Next Article in Journal
Resistance to Chemotherapeutic 5-Fluorouracil Conferred by Modulation of Heterochromatic Integrity through Ino80 Function in Fission Yeast
Previous Article in Journal
Investigation of Molecular Interactions Mechanism of Pembrolizumab and PD-1
Previous Article in Special Issue
Protein Posttranslational Modification in Stemness Remodeling and Its Emerging Role as a Novel Therapeutic Target in Gastrointestinal Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Deciphering Common Traits of Breast and Ovarian Cancer Stem Cells and Possible Therapeutic Approaches

1
Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
2
Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(13), 10683; https://doi.org/10.3390/ijms241310683
Submission received: 6 May 2023 / Revised: 21 June 2023 / Accepted: 23 June 2023 / Published: 26 June 2023
(This article belongs to the Special Issue New Biomarkers and Therapy for Cancer Stem Cells)

Abstract

:
Breast cancer (BC) and ovarian cancer (OC) are among the most common and deadly cancers affecting women worldwide. Both are complex diseases with marked heterogeneity. Despite the induction of screening programs that increase the frequency of earlier diagnosis of BC, at a stage when the cancer is more likely to respond to therapy, which does not exist for OC, more than 50% of both cancers are diagnosed at an advanced stage. Initial therapy can put the cancer into remission. However, recurrences occur frequently in both BC and OC, which are highly cancer-subtype dependent. Therapy resistance is mainly attributed to a rare subpopulation of cells, named cancer stem cells (CSC) or tumor-initiating cells, as they are capable of self-renewal, tumor initiation, and regrowth of tumor bulk. In this review, we will discuss the distinctive markers and signaling pathways that characterize CSC, their interactions with the tumor microenvironment, and the strategies they employ to evade immune surveillance. Our focus will be on identifying the common features of breast cancer stem cells (BCSC) and ovarian cancer stem cells (OCSC) and suggesting potential therapeutic approaches.

1. Introduction

Breast cancer (BC) and ovarian cancer (OC) are the most common and sixth most common cancers, respectively, and the first and fourth leading causes, respectively, of cancer-related deaths among women under the age of 60 [1,2]. Both are also the deadliest women’s cancers, BC altogether and OC among gynecologic malignancies.
Interestingly, although BC and OC are two different cancers that arise in different tissues of the body, women with breast cancer susceptibility gene type 1 and type 2 (BRCA1 and BRCA2) mutations have a higher risk of developing BC and/or OC. The specific factors that determine whether a woman with a BRCA1/2 mutation will develop breast or ovarian cancer are not fully understood. Since BRCA1 and BRCA2 are involved in DNA repair, mutations in these genes can cause genetic errors leading to cancer. Yet, the specific effects of BRCA1/2 mutations on cancer development can vary depending on the type of mutation, its location within the gene, and other genetic and environmental factors [3,4]. Cancers related to BRCA1/2 mutations comprise about 5–7% of BC and 20–25% of OC cases [5]. The cumulative risk of developing BC or OC by age 80 is 72% for BRCA1 and 69% for BRCA2 mutation carriers in BC, whereas for OC it is 44% for BRCA1 and 17% for BRCA2 mutation carriers [6] and estimated to be even higher [7]. While these women may never develop cancer, as women without a BRCA1/2 mutation may also develop BC or OC, risk-reducing surgeries such as bilateral mastectomy, bilateral salpingo-oophorectomy, or a combination of both procedures are suggested to reduce the risk of developing BC or OC. They have been shown to reduce the risk of cancer that arises in the removed tissue by approximately 90%. While salpingo-oophorectomy has also been shown to reduce the risk of BC [8,9,10,11,12], the data should be evaluated cautiously [13]. Indeed, women over 45 with BRCA1/2 mutations have a three-fold increased risk of BC if taking hormone replacement therapy after salpingo-oophorectomy [14]. Therefore, the decision for women with a BRCA1/2 mutation to undergo these surgeries should be based on individual factors such as age, family history, personal preferences, and overall health, which requires a patient-oriented approach.
The majority of BC (~99%) and OC (~90%) are of epithelial origin, arising from epithelial cells lining the lobules and terminal ducts in the breast or surrounding the ovaries. OC is histopathologically classified into five main types: high-grade serous carcinoma (HGSOC), low-grade serous carcinoma, endometrioid carcinoma, clear-cell carcinoma, and mucinous carcinoma [15]. BC is highly heterogeneous and mainly classified according to molecular features (hormone receptors, HER2, and Ki67) into luminal A, luminal B, human epidermal growth factor receptor 2 (HER2)- enriched, and basal-like, which is a subtype of triple negative BC (TNBC) [16]. Expression of hormone receptors can also be used for clinically applicable classification of HGSOC [17].
