Open Access

Cancer stem cells markers in ovarian cancer: Clinical and therapeutic significance (Review)

  • Authors:
    • Patrycja Królewska-Daszczyńska
    • Dominika Wendlocha
    • Marta Smycz-Kubańska
    • Sebastian Stępień
    • Aleksandra Mielczarek-Palacz
  • View Affiliations

  • Published online on: November 7, 2022     https://doi.org/10.3892/ol.2022.13585
  • Article Number: 465
  • Copyright: © Królewska-Daszczyńska et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ovarian cancer is a gynecological neoplasm that can be found in women, which, due to diagnostic difficulties, is often detected at advanced stages when treatment becomes a significant problem. Moreover, in a number of cases there is a cancer recurrence and resistance to standard chemotherapy treatment. It has been suggested that cancer stem cells (CSCs) that were not eradicated during therapy may be responsible for this. For this reason, effective therapeutic methods eliminating CSCs are being studied, such as therapy targeting CSCs markers. In addition, numerous studies have also drawn attention to the usefulness of CSCs markers in predicting disease progression and assessing patient's prognosis as well as their importance in the development of treatment resistance. The present review presented research on selected CSCs markers, which may be of significant prognostic and therapeutic importance in ovarian cancer.

Introduction

Ovarian cancer (OC) is one of the 10 most common types of cancer in women in the world. In 2020, ovarian cancer was ranked eighth in terms of incidence and mortality with over 313 000 new cases and over 207 000 deaths (1).

Among ovarian cancers (carcinomas), there are cancers originating from epithelial cells (the most common), germ cells and stromal cells (2). According to the World Health Organization (WHO) classification of female genital tumours from 2020, at least five main types of ovarian carcinomas are identified based on histopathology, immunoprofile and molecular analysis. Among them, high-grade serous carcinoma (HGSC) is the most common ovarian cancer accounting for about 70% of all ovarian carcinomas, the second most common histotype is endometroid carcinoma (EC, 10%), clear cell carcinoma (CCC, 6–10%), low-grade serous carcinoma (LGSC, 5%) and mucinous carcinoma (MC, 3–4%) (3).

An important diagnostic and therapeutic problem is a diagnosis of ovarian cancer patients at advanced stages with metastatic sites within the peritoneal cavity, retroperitoneum and even in distant organs (4). In such cases patients have a much lower chance of recovery and the five-year survival rate is less than 30% (5,6). Early diagnosis of ovarian cancer is difficult due to the lack of appropriate markers and definitive screening tools as well as non-specific symptoms accompanying this cancer. These include: bloating and abdominal pain, early satiety or fullness, changes in bowel habits or frequent urination. Women with those symptoms may seek medical help too late and even be treated without identifying the specific causes of their symptoms (7).

Treatment depends on the diagnosed stage of ovarian cancer (8). The standard therapy involves surgical treatment, which is maximal cytoreductive debulking, and the platinum-based chemotherapy (8,9). Unfortunately, tumours relapse in over 70% of cases, despite an initially good response to treatment by the majority of patients (4). In recent years, Poly(ADP-ribose) polymerase inhibitors (PARPi) have been approved for the treatment of ovarian cancer as drugs that maintain therapy following the completion of first-line platinum-based chemotherapy (10,11).

It is believed that cancer stem cells (CSCs) that have not been eliminated during treatment and are responsible for the development of resistance to chemotherapy and are able to replenish their population, may contribute to recurrence of the cancer, which might be even more aggressive (4). Hence, effective methods of cancer treatment based on the elimination of CSCs are being sought. High hopes are raised by CSCs-targeting therapies, which in combination with traditional methods of treatment may give a better therapeutic effect (12).

Cancer stem cells (CSCs)

Models of tumorigenesis

The two main models try to explain the origin, progression and heterogeneity of tumours: the stochastic model (or clonal evolution model) and the hierarchical (or CSC) model (1315). According to the stochastic model, each tumour cell is biologically homogeneous and has the same developmental potential as well as the ability to promote tumour progression (16,17). This model assumes that the acquisition of oncogenic mutations in normal differentiated somatic cells results in hyperplasia and contributes to clonal expansion (14,17). The accumulation of genetic and epigenetic alterations in cells may increase tumour aggressiveness, invasiveness and treatment resistance which in turn leads to tumour progression and increases tumour heterogeneity (13).

The hierarchical model states that only a distinct population of cancer cells features tumorigenic potential-these cells are referred to as cancer stem cells (CSCs). According to this model, tumour initiation starts when a normal stem cell escapes regulation and becomes cancer stem cell-the first abnormal cell assumed to be the cell-of-origin (14,17). Moreover, this model says that there is a differentiation hierarchy of cells in tumour that includes CSCs responsible for maintaining the whole populations of cells in tumour (13,15,16,18).

However, there is an alternative model of cellular plasticity that combines these two models by assuming that cancer cells can interconvert between stem cell and differentiated states (13). Cell dedifferentiation capacity may be inherited (hierarchical model) or acquired through mutations (stochastic model) (14). According to the plasticity model, differentiated tumour cells can reacquire stem cell characteristics by intrinsic processes of these cells and/or stimuli within the tumour microenvironment (13).

The origin of cancer stem cells

Cancer stem cells (CSCs) represent a small subpopulation of cells in tumour mass (19). The origin of these cells has still not been clearly elucidated. One of the hypotheses suggests that CSCs originate from normal adult stem cells that have acquired epigenetic and genetic changes (20). On the other hand, CSCs may derive from mature differentiated cells through various mechanisms, including genomic instability, horizontal gene transfer and microenvironmental changes (21). A differentiated cancer cells may de-differentiate into CSCs in response to different factors, such as stress and hypoxia, wounding or ionizing radiation (22). Various studies suggest that the epithelial-mesenchymal transition (EMT) is involved in dedifferentiation and cells that have undergone this process exhibit a more CSC-like phenotype allowing them to self-renew and differentiate into all cell types in the tumour (23). It is believed that CSCs can also arise as a result of cell fusion and metabolic reprogramming of non-CSCs into CSCs during the cancer development (21,22).

Characteristics of cancer stem cells

The assumption that CSCs originate from normal stem cells (NSCs) that have accumulated transforming mutations may be supported by the fact that these cells share many features (13). Both CSCs and NSCc have the capacity for self-renewal through mitotic divisions. Symmetric divisions give rise to two sister stem cells, while asymmetric divisions give rise to one daughter stem cell and one differentiated cell (24). The ability of CSCs to divide asymmetrically enables these cells to both self-renew their population and initiate a neoplastic process (25). However, NSCs are able to control and regulate self-renewal, while CSCs have lost this capacity (26). Moreover, CSCs and NSCs are regulated by similar signalling pathways such as Wnt, Hedgehog or Notch (26). Most of these pathways are essential for stemness properties of NSCs, such as the ability to self-renew, differentiate, proliferate and develop various organs during embryogenesis. However, genetic mutations and epigenetic changes may cause dysregulation of these pathways in the CSCs, leading to uncontrolled self-renewal and impaired differentiation of these cells (27).

