Proceedings of Stem Cell Research and Oncogenesis (2020), 8:e1001 © 2020 Creative Commons. All rights reserved ISSN 2641-5070 DOI: 14343/PSCRO:2020.8e1001 http://stemonco.com |
![]() |
Review Article | Open Access ![]() |
Stem Cells in Ovarian Cancer and Potential Therapies | |
Elena Zuber1, Diana Schweitzer1, Dominick Allen1, Seema Parte2, and Sham S. Kakar 1,3,* | |
1Department of Physiology, University of Louisville, Louisville, KY40202, | |
2Department of Biochemistry and Molecular Biology, University of Nebraska, Omaha, NE-68198-5870, | |
3James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202. | |
*Correspondence: Sham S. Kakar, Ph.D., MBA, Department of Physiology, 500 South Floyd Street, University of Louisville, Louisville, KY 40202. Phone: (502) 852-0812; E-mail: sham.kakar@louisville.edu Received: March 1, 2020; Revised: March 20, 2020; Accepted: March 22, 2020 |
Ovarian cancer represents the fifth cause of deaths from cancer accounting to 21,750 new cases and 13,940 deaths expected in the USA in 2020 [1, 2]. Due to late diagnosis of ovarian cancer, the cancer cells are already disseminated to the peritoneal cavity at diagnosis and thus impose a serious clinical challenge [3–5]. Ovarian tumors may be divided into three types in particular: epithelial, germ cell, and stromal cells—epithelial carcinoma being the most abundant [6]. Furthermore, within epithelial tumors there are four subtypes, serous, endometrioid, clear cell, and mucinous—wherein serous tumors are the most lethal [6–8]. The first line treatment for ovarian cancer is cytoreductive surgery followed by chemotherapy mainly combination of carboplatin and paclitaxel [9, 10]. Initial response of this combination therapy is very high (70%), however after few treatments, patients develop cisplatin resistance leading to tumor relapse and recurrence [11, 12]. Recently, several investigators have indicated the presence of cancer stem cells (CSCs) also known as cancer initiating cells in tumors as putative entities responsible for cancer initiation and progression [13, 14]. These CSCs have been reported to be chemo- and radio- resistant, and ultimately leading to cancer recurrence [15–19]. Therefore, it is crucial to understand the biology of CSCs including their regulation in order to develop therapies that can target both the cancer cells and CSCs and thus provide highly effective therapy for the treatment of cancer. Present review article briefly covers the biology of different populations of CSCs in ovarian cancer based upon several reported CSC specific biomarkers and cell surface markers and potential therapies being developed recently to target CSCs.
Cancer arises from a cell type within the tumors that can undergo self-renewal and promotes tumorigenesis—these cells are known as tumor initiating cells or cancer stem cells (CSCs). Various specific markers including but not limited to ALDH1/2, CD133, CD117, CD24, CD34, CD44, EpCAM, NANOG, OCT 3/4, LGR5 and LY6A have been reported and used in isolation and characterization of CSCs from ovarian cancer cell lines, ovarian cancer, and ascites collected from patients with recurrent ovarian cancer [20, 21]. Currently, it is accepted that CSCs are not only responsible for the development of chemotherapeutic and cytostatic resistances, but also for primary tumor growth, metastasis and tumor relapse [22–24].
In addition to their origin and morphologies, these malignant populations also vary in their biological behavior [24]. A tiny population of stem cells with embryonic characteristics from normal human ovaries [25–29] have been suggested as progenitors [28–30], however, this has yet to be elucidated. The high level of non-consistent gene mutations giving rise to heterogeneous populations making a daunting task in identifying a suitably effective target. Even though the existence of CSCs has been identified in a variety of tumors, their origin is not well understood. Owing to the common characteristics and self-renewal mechanisms shared between stem as well as CSCs, it is speculated that cancer may be originating from the transformation of normal tissue specific stem cells [31] i.e. ovarian stem cells in this instance. High levels of expression of several oncogenes and transforming genes in CSCs support the hypothesis that CSCs could be a result of transformation of normal stem cells present in adult tissues [31]. However, this hypothesis needs to be tested. Ovarian CSCs have been attributed with characteristics of self-renewal, tumor-initiation, growth, differentiation, drug resistance, and tumor relapse [31]. In this study an overlooked and unconventional role of PTTG1 as a marker of CSCs (in normal ovaries, benign, borderline, high grade tumors and ascites derived tumors) and its ability to modulate CSCs via the ovarian germline and stemness-related genes was dissected very intricately and reported for the first time by exploring the self-renewal and epithelial-mesenchymal transition pathways regulated by PTTG1.
Recently, our group have also identified and characterized ovarian stem cells and CSC compartments on basis of unique germline stem cell specific marker VASA with the help of co-expression studies. Non-proliferating and quiescent stem cell populations were identified in normal ovaries besides benign, borderline and high-grade ovarian tumors. Typically, two distinct stem like/cancer stem-like cells expressing various combination of markers were detected in the samples including normal ovaries [31–33]. In a quest towards identifying several heterogeneous CSC populations in ovarian tumors and metastatic ascites derived fluid, our group has extensively characterized these cells with the aid of several biomarkers and stemness associated genes. In one study, the germline stem cell marker associated with the normal ovarian stem cells was found to be co-expressed with most of the CSC specific surface markers with their prominent localization in the ovarian surface epithelium (OSE) layer and the adjacent ovarian cortex [33]. An interesting localization, distribution and predominance of specific combination of markers were detected across the normal ovaries, benign, borderline and high-grade ovarian tumor samples from patients. At the same time, other study revealed the proliferating and quiescent populations of CSCs with the aid of Ki67 within similar samples [33]. Further, we reported the exclusive localization of an oncogene PTTG in not only normal ovaries, benign, borderline and high-grade ovarian tumors but also the ascites fluid [31]. We reported for the first time a systematic expression profiling of various CSC markers and their co-expression with the oncogene PTTG. Our study highlighted that the stem cell specific self-renewal related and EMT specific pathways [31] executed CSC regulation by PTTG. An interesting network of stemness pathways governing the various CSC populations in ovarian tumors and similar cellular signaling networks operating in ascites derived CSCs was anticipated based on persistence of similar CSC populations. Hence, a thorough understanding of the stem cell contribution to tumor pathogenesis (i.e. tumor initiation, dissemination and eventual culmination into therapy resistance) attributed to CSCs warrant a thorough investigation while not overlooking real time patient-level clinical scenario of treatment regimen administered. Some of the commonly identified cancer stem cells in ovarian cancer are listed in Table 1.
Marker | Type of protein | References |
---|---|---|
CD24 | Cell surface transmembrane glycoprotein | [31], [104–110] |
CD34 | Cell surface transmembrane glycoprotein | [31], [111], [112] |
CD44 | Cell surface transmembrane glycoprotein | [31], [113–116] |
CD105 | Cell surface transmembrane glycoprotein | [112] |
CD117 | Tyrosine kinase receptor | [31], [117–122] |
CD133 | Cell surface transmembrane glycoprotein | [31], [117], [123–127] |
EpCam | Cell surface transmembrane glycoprotein | [31], [128], [129] |
ROR1 | Tyrosine kinase receptor | [37], [130] |
ALDH | Cytosolic aldehyde dehydrogenase enzyme | [31], [131–134] |
SOX2 | Transcription factor | [31–33], [131–134] |
OCT4 | Transcription factor | [31–33], [80] |
NANOG | Transcription factor | [31–33], [138–141] |
MYC | Transcription factor | [23], [142] |
ABCG1, ABCG2 | ATP binding cassette transporter | [143–145] |
PTTG1 | Cytosolic/nuclear protein | [31] |
LGR5 | Cell surface membrane protein | [31], [32], [51] |
DDX4/VASA | ATP-dependent RNA helicases | [29], [31], [32], [146] |
IFITM3/FRAGILIS | Cytosolic protein | [31], [32], [147] |
SSEA4 | Cytosolic protein | [31], [32] |
STELLA | Cytosolic and nuclear protein | [32], [148], [149] |
Note: Some of the information are taken from Kenda Suster N, Virant-Klun, I [28].
