. Wong, M.; Tan, N.; Zha, J.; Peale, F.V.; Yue, P.; Fairbrother, W.J.; Belmont, L.D. Navitoclax (ABT-263) Reduces Bcl-x L –Mediated Chemoresistance in Ovarian Cancer Models. Mol. Cancer Ther. 2012, 11, 1026–1035.
  86. Pastò, A.; Bellio, C.; Pilotto, G.; Ciminale, V.; Silic-Benussi, M.; Guzzo, G.; Rasola, A.; Frasson, C.; Nardo, G.; Zulato, E.; et al. Cancer Stem Cells from Epithelial Ovarian Cancer Patients Privilege Oxidative Phosphorylation, and Resist Glucose Deprivation. Oncotarget 2014, 5, 4305–4319.
  87. Jung, J.-G.; Stoeck, A.; Guan, B.; Wu, R.-C.; Zhu, H.; Blackshaw, S.; Shih, I.-M.; Wang, T.-L. Notch3 Interactome Analysis Identified WWP2 as a Negative Regulator of Notch3 Signaling in Ovarian Cancer. PLoS Genet. 2014, 10, e1004751.
  88. Testa, U.; Petrucci, E.; Pasquini, L.; Castelli, G.; Pelosi, E. Ovarian Cancers: Genetic Abnormalities, Tumor Heterogeneity and Progression, Clonal Evolution and Cancer Stem Cells. Medicines 2018, 5, 16.
  89. Alvero, A.B.; Fu, H.-H.; Holmberg, J.; Visintin, I.; Mor, L.; Marquina, C.C.; Oidtman, J.; Silasi, D.-A.; Mor, G. Stem-Like Ovarian Cancer Cells Can Serve as Tumor Vascular Progenitors. Stem Cells 2009, 27, 2405–2413.
  90. Yin, G.; Alvero, A.B.; Craveiro, V.; Holmberg, J.C.; Fu, H.-H.; Montagna, M.K.; Yang, Y.; Chefetz-Menaker, I.; Nuti, S.; Rossi, M.; et al. Constitutive Proteasomal Degradation of TWIST-1 in Epithelial–Ovarian Cancer Stem Cells Impacts Differentiation and Metastatic Potential. Oncogene 2013, 32, 39–49.
  91. Ayub, T.H.; Keyver-Paik, M.-D.; Debald, M.; Rostamzadeh, B.; Thiesler, T.; Schröder, L.; Barchet, W.; Abramian, A.; Kaiser, C.; Kristiansen, G.; et al. Accumulation of ALDH1-Positive Cells after Neoadjuvant Chemotherapy Predicts Treatment Resistance and Prognosticates Poor Outcome in Ovarian Cancer. Oncotarget 2015, 6, 16437–16448.
  92. Meyer, L.A.; Cronin, A.M.; Sun, C.C.; Bixel, K.; Bookman, M.A.; Cristea, M.C.; Griggs, J.J.; Levenback, C.F.; Burger, R.A.; Mantia-Smaldone, G.; et al. Use and Effectiveness of Neoadjuvant Chemotherapy for Treatment of Ovarian Cancer. J. Clin. Oncol. 2016, 34, 3854–3863.
  93. Bapat, S.A.; Mali, A.M.; Koppikar, C.B.; Kurrey, N.K. Stem and Progenitor-Like Cells Contribute to the Aggressive Behavior of Human Epithelial Ovarian Cancer. Cancer Res. 2005, 65, 3025–3029.
  94. Boesch, M.; Zeimet, A.G.; Reimer, D.; Schmidt, S.; Gastl, G.; Parson, W.; Spoeck, F.; Hatina, J.; Wolf, D.; Sopper, S. The Side Population of Ovarian Cancer Cells Defines a Heterogeneous Compartment Exhibiting Stem Cell Characteristics. Oncotarget 2014, 5, 7027–7039.
  95. House, C.D.; Jordan, E.; Hernandez, L.; Ozaki, M.; James, J.M.; Kim, M.; Kruhlak, M.J.; Batchelor, E.; Elloumi, F.; Cam, M.C.; et al. NFκB Promotes Ovarian Tumorigenesis via Classical Pathways That Support Proliferative Cancer Cells and Alternative Pathways That Support ALDH+ Cancer Stem–like Cells. Cancer Res. 2017, 77, 6927–6940.