Both CSCs and NSCs have the ability to differentiate into multiple progenitor cell types. However, CSCs can replicate and differentiate in an uncontrolled manner into populations of molecularly and phenotypically altered progenitor cells that may have limitless proliferative and survival potential with more plasticity than progeny of NSCs (13). Additionally, CSCs and NSCs possess high telomerase activity that prolongs their life span, express similar surface receptors and can stimulate angiogenesis (28).

In addition to impaired self-renewal and differentiation abilities, CSCs also have other characteristics that distinguish them from NSCs, such as the ability to form tissues and organs. NSCs develop through organogenesis to form internal organs, while CSCs have tumorigenic properties and form tumour tissues. Moreover, NSCs have normal karyotyping, whereas CSCs have abnormal karyotyping with genetic alterations (28). Characteristics of CSCs are summarised in Fig. 1.

The importance of tumour microenvironment for CSCs

Stem cells division and differentiation take place in a specialised microenvironment (the niche) that regulates self-renew of these cells through cell-cell communication or secretion of paracrine factors (28,29). CSCs are population of cancer cells within the cancer microenvironment that consists of various cells, including cancer-associated fibroblasts (CAFs), mesenchymal stem cells (MSCs), endothelial cells (ECs) and immune cells (the macrophages, T-cells and natural killer (NK) cells, factors secreted by these cells, such as cytokines and growth factors as well as the extracellular matrix (ECM) (28,29). ECM is a noncellular component of tumour microenvironment composed of glycosaminoglycans, collagens, metalloproteases, hyaluronic acid, polysaccharides, glycoproteins, proteoglycans and other proteins (30).

The components of tumour microenvironment provide ideal conditions for maintaining the properties of CSCs, such as self-renewal, proliferation and differentiation as well as generation of heterogeneous cancer population (30). Moreover, the tumour niche supports initiation, growth, invasion and metastasis of tumour cells and also plays an important role in therapy resistance, mostly by supporting stem-related signaling pathway maintenance in CSCs (29). The CSCs are surrounded by cancer niche cells, which secrete factors promoting survival and plasticity of CSCs, as well as increasing drug resistance. Additionally, ECM is a physical barrier that protects CSCs from chemotherapeutic agents (29).

However, the relation between CSCs and their niche can be bidirectional. It is suggested that CSCs may promote the recruitment and activation of niche components by producing factors such as proinflammatory cytokines and chemokines. Furthermore, it has been shown that CSCs may differentiate into functional ECs, which in turn may transdifferentiate into MSCs (29).

Within tumours CSCs are usually located near hypoxic regions. Hypoxia plays a role in the maintenance of CSCs characteristics. It is also involved in chemo- and radioresistance (29). In hypoxic conditions CSCs produce vascular endothelial growth factor (VEGF), which further induces angiogenesis (14).

Methods of CSCs identification and isolation

The specific properties of CSCs are used in methods of isolating these cells from a tumour mass or cell culture (31). There are several in vitro assays to identify CSCs, such as the detection of surface markers, assessment of the activity of aldehyde dehydrogenase (ALDH)-Aldefluor assay, sphere-forming assay or Hoechst dye exclusion assay (32). Methods that use the expression of surface markers of CSCs to isolate these cells include fluorescence-activated cell sorting (FACS), based on flow cytometry and using fluorescently labeled antibodies, and magnetic activated cell sorting (MACS) using antibodies coupled with superparamagnetic nanoparticles (31). Moreover, polymerase chain reaction analysis is also used to isolate CSCs by identifying the markers expressed on the cell surface (21,32).

On the other hand, CSCs may also be identified by the flow cytometry-based ALDEFLUOR assay which measure the activity of intracellular marker-ALDH, which is increased in these cells (3133).

The ability of CSCs to form spheres is also used to identify and isolate these cells. Culturing cells harvested from tumour specimens in a serum-free medium supplemented with basic fibroblast growth factor (bFGF) and epithelial growth factor (EGF) results in the formation of non-adherent spheres by immature cells (21,31,32). Another method of isolating CSCs is based on the ability of cells, termed as the side population (SP), to export a fluorescent dye, such as Hoechst 33342 or Rhodamine 123, by ABC transporters (21,31,32). However, some ABC transporters expressed on CSCs, such as ABCB1 or ABCG2, are also expressed on non-CSCs (21). Moreover, this method is limited by the toxicity of the dyes (21,32).

When identifying and isolating CSCs, it should be kept in mind, that CSCs share many features with NSCs and are similar e.g., in the expression of specific surface markers or the utilization of common signaling pathways. In contrast, when CSCs are transplanted into animals they can form a tumour, while NSCs do not have this ability (32). In the method of isolating CSCs in vivo, that is serial transplantation assay in animal model, tumour cells are transplanted into immunocompromised mouse. Such in vivo assays are regarded as the gold standard in the identification of CSCs (32).

CSCs in ovarian cancer

Cancer stem cells (CSCs) are a small subpopulation of cancer cells within ovarian tumour tissue (24). Bapat et al (34) were the first to confirm the existence of cells with the characteristics of NSCs in ovarian cancer, which are capable of driving tumorigenesis. Ovarian CSCs are thought to be responsible for tumour growth, metastasis and recurrence, as well as resistance to standard treatments such as chemotherapy (35). It is believed that the involvement of CSCs in metastasis in ovarian cancer is related to their ability to resist anoikis, which allows them to survive in non-adherent conditions and then to adhere in other than primary locations and create secondary tumours there (36). Moreover, the metastasis formation may be influenced by the ability of CSCs to undergo the process of EMT which is an example of the plasticity of these cells (36).

An important therapeutic problem in patients with ovarian cancer is the frequent recurrence of the disease, even if an initial response to the treatment is promising. Moreover, patients may become resistant to chemotherapy, which results in treatment failure or even death (37). The mechanisms underlying the development of chemoresistance are not entirely clear, but it is suggested that CSCs may play a role in cancer recurrence following chemotherapy. There are several mechanisms implicated in chemoresistance of ovarian CSCs, including increased drug effects, CSCs quiescence (essential for self-renewal function), enhanced DNA repair, autophagy, etc. (37).

Various markers are used to identify CSCs in ovarian cancer. However, due to a tumour heterogeneity it is difficult to describe ovarian CSCs phenotype. Among the characteristic markers of CSCs in ovarian cancer, there are: CD133, CD44, CD24, CD117, or ALDH1 (4,38). Recent findings indicate that some markers may be of diagnostic and prognostic importance in ovarian cancer (38). In addition, scientists' attention is drawn to the use of CSCs markers in targeted personalised therapies (39).

Clinical significance of CSCs markers in ovarian cancer

This review presents selected CSCs markers used in ovarian cancer research with particular emphasis on their prognostic value and association with chemoresistance in this cancer.