Ovarian cancers are difficult to treat due to their complexity and heterogeneity from patient to patient. Presence of CSCs in ovarian cancer and ascites are resistant to chemotherapy, and their amplification occurs upon treatment with currently used chemotherapies. Investigators have shown enrichment of these CSC populations when treated with chemotherapy, in vitro and in experimental models, resulting in increased drug resistance [34, 35].
ALDH1A1+ CSCs have been implicated in rendering resistance to the PARPi in BRCA2-mutated epithelial ovarian cancer [36]. A type I receptor tyrosine kinase-like orphan receptor (ROR1) expressed during embryogenesis and in several cancers is expressed in ovarian CSCs and contributes towards migration/invasion or spheroid formation in vitro and tumor engraftment in immune-deficient mice thus serving as a potential targetable molecule [37]. Recently propagation of primary ovarian CSCs specific for individual patient, followed by 3D hanging drop suspension culture to establish spheroidal cultures and xenografting in immuno-compromised mice for pre-clinical drug screening, especially for assessing patient-specific (personalized) responses to chemotherapeutics and for delving deeper into understanding therapeutic resistance from a CSC standpoint have been established. Such platforms may enable testing of CSC heterogeneity and differential suppression as well as enrichment of sensitive and resistant populations respectively [38]. Since phenomena of EMT, chemoresistance, tumor invasion, metastasis, recurrence and CSCs are inter-related, development of multi-targeted approaches in the future may help to eradicate various CSC populations expressing CD117, CD133, Notch and targeting of EMT/CSCs pathways such as PI3K/mTOR, JAK2/STAT3, DNA methyltransferase, p53 mediated apoptosis, EGFR/Stat3, TGF-β, Notch3/ERK etc. may prove highly promising. EMT targeting approaches to overcome resistance feature are worth pursuing and systematic evaluation of clinical efficacy in clinical trials are further warranted [39, 40]. In recent years, varied therapies are being tested to target CSCs in order to reduce or eliminate recurrence of cancer which are addressed in following sections.
Mutations, overexpression and dysregulation of several CSC self-renewal pathways have been implicated in the progression, metastasis, and recurrence of ovarian cancer. These pathways often lead to growth and maintenance of ovarian CSCs. Some of these pathways include Notch, PI3K/PTEN/AKT, JAK/STAT, Wnt/β-catenin, and Sonic Hedgehog (SHH) (Fig. 1). Current research has focused on these signaling pathways in the context of targeting of CSCs.
![]() |
Figure 1. Schematic representation of various factors regulating the signaling pathways involved in cancer stem cell self-renewal, differentiation and metastasis. |
The Notch signaling pathway is an evolutionarily conserved signaling cascade involved in embryonic development. Additionally, increased expression of Notch3 has correlated with poor prognosis in patients with ovarian cancer. It plays a critical role in maintenance, differentiation, proliferation, communication, and apoptosis of progenitor cells; and inextricably intertwined for many cancers, including ovarian cancer [41, 42]. More specifically, it contributes toward the maintenance of ovarian CSCs and their resistance to chemotherapy.
The protein coding gene Notch3 is amplified and overexpressed in the majority of high-grade serious ovarian carcinoma (HGSOC) [43]. The most highly expressed Notch3 ligand is Jagged1. This is expressed primarily in mesothelial cells in a tumor microenvironment, indicating that this pathway likely contributes to the signaling that regulates cell adhesion and tumor proliferation [42, 43]. Notch3 has also been shown to play a critical role in chemoresistance and stemness of ovarian CSCs, and it can be used as a prognostic indicator. Activation of Notch receptors leads to proteolytic cleavage of the intracellular Notch domain mediated by γ-secretase. This cleaved domain then translocates to the nucleus, where it regulates gene transcription [44]. It has been reported that overexpression of Notch3 results in increased expression of stem cell markers such as OCT4, SOX2, and NANOG. OCT4 promotes self-renewal of CSCs, while SOX2 is required for their maintenance [43]. Notch overexpression also causes enhanced expression of a transport protein (ABCG1) that increases chemo-resistance, specifically to platinum and carboplatin. In addition, it leads to expansion of the side population of CSCs, resulting in increased chemoresistance [43, 44].
Downstream effects of Notch were improved by knockdown of Notch3. Inhibition of Notch led to depletion of total number of ovarian CSCs along with increased tumor sensitivity to platinum [44]. Additionally, down-regulation of Jagged1 led to increased sensitivity to chemotherapy, further supporting the evidence that the Notch pathway contributes to stemness and chemoresistance [43]. One study used a γ-secretase inhibitor in conjunction with cisplatin to induce DNA damage, cell cycle arrest, and apoptotic cell death [42]. Notch3 protein when examined in primary and recurrent tumors from the same patients showed elevated expression levels in the recurrent tumors as compared to the primary tumors indicating its relationship with poor prognosis [43, 44], suggesting that the Notch pathway is a promising candidate for targeted therapies.
Another important pathway relevant to both embryogenesis and ovarian cancer is the Wnt/β-catenin pathway. During embryogenesis, it regulates cell fate and is involved in the normal development of ovaries and fallopian tubes. In adults, it is critical for self-renewal in tissues, and plays a role in maintenance, quiescence, and chemoresistance of stem cells [45, 46]. Since, this pathway is so complex, its signaling appears to differ among different histotypes of ovarian cancer.
In its deactivated form, β-catenin is degraded within proteasomes. However, on activation of Wnt/β-catenin pathway, β-catenin is not phosphorylated and it is released from the destruction complex and translocates to the nucleus, where it acts as a transcription factor, thereby activating Wnt target genes – including those involved in stemness and chemoresistance [47–50]. These target genes include those that code for leucine-rich-repeat-containing G protein-coupled receptors (LGR), which is not only expressed in ovarian cancer but also in several cancer types [51]. LGR5 is a stem cell marker for ovarian stem cells and LGR6 is a cancer stem cell marker for fallopian tube, and expression of either one is a sign of elevated Wnt significantly [51–54]. Elevated Wnt signaling in tumors indicates that Wnt signaling is necessary for the expression of the stem cell factors that support tumor growth and maintenance [51]. Down-regulation of LGR5 has been found to be associated with lower CSC-like phenotypes and lower chemo-resistance properties in vitro. Additionally, silencing of LGR5 inhibited stemness and chemo-resistance in vivo, specifically through inhibition of the Wnt/β-catenin signaling pathway [53–55].