Cancer recurrence is a major concern for patients with BC and OC, and several factors influence its occurrence, such as age, tumor grade, tumor size, axillary nodal involvement, and hormone receptor status [18]. Recurrence is discovered in 25% of early detected OC and in 80% of more advanced OC [19], while in BC it varies between 10 and 41%, depending on subtype and other factors [20,21]. Additionally, the five-year survival rate for recurrent OC is less than 30% [22], and 24% for metastatic BC [23]. Therefore, it is crucial to gain a better understanding of the mechanisms that drive tumor survival and expansion. Cancer progression and recurrence are mainly attributed to subpopulations of cancer cells that are resistant to cancer therapy and have the ability to grow the tumor. These cells are referred to as cancer stem cells (CSC) or tumor-initiating cells.
In this review, we will discuss the distinctive markers and signaling pathways that characterize CSC, as well as their interactions with the tumor microenvironment (TME) and the strategies they employ to evade immune surveillance. Our focus will be on identifying the common traits of breast cancer stem cells (BCSC) and ovarian cancer stem cells (OCSC), along with potential therapeutic approaches.

2. Cancer Development and Progression

Ovarian cancer is characterized by rapid proliferative growth and great metastatic potential. Indeed, epithelial–mesenchymal transition (EMT) is a characteristic of ovarian epithelium cells. During this process, epithelial cells depolarize, disassemble cell–cell contacts, acquire fibroblast-like morphological features, and adopt an invasive, migratory phenotype [24]. It was suggested that mutations in tumor protein P53 (TP53), RB transcriptional corepressor 1 (RB1), and phosphatase and tensin homolog PTEN, are necessary for initiating the transformation of ovarian surface epithelium stem cells [25].
If OC is not detected at an early stage (Stage I), before cancer spreads beyond the ovaries, the survival rate of patients decreases significantly. OC can reach stage II—metastasized to pelvic organs (the uterus, fallopian tube, bladder, and rectum), stage III—metastasized across the pelvic cavity to abdominal organs (the omentum, small intestine, and retroperitoneal lymph nodes), or stage IV—metastasized beyond the peritoneal cavity to distant parenchymal organs (liver and lung), lymph nodes, bones, and brain [15,26]. Non-specific symptoms, which usually occur when cancer has already progressed to an advanced stage, are the main reason for the high mortality rates of OC. Unfortunately, more than 70% of OC cases are diagnosed at a late stage [27]. In the case of epithelial ovarian cancer (EOC), metastasis can occur via three different routes: the transcoelomic, hematogenous, or lymphatic routes. Transcoelomic metastasis is the most common [27,28]. Transcoelomic metastasis occurs through the build-up of ascites fluid within the peritoneal cavity. In this process, metastatic OC cells undergo EMT and enter the ascites fluid by shedding from the primary tumor as single cells or in groups as spheroids and spread passively [29,30]. Once a colony is established at a secondary site, the cells can undergo a mesenchymal-to-epithelial transition (MET) and begin to grow rapidly. Research has shown that ascites is enriched in OCSC [31,32], which have the ability to self-renew, differentiate, and resist anoikis [33], thus playing a pivotal role in the formation of multicellular spheroids during the transcoelomic peritoneal dissemination [29].
Breast cancer typically originates from the epithelial cells within the ducts (85%) or lobules (15%) of the breast glandular tissue [34]. Recent studies suggest the existence of a common epithelial progenitor or stem cell located at the terminal duct lobular units (TDLUs), which exhibits a high degree of phenotypic plasticity and is responsible for the development of BC [35]. Initially, BC growth is limited to the duct or lobule and is considered “in situ”. At this stage, the cancer is typically asymptomatic and has minimal potential for metastasis. However, over time, these in situ cancers may progress and invade the surrounding breast tissue, leading to invasive BC. If left untreated, invasive BC can spread to nearby lymph nodes (regional metastasis) or other organs in the body (distant metastasis). Cancer metastasis is a complex process that involves multiple steps, including tumor cell dissociation, neoangiogenesis, intravasation, survival and diffusion through the circulation, adhesion to target tissues, extravasation, and establishment of metastatic foci. BC metastasis follows an organ-specific pattern, with a preference for bone, liver, lung, and brain. Metastatic BC is highly heterogeneous, making it challenging to identify risk factors and treatments [36]. Genomic analysis of synchronous primary BC and metastases which were not exposed to therapy revealed differences in their repertoire of somatic mutations. While the driver genes including TP53, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), and GATA binding protein 3 (GATA3) were common in both sites, EMT-related genes were enriched in or restricted to metastases. This mutational difference contributes to spatial intratumor heterogeneity [37].