CD133

CD133 is one of the most well-known markers of CSCs, used to isolate and study these cells in different types of cancer, including ovarian cancer (40). Zhou et al (41) performed meta-analysis of eight studies including a total of 1051 women with ovarian cancer to investigate the association between the expression of CD133 and clinicopathological outcomes as well as to determine the prognostic value of CD133 in ovarian cancer. Their analysis showed that the presence of CD133 expression was highly correlated with poor two-year overall survival (OS), which may indicate the prognostic importance of this marker related to the worse prognosis in patients with ovarian cancer. Moreover, they showed that the expression of CD133 correlated with tumour stage, but was not associated with other clinical parameters, such as patients' age, tumour grade, histological type and response to treatment (41). Another meta-analysis performed by Tao et al (42) indicated that the expression of CD133 correlated with FIGO stage and was statistically associated with tumour differentiation grade, which may suggest the involvement of CD133 in the malignant progression of ovarian cancer.

Different results were obtained in the study of Onisim et al (43), who did not observe an association between the expression of CD133 and progression free survival (PFS) or OS in patients with serous ovarian carcinoma. They also found that the expression of CD133 in tumour cells was not significantly associated with clinicopathological parameters, such as age, serum CA125, peritoneal carcinomatosis, malignant ascites or tumour grade (43).

In the study by Ruscito et al (44) it was shown that there was a significant shift from higher frequency of CD133+ cells in patients with primary high-grade serous ovarian cancer (HGSOC) to lower levels in the paired recurrent samples. Moreover, all primary ovarian cancer CD133+ patients were diagnosed at FIGO III/IV stage and had significantly worse progression-free survival (PFS) as well as OS (44). In turn, in the study by Steg et al (45), who examined matched primary and recurrent tumour pairs from patients with high grade ovarian adenocarcinomas, it was shown that the average number of CD133-positive cells was significantly higher in the samples of recurrent tumours than in primary tumours. Moreover, the expression of CD133 was significantly increased in tumours collected from recurrent platinum-resistant patients (45). Liu et al (46) showed that the absence of CD133 expression in patients with primary epithelial ovarian cancer was significantly associated with high platinum sensitivity in patients with and without central nervous system (CNS) metastases. Their results also indicated a positive association between the expression of CD133 in primary tumours and increased risk of CNS metastases (46). The association between the expression of CD133 and chemoresistance was also shown in another study (47).

The presented results may indicate a relationship between the expression of CD133 and chemoresistance in women with ovarian cancer and the potential use of this marker in personalized targeted therapy.

CD44

The prognostic value and clinical significance of CSCs surface marker CD44 in patients with ovarian cancer is controversial. Different authors in their reviews point out that there are some conflicted data on CD44 expression and its correlation with prognosis in ovarian cancer (48,49).

The meta-analysis performed by Lin and Ding (50) included 18 studies conducted in total on over 2,000 patients with ovarian cancer. Their study revealed that the expression of CD44 in ovarian cancers was significantly associated with a high TMN stage and with a poor five-year OS, while was not significantly correlated with disease-free survival (DFS). They also showed that there was no significant correlation between the expression of CD44 and tumour grade, lymphatic metastasis, patients' age, residual tumour size, ascites volume as well as response to chemotherapy (50). Another meta-analysis conducted by Tao et al (42) showed that overexpression of isoform CD44s was associated with poor OS and worse DFS as well as with chemotherapy resistance in ovarian cancer patients. However, there was no association between overexpression of isoform CD44v6 and poor OS (42).

In the studies of Zhou et al (51) it was found that in patients with ovarian cancer the high expression of CD44 was associated with higher histological grade and more advanced FIGO stage. Moreover, they showed that high expression of CD44 was significantly associated with worse OS and DFS suggesting that CD44 may be a potential prognostic marker (51). High expression of CD44 has also been demonstrated in the samples of chemotherapy resistant epithelial ovarian cancer tissue, which may indicate the usefulness of this marker in targeted therapy (52).

Zhu et al (53) showed that CD44/myeloid differentiation factor 88 (MyD88) co-expression in patients with epithelial ovarian carcinoma (EOC) was associated with tumour progression, metastasis and recurrence. Moreover, the authors' findings suggest that CD44/MyD88 co-expression is an independent prognostic factor related to poor DFS and OS (53).

The researchers' attention is also focused on the clinical significance of CD44 variant 6 (CD44v6). It was found that CD44v6 is highly expressed in ovarian cancer patients, suggesting that CD44v6 may promote incidence and progression of this cancer (54). In addition, the study by Tjhay et al (55) showed that an increased number of CD44v6-positive cancer cells in primary tumours was associated with a shortened OS in patients with advanced epithelial ovarian cancer (stage III–IV). The authors also found that CD44v6-positive cancer cells show metastatic potential and they are associated with tumour chemoresistance (55). Motohara et al (56) found that the expression of CD44v6 was an independent risk factor for distant metastatic recurrence in patients with ovarian cancer. Moreover, increased expression of this marker in primary ovarian tumours was associated with shorter OS (56).

ALDH1

Different studies results indicate a relationship between high expression of ALDH1 and poor prognosis and clinical outcome in patients with ovarian cancer (5759). However, there is also a study in which the expression of ALDH1 was associated with favourable prognosis in ovarian cancer (60). The long-term follow-up retrospective study by Huang et al (61) showed that high expression of ALDH1 in ovarian cancer cells was associated with histological subtypes, early FIGO stage, well differentiation grade and better survival. However, in multivariate analysis, the expression of ALDH1 in tumour cells was not an independent risk factor for OS. Their study revealed that high expression of ALDH1 in ovarian cancer cells may portends favourable prognosis (61).

The clinicopathological characteristics and prognostic significance of ALDH1 in ovarian cancer were evaluated by Zhao et al (62) in a meta-analysis of 18 studies including over 2 500 patients. Their results indicated that elevated expression of ALDH1 was significantly associated with poor OS but not with DFS. They also found that ALDH1 was most frequently elevated in patients with poor clinicopathological characteristics and was associated with FIGO stage, lymph node metastasis and distant metastasis (62). Another meta-analysis, published in the same year, showed that overexpression of ALDH1 was correlated with poor OS as well as with worse DFS (42).

Ayub et al (63) demonstrated that in patients with advanced epithelial ovarian cancer the enrichment of ALDH1 expression after treatment was associated with poor response to chemotherapy. Another study showed that the expression of isoform ALDH1A1 was associated with poor response to platinum-based therapy in patients with high-grade ovarian serous carcinoma (64).

CD133/ALDH1

The study conducted by Ricci et al (65) found that neither CD133 expression nor ALDH enzymatic activity were correlated with response to therapy, PFS and OS in ovarian cancer. The authors suggest that those markers do not provide additional predictive/prognostic information in ovarian cancer patients (65). On the other hand, Silva et al (66) showed that the presence of ALDH+CD133+ cells in debulked primary tumour specimens correlated with reduced disease-free survival and OS in ovarian cancer patients. Similarly, in the aforementioned study by Ruscito et al (44) it was found that the co-expression of CD133/ALDH1 in patients with primary HGSOC, rather than the expression of a single marker, was an independent prognostic factor associated with poor PFS and OS.