Evidence show that, when Wnt target genes are activated, a hypoxic niche leads to CD117 expression, leading to Akt activation and nuclear β-catenin expression, which induces a drug transporter that leads to chemoresistance [43]. Knockdown of CD117 by using specific siRNA results in lower numbers and size of ovarian CSC subpopulations and their pro-tumorigenic activity, indicating that CD117 contributes to the tumorigenic and chemoresistant properties of ovarian CSCs through Wnt activation [48]. This is further supported by the finding that CD117 overexpression leads to the upregulation of ATP-binding cassette G2 (ABCG2) in the Wnt/β-catenin pathway, enhancing chemoresistance. Similarly, expression of Wnt/β-catenin target genes and ligands were upregulated in chemoresistant ovarian cancer cells, and these cells showed high expression of CSC markers [54, 55]. More importantly, it was found that Wnt/β-catenin specific inhibitors sensitize ovarian CSCs to chemotherapy and reduce the number of CSC subpopulations, indicating that inhibition of this pathway can stifle CSC characteristics and enhance chemosensitivity [55].
Raghavan et al [56] found that tumor-associated macrophages are responsible for promoting metastasis, angiogenesis, and tumorigenesis within the tumor microenvironment, so these are likely to play a role in recurrence, metastasis, resistance, and preservation of stem-like phenotypes through Wnt signaling. These investigators also found that paracrine Wnt activation during interactions between CSCs and M2 macrophages make up a positive feedback loop, which likely contributes to phenotypes that are more aggressive. This evidence further supports that Wnt pathway as a potential target, particularly for the reduction/elimination of CSC and M2 macrophages in the tumor microenvironment. Another promising therapy was found to be theaflavin-3, 3’digallate (TF3). This compound was shown to inhibit ovarian CSCs by inducing apoptosis and cell cycle arrest and inhibiting angiogenesis through the Wnt/β-catenin pathway [57].
During embryogenesis, the Hedgehog pathway regulates tissue polarity and pattern along with stem cell development and maintenance [58]. This pathway consists of secreted ligands: Sonic hedgehog (Shh), Indian hedgehog (Ihh), and Desert hedgehog (Dhh); Hedgehog receptors: Patched (PTCH1, PTCH2) and Smoothened (SMO); and Gli transcription factors 1, 2, and 3 [59]. Activation of this pathway is associated with regulation of CSC phenotypes like self-renewal, differentiation, and tumor initiation [59]. This pathway can become disrupted in two ways in ovarian cancer: 1) disrupted by the overexpression of endogenous pathway ligands such as Shh; 2) disrupted through using inhibitor vismodegib [60].
It has been reported that ovarian CSCs exhibit increased intracellular Gli1 expression, which correlates with increased formation of spheroids with CSC properties. Gli1 has also been found to play a role in expression of ABC transporters ABCG1 and ABCG2 by directly binding to their promoter regions resulting in increased resistance to both cisplatin and paclitaxel in spheroid forming of ovarian cancer cells [61, 62]. Additionally, there is higher expression of SMO and Gli1 in borderline and malignant tumor tissues than in benign tumor and normal ovarian tissues [63, 64], indicating their importance in progression of ovarian cancer.
Investigators found that the inhibition of the hedgehog pathway by cyclopamine resulted in a decrease in Gli concentrations followed by decreased cancer cell proliferation [65]. Furthermore, it was demonstrated that cyclopamine also inhibited tumor growth and impaired spheroid function. Exogenous expression of Gli was found to increase both cell proliferation and invasiveness by at least 200%, demonstrating the effect of Gli on ovarian cancer cells proliferation [66]. Hh pathway components SMO, PTCH and GLI1 were reported to be activated in benign, borderline and malignant ovarian epithelial tumors and implicated its association with cisplatin resistance [66–68]. This clearly implicates importance of Hedgehog signaling in malignancy and chemoresistance of ovarian CSCs. GDC-0449, a potent hedgehog inhibitor, is in the clinical phases for its application in treating ovarian cancer patients [68].
Phosphatidylinositol 3-kinases (PI3K) is an enzyme involved generally in cell growth, proliferation, differentiation, motility, survival, and intracellular trafficking. It appears that the PI3K/PTEN/AKT is activated in approximately half of high-grade serious ovarian carcinoma (HGSOC), making it a potential therapeutic target for ovarian cancer. Additionally, high expression of activated Akt is associated with poor prognosis and survival, due to mutations that could occur in several components of this pathway [69, 70].
This pathway has been found to regulate enrichment of ovarian CSCs, maintenance of the stem cell (SC) phenotype, and chemoresistance. Ovarian spheroids show high expression of pAKT1 and low expression of PTEN, along with increased resistance to paclitaxel [71–74]. Additionally, inhibition of AKT1 results in decreased spheroid formation and migration. Furthermore, knockdown of AKT1 using siRNA led to loss of CSC marker expression, loss of spheroid formation, and loss of paclitaxel resistance [75, 76]. In cisplatin-resistant CSCs, AKT regulates expression of PPM1D, a gene that codes for a protein, which inhibits DNA damage and apoptotic response after DNA damage. Moreover, the down-regulation of AKT activity led to loss of PPM1D stability and an increase in its degradation [77, 78]. This, in turn, led to increased response to cisplatin, indicating the importance of this pathway in initiation and maintenance of CSCs and their chemoresistance, making this pathway an ideal target for ovarian cancer therapy.
PI3K/PTEN/AKT pathway also appears to play a crucial role through C-Kit, a well-known proto-oncogene of ovarian CSCs [79]. Its ligand, stem cell factor (SCF) can exist either in soluble form or as a transmembrane protein. SCF interacts with C-kit to regulate cell viability, proliferation, and differentiation. SCF was found in high levels in epithelial ovarian cancer, but it appears to be only membrane bound in tumor cells [79]. Both secretory and membrane bound forms of SCF are observed in tumor associated macrophages (TAM) and fibroblasts (TAF). Interestingly, in the circulating monocytes of healthy patients neither of these forms were observed. However, both forms were produced upon differentiation into macrophages, with M1 and M2 polarization having no effect [79]. Both of these isoforms were then able to activate the AKT pathway in cells with c-Kit receptor. This effect was also counteracted by imatinib, a tyrosine kinase inhibitor. These findings are thus an evidence of a juxtacrine/paracrine circuit in ovarian cancer that may exist and act through the AKT pathway [79].
The JAK/STAT signaling pathway is a universally expressed intracellular signal transduction pathway and is involved in many crucial biological processes, including cell proliferation, differentiation, apoptosis, and immune regulation. JAK/STAT pathway also generally regulates differentiation, proliferation, and survival of stem cells. In ovarian cancer, the JAK/STAT pathway almost is always active, with no inhibition or regulation of its activity [80]. This pathway has been reported as an important pathway in maintaining CSC population especially OCT4 in ovarian cancer [80, 81]. There are four JAK family non-receptor tyrosine kinases, JAK1, JAK2, JAK3 and TYK2. It has been reported that JAK1, JAK2 and TYK2 are ubiquitously expressed, whereas JAK3 is predominantly expressed in hematopoietic cells. There are several types of STAT (STAT1, STAT2, STAT3, STAT4, STAT5a and b). During the activation of the signaling cascade, these JAK and STAT molecules assemble into homo- and hetero-dimers, or even into more complex multi-mers [81]. Jin [82] showed that activation of JAK/STAT3 pathway leads to increased tumorigenesis and metastasis ability, the transition of cancer stem cells (CSCs), and chemoresistance in cancer through the regulation of epithelial-mesenchymal transition (EMT) [82] inducing transcription factors such as Snail, Zeb1, JUNB, and Twist1 [83–85]. Activation of JAK/STAT signaling is triggered by various hormones, cytokines (including the IL-6 family) [83], and growth factor through a variety of molecular mechanisms leading to tumorigenesis, tumor progression and metastasis. Furthermore, the JAK/STAT pathway regulates expression of several genes needed for the maintenance of CSC phenotypes and acquisition of drug resistance [86, 87]. JAK/STAT pathway is a potential target for therapeutic purpose to treat several disorders including cancer. Several inhibitors have been synthesized and tested for their activity in vitro and in vivo. Some of the inhibitors are in clinical trials for their application for the treatment of cancer [88].