BC and OC share approximately 50% of their top mutated genes. Integrated molecular analysis revealed a great difference between BC subtypes and many commonalities between HGSOC and basal-like TNBC, indicating related etiology [38]. The main commonalities include alterations in TP53 and BRCA1 with additional PTEN loss, which is an early initiating event in BRCA1-associated basal-like BC [39] and a known putative driver in OC [40]. In addition, the synergistic effect of two BRCA1 associated RING domain 1 (BARD1) mutations contributes to the tumorigenesis of BRCA1-associated BC and OC [41], and four new shared driver genes for BC and OC have recently been proposed [42].

3. Biomarkers of Breast Cancer Stem Cells (BCSC) and Ovarian Cancer Stem Cells (OCSC)

BC and OC are highly heterogenous, comprising genetically and molecularly diverse cells. Different theories explain how cancer develops and progresses. According to the stochastic theory, all cancer cells can contribute by accumulating random mutations, while the hierarchy theory suggests that CSC control proliferation and progression [43].
CSC possess the ability to self-renew and undergo symmetrical/asymmetrical division, which contributes to tumor heterogeneity, tumorigenicity, dissemination, and resistance to therapy [44]. While chemotherapy or radiotherapy can eliminate a large proportion of tumor cells, the presence of resistant CSC underscores the need for more extensive research to discover more effective treatments. Due to low abundance in tumor bulk (estimated 0.1–1% in BC), identifying CSC depends largely on specific markers [45].
CD44, CD24, CD133, aldehyde dehydrogenase 1 (ALDH1), CD326 (EpCAM), CD338, and the pluripotency markers NANOG, OCT4, and SOX2 are shared among BCSC and OCSC, although there are others (Table 1).
In a seminal research study, Al-Hajj et al. identified BCSC by showing that 100 CD44+/CD24−/low-BC cells formed tumors in immune-deficient NOD/SCID mice, while even 100 times more cells of a different phenotype failed to do so. While these BCSC are a minority, they can generate a diverse range of differentiated cells that form the bulk of the tumor [45]. CD44 is a transmembrane glycoprotein that binds hyaluronic acid and other extracellular ligands, facilitating activation of diverse signaling pathways to regulate cell proliferation, adhesion, migration, survival, invasion, and EMT [46,47]. CD24 is a cell-surface protein involved in cell-matrix and cell–cell communication [48], the expression of which is a prognostic marker in BC as well as in OC [49,50]. While in BC, the expression of CD24 correlates with more differentiated, epithelial characteristics and a lower expression is characteristic of basal-like BC [51], in OC, the expression of CD24 is associated with higher metastatic traits, chemoresistance, and poor prognosis [52,53]. Differential expression of CD24 and CD44 between OC and normal epithelium highlights CD24 as a specific marker for the recurrence of OC [54], whereas CD44+ cells have been shown to be resistant to standard chemotherapy used for OC treatment, such as carboplatin and paclitaxel [55]. High expression of CD44 in BC is mainly linked with metastasis and not tumor initiation [56]. Therefore, inhibition of CD24 and CD44 might be therapeutically relevant. Knockdown of CD24 in SKOV3 cells suppressed OC growth in mice in vivo [54]. An anti-CD44 antibody approach reduced invasion and cell migration of ER+ BC cell lines and TNBC cell lines [57]. Indeed, BIWA-4 (bivatuzumab), an anti-CD44 antibody, is currently in preclinical trials for patients with head and neck tumors, indicating the interest in CD44 inhibition as a potential cancer therapy [58].