  96. Bonneau, C.; Rouzier, R.; Geyl, C.; Cortez, A.; Castela, M.; Lis, R.; Daraï, E.; Touboul, C. Predictive Markers of Chemoresistance in Advanced Stages Epithelial Ovarian Carcinoma. Gynecol. Oncol. 2015, 136, 112–120.
  97. Condello, S.; Morgan, C.A.; Nagdas, S.; Cao, L.; Turek, J.; Hurley, T.D.; Matei, D. β-Catenin-Regulated ALDH1A1 Is a Target in Ovarian Cancer Spheroids. Oncogene 2015, 34, 2297–2308.
  98. Januchowski, R.; Wojtowicz, K.; Sterzyſska, K.; Sosiſska, P.; Andrzejewska, M.; Zawierucha, P.; Nowicki, M.; Zabel, M. Inhibition of ALDH1A1 Activity Decreases Expression of Drug Transporters and Reduces Chemotherapy Resistance in Ovarian Cancer Cell Lines. Int. J. Biochem. Cell Biol. 2016, 78, 248–259.
  99. Kuroda, T.; Hirohashi, Y.; Torigoe, T.; Yasuda, K.; Takahashi, A.; Asanuma, H.; Morita, R.; Mariya, T.; Asano, T.; Mizuuchi, M.; et al. ALDH1-High Ovarian Cancer Stem-Like Cells Can Be Isolated from Serous and Clear Cell Adenocarcinoma Cells, and ALDH1 High Expression Is Associated with Poor Prognosis. PLoS ONE 2013, 8, e65158.
  100. Meng, E.; Mitra, A.; Tripathi, K.; Finan, M.A.; Scalici, J.; McClellan, S.; da Silva, L.M.; Reed, E.; Shevde, L.A.; Palle, K.; et al. ALDH1A1 Maintains Ovarian Cancer Stem Cell-Like Properties by Altered Regulation of Cell Cycle Checkpoint and DNA Repair Network Signaling. PLoS ONE 2014, 9, e107142.
  101. Ottevanger, P.B. Ovarian Cancer Stem Cells More Questions than Answers. Semin. Cancer Biol. 2017, 44, 67–71.
  102. Pylväs-Eerola, M.; Liakka, A.; Puistola, U.; Koivunen, J.; Karihtala, P. Cancer Stem Cell Properties as Factors Predictive of Chemoresistance in Neoadjuvantly-Treated Patients with Ovarian Cancer. Anticancer Res. 2016, 36, 3425–3431.
  103. Motohara, T.; Katabuchi, H. Ovarian Cancer Stemness: Biological and Clinical Implications for Metastasis and Chemotherapy Resistance. Cancers 2019, 11, 907.
  104. Deng, S.; Yang, X.; Lassus, H.; Liang, S.; Kaur, S.; Ye, Q.; Li, C.; Wang, L.-P.; Roby, K.F.; Orsulic, S.; et al. Distinct Expression Levels and Patterns of Stem Cell Marker, Aldehyde Dehydrogenase Isoform 1 (ALDH1), in Human Epithelial Cancers. PLoS ONE 2010, 5, e10277.
  105. Liebscher, C.A.; Prinzler, J.; Sinn, B.V.; Budczies, J.; Denkert, C.; Noske, A.; Sehouli, J.; Braicu, E.I.; Dietel, M.; Darb-Esfahani, S. Aldehyde Dehydrogenase 1/Epidermal Growth Factor Receptor Coexpression Is Characteristic of a Highly Aggressive, Poor-Prognosis Subgroup of High-Grade Serous Ovarian Carcinoma. Hum. Pathol. 2013, 44, 1465–1471.
  106. Mizuno, T.; Suzuki, N.; Makino, H.; Furui, T.; Morii, E.; Aoki, H.; Kunisada, T.; Yano, M.; Kuji, S.; Hirashima, Y.; et al. Cancer Stem-like Cells of Ovarian Clear Cell Carcinoma Are Enriched in the ALDH-High Population Associated with an Accelerated Scavenging System in Reactive Oxygen Species. Gynecol. Oncol. 2015, 137, 299–305.
  107. Li, Y.; Chen, K.; Li, L.; Li, R.; Zhang, J.; Ren, W. Overexpression of SOX2 Is Involved in Paclitaxel Resistance of Ovarian Cancer via the PI3K/Akt Pathway. Tumor Biol. 2015, 36, 9823–9828.