Treating ovarian CSCs with phenethyl isothiocyanate (PEITC) revealed an anti-CSC activity, defined as reduced expression of biomarkers for stemness [89]. Koschorke and colleagues discovered that treating ovarian CSCs with PEITC impaired the spheroid forming efficiency of the cells by decreasing their ALDH-positive compartments. ALDH is a marker for stemness, so this observation implies that the CSCs are being directly targeted and the ALDH+ population is decreased resulting in decreased tumor growth and increased responsiveness to chemotherapies [89].
One study demonstrated the therapeutic potential of graphene oxide-silver nanoparticle nano-composites for ovarian cancer by targeting the ovarian CSCs. Ovarian CSCs were harvested and incubated with the nanocomposite. After 3 weeks of incubation, the number of stem cell colonies were reduced which hinted to the toxicity of the nanocomposite towards the ovarian CSCs [90]. The mechanism for the composite was to generate reactive oxygen species, leaking of lactate dehydrogenase, reduced mitochondrial membrane potential, and increased expression of apoptotic genes, all of which contribute to reduce stem cell viability. Choi and colleagues [90] added salinomycin to the media, which, in tandem with the graphene oxide-silver nanoparticle nanocomposites, and showed an increased rate of apoptosis. This therapy is advantageous in that it requires very low concentrations to be effective, which could result in decreased pricing for therapies along with the conservation of resources [90].
Recently, Udoh et al [91] and Carter et al [92] showed that a fungal metabolite from myrothecium verrucaria known as “Verrucarin J” (VJ) targets both cancer cells and CSCs in lung and ovarian cancers. Such pronounced effects of VJ were achieved through the inhibition of Wnt/β-catenin and Notch1 signaling pathways. In independent studies, Kakar and his group [93, 94] showed inhibition of tumor growth and metastasis of ovarian cancer by Withaferin A (WFA), a product from
Several strides have been performed in order to target ovarian cancer and the CSCs in particular which were recently summarized during the American Association of Cancer Research/Rivkin Center Ovarian Cancer Research Symposium held at the University of Washington in September 2018. Current state of the art of ovarian cancer treatment were highlighted and manifold efforts of researchers towards multiple targeting modalities such as clinical trials employing novel agents, including poly-ADP-ribose polymerase (PARP) inhibitors, other DNA-damaging agents, vascular endothelial growth factor receptor inhibitors, mirvetuximab soravtansine, and immune checkpoint blockade. Innovative novel technologies such as antibody-drug conjugates targeting surface receptors in ovarian cancer either alone or in combination with immune checkpoint blockade exhibited strong translational potential. Potential therapeutic combination partners such as DNA repair inhibiting agents, those targeting cellular checkpoints, and drugs with potential against CSCs were identified. Novel therapeutic strategies based upon endoplasmic reticulum stress response, epithelial to mesenchymal transition, and targeting of surface molecules using novel antibody drug conjugates revealed anti-tumor potential in pre-clinical models and its tremendous clinical potential. Study highlights pronounced the significance of developing effective and combinatorial treatment modalities such as immune checkpoint inhibitors with other immunotherapies, PARP inhibitors, and standard chemotherapeutic regimen, rather than a single drug based treatment modality [95–101]. Besides the primary ovarian tumor, peritoneal tumor microenvironment supporting the ascites derived CSCs and the transition into an aggressive metastatic form of ovarian cancer is implicated rather than the potential primary tumor properties and thus targeting ascites derived CSCs is equally pertinent. Some of the key and novel treatment strategies addressing therapy-induced resistance arising out of putative CSC dormancy and plasticity, highly efficient drug efflux pumps and DNA repair mechanisms have been recently summarized by Ahmed and her group [102].
Owing to the complexity of the mechanisms of regulation of ovarian cancer growth and metastasis, there are many opportunities for disrupting these cancer-causing processes. However, there is still significant research to be performed in relation to implementation of these therapies and the best ways to tailor them to individual cases of ovarian cancer. Precision medicine with targeted treatment for ovarian cancer patients based on their individual genetic susceptibility and molecular signatures requires patient stratification and further efforts in terms of clinical studies [101]. The signaling cascades involved in stemness, chemoresistance, and metastasis, such as Wnt, Notch, and Hedgehog, etc. provide many potential points of inhibition of growth of ovarian cancer as aptly reviewed recently by Keyvani and group [103]. In addition, better classification and understanding of CSCs in ovarian cancer, will hopefully lead to better understanding of the origin of ovarian cancer malignancies and even more potential sites of inhibition may be revealed.
The authors do not have any potential conflict of interest to disclose.
E Zuber, D Schweitzer and D Allen wrote the first draft of the manuscript. S Parte and SS Kakar made substantial contribution towards drafting and inclusion of appropriate information and made final edits to the manuscript. All authors have read and agreed to the final version of this manuscript.
This work was supported by NIH grants U01CA185148 (subcontract), T32 HL134644 and R25 GM133328 to SSK.
We regret for missing the work of some of the investigators due to our over-sight and space constraints.