The CD44+/CD24−/low phenotype is shown to distinguish both BCSC and OCSC. Meng et al. have demonstrated that the CD44+/CD24− phenotype serves as a marker for OCSC, as these cells exhibit increased differentiation, invasion, and resistance to chemotherapy [59]. Similarly, CD44+/CD24−/low cells show increased chemoresistance [60] and radioresistance [61] in BC. In addition, the CD44+/CD24−/low phenotype may not necessarily correlate with clinical outcomes due to the tumor-type-dependent frequency of BCSC [51,62,63]. However, when a tumor has more than 10% of BCSC, it is correlated with worse clinical outcomes [62]. High CSC plasticity, the ability to transform and change in response to various signals from the TME, including hypoxia, transforming growth factor beta (TGFβ, and epidermal growth factor (EGF)) contribute to tumor growth [64]. The shift from CD44−/low to CD44+/high is possible and has been linked to a more aggressive phenotype [65,66,67]. CD44+/CD24−/low BCSC play a crucial role in cancer invasion, as cell lines with a higher proportion of CD44+/CD24−/low cells exhibit greater expression of pro-invasive genes, such as interleukin-1 alpha (IL1α), interleukin-6 (IL6), and interleukin-8 (IL8) [65]. Though CD44 was implied as a homing receptor [56], Sheridan et al. found that the CD44+/CD24− phenotype is not enough for homing of cancer cells at the site of metastasis and its proliferation, implying the need for other factors for successful metastasis [65]. Among them, and shared by BCSC and OCSC, are ALDH1, CD133, CD326 (EpCAM), CD338, and others listed in Table 1.
ALDH1 is an enzyme that oxidizes aldehydes and retinol to retinoic acid (RA). The oxidation of toxic aldehydes protects cell while the activation of RA signaling generally affects cell development, differentiation, and apoptosis, and promotes cancer cell proliferation, drug resistance, and inhibition of apoptosis [68]. ALDH1 is a marker of OCSC and BCSC associated with cell proliferation, migration, poor survival, and chemoresistance [66,69,70,71,72].
CD133 is a transmembrane single-chain glycoprotein also known as prominin, mainly located in cholesterol-rich membrane domains (lipid rafts) [73]. In normal breast tissue, CD133 is not recognized as a stem cell marker [74], but expression of CD133 in BRCA1-associated BC is correlated with higher tumorigenicity in mouse mammary tumors [75]. Perturbations in TME, namely hypoxia, increase the expression of CD133 [76]. CD133+ cells in TNBC show higher self-renewal properties as well as a capacity of transdifferentiation, thus contributing to vasculogenic mimicry [77]. CD133 is also a widely described OCSC marker associated with chemoresistance, elevated migration, and invasion ability [78,79].
CD326, known as the epithelial cell adhesion molecule (EpCAM), is a transmembrane glycoprotein involved in cell adhesion. The higher expression of CD326 in BC is associated with the stem-like phenotype, increased invasiveness, bone metastasis, and radioresistance [80,81]. Similarly, the overexpression of CD326, mainly observed in metastatic OC, contributes to EMT leading to metastasis [82]. Thus, CD326 is considered as a possible therapeutic target for EOC and metastatic BC. Zheng et al. showed that simultaneous targeting of CD326 and CD44 blocks ovarian intraperitoneal tumor outgrowth more efficiently than single targeting [83]. While adecatumumab (MT201), a fully human anti-EpCAM antibody, showed significant inhibition of MCF-7 breast cancer cell proliferation even without a complement and immune cells [84], in a randomized phase II trial, its monotherapy use in patients with metastatic BC, albeit showing some benefit in patients overexpressing EpCAM, did not lead to tumor regression [85]. Catumaxomab, an antibody against CD326, entered clinical trials in patients with advanced OC [86].
CD338 is an ABC transporter often overexpressed in OCSC and associated with chemoresistance [87,88,89]. Overexpression of CD338 was shown to be unique for the luminal progenitor subpopulation of BRCA1-mutated cells [90]. Other common markers, such as receptor tyrosine kinase-like orphan receptor 1 (ROR1), LIN28, CD90, leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5), CD49f, Spalt-like transcription factor 4 (SALL4), and B-cell-specific Moloney murine leukemia virus integration site 1 (BMI1), also emerged and are shown in Table 1.
A more accurate identification of CSC relies on a combination of various markers, particularly due to their distinct distribution patterns and observed stemness and tumorigenic potential. The percentages of BCSC as well as expression of their markers, such as CD44, CD24, ALDH1, and CD133, are tumor-subtype dependent. Basal-like and HER2+ tumors exhibited a higher frequency of ALDH1+ cells compared to luminal tumors, and a higher percentage of CD44+/CD24− cells is mainly attributed to TNBC and to BRCA1-mutated BC [51,91]. TNBC cell lines with the ALDH1+/CD44+/CD24− and ALDH1+/CD44+/CD133+ phenotypes exhibit increased malignant characteristics, both in vitro and in vivo [92]. Similarly, Kryzcek et al. demonstrated that ALDH1+/CD133+ OCSC formed three-dimensional spheres more efficiently than single positive ALDH1+ or CD133+ cells [93].