  108. Wen, Y.; Hou, Y.; Huang, Z.; Cai, J.; Wang, Z. SOX2 Is Required to Maintain Cancer Stem Cells in Ovarian Cancer. Cancer Sci. 2017, 108, 719–731.
  109. Levasseur, D.N.; Wang, J.; Dorschner, M.O.; Stamatoyannopoulos, J.A.; Orkin, S.H. Oct4 Dependence of Chromatin Structure within the Extended Nanog Locus in ES Cells. Genes Dev. 2008, 22, 575–580.
  110. Peng, S.; Maihle, N.J.; Huang, Y. Pluripotency Factors Lin28 and Oct4 Identify a Sub-Population of Stem Cell-like Cells in Ovarian Cancer. Oncogene 2010, 29, 2153–2159.
  111. Di, J.; Duiveman-de Boer, T.; Zusterzeel, P.L.M.; Figdor, C.G.; Massuger, L.F.A.G.; Torensma, R. The Stem Cell Markers Oct4A, Nanog and c-Myc Are Expressed in Ascites Cells and Tumor Tissue of Ovarian Cancer Patients. Cell. Oncol. 2013, 36, 363–374.
  112. Muñoz-Galván, S.; Felipe-Abrio, B.; Verdugo-Sivianes, E.M.; Perez, M.; Jiménez-García, M.P.; Suarez-Martinez, E.; Estevez-Garcia, P.; Carnero, A. Downregulation of MYPT1 Increases Tumor Resistance in Ovarian Cancer by Targeting the Hippo Pathway and Increasing the Stemness. Mol. Cancer 2020, 19, 7.
  113. Fan, Q.; Cai, Q.; Xu, Y. FOXM1 Is a Downstream Target of LPA and YAP Oncogenic Signaling Pathways in High Grade Serous Ovarian Cancer. Oncotarget 2015, 6, 27688–27699.
  114. Nagaraj, A.B.; Joseph, P.; Kovalenko, O.; Singh, S.; Armstrong, A.; Redline, R.; Resnick, K.; Zanotti, K.; Waggoner, S.; DiFeo, A. Critical Role of Wnt/β-Catenin Signaling in Driving Epithelial Ovarian Cancer Platinum Resistance. Oncotarget 2015, 6, 23720–23734.
  115. Zhou, J.; Wang, Y.; Wang, Y.; Yin, X.; He, Y.; Chen, L.; Wang, W.; Liu, T.; Di, W. FOXM1 Modulates Cisplatin Sensitivity by Regulating EXO1 in Ovarian Cancer. PLoS ONE 2014, 9, e96989.
  116. Auersperg, N. The Stem-Cell Profile of Ovarian Surface Epithelium Is Reproduced in the Oviductal Fimbriae, with Increased Stem-Cell Marker Density in Distal Parts of the Fimbriae. Int. J. Gynecol. Pathol. 2013, 32, 444–453.
  117. Flesken-Nikitin, A.; Hwang, C.-I.; Cheng, C.-Y.; Michurina, T.V.; Enikolopov, G.; Nikitin, A.Y. Ovarian Surface Epithelium at the Junction Area Contains a Cancer-Prone Stem Cell Niche. Nature 2013, 495, 241–245.
  118. Toledo-Guzmán, M.E.; Bigoni-Ordóñez, G.D.; Hernández, M.I.; Ortiz-Sánchez, E. Cancer Stem Cell Impact on Clinical Oncology. World J. Stem Cells 2018, 10, 183–195.
  119. Wang, L.; Xu, T.; Cui, M. Are Ovarian Cancer Stem Cells the Target for Innovative Immunotherapy? OncoTargets Ther. 2018, 11, 2615–2626.
  120. Majzner, R.G.; Mackall, C.L. Tumor Antigen Escape from CAR T-Cell Therapy. Cancer Discov. 2018, 8, 1219–1226.
  121. Crupi, M.J.F.; Bell, J.C.; Singaravelu, R. Concise Review: Targeting Cancer Stem Cells and Their Supporting Niche Using Oncolytic Viruses. Stem Cells 2019, 37, 716–723.
More
Upload a video for this entry
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 637
Revisions: 2 times (View History)
Update Date: 03 Mar 2022
1000/1000
Hot Most Recent
Academic Video Service