CD24

CD24 is a sialoglycoprotein that has been identified as an independent prognostic marker of survival in patients with ovarian cancer (67). CD24 is localised in lipid rafts through its glycosylphosphatidylinositol anchor, but also its diffuse cytoplasmic accumulation is observed in cancer cells (67). Kristiansen et al (68) found that cytoplasmic expression of CD24 was a prognostic factor for poor survival in ovarian cancer, while membranous expression had no influence on patients survival. In the study by Nakamura et al (69) it was shown that the expression of CD24 was significantly associated with progression-free survival and overall survival in patients with ovarian cancer. Moreover, the authors found that the expression of CD24 was correlated with the FIGO stage and the presence of peritoneal and lymph node metastasis. Additionally, CD24 induced the EMT phenomenon in ovarian cancer, which was involved in resistance to chemotherapy (69). Also, according to Soltész et al (70) high expression of CD24 in serous ovarian cancer patients' tissue samples was associated with advanced FIGO stages.

CD117

Meta-analysis conducted by Yang et al (71) included seven studies enrolling over 1200 patients with epithelial ovarian cancer. They showed that the expression of CD117 was significantly correlated with FIGO stage, histological type, tumour differentiation grade and age. Moreover, high expression of CD117 was significantly correlated with poor OS, but there was no statistically significant association between this marker expression and DFS (71). The study by Luo et al (72) showed that the expression of CD117 is also statistically correlated with response to chemotherapy and CD117+ patients were less sensitive to chemotherapy than CD117 patients.

CD105 (endoglin)

It has been shown that the expression of CD105 was associated with poor survival in patients with ovarian cancer (73). Furthermore, it is suggested that CD105 plays a role in ovarian cancer metastasis (74). Zhang et al (52) found that moderately and highly differentiated ovarian cancer tissue samples exhibited decreased expression of CD105 compared with poorly differentiated samples. Moreover, early-stage (I and II) ovarian cancer tissue samples exhibited decreased expression of CD105 compared with advanced stage (III) samples. Additionally, there were increased protein expression of CD105 in drug-resistant epithelial ovarian cancer tissue samples compared with drug-sensitive samples (52). Ziebarth et al (75) found that inhibition of CD105 increased cisplatin sensitivity in epithelial ovarian cancer.

CD106 (VCAM-1)

The study conducted by Huang et al (76) showed that overexpression of VCAM-1 in high grade serous ovarian cancer cells was associated with poor prognosis. Moreover, the authors found that high expression of VCAM-1 was related to advanced age at diagnosis and poor response to surgery and chemotherapy. Their data suggest that VCAM-1 may be a prognostic factor and novel therapeutic target for ovarian cancer (76). Scalici et al (77) found that mesothelium expression of VCAM-1 in patients with epithelial ovarian cancer was associated with shorter PFS and OS. In the study by Zhang et al (52) it was shown that high expression of CD106 was associated with drug resistance.

EpCAM

The study by Tayama et al (78) showed that an increased expression of EpCAM was associated with poor prognosis in patients with ovarian cancer and correlated with shortened PFS and OS. Moreover, they also found that EpCAM was associated with chemoresistance to platinum-based chemotherapy (78). Spizzo et al (79) also showed that overexpression of EpCAM was significantly correlated with decreased OS in patients with epithelial ovarian cancer. However, different results were obtained by Woopen et al (80) who showed that epithelial ovarian cancer patients with overexpression of EpCAM had better prognosis than patients with a weak or no expression of this marker. EpCAM overexpression was associated with a more favourable OS, better PFS and high response to platinum-based chemotherapy (80).

SOX2

The association between the expression of SOX2 and poor prognosis in ovarian cancer was shown by Zhang et al (81). They found that the expression of SOX2 was associated with decreased DFS durations, but there was no association between SOX2 expression and OS. Moreover, there was significant association between the expression of SOX2 and high-grade serous carcinoma. Their data showed that there was no significant correlation between the expression of SOX2 and response to chemotherapy (81). Bååth et al (82) found that within the group of patients with non-radical debulking surgery, there were shorter OS and PFS for patients with SOX2-positive tumours. Moreover, Li et al (83) investigated that the SOX2 was overexpressed in paclitaxel-resistant cells.

Nestin

The study by Onisim et al (43) showed that the expression of nestin in tumour cells was associated with poorer PFS and OS in patients with ovarian cancer. In another study by Czekierdowski et al (84) it was found that in high grade serous ovarian cancer patients with high expression of nestin had worse OS and DFS rates than patients with low expression of nestin. Qin et al (85) found that in serous ovarian cancer nestin-positive patients had significantly shorter OS. Moreover, overexpression of nestin was associated with the cisplatin-based chemotherapy resistance (85).

SSEA1

SSEA1 was studied by Davidson et al (86) in metastatic high grade serous carcinoma. They found that higher expression of SSEA1 was significantly associated with shorter OS and poorer PFS. Moreover, SSEA1 was significantly overexpressed in post-chemotherapy effusions compared with pre-chemotherapy specimens tapped at diagnosis (86).

Thy-1 (CD90)

In the study conducted by Chen et al (87) it was found that the expression of CD90 was significantly decreased in ovarian tumour tissues and lower expression of CD90 was correlated with poor survival rate. Moreover, the authors investigated that CD90 decreased the expression of other CSCs markers, such as CD133 and CD24 (87). Different results were obtained by Connor et al (88), who found that the expression of Thy-1 (CD90) was associated with poorer clinical outcome in women with ovarian cancer. Their study showed that in high expression of Thy-1 was associated with poorer OS and PFS in women with serous ovarian cancer, while the expression of Thy-1 in endometroid ovarian cancer was associated only with poorer PFS. Moreover, they demonstrated that the expression of Thy-1 is associated with increased proliferative and self-renewal capacity of ovarian cancer cells (88).

All CSCs markers selected for this review are also presented in Fig. 2, according to their surface or intracellular presence. Additionally, association of chemoresistance with type of ovarian cancer is presented in Table I.

Table I.

Association of chemoresistance with type of ovarian cancer.

Table I.

Association of chemoresistance with type of ovarian cancer.

CSCs marker(Refs.)Type of ovarian cancer
CD133Steg et al (45)High grade ovarian adenocarcinomas
Liu et al (46)Epithelial ovarian cancer (serous, mucinous, endometroid, clear cell, mixed epithelial, undifferentiated) with and without CNS metastases
Liu et al (47)Ovarian cancer cell lines (CSC-like SKOV3 spheres, CSC-like IGROV1-spheres)
CD44Tao et al (42)Meta-analysis (patients with different types of ovarian cancer)
Zhang et al (52)Epithelial ovarian cancer, OVCAR3 cell line, PTX-resistant OC3/TAX300 cells
ALDH1Ayub et al (63)Epithelial ovarian cancer
Roy et al (64)High grade serous ovarian cancer
CD24Nakamura et al (69)Caov-3 (human ovarian mucinous adenocarcinoma cancer cell line)
CD117Luo et al (72)Ovarian serous adenocarcinoma
CD105Zhang et al (52)Epithelial ovarian cancer, OVCAR3 cell line, PTX-resistant OC3/TAX300 cells
Ziebarth et al (75)Epithelial ovarian cancer (cell lines)
CD106Zhang et al (52)Epithelial ovarian cancer, OVCAR3 cell line, PTX-resistant OC3/TAX300 cells
Huang et al (76)High grade serous ovarian cancer
EpCAMTayama et al (78)Epithelial ovarian cancer-tissue samples (serous, clear cell, endometroid, mucinous, other). Human ovarian cancer cell lines. Animal study
SOX2Li et al (83)Tissue specimens (patients diagnosed with ovarian cancer). SKOV3 and SKOV3/TAX cells (paclitaxel-resistant human ovarian adenocarcinoma cell line)
NestinQin et al (85)Serous ovarian cancer

[i] SSEA1, stage-specific embryonic antigen-1; EpCAM, epithelial cell adhesion molecule; ALDH1, aldehyde dehydrogenase 1; SOX2, sex-determining region Y-box 2.