NANOG, SOX2, and OCT4 are well-known transcription factors essential for maintaining self-renewal and pluripotency in both normal and cancer stem cells, including BCSC and OCSC. Overexpression of NANOG promotes EMT, cell migration, and invasion in OC and is often associated with high-grade cancers, serous histological subtypes, reduced chemosensitivity, and poor overall and disease-free survival [94]. Furthermore, inhibition of NANOG has been shown to reduce proliferation and migration in BC cells [95]. SOX2 expression is associated with sphere formation, tumor initiation, cell proliferation, migration, drug resistance, and the expression of stemness-related and EMT-related genes in BC and OC [96,97,98]. OCT4 expression is linked to chemoresistance, tumorigenicity, and poor outcomes in BC and OC [99,100]. These factors are commonly co-expressed in CSC and play significant roles in promoting cancer progression, including EMT, migration, and invasion.
Aside from common markers, some divergent markers of BCSC and OCSC can be found in Table 1.
Table 1. Markers of BCSC and OCSC.
Table 1. Markers of BCSC and OCSC.
Common MarkersBiological Function in BCSC and OCSCReference
CD44Transmembrane glycoprotein associated with chemoresistance.[45,101]
CD24Cell-surface glycoprotein associated with metastasis, chemoresistance, and poor prognosis.[49,52]
ALDH1Aldehyde dehydrogenase, an enzyme involved in oxidation of aldehydes, associated with cell proliferation, migration, poor survival, and chemoresistance.[69,72]
CD133Transmembrane glycoprotein associated with chemoresistance, elevated migration, and invasiveness.[75,78]
CD326Glycoprotein important for calcium-independent cell adhesion.[80,83]
CD338Adenosine triphosphate (ATP)-binding cassette transporter associated with chemoresistance.[90,102]
NANOGTranscription factor essential for maintaining self-renewal and pluripotency.[94,95]
SOX2Transcription factor important for cell proliferation, migration, drug resistance, and expression of stemness-related and EMT-related genes.[96,98]
OCT4Transcription factor often co-expressed with NANOG and SOX2 associated with increased drug resistance and tumorigenicity.[31,103]
ROR1Receptor tyrosine kinase important for proliferation, migration, and invasion.[104,105]
LIN28RNA-binding protein important for reprogramming of somatic cells to induced pluripotent stem cells.[106,107]
CD90Membrane glycosylphosphatidyl inositol (GPI)-anchored protein, related to tumor initiation and aggressiveness, and poorer patient prognosis.[108,109]
LGR5Transmembrane receptor, increases the stemness of BC cells, BC recurrence, poor outcome, and high tumorigenicity. It promotes EOC proliferation, metastasis, and EMT. Yet, its high expression in HGSOC is linked with improved progression-free survival.[110,111,112]
CD49fIncreases tumorsphere-formation ability, enhances tumorigenicity, drug resistance, and self-renewal properties.[113,114]
SALL4Transcription factor, involved in cancer cell stemness, invasion, proliferation, tumor aggressiveness, and poor survival. [115,116]
BMI1Regulator of gene expression, necessary for self-renewal properties of stem cells, plays a role in tumor aggressiveness, invasion, EMT, and drug resistance.[117,118]
BCSC
Markers
Biological Function in BCSCReference
CD61, ESAIncreases tumorsphere-formation ability, enhances tumorigenicity, drug resistance, and self-renewal properties.[113]
Sca-1Enhances cancer cell tumorigenic ability.[119]
CD70BCSC self-renewal, metastasis, and tumorgenicity. [120]
CD29Integrin protein, enhances metastatic potential, EMT, and cell migration.[121]
KLF4Transcription factor, maintenance of breast cancer stem cells, cell migration, and invasion.[122]
OCSC
Markers
Biological Function in OCSCReference
CD117Receptor tyrosine kinase, promotes self-renewal, differentiation, and regeneration of tumor in xenograft model, chemoresistance, and metastasis.[123]
CD166Glycoprotein, increases sphere-forming ability, adhesion, cell migration, and chemoresistance, high tumorigenic potential.[124]
CD184Chemokine receptor, important for migration and proliferation.[125]
CD243ABC transporter, responsible for paclitaxel resistance in OCSC.[126]
ETRAEndothelin receptor-A, ETRA inhibition prevents chemotherapy-induced increase in CSCs, reduces the formation of tumor spheres.[127]
NPRAAtrial natriuretic peptide receptor, associated with CSCs induced tumorigenesis.[128]
ZIP4Transmembrane zinc transporter, marker for tumor formation in vivo, self-renewal, and differentiation abilities in vitro. [129]
IL-17RPromotes self-renewal of CD133+ CSC in OC by binding IL-17 produced by the tumor microenvironment.[130]
MISRsMüllerian-inhibiting substance receptors, overexpressed in CD44+CD24+EpCAM+ cells of various OC cell lines.[131]