This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) http://creativecommons.org/licenses/by/4.0/
[1] | American Cancer Society Facts and Figures, 2020. |
[2] | Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7–30. |
[3] | Auersperg N, Edelson MI, Mok SC, Johnson SW, Hamilton TC. The biology of ovarian cancer. Semin Oncol 1998;25:281–304. |
[4] | Ahmed N, Stenvers KL. Getting to know ovarian cancer ascites: Opportunities for targeted therapy-based translation-al research. Frontiers in Oncology 2013;3:256. |
[5] | Lengyel E. Ovarian cancer development and metastasis. Am J Pathol 2010;177:1053–64. |
[6] | Chen VW, Ruiz B, Killeen JL, Cote T, Wu XC, Correa CN. Pathology and Classification of Ovarian Tumors. Pathology and classification of ovarian tumors. Cancer 2003;97(10 Suppl):2631–42. |
[7] | Koshiyama M, Matsumura N, Konishi I. Recent concepts of ovarian carcinogenesis: Type I and Type II. Biomed Res Int 2014;934261. |
[8] | Koshiyama M, Matsumura N, Konishi I. Subtypes of Ovarian Cancer and Ovarian Cancer Screening. Diagnostics 2017;7:pii.E12. |
[9] | Cannistra SA. Cancer of the ovary. N Engl J Med 2004;351:2519–29. |
[10] | Ozols RF, Bundy BN, Greer BE, Fowler JM, Clarke-Pearson D, Burger RA, Mannel RS, DeGeest K, Hartenbach EM, Baergen R. Gynecologic Oncology Group. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol 2003;21:3194–200. |
[11] | Matsuo K, Lin YG, Roman LD, Sood AK. Overcoming platinum resistance in ovarian carcinoma. Expert Opin Investing Drugs 2010;9:1339–54. |
[12] | Martin LP, Schilder RJ. Management of recurrent ovarian carcinoma: current status and future directions. Semin Oncol 2009;36:112–25. |
[13] | Dyall S, Gayther SA, Dafou D. Cancer stem cells and epithelial ovarian cancer. J Oncol 2010;2010:105269. |
[14] | Hermann PC, Huber SL, Heeschen C. Metastatic cancer stem cells: a new target for anti-cancer therapy? Cell Cycle 2008;7:188–93. |
[15] | Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer 2005;5:275–84. |
[16] | Rich JN. Cancer stem cells in radiation resistance. Cancer Res 2007;67:8980–4. |
[17] | Blagosklonny MV. Why therapeutic response may not prolong the life of a cancer patient: selection for oncogenic resistance. Cell Cycle 2005;4:1693–8. |
[18] | Vlashi E, McBride WH, Pajonk F. Radiation responses of cancer stem cells. J Cell Biochem 2009;108:339–42. |
[19] | Jordan CT. Cancer stem cells: controversial or just misunderstood? Cell Stem Cell 2009;4:203–5. |
[20] | Bapat SA, Mali AM, Koppikar CB, Kurrey NK. Stem and progenitor-like cells contribute to the aggressive behavior of human epithelial ovarian Cancer. Cancer Res 2005;65:3025–9. |
[21] | Latifi A, Luwor RB, Bilandzic M, Nazaretian S, Stenvers K, Pyman J, Zhu H, Thompson EW, Quinn MA, Findlay JK, Ahmed N. Isolation and characterization of tumor cells from the ascites of ovarian Cancer patients: molecular phenotype of Chemoresistant ovarian tumors. PLoS One 2012;7:e46858. |
[22] | Foster R, Buckanovich RJ, Rueda BR. Ovarian cancer stem cells: working towards the root of stemness. Cancer Letts 2013;338:147–57. |
[23] | Garson K, Vanderhyden BC. Epithelial ovarian cancer stem cells: under lyingcomplexity of a simple paradigm. Reproduction 2015;149:R59–R70. |
[24] | Kurman RJ, Shih IeM. The Dualistic Model of Ovarian Carcinogenesis: Revisited, Revised, and Expanded. Am J Pathol 2016;186:733–47. |
[25] | Virant-Klun I. Functional Testing of Primitive Oocyte-like Cells Developed in Ovarian Surface Epithelium Cell Culture from Small VSEL-like Stem Cells: Can They Be Fertilized One Day? Stem Cell Rev Rep 2018;14:715–21. |
[26] | Parte S, Bhartiya D, Telang J, Daithankar V, Salvi V, Zaveri K, Hinduja I. Detection, characterization, and spontaneous differentiation in vitro of very small embryonic-like putative stem cells in adult mammalian ovary. Stem Cells Dev 2011;20:1451–64. |
[27] | Parte S, Patel H, Sriraman K, Bhartiya D. Isolation and characterization of stem cells in the adult mammalian ovary. Methods Mol Biol 2015;1235:203–29. |
[28] | Kenda Suster N, Virant-Klun I. Presence and role of stem cells in ovarian cancer. World J Stem Cells 2019;11:383–97. |
[29] | Virant-Klun I, Skerl P, Novakovic S, Vrtacnik-Bokal E, Smrkolj S. Similar Population of CD133+ and DDX4+ VSEL-Like Stem Cells Sorted from Human Embryonic Stem Cell, Ovarian, and Ovarian Cancer Ascites Cell Cultures: The Real Embryonic Stem Cells? Cells 2019;8:pii.E706. |
[30] | Ng A, Barker N. Ovary and fimbrial stem cells: biology, niche and cancer origins. Nat Rev Mol Cell Biol 2015;16:625–38. |
[31] | Parte S, Virant-Klun I, Patankar M, Batra SK, Straughn A, Kakar SS. PTTG1: a Unique Regulator of Stem/Cancer Stem Cells in the Ovary and Ovarian Cancer. Stem Cell Rev Rep 2019;15:866–79. |
[32] | Parte SC, Smolenkov A, Batra SK, Ratajczak MZ, Kakar SS. Ovarian Cancer Stem Cells: Unraveling a Germline Connection. Stem Cells Dev 2017;26:1781–803. |
[33] | Parte SC, Batra SK, Kakar SS. Characterization of stem cell and cancer stem cell populations in ovary and ovarian tumors. J Ovarian Res 2018;11:69. |
[34] | Kakar SS, Ratajczak MZ, Powell KS, Moghadamfalahi M, Miller DM, Batra SK, Singh SK. Withaferin A alone and in combination with cisplatin suppresses growth and metastasis of ovarian cancer by targeting putative cancer stem cells. PLoS One 2014;9:e107596. |
[35] | Abubaker K, Latifi A, Luwor R, Nazaretian S, Zhu H, Quinn MA, Thompson EW, Findlay JK, Ahmed N. Short-term single treatment of chemotherapy results in the enrichment of ovarian cancer stem cell-like cells leading to an increased tumor burden. Mol Cancer 2013;12:24. |
[36] | Liu L, Cai S, Han C, et al. ALDH1A1 Contributes to PARP Inhibitor Resistance via Enhancing DNA Repair in BRCA2-/- Ovarian Cancer Cells. Mol Cancer Ther 2020;19:199–210. |
[37] | Zhang S, Cui B, Lai H, et al. Ovarian cancer stem cells express ROR1, which can be targeted for anti-cancer-stem-cell therapy. Proc Natl Acad Sci U S A 2014;111:17266–71. |
[38] | Raghavan S, Mehta P, Ward MR, et al. Personalized Medicine-Based Approach to Model Patterns of Chemoresistance and Tumor Recurrence Using Ovarian Cancer Stem Cell Spheroids. Clin Cancer Res 2017;23:6934–45. |
[39] | Roy L, Cowden Dahl KD. Can Stemness and Chemoresistance Be Therapeutically Targeted via Signaling Pathways in Ovarian Cancer? Cancers 2018;10:241 pii.E241. |