Therapeutic importance of CSCs markers

Targeting CSCs markers remains a challenge. Most of currently known CSCs surface markers are also expressed on normal stem cells (embryonic and/or adult stem cells) and they are rarely or considerably expressed on various normal tissue cells (89,90). Markers CD133, CD24, CD117, CD90 are expressed on the surface of human embryonic stem cells (hESC) and adult stem cells (89). CD133 is also expressed in epithelial and non-epithelial cells as well as it can be found in many cancers such as breast, lung, ovarian, melanoma, pancreatic, colon, prostate, glioma and hepatocellular cancers (91). EpCAM has been used as an undifferentiated hESC marker and it is also expressed on some normal epithelial cells (89). SSEA-1 is a surface marker for neural stem cells and is related to lung and renal tumours (89). Marker CD44 has been detected in human hematopoietic, mesenchymal and adipose-derived stem cells. Moreover, it is ubiquitously expressed in many normal tissue cells (89). CD106 is expressed by mesenchymal and neural stem cells (52).

Monoclonal antibodies (mAb) that target specific CSCs markers are a promising therapeutic option. Yang et al (92) reviewed agents that have been used to target CSCs markers in recent years. For example, anti-CD44mAb (bivatuzumab) was used for the treatment of head and neck squamous cell carcinoma, and EpCAM antibody (adecatumumab) was used in patients with hormone-resistant prostate cancer (92). CSCs markers could also be a target for chimeric antigen receptor (CAR)-T cell therapy (93,94).

Conclusion

The role of CSCs in the development and progression of ovarian cancer as well as their association with therapy resistance is still the subject of numerous studies. Unfortunately, due to the heterogeneity and plasticity of these cells, finding a specific phenotype of CSCs that would allow for their better identification remains a challenge. Moreover, identification of such phenotypes could also be helpful in developing new diagnostic and therapeutic strategies in ovarian cancer.

Despite the ambiguous results, the usefulness of CSCs markers in the assessment of prognosis and their relationship with the development of chemoresistance in ovarian cancer patients has been demonstrated. In our review we found that the expression of ovarian CSCs markers CD133, CD44, ALDH1, CD24, CD117, CD105, CD106, SOX2, Nestin and SSEA1 may have a prognostic significance associated with poor prognosis for patients with ovarian cancer. Moreover, the expression of CD133, CD44, ALDH1, CD24, CD117, CD105, CD106, EpCAM, SOX2 and Nestin could be associated with resistance to chemotherapy in ovarian cancer. However, it is advisable to perform further studies that will allow the use of CSCs markers especially in the aspect of tumour recurrence and in the development of personalised targeted therapies.

Acknowledgements

Not applicable.

Funding

The present review was funded by the Medical University of Silesia in Katowice, Poland (grant no. PCN-1-069/K/1/O).

Availability of data and materials

Not applicable.

Authors' contributions

AMP and PKD conceptualised this review. PKD, DW, MSK and SS searched and selected literature. AMP and PKD prepared and reviewed the original draft. PKD, DW, MSK and SS designed the table and figures. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Kroeger PT Jr and Drapkin R: Pathogenesis and heterogeneity of ovarian cancer. Curr Opin Obstet Gynecol. 29:26–34. 2017. View Article : Google Scholar : PubMed/NCBI

3 

De Leo A, Santini D, Ceccarelli C, Santandrea G, Palicelli A, Acquaviva G, Chiarucci F, Rosini F, Ravegnini G, Pession A, et al: What is new on ovarian carcinoma: Integrated morphologic and molecular analysis following the new 2020 World Health Organization classification of female genital tumors. Diagnostics (Basel). 11:6972021. View Article : Google Scholar : PubMed/NCBI

4 

Kenda Suster N and Virant-Klun I: Presence and role of stem cells in ovarian cancer. World J Stem Cells. 11:383–397. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Nebgen DR, Lu KH and Bast RC Jr: Novel approaches to ovarian cancer screening. Curr Oncol Rep. 21:752019. View Article : Google Scholar : PubMed/NCBI

6 

Kujawa KA and Lisowska KM: Ovarian cancer-from biology to clinic. Postepy Hig Med. Dosw (online). 69:1275–1290. 2015.(In Polish). View Article : Google Scholar : PubMed/NCBI

7 

Stewart C, Ralyea C and Lockwood S: Ovarian cancer: An integrated review. Semin Oncol Nurs. 35:151–156. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Ottevanger PB: Ovarian cancer stem cells more questions than answers. Semin Cancer Biol. 44:67–71. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Cortez AJ, Tudrej P, Kujawa KA and Lisowska KM: Advances in ovarian cancer therapy. Cancer Chemother Pharmacol. 81:17–38. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Valabrega G, Scotto G, Tuninetti V, Pani A and Scaglione F: Differences in PARP inhibitors for the treatment of ovarian cancer: Mechanisms of action, pharmacology, safety, and efficacy. Int J Mol Sci. 22:42032021. View Article : Google Scholar : PubMed/NCBI

11 

Liu L, Cai S, Han C, Banerjee A, Wu D, Cui T, Xie G, Zhang J, Zhang X, McLaughlin E, et al: ALDH1A1 contributes to PARP inhibitor resistance via enhancing DNA repair in BRCA2−/− ovarian cancer cells. Mol Cancer Ther. 19:199–210. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Pan Y, Ma S, Cao K, Zhou S, Zhao A, Li M, Qian F and Zhu C: Therapeutic approaches targeting cancer stem cells. J Cancer Res Ther. 14:1469–1475. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Rich JN: Cancer stem cells: Understanding tumour hierarchy and heterogeneity. Medicine (Baltimore). 95 (Suppl 1):S2–S7. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Plaks V, Kong N and Werb Z: The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 16:225–238. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Afify SM and Seno M: Conversion of stem cells to cancer stem cells: Undercurrent of cancer initiation. Cancers (Basel). 11:3452019. View Article : Google Scholar : PubMed/NCBI

16 

Szaryńska M and Kmieć Z: The role of cancer stem cells in pathogenesis and therapy of cancer. Forum Med Rodz. 5:47–56. 2011.