c-MYCTranscription factor, involved in reprogramming of OCSC through interaction with the tumor microenvironment.[31]
MyD88Adaptor protein associated with Toll-like receptor (TLR) signaling, resistance to proapoptotic signals, and the ability to create a pro-inflammatory microenvironment. [132]
SNORA72Highly expressed in ovarian sphere cells with CSC-like characteristics.[133]

4. Signaling Pathways in BCSC and OCSC

A great number of oncogenic signaling pathways has been shown to be implicated in the maintenance of BCSC and OCSC (Figure 1); therefore, targeting these pathways represents a promising therapeutic approach to combating the CSC population.
WNT signaling is an evolutionary conserved signaling pathway that plays a crucial role in normal embryonic development and adult homeostasis of various organs, including the mammary gland and ovary [134,135]. WNT signaling involves the binding of the WNT ligand to LRP5/6 and Frizzled receptor, which inhibits β-catenin phosphorylation by glycogen synthase kinase 3 (GSK-3) and subsequent proteasomal degradation. The accumulated β-catenin translocates from the cytoplasm to the nucleus, where it regulates cyclin D1 expression [136]. Aberrant regulation of this pathway due to mutations and deletions of genes is common in cancer. WNT promotes EMT and chemoresistance in OCSC through induction of SNAIL and ATP-binding cassette super-family G member 2 (ABCG2) transporter pump expression, respectively [137,138]. Additionally, WNT signaling, activated by WNT5b ligand secretion from macrophages, increases the ALDH+ CSC and immunosuppressive traits of macrophages [139]. The pathway is overactive in 60% of BC [140] and can affect self-renewal, differentiation, metastasis [141], and therapy resistance [137,142]. In HER2+ BC, HER2 interacts with a serine/threonine protein kinase B (AKT) and extracellular signal-regulated kinase (ERK) to inhibit β-catenin, leading to trastuzumab resistance and EMT promotion [142]. Suppression of the pathway reduces BCSC properties, migration, and growth in TNBC [143]. Silencing β-catenin reduces ALDH1+ BCSC and increases sensitivity to chemotherapeutics [144,145]. Hypoxia-inducible factor (HIF)-2α overexpression, induced by chronic hypoxia, supports the BCSC phenotype and induces paclitaxel resistance through activation of the WNT/β-catenin pathway, which can be reversed by DKK, a WNT pathway inhibitor [146].
The PI3K/AKT/mTOR pathway plays a crucial role in regulating cell proliferation, metabolism, angiogenesis, and differentiation. Phosphatidylinositol-3-kinase (PI3K) is activated by the binding of growth factors and cytokines to the receptors on a cellular membrane, such as epidermal growth factor receptor (EGFR), insulin-like growth factor I receptor (IGF-IR), and fibroblast growth factor receptor (FGFR), leading to the autophosphorylation of tyrosine residues in the receptors’ cytoplasmic region. PI3K can then subsequently activate AKT, which further activates downstream effector molecules, mainly mammalian target of rapamycin (mTOR). PTEN antagonizes the action of PI3K [147]. Mutations in PI3K, AKT, and PTEN can lead to the abnormal activation of this pathway, which is observed in approximately 70% of BC and OC cases. PIK3CA and PTEN mutations are the most common and subtype dependent in both BC and OC. The highest percentage of PIK3CA mutations occurs in hormone receptor-positive (HR+)/HER2− subtypes of BC and endometriosis-associated OC (endometrioid ovarian carcinoma (EnOC) and ovarian clear-cell carcinoma (OCCC)) while the lowest percentage is found in TNBC and HGSOC [148,149]. In addition, PTEN mutations are more frequently associated with more aggressive types of BC and OC [39,150], although they can coexist [151]. This aberrant pathway activation contributes to resistance to chemotherapy and poor prognosis [152,153,154,155,156,157]. PTEN mutations or inhibition contribute to activation of the EGFR/PI3K/AKT pathway, leading to increased mTOR activation, induction of EMT, enrichment of the stem-like cell fraction, tumorigenicity, and chemoresistance [155,158,159]. Inhibition of the PI3K/AKT/mTOR pathway reduces the survival and stem-like phenotype of BCSC and OCSC [160]. The inhibition of mTOR, a signaling pathway important for maintaining the stem-like properties of cancer cells [158], negatively influences the expression of ALDH1 in CSC [161], while inhibition by metformin (an adenosine monophosphate (AMP)-activated protein kinase (AMPK) activator) leads to the suppression of proliferation in BCSC and OCSC [162,163]. The overexpression of steroid receptor coactivator 3 (SRC-3) and tumor necrosis factor (TNF)-receptor-associated factor 4 (TRAF4), activators of PI3K/AKT signaling, which is associated with a more aggressive phenotype, enrichment of CSC population, and therapy resistance, is frequently found in BC and OC [164,165,166].