[40] | Deng J, Wang L, Chen H, et al. Targeting epithelial-mesenchymal transition and cancer stem cells for chemoresistant ovarian cancer. Oncotarget 2016;7:55771–88. |
[41] | Hu W, Liu T, Ivan C, Sun Y, Huang J, et al. Notch3 pathway alterations in ovarian cancer. Cancer Res 2014;74:3282–93. |
[42] | Hoarau-Véchot J, Touboul C, Halabi N, et al. Akt-activated endothelium promotes ovarian cancer proliferation through notch activation. J Transl Med 2019;17:194. |
[43] | Motohara T, Katabuchi H. Ovarian Cancer Stemness: Biological and Clinical Implications for Metastasis and Chemotherapy Resistance. Cancers 2019;11:907. |
[44] | Zong X, Nephew KP. Ovarian Cancer Stem Cells: Role in metastasis and opportunity for therapeutic targeting. Cancers 2019;11:pii.E934. |
[45] | Bothun AM, Woods DC. Dynamics of WNT signaling components in the human ovary from development to adulthood. Histochem Cell Biol 2019;151:115–23. |
[46] | Hernandez Gifford JA. The role of WNT signaling in adult ovarian folliculogenesis. Reproduction 2015;150:R137–48. |
[47] | Coopes A, Henry CE, Llamosas E, Ford CE. An update of Wnt signalling in endometrial cancer and its potential as a therapeutic target. Endocrine Relat Cancer 2018;25:R647–62. |
[48] | Yang L, Shi P, Zhao G, Xu J, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther 2020;5:8. |
[49] | Ramakrishnan AB, Cadigan KM. Wnt target genes and where to find them. F1000 Res 2017;6:746. |
[50] | Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene 2017;36:1461–73. |
[51] | Schindler AJ, Watanabe A, Howell SB. LGR5 and LGR6 in stem cell biology and ovarian cancer. Oncotarget 2017;9:1346–55. |
[52] | Ng A, Tan S, Singh G, Rizk P, et al. Lgr5 marks stem/progenitor cells in ovary and tubal epithelia. Nat Cell Biol 2014;16:745–57. |
[53] | de Sousa E Melo F, Vermeulen L. Wnt Signaling in Cancer Stem Cell Biology. Cancers 2016;8:pii.E60. |
[54] | Ruan X, Liu A, Zhong M, Wei J, Zhan W, Rong Y, Liu W, Li M, Qing X, Chen G, Li R, Liao Y, Liu Q, Zhang X, Ren D, Wang Y. Silencing LGR6 Attenuates Stemness and Chemoresistance via Inhibiting Wnt/β-Catenin Signaling in Ovarian Cancer. Mol Ther Oncolytics 2019;14:94–06. |
[55] | Anastas JN, Moon RT. WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer 2013;13:11–26. |
[56] | Raghavan S, Mehta P, Xie Y, Lei YL, Mehta G. Ovarian cancer stem cells and macrophages reciprocally interact through the WNT pathway to promote pro-tumoral and malignant phenotypes in 3D engineered microenvironments. J Immunother Cancer 2019;7:190. |
[57] | Pan H, Kim E, Rankin G, Rojanasakul Y, Tu Y, Chen Y. Theaflavin-3,3′-digallate inhibits ovarian cancer stem cells via suppressing Wnt/β-Catenin signaling pathway. Journal of Funct Foods 2018;50:1–7. |
[58] | Bhateja P, Cherian M, Majumder S, Ramaswamy B. The Hedgehog Signaling Pathway: A Viable Target in Breast Cancer? Cancers 2019;11:pii.E1126. |
[59] | Zhu Q, Shen Y, Chen X, He J, Liu J, Zu X. Self-Renewal Signalling Pathway Inhibitors: Perspectives on Therapeutic Approaches for Cancer Stem Cells. Onco Targets Ther 2020;13:525–40. |
[60] | Wang D, Nagle PW, Wang HH, et al. Hedgehog Pathway as a Potential Intervention Target in Esophageal Cancer. Cancers 2019;11:pii.E821. |
[61] | Chen Y, Bieber MM, Teng NN. Hedgehog signaling regulates drug sensitivity by targeting ABC transporters ABCB1 and ABCG2 in epithelial ovarian cancer. Mol Carcinog 2014;53:625–34. |
[62] | Zeng C, Chen T, Zhang Y, et al. Hedgehog signaling pathway regulates ovarian cancer invasion and migration via adhesion molecule CD24. J Cancer 2017;8:786–92. |
[63] | Liao X, Siu MK, Au CW, et al. Aberrant activation of hedgehog signaling pathway in ovarian cancers: effect on prognosis, cell invasion and differentiation. Carcinogenesis 2009;30:131–40. |
[64] | Chen X, Horiuchi A, Kikuchi N, et al. Hedgehog signal pathway is activated in ovarian carcinomas, correlating with cell proliferation: it's inhibition leads to growth suppression and apoptosis. Cancer Sci 2007;98:68–77. |
[65] | Liu HY, Dong Z. Gli3 silencing enhances cyclopamine suppressive effects on ovarian cancer. Onco Targets Ther 2014;7:2007–11. |
[66] | Bhattacharya R, Kwon J, Ali B, Wang E, Patra S, Shridhar V, Mukherjee P. Role of hedgehog signaling in ovarian cancer. Clin Cancer Res 2008;14:7659–66. |
[67] | Cortes JE, Gutzmer R, Kieran MW, Solomon JA. Hedgehog signaling inhibitors in solid and hematological cancers. Cancer Treat Rev 2019;76:41–50. |
[68] | Song X, Yan L, Lu C, et al. Activation of hedgehog signaling and its association with cisplatin resistance in ovarian epithelial tumors. Oncol Lett 2018;15:5569–76. |
[69] | Lengyel CG, Altuna SC, Habeeb BS, Trapani D, Khan SZ. The Potential of PI3K/AKT/mTOR signaling as a druggable target for endometrial and ovarian carcinomas. Curr Drug Targets 2019; Nov 20. |
[70] | Wei X, Jia Y, Lou H, et al. Targeting YAP suppresses ovarian cancer progression through regulation of the PI3K/Akt/mTOR pathway. Oncol Rep 2019;42:2768–76. |
[71] | Etemadmoghadam D, Bowtell D. AKT1 gene amplification as a biomarker of treatment response in ovarian cancer: mounting evidence of a therapeutic target. Gynecol Oncol 2014;135:409–10. |
[72] | Lee S, Choi EJ, Jin C, Kim DH. Activation of PI3K/Akt pathway by PTEN reduction and PIK3CA mRNA amplification contributes to cisplatin resistance in an ovarian cancer cell line. Gynecol Oncol 2005;97:26–34. |
[73] | Sun M, Wang G, Paciga JE, et al. AKT1/PKBalpha kinase is frequently elevated in human cancers and its constitutive activation is required for oncogenic transformation in NIH3T3 cells. Am J Pathol 2001;159:431–37. |
[74] | Altomare DA, Wang HQ, Skele KL, et al. AKT and mTOR phosphorylation is frequently detected in ovarian cancer and can be targeted to disrupt ovarian tumor cell growth. Oncogene 2004;23:5853–57. |
[75] | Deng J, Bai X, Feng X, Ni J, Beretov J, Graham P, Li Y. Inhibition of PI3K/Akt/mTOR signaling pathway alleviates ovarian cancer chemoresistance through reversing epithelial-mesenchymal transition and decreasing cancer stem cell marker expression. BMC Cancer 2019;19:618. |
[76] | Thakur B, Ray P. Cisplatin triggers cancer stem cell enrichment in platinum-resistant cells through NF-κB-TNFα-PIK3CA loop. J Exp Clin Cancer Res 2017;36:164 |
[77] | Ali AY, Kim JY, Pelletier JF, Vanderhyden BC, Bachvarov DR, Tsang BK. Akt confers cisplatin chemoresistance in human gynecological carcinoma cells by modulating PPM1D stability. Mol Carcinog 2015;54:1301–14. |