17 

Melzer C, von der Ohe J, Lehnert H, Ungefroren H and Hass R: Cancer stem cell niche models and contribution by mesenchymal stroma/stem cells. Mol Cancer. 16:282017. View Article : Google Scholar : PubMed/NCBI

18 

Wang T, Shigdar S, Gantier MP, Hou Y, Wang L, Li Y, Shamaileh HA, Yin W, Zhou SF, Zhao X and Duan W: Cancer stem cell targeted therapy: Progress amid controversies. Oncotarget. 6:44191–44206. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Markowska J, Kojs Z and Twardawa D: Cancer stem cells in targeted therapy. Curr Gynecol Oncol. 16:96–100. 2018. View Article : Google Scholar

20 

Islam F, Qiao B, Smith RA, Gopalan V and Lam AK: Cancer stem cell: fundamental experimental pathological concepts and updates. Exp Mol Pathol. 98:184–191. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Atashzar MR, Baharlou R, Karami J, Abdollahi H, Rezaei R, Pourramezan F and Zoljalali Moghaddam SH: Cancer stem cells: A review from origin to therapeutic implications. J Cell Physiol. 235:790–803. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Nimmakayala RK, Batra SK and Ponnusamy MP: Unraveling the journey of cancer stem cells from origin to metastasis. Biochim Biophys Acta Rev Cancer. 1871:50–63. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Wang H and Unternaehrer JJ: Epithelial-mesenchymal transition and cancer stem cells: At the crossroads of differentiation and dedifferentiation. Dev Dyn. 248:10–20. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Bar JK, Grelewski P, Lis-Nawara A and Drobnikowska K: The role of cancer stem cells in progressive growth and resistance of ovarian cancer: True or fiction? Postepy Hig Med Dosw (Online). 69:1077–1086. 2015.(In Polish). PubMed/NCBI

25 

Huang R and Rofstad EK: Cancer stem cells (CSCs), cervical CSCs and targeted therapies. Oncotarget. 8:35351–35367. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Al-Alem LF, Pandya UM, Baker AT, Bellio C, Zarrella BD, Clark J, DiGloria CM and Rueda BR: Ovarian cancer stem cells: What progress have we made? Int J Biochem Cell Biol. 107:92–103. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Lathia JD and Liu H: Overview of cancer stem cells and stemness for community oncologists. Target Oncol. 12:387–399. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Wan Kamarul Zaman WS, Nurul AA and Nordin F: Stem cells and cancer stem cells: The Jekyll and Hyde scenario and their implications in stem cell therapy. Biomedicines. 9:12452021. View Article : Google Scholar : PubMed/NCBI

29 

Prieto-Vila M, Takahashi RU, Usuba W, Kohama I and Ochiya T: Drug resistance driven by cancer stem cells and their niche. Int J Mol Sci. 18:25742017. View Article : Google Scholar : PubMed/NCBI

30 

Bighetti-Trevisan RL, Sousa LO, Castilho RM and Almeida LO: Cancer stem cells: Powerful targets to improve current anticancer therapeutics. Stem Cells Int. 2019:96180652019. View Article : Google Scholar : PubMed/NCBI

31 

Helbrecht I, Szymanski Ł, Fiedorowicz M, Matak D, Bartnik E, Golik P, Szczylik C and Czarnecka AM: Isolation of renal cancer stem cells. Postępy Biologii Komórki. 45:115–134. 2018.

32 

Bandhavkar S: Cancer stem cells: A metastasizing menace! Cancer Med. 5:649–655. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Codd AS, Kanaseki T, Torigo T and Tabi Z: Cancer stem cells as targets for immunotherapy. Immunology. 153:304–314. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Bapat SA, Mali AM, Koppikar CB and Kurrey NK: Stem and progenitor-like cells contribute to the aggressive behavior of human epithelial ovarian cancer. Cancer Res. 65:3025–3029. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Lupia M and Cavallaro U: Ovarian cancer stem cells: Still an elusive entity? Mol Cancer. 16:642017. View Article : Google Scholar : PubMed/NCBI

36 

Bregenzer ME, Horst EN, Mehta P, Novak CM, Repetto T and Mehta G: The role of cancer stem cells and mechanical forces in ovarian cancer metastasis. Cancers (Basel). 11:10082019. View Article : Google Scholar : PubMed/NCBI

37 

Li SS, Ma J and Wong AST: Chemoresistance in ovarian cancer: Exploiting cancer stem cell metabolism. J Gynecol Oncol. 29:e322018. View Article : Google Scholar : PubMed/NCBI

38 

Klemba A, Purzycka-Olewiecka JK, Wcisło G, Czarnecka AM, Lewicki S, Lesyng B, Szczylik C and Kieda C: Surface markers of cancer stem-like cells of ovarian cancer and their clinical relevance. Contemp Oncol (Pozn). 22:48–55. 2018.PubMed/NCBI

39 

Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, Blaudszun AR, Yevsa T, Fricke S and Kossatz-Boehlert U: Cancer stem cells-origins and biomarkers: Perspectives for targeted personalized therapies. Front Immunol. 11:12802020. View Article : Google Scholar : PubMed/NCBI

40 

Liou GY: CD133 as a regulator of cancer metastasis through the cancer stem cells. Int J Biochem Cell Biol. 106:1–7. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Zhou Q, Chen A, Song H, Tao J, Yang H and Zuo M: Prognostic value of cancer stem cell marker CD133 in ovarian cancer: A meta-analysis. Int J Clin Exp Med. 8:3080–3088. 2015.PubMed/NCBI

42 

Tao Y, Li H, Huang R, Mo D, Zeng T, Fang M and Li M: Clinicopathological and prognostic significance of cancer stem cell markers in ovarian cancer patients: Evidence from 52 studies. Cell Physiol Biochem. 46:1716–1726. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Onisim A, Iancu M, Vlad C, Kubelac P, Fetica B, Fulop A, Achimas-Cadariu A and Achimas-Cadariu P: Expression of Nestin and CD133 in serous ovarian carcinoma. J BUON. 21:1168–1175. 2016.PubMed/NCBI

44 

Ruscito I, Cacsire Castillo-Tong D, Vergote I, Ignat I, Stanske M, Vanderstichele A, Ganapathi RN, 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. 79:214–225. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Steg AD, Bevis KS, Katre AA, Ziebarth A, Dobbin ZC, Alvarez RD, Zhang K, Conner M and Landen CN: Stem cell pathways contribute to clinical chemoresistance in ovarian cancer. Clin Cancer Res. 18:869–881. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Liu BL, Liu SJ, Baskys A, Cheng H, Han Y, Xie C, Song H, Li J and Xin XY: Platinum sensitivity and CD133 expression as risk and prognostic predictors of central nervous system metastases in patients with epithelial ovarian cancer. BMC Cancer. 14:8292014. View Article : Google Scholar : PubMed/NCBI

47 

Liu CL, Chen YJ, Fan MH, Liao YJ and Mao TL: Characteristics of CD133-sustained chemoresistant cancer stem-like cells in human ovarian carcinoma. Int J Mol Sci. 21:64672020. View Article : Google Scholar : PubMed/NCBI