NOTCH signaling is also associated with stemness maintenance, which is important in adult organisms and pathologies such as cancer. NOTCH1–4 is a family of transmembrane protein receptors that can bind five ligands—Jagged canonical Notch ligands 1 and 2 (JAG1, JAG2) and Delta-like canonical Notch ligands 1, 3, and 4 (DLL1, DLL3, and DLL4). When a ligand interacts with the receptor, it triggers proteolytic cleavages of the receptor by ADAM metallopeptidase domain 10 (ADAM10) and γ-secretase enzymes. This cleavage releases the intracellular portion of the NOTCH receptor, which then initiates the downstream signaling and transcription of target genes [167,168]. Hyperactivation of NOTCH signaling is crucial for the maintenance of CSC, invasiveness, and resistance to therapy in BC and OC [169,170,171]. NOTCH activity is positively correlated with ALDH1 expression in both OC [172] and BC, and NOTCH downregulation inhibits growth and induces apoptosis in ALDH1+cells [173]. Hypoxic TME induces JAG1 and activates NOTCH signaling, promoting cell proliferation, migration, invasion, chemoresistance, and an increase in the percentage of BCSC and OCSC [171,174,175]. RAS can activate NOTCH1 and upregulate DLL1, and its overexpression is correlated with the upregulation of NOTCH1 in BC [144,176]. NOTCH-positive BC cells have a higher possibility for tumor initiation as NOTCH is implicated in the transition of healthy stem cells to CSC [174,177]. Targeting the NOTCH signaling pathway with antibodies or inhibitors is shown to be an effective way of reducing the population of CD44+/CD24low BCSC, causing a decrease in mammosphere formation and brain metastasis in TNBC [178,179]. It is also effective for OCSC [180]. For example, treatment with γ-secretase inhibitor 1 (GSI), a NOTCH pathway inhibitor, has shown promise in eradicating BCSC and OCSC [181,182].
The Hedgehog–GLI (HH-GLI) pathway plays a major role in embryogenesis. Although its activity is reduced in the adult stage, it is highly required for the maintenance of the stem cell population, tissue repair, and regeneration [183]. In cancer, it is crucial for the regulation of stem-like genes [184] and contributes to the proliferation, drug resistance, and metastasis of CSC [185]. The pathway is activated when HH ligands bind to Patched 1 (PTCH1), derepressing Smoothened (SMO) and activating an intracellular cascade that leads to the activation of three GLI transcription factors, which are the final effectors of the pathway [186]. In OC, the overexpression of PTCH1 and GLI1 correlates with poor prognosis and survival [187]. The HH-GLI signaling was also shown to activate stemness-related transcription factors, such as NANOG, OCT4, SOX2, and BMI1 in gliomas [188], while its inhibition reduces NANOG expression, decreases cell proliferation and colony formation, and abolishes cisplatin resistance in OC cells [189]. Ray et al. found that, compared to normal immortalized epithelial cells, OC cells exhibited increased GLI1 expression, spheroid-forming ability, and CSC traits (self-renewal, differentiation, and chemoresistance) [190]. The pathway is upregulated in TNBC and Luminal B BC and is associated with stem-like cell expansion [191] and vascular endothelial growth factor (VEGF)-independent angiogenesis [185]. The pathway was found to be more active in less differentiated cells, such as CD44+/CD24− cells [144,191]. As in OC, its activation is positively correlated with the expression of NANOG and OCT4 [185]. The HH signaling pathway activation contributes to enrichment of the CD44+/CD24− population [192] and chemoresistance associated with BCSC [144], while its dysregulation, observed as the overexpression of Sonic hedgehog (SHH), Desert hedgehog (DHH), and GLI1, is correlated with a worse prognosis and more advanced stages of BC [192,193]. The knockdown of GLI2 can decrease ALDH activity and influence mammosphere growth, while its overexpression has a positive effect on mammosphere growth [194]. Inhibition of the pathway using inhibitors such as cyclopamine has shown negative effects on tumor progression in BC and OC mouse models [191,195]. In addition, inhibition with a GLI protein antagonist, GANT-61, affects cellular growth, stemness, and migration in OC by suppressing its target genes transcription [196] and inducing apoptosis and decreased BCSC population in TNBC cell lines [197].