[78] | Regad T. Targeting RTK signaling pathways in cancer. Cancers 2015;7:1758–84. |
[79] | Mazzoldi EL, Pavan S, Pilotto G, et al. 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. |
[80] | Ruan Z, Yang X, Cheng W. OCT4 accelerates tumorigenesis through activating JAK/STAT signaling in ovarian cancer side population cells. Cancer Manag Res 2018;11:389–99. |
[81] | Muller R. JAK inhibitors in 2019, synthetic review in 10 points. European Journal of Internal Medicine 2019;66:9–17. |
[82] | Jin W. Role of JAK/STAT3 signaling in the regulation of metastasis, the transition of cancer stem cells, and chemoresistance of cancer by epithelial-mesenchymal transition. Cells 2020;9:pii.E217. |
[83] | Yadav A, Kumar B, Datta J, Teknos TN, Kumar P. IL-6 promotes head and neck tumor metastasis by inducing epithelial-mesenchymal transition via the JAK-STAT3-SNAIL signaling pathway. Mol Cancer Res 2011;9:1658–67. |
[84] | Xiong H, Hong J, Du W, Lin YW, et al. Roles of STAT3 and ZEB1 proteins in E-cadherin down-regulation and human colorectal cancer epithelial-mesenchymal transition. J Biol Chem 2012;287:5819–32. |
[85] | Gong C, Shen J, Fang Z, Qiao L, Feng R, Lin X, Li S. Abnormally expressed JunB transactivated by IL-6/STAT3 signaling promotes uveal melanoma aggressiveness via epithelial-mesenchymal transition. Biosci Rep 2018;38:pii.BSR20180532. |
[86] | Shibue T, Weinberg RA. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat Rev Clin Oncol 2017;14:611–29. |
[87] | Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol 2019;20:69–84. |
[88] | Musumeci F, Greco C, Giacchello I, et al. An Update on JAK Inhibitors. Curr Med Chem 2019;26:1806–32. |
[89] | Koschorke A, Faraci S, Giani D, et al. Phenethyl isothiocyanate hampers growth and progression of HER2-positive breast and ovarian carcinoma by targeting their stem cell compartment. Cell Oncol 2019;42:815–28. |
[90] | Choi YJ, Gurunathan S, Kim JH. Graphene Oxide–Silver Nanocomposite Enhances Cytotoxic and Apoptotic Potential of Salinomycin in Human Ovarian Cancer Stem Cells (OvCSCs): A Novel Approach for Cancer Therapy. Int J Mol Sci 2018;19:710. |
[91] | Udoh K, Parte S, Carter K, Mack A, Kakar SS. Targeting of Lung Cancer Stem Cell Self-Renewal Pathway by a Small Molecule Verrucarin J. Stem Cell Rev Rep 2019;15:601–11. |
[92] | Carter K, Rameshwar P, Ratajczak MZ, Kakar SS. Verrucarin J inhibits ovarian cancer and targets cancer stem cells. Oncotarget 2017;8:92743–56. |
[93] | Kakar SS, Worth CA, Wang Z, Carter K, Ratajczak M, Gunjal P. DOXIL when combined with Withaferin A (WFA) targets ALDH1 positive cancer stem cells in ovarian cancer. J Cancer Stem Cell Res 2016;4:e1002. |
[94] | Kakar SS, Parte S, Carter K, et al. Withaferin A (WFA) inhibits tumor growth and metastasis by targeting ovarian cancer stem cells. Oncotarget 2017;8:74494–05. |
[95] | Zamarin D. Novel therapeutics: response and resistance in ovarian cancer. Int J Gynec Cancer 2019;29:s16–s21. |
[96] | Lee J-M, Nair J, Zimmer A, et al. Prexasertib, a cell cycle checkpoint kinase 1 and 2 inhibitor, in BRCA wild-type recurrent high-grade serous ovarian cancer: a first-in-class proof-of-concept phase 2 study. Lancet Oncol 2018;19:207–15. |
[97] | Matulonis UA, Shapira-Frommer R, Santin A, et al. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase 2 KEYNOTE-100 study. Ann Oncol 2019;30:1080–87. |
[98] | Hamanishi J, Mandai M, Ikeda T, et al. Safety and antitumor activity of anti-PD-1 antibody, nivolumab, in patients with platinum-resistant ovarian cancer. J Clin Oncol 2015;33:4015–22. |
[99] | Pujade-Lauraine E, Hilpert F, Weber B, et al. Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: the AURELIA open-label randomized phase III trial. J Clin Oncol 2014;32:1302–8. |
[100] | Young AN, Herrera D, Huntsman AC, et al. Phyllanthusmin derivatives induce apoptosis and reduce tumor burden in highgrade serous ovarian cancer by late-stage autophagy inhibition. Mol Cancer Ther 2018;17:2123–35. |
[101] | Aust S, Schwameis R, Gagic T, et al. Precision medicine tumor boards: clinical applicability of personalized treatment concepts in ovarian cancer. Cancers 2020;12:pii:E548. |
[102] | Ahmed N, Kadife E, Raza A, Short M, Jubinsky PT, Kannourakis G. Ovarian Cancer, Cancer Stem Cells and Current Treatment Strategies: A Potential Role of Magmas in the Current Treatment Methods. Cells 2020;9:719. |
[103] | Keyvani V, Farshchian M, Esmaeili SA, et al. Ovarian cancer stem cells and targeted therapy. J Ovarian Res 2019;12:120. Published 2019 Dec 6. doi:10.1186/s13048-019-0588-z. |
[104] | Meirelles K, Benedict LA, Dombkowski D, et al. Human ovarian cancer stem/progenitor cells are stimulated by doxorubicin but inhibited by Mullerian inhibiting substance. Proc Natl Acad Sci U S A 2012;109:2358–63. |
[105] | Burgos-Ojeda D, Rueda BR, Buckanovich RJ. Ovarian cancer stem cell markers: prognostic and therapeutic implications. Cancer Lett 2012;322:1–7. |
[106] | Nakamura K, Terai Y, Tanabe A, 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;37:3189–200. |
[107] | Jaggupilli A, Elkord E. Significance of CD44 and CD24 as cancer stem cell markers: an enduring ambiguity. Clin Dev Immunol 2012;2012:708036. |
[108] | Choi YL, Kim SH, Shin YK, Hong YC, Lee SJ, Kang SY, Ahn G. Cytoplasmic CD24 expression in advanced ovarian serous borderline tumors. Gynecol Oncol 2005;97:379–86. |
[109] | Su D, Deng H, Zhao X, Zhang X, Chen L, Chen X, Li Z, Bai Y, Wang Y, Zhong Q, Yi T, Qian Z, Wei Y. Targeting CD24 for treatment of ovarian cancer by short hairpin RNA. Cytotherapy 2009;11:642–52. |
[110] | Gao MQ, Choi YP, Kang S, Youn JH, Cho NH. CD24+ cells from hierarchically organized ovarian cancer are enriched in cancer stem cells. Oncogene 2010;29:2672–80. |
[111] | Hollingsworth HC, Kohn EC, Steinberg SM, Rothenberg ML, Merino MJ. Tumor angiogenesis in advanced stage ovarian carcinoma. Am J Pathol 1995;147:33–41. |
[112] | Cwiklnska A, Sobstyl M, Kwasniewski W, Bednarek W. Microtissue density prognostic factor evaluation based on antigens CD34 and CD 105 in ovarian cancer patients. Ann Agric Enviorn Med 2013;20:838–42. |
[113] | Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, Yan PS, Huang TH, Nephew KP. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res 2008;68:4311–20. |