48 

Ween MP, Oehler MK and Ricciardelli C: Role of versican, hyaluronan and CD44 in ovarian cancer metastasis. Int J Mol Sci. 12:1009–1029. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Sacks JD and Barbolina MV: Expression and function of CD44 in epithelial ovarian carcinoma. Biomolecules. 5:3051–3066. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Lin J and Ding D: The prognostic role of the cancer stem cell marker CD44 in ovarian cancer: A meta-analysis. Cancer Cell Int. 17:82017. View Article : Google Scholar : PubMed/NCBI

51 

Zhou J, Du Y, Lu Y, Luan B, Xu C, Yu Y and Zhao H: CD44 expression predicts prognosis of ovarian cancer patients through promoting epithelial-mesenchymal transition (EMT) by regulating snail, ZEB1, and caveolin-1. Front Oncol. 9:8022019. View Article : Google Scholar : PubMed/NCBI

52 

Zhang J, Yuan B, Zhang H and Li H: Human epithelial ovarian cancer cells expressing CD105, CD44 and CD106 surface markers exhibit increased invasive capacity and drug resistance. Oncol Lett. 17:5351–5360. 2019.PubMed/NCBI

53 

Zhu Y, Zhang H, Zhang G, Shi Y and Huang J: Co-expression of CD44/MyD88 is a poor prognostic factor in advanced epithelial ovarian cancer. Ann Transl Med. 7:912019. View Article : Google Scholar : PubMed/NCBI

54 

Zhang HF, Hu P and Fang SQ: Understanding the role of CD44V6 in ovarian cancer. Oncol Lett. 14:1989–1992. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Tjhay F, Motohara T, Tayama S, Narantuya D, Fujimoto K, Guo J, Sakaguchi I, Honda R, Tashiro H and Katabuchi H: CD44 variant 6 is correlated with peritoneal dissemination and poor prognosis in patients with advanced epithelial ovarian cancer. Cancer Sci. 106:1421–1428. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Motohara T, Fujimoto K, Tayama S, Narantuya D, Sakaguchi I, Tashiro H and Katabuchi H: CD44 variant 6 as a predictive biomarker for distant metastasis in patients with epithelial ovarian cancer. Obstet Gynecol. 127:1003–1011. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Deng S, Yang X, Lassus H, Liang S, Kaur S, Ye Q, Li C, Wang LP, Roby KF, Orsulic S, et al: Distinct expression levels and patterns of stem cell marker, aldehyde dehydrogenase isoform 1 (ALDH1), in human epithelial cancers. PLoS One. 5:e102772010. View Article : Google Scholar : PubMed/NCBI

58 

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. 8:e651582013. View Article : Google Scholar : PubMed/NCBI

59 

Wang YC, Yo YT, Lee HY, Liao YP, Chao TK, Su PH and Lai HC: ALDH1-bright epithelial ovarian cancer cells are associated with CD44 expression, drug resistance, and poor clinical outcome. Am J Pathol. 180:1159–1169. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Chang B, Liu G, Xue F, Rosen DG, Xiao L, Wang X and Liu J: ALDH1 expression correlates with favorable prognosis in ovarian cancers. Mod Pathol. 22:817–823. 2009. View Article : Google Scholar : PubMed/NCBI

61 

Huang R, Li X, Holm R, Trope CG, Nesland JM and Suo Z: The expression of aldehyde dehydrogenase 1 (ALDH1) in ovarian carcinomas and its clinicopathological associations: A retrospective study. BMC Cancer. 15:5022015. View Article : Google Scholar : PubMed/NCBI

62 

Zhao W, Zang C, Zhang T, Li J, Liu R, Feng F, Lv Q, Zheng L, Tian J and Sun C: Clinicopathological characteristics and prognostic value of the cancer stem cell marker ALDH1 in ovarian cancer: A meta-analysis. Onco Targets Ther. 11:1821–1831. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Ayub TH, Keyver-Paik MD, 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. 6:16437–16448. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Roy M, Connor J, Al-Niaimi A, Rose SL and Mahajan A: Aldehyde dehydrogenase 1A1 (ALDH1A1) expression by immunohistochemistry is associated with chemo-refractoriness in patients with high-grade ovarian serous carcinoma. Hum Pathol. 73:1–6. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Ricci F, Bernasconi S, Porcu L, Erba E, Panini N, Fruscio R, Sina F, Torri V, Broggini M and Damia G: ALDH enzymatic activity and CD133 positivity and response to chemotherapy in ovarian cancer patients. Am J Cancer Res. 3:221–229. 2013.PubMed/NCBI

66 

Silva IA, Bai S, McLean K, Yang K, Griffith K, Thomas D, Ginestier C, Johnston C, Kueck A, Reynolds RK, et al: Aldehyde dehydrogenase in combination with CD133 defines angiogenic ovarian cancer stem cells that portend poor patient survival. Cancer Res. 71:3991–4001. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Tarhriz V, Bandehpour M, Dastmalchi S, Ouladsahebmadarek E, Zarredar H and Eyvazi S: Overview of CD24 as a new molecular marker in ovarian cancer. J Cell Physiol. 234:2134–2142. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Kristiansen G, Denkert C, Schlüns K, Dahl E, Pilarsky C and Hauptmann S: CD24 is expressed in ovarian cancer and is a new independent prognostic marker of patient survival. Am J Pathol. 161:1215–1221. 2002. View Article : Google Scholar : PubMed/NCBI

69 

Nakamura K, Terai Y, Tanabe A, Ono YJ, 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. 37:3189–3200. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Soltész B, Lukács J, Szilágyi E, Márton É, Szilágyi Bónizs M, Penyige A, Póka R and Nagy B: Expression of CD24 in plasma, exosome and ovarian tissue samples of serous ovarian cancer patients. J Biotechnol. 298:16–20. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Yang B, Yan X, Liu L, Jiang C and Hou S: Overexpression of the cancer stem cell marker CD117 predicts poor prognosis in epithelial ovarian cancer patients: Evidence from meta-analysis. Onco Targets Ther. 10:2951–2961. 2017. View Article : Google Scholar : PubMed/NCBI

72 

Luo L, Zeng J, Liang B, Zhao Z, Sun L, Cao D, Yang J and Shen K: Ovarian cancer cells with the CD117 phenotype are highly tumorigenic and are related to chemotherapy outcome. Exp Mol Pathol. 91:596–602. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Taskiran C, Erdem O, Onan A, Arisoy O, Acar A, Vural C, Erdem M, Ataoglu O and Guner H: The prognostic value of endoglin (CD105) expression in ovarian carcinoma. Int J Gynecol Cancer. 16:1789–1793. 2006. View Article : Google Scholar : PubMed/NCBI

74 

Bai S, Zhu W, Coffman L, Vlad A, Schwartz LE, Elishaev E, Drapkin R and Buckanovich RJ: CD105 is expressed in ovarian cancer precursor lesions and is required for metastasis to the ovary. Cancers (Basel). 11:17102019. View Article : Google Scholar : PubMed/NCBI