The JAK/STAT pathway regulates cell proliferation, differentiation, apoptosis, and immune response by being stimulated mainly by growth factors (EGF, IGF, and FGF) and cytokines (IL6, IL5, IL8, IL10, and IL23) [198,199]. There are three Janus Kinase (JAK) protein (1–3) and seven signal transducer and activator of transcription (STAT) proteins (0–6). Ligands binding to membrane receptors initiate JAK transphosphorylation, resulting in the phosphorylation of tyrosine residues on the receptor. These phosphorylated sites serve as docking sites for STAT proteins. JAK phosphorylates STATs, which then dissociate from the receptor and form dimers. These dimers subsequently translocate into the nucleus, where they regulate the transcription of specific target genes [200]. The JAK/STAT pathway has been implicated in CSC biology in several cancer types, including BC and OC, where it plays a role in cancer initiation, progression, multidrug resistance, and metastasis. In OC, inhibition of the JAK/STAT pathway resulted in a loss of CSC-like markers and CA125 expression [201]. Moreover, increased phosphorylation of STAT3 and expression of STAT3 target genes, NANOG and C-MYC in CD24+ OCSC, when inhibited with JAK2, induces cytotoxicity in CD24+ cells, reduces tumor metastasis, and prolongs overall survival [202]. Ruan and colleagues demonstrated that the inhibition of JAK/STAT signaling attenuated the tumor-feeding effects caused by the upregulation of OCT4 in OCSCs [203]. Moreover, IL23 has been shown to maintain the tumorigenic potential of CD133+ OCSCs in vivo and mediate their ability to self-renew through activation of STAT3 [204]. Overexpression of STAT3 is found in more than 40% of BC, mainly in TNBC [144]. Similar to OC, activation of JAK/STAT is important for the occurrence of BCSC, such as an increase in the CD44+/CD24low BCSC population observed in hypoxia, leading to the upregulation of genes that influence angiogenesis, proliferation, and EMT, further promoting BC development and chemoresistance [144,199]. STAT3 upregulates matrix metalloproteinases (MMP), especially MMP-2 and MMP-9, which promote cancer invasion and metastasis [199]. ROS- and IL6-induced activation of STAT3 promotes BCSC occurrence, BC progression, and inflammation [199]. In addition, IL6- and IL10-induced activation of STAT3 is involved in BC metastasis to the liver, bones, and lungs [205]. STAT3 interacts with CD44, nuclear factor kappa B (NF-κB), and catalytic subunit of telomerase (hTERT) to promote the BCSC phenotype [199]. BCSC with ALDH+ and ALDH+/CD44+/CD24− phenotype were associated with the activation of JAK/STAT signaling, and the use of STAT3 inhibitors (Stattic or LLL12) reduced the growth and invasiveness of BCSC while inducing apoptosis [206,207]. In both BCSC and OCSC, activation of JAK/STAT signaling promotes macrophage M2 polarization and cancer cell invasion [208], which negatively correlates with patient survival [209].
The transforming growth factor-β (TGFβ) signaling pathway plays a crucial role in maintaining normal tissue homeostasis, but its dysregulation is implicated in tumorigenesis, including BC and OC. TGFß binds to the specific receptor complex consisting of the TGF-β type II receptor (TGFβRII) and the TGF-β type I receptor (TGFβRI). Ligand binding with TGFβRII leads to the receptor phosphorylation forming a binding site for TGFβRI, in which phosphorylation triggers the activation of the receptor-activated suppressor of mothers against decapentaplegic