[114] | Casagrande F, Cocco E, Bellone S, et al. Eradication of chemotherapy-resistant CD44+ human ovarian cancer stem cells in mice by intraperitoneal administration of clostridium perfringens enterotoxin. Cancer 2011;117:5519–28. |
[115] | 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;44:1882–9. |
[116] | Meng E, Long B, Sullivan P, McClellan S, et al. CD44+/CD24- ovarian cancer cells demonstrate cancer stem cell properties and correlate to survival. Clin Exp Metastasis 2012;29:939–48. |
[117] | Stemberger-Papić S, Vrdoljak-Mozetic D, Ostojić DV, et al. Expression of CD133 and CD117 in 64 Serous Ovarian Cancer Cases. Coll Antropol 2015;39:745–53. |
[118] | 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. |
[119] | Conic I, Stanojevic Z, Jankovic Velickovic L, et al. Epithelial ovarian cancer with CD117 phenotype is highly aggressive and resistant to chemotherapy. J Obstet Gynaecol Res 2015;41:1630–7. |
[120] | Helland Å, Anglesio MS, George J, et al. Deregulation of MYCN, LIN28B and LET7 in a molecular subtype of aggressive high-grade serous ovarian cancers. PLoS One 2011;6:e18064. |
[121] | Schilder RJ, Sill MW, Lee RB, et al. Phase II evaluation of imatinib mesylate in the treatment of recurrent or persistent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group Study. J Clin Oncol 2008;26:3418–25. |
[122] | Raspollini MR, Amunni G, Villanucci A, et al. c-KIT expression and correlation with chemotherapy resistance in ovarian carcinoma: an immunocytochemical study. Ann Oncol 2004;15:594–7. |
[123] | Curley MD, Therrien VA, Cummings CL, et al. CD133 expression defines a tumor initiating cell population in primary human ovarian cancer. Stem Cells 2009;27:2875–83. |
[124] | Skubitz AP, Taras EP, Boylan KL, et al. Targeting CD133 in an in vivo ovarian cancer model reduces ovarian cancer progression. Gynecol Oncol 2013;130:579–87. |
[125] | Baba T, Convery PA, Matsumura N, et al. Epigenetic regulation of CD133 and tumorigenicity of CD133+ ovarian cancer cells. Oncogene 2009;28:209–18. |
[126] | Zhang J, Guo X, Chang DY, Rosen DG, Mercado-Uribe I, Liu J. CD133 expression associated with poor prognosis in ovarian cancer. Mod Pathol 2012;25:456–64. |
[127] | Klapdor R, Wang S, Hacker U, et al. Improved Killing of Ovarian Cancer Stem Cells by Combining a Novel Chimeric Antigen Receptor-Based Immunotherapy and Chemotherapy. Hum Gene Ther 2017;28:886–96. |
[128] | Zheng J, Zhao S, Yu X, Huang S, Liu HY. Simultaneous targeting of CD44 and EpCAM with a bispecific aptamer effectively inhibits intraperitoneal ovarian cancer growth. Theranostics 2017;7:1373–88. |
[129] | Tayama S, Motohara T, Narantuya D, et al. The impact of EpCAM expression on response to chemotherapy and clinical outcomes in patients with epithelial ovarian cancer. Oncotarget 2017;8:44312–25. |
[130] | Zhang H, Qiu J, Ye C, Yang D, Gao L, Su Y, Tang X, Xu N, Zhang D, Xiong L, Mao Y, Li F, Zhu J. ROR1 expression correlated with poor clinical outcome in human ovarian cancer. Sci Rep 2014;4:5811. |
[131] | Wang Y, Shao F, Chen L. ALDH1A2 suppresses epithelial ovarian cancer cell proliferation and migration by downregulating STAT3. Onco Targets Ther 2018;11:599–608. |
[132] | Wang YC, Yo YT, Lee HY, et al. ALDH1-bright epithelial ovarian cancer cells are associated with CD44 expression, drug resistance, and poor clinical outcome. Am J Pathol 2012;180:1159–69. |
[133] | Januchowski R, Wojtowicz K, Sterzyſska K, et al. Inhibition of ALDH1A1 activity decreases expression of drug transporters and reduces chemotherapy resistance in ovarian cancer cell lines. Int J Biochem Cell Biol 2016;78:248–59. |
[134] | Landen CN, Goodman B, Katre AA, et al. Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer. Mol Cancer Ther 2010;9:3186–99. |
[135] | Ng PM, Lufkin T. Embryonic stem cells: protein interaction networks. Biomol Concepts 2011;2:13–25. |
[136] | Yu Z, Pestell TG, Lisanti MP, Pestell RG. Cancer stem cells. Int J Biochem Cell Biol 2012;44:2144–51. |
[137] | Wen Y, Hou Y, Huang Z, Cai J, Wang Z. SOX2 is required to maintain cancer stem cells in ovarian cancer. Cancer Sci 2017;108:719–31. |
[138] | Lee M, Nam EJ, Kim SW, Kim S, Kim JH, Kim YT. Prognostic impact of the cancer stem cell-related marker NANOG in ovarian serous carcinoma. Int J Gynecol Cancer 2012;22:1489–96. |
[139] | Pan Y, Jiao J, Zhou C, Cheng Q, Hu Y, Chen H. Nanog is highly expressed in ovarian serous cystadenocarcinoma and correlated with clinical stage and pathological grade. Pathobiology 2010;77:283–8. |
[140] | Qin S, Li Y, Cao X, Du J, Huang X. NANOG regulates epithelial-mesenchymal transition and chemoresistance in ovarian cancer. Biosci Rep 2017;37:pii. BSR20160247. |
[141] | Di J, Duiveman-de Boer T, Zusterzeel PL, Figdor CG, Massuger LF, 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 (Dordr) 2013;36:363–74. |
[142] | Ning YX, Luo X, Xu M, Feng X, Wang J. Let-7d increases ovarian cancer cell sensitivity to a genistein analog by targeting c-Myc. Oncotarget 2017;8:74836–45. |
[143] | Hu L, McArthur C, Jaffe RB. Ovarian cancer stem-like side-population cells are tumourigenic and chemoresistant. Br J Cancer 2010;102:1276–83. |
[144] | Dou J, Jiang C, Wang J, et al. Using ABCG2-molecule-expressing side population cells to identify cancer stem-like cells in a human ovarian cell line. Cell Biol Int 2011;35:227–34. |
[145] | Kobayashi Y, Seino K, Hosonuma S, et al. Side population is increased in paclitaxel-resistant ovarian cancer cell lines regardless of resistance to cisplatin. Gynecol Oncol 2011;121:390–4. |
[146] | Hashimoto H, Sudo T, Mikami Y, et al. Germ cell specific protein VASA is over-expressed in epithelial ovarian cancer and disrupts DNA damage-induced G2 checkpoint. Gynecol Oncol 2008;111:312–9. |
[147] | Fraunhoffer NA, Meilerman Abuelafia A, Stella I, Galliano S, Barrios M, Vitullo AD. Identification of germ cell-specific VASA and IFITM3 proteins in human ovarian endometriosis. J Ovarian Res 2015;8:66. |
[148] | Wolfe AD, Rodriguez AM, Downs KM. STELLA collaborates in distinct mesendodermal cell subpopulations at the fetal-placental interface in the mouse gastrula. Dev Biol 2017;425:44–57. |
[149] | Virant-Klun I, Stimpfel M. Novel population of small tumour-initiating stem cells in the ovaries of women with borderline ovarian cancer. Sci Rep 2016;6:34730. |