75 

Ziebarth AJ, Nowsheen S, Steg AD, Shah MM, Katre AA, Dobbin ZC, Han HD, Lopez-Berestein G, Sood AK, Conner M, et al: Endoglin (CD105) contributes to platinum resistance and is a target for tumor-specific therapy in epithelial ovarian cancer. Clin Cancer Res. 19:170–182. 2013. View Article : Google Scholar : PubMed/NCBI

76 

Huang J, Zhang J, Li H, Lu Z, Shan W, Mercado-Uribe I and Liu J: VCAM1 expression correlated with tumorigenesis and poor prognosis in high grade serous ovarian cancer. Am J Transl Res. 5:336–346. 2013.PubMed/NCBI

77 

Scalici JM, Arapovic S, Saks EJ, Atkins KA, Petroni G, Duska LR and Slack-Davis JK: Mesothelium expression of vascular cell adhesion molecule-1 (VCAM-1) is associated with an unfavorable prognosis in epithelial ovarian cancer (EOC). Cancer. 123:977–984. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Tayama S, Motohara T, Narantuya D, Li C, Fujimoto K, Sakaguchi I, Tashiro H, Saya H, Nagano O and Katabuchi H: The impact of EpCAM expression on response to chemotherapy and clinical outcomes in patients with epithelial ovarian cancer. Oncotarget. 8:44312–44325. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Spizzo G, Went P, Dirnhofer S, Obrist P, Moch H, Baeuerle PA, Mueller-Holzner E, Marth C, Gastl G and Zeimet AG: Overexpression of epithelial cell adhesion molecule (Ep-CAM) is an independent prognostic marker for reduced survival of patients with epithelial ovarian cancer. Gynecol Oncol. 103:483–488. 2006. View Article : Google Scholar : PubMed/NCBI

80 

Woopen H, Pietzner K, Richter R, Fotopoulou C, Joens T, Braicu EI, Mellstedt H, Mahner S, Lindhofer H, Darb-Esfahani S, et al: Overexpression of the epithelial cell adhesion molecule is associated with a more favorable prognosis and response to platinum-based chemotherapy in ovarian cancer. J Gynecol Oncol. 25:221–228. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Zhang J, Chang DY, Mercado-Uribe I and Liu J: Sex-determining region Y-box 2 expression predicts poor prognosis in human ovarian carcinoma. Hum Pathol. 43:1405–1412. 2012. View Article : Google Scholar : PubMed/NCBI

82 

Bååth M, Westbom-Fremer S, Martin de la Fuente L, Ebbesson A, Davis J, Malander S, Måsbäck A, Kannisto P and Hedenfalk I: SOX2 is a promising predictor of relapse and death in advanced stage high-grade serous ovarian cancer patients with residual disease after debulking surgery. Mol Cell Oncol. 7:18050942020. View Article : Google Scholar : PubMed/NCBI

83 

Li Y, Chen K, Li L, Li R, Zhang J and Ren W: Overexpression of SOX2 is involved in paclitaxel resistance of ovarian cancer via the PI3K/Akt pathway. Tumour Biol. 36:9823–9828. 2015. View Article : Google Scholar : PubMed/NCBI

84 

Czekierdowski A, Stachowicz N, Czekierdowska S, Łoziński T, Gurynowicz G and Kluz T: Prognostic significance of TEM7 and nestin expression in women with advanced high grade serous ovarian cancer. Ginekol Pol. 89:135–141. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Qin Q, Sun Y, Fei M, Zhang J, Jia Y, Gu M, Xia R, Chen S and Deng A: Expression of putative stem marker nestin and CD133 in advanced serous ovarian cancer. Neoplasma. 59:310–315. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Davidson B, Holth A and Dong HP: Expression of the cancer stem cell marker SSEA1 is associated with poor survival in metastatic high-grade serous carcinoma. Virchows Arch. 477:677–685. 2020. View Article : Google Scholar : PubMed/NCBI

87 

Chen WC, Hsu HP, Li CY, Yang YJ, Hung YH, Cho CY, Wang CY, Weng TY and Lai MD: Cancer stem cell marker CD90 inhibits ovarian cancer formation via β3 integrin. Int J Oncol. 49:1881–1889. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Connor EV, Saygin C, Braley C, Wiechert AC, Karunanithi S, Crean-Tate K, Abdul-Karim FW, Michener CM, Rose PG, Lathia JD and Reizes O: Thy-1 predicts poor prognosis and is associated with self-renewal in ovarian cancer. J Ovarian Res. 12:1122019. View Article : Google Scholar : PubMed/NCBI

89 

Kim WT and Ryu CJ: Cancer stem cell surface markers on normal stem cells. BMB Rep. 50:285–298. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Yang W, Kim D, Kim DK, Choi KU, Suh DS and Kim JH: Therapeutic strategies for targeting ovarian cancer stem cells. Int J Mol Sci. 22:50592021. View Article : Google Scholar : PubMed/NCBI

91 

Barzegar Behrooz A, Syahir A and Ahmad S: CD133: Beyond a cancer stem cell biomarker. J Drug Target. 27:257–269. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, Zhang G, Wang X, Dong Z, Chen F and Cui H: Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther. 5:82020. View Article : Google Scholar : PubMed/NCBI

93 

Masoumi J, Jafarzadeh A, Abdolalizadeh J, Khan H, Philippe J, Mirzaei H and Mirzaei HR: Cancer stem cell-targeted chimeric antigen receptor (CAR)-T cell therapy: Challenges and prospects. Acta Pharm Sin B. 11:1721–1739. 2021. View Article : Google Scholar : PubMed/NCBI

94 

Huang B, Miao L, Liu J, Zhang J and Li Y: A promising antitumor method: Targeting CSC with immune cells modified with CAR. Front Immunol. 13:9373272022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2022
Volume 24 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Królewska-Daszczyńska P, Wendlocha D, Smycz-Kubańska M, Stępień S and Mielczarek-Palacz A: Cancer stem cells markers in ovarian cancer: Clinical and therapeutic significance (Review). Oncol Lett 24: 465, 2022
APA
Królewska-Daszczyńska, P., Wendlocha, D., Smycz-Kubańska, M., Stępień, S., & Mielczarek-Palacz, A. (2022). Cancer stem cells markers in ovarian cancer: Clinical and therapeutic significance (Review). Oncology Letters, 24, 465. https://doi.org/10.3892/ol.2022.13585
MLA
Królewska-Daszczyńska, P., Wendlocha, D., Smycz-Kubańska, M., Stępień, S., Mielczarek-Palacz, A."Cancer stem cells markers in ovarian cancer: Clinical and therapeutic significance (Review)". Oncology Letters 24.6 (2022): 465.
Chicago
Królewska-Daszczyńska, P., Wendlocha, D., Smycz-Kubańska, M., Stępień, S., Mielczarek-Palacz, A."Cancer stem cells markers in ovarian cancer: Clinical and therapeutic significance (Review)". Oncology Letters 24, no. 6 (2022): 465. https://doi.org/10.3892/ol.2022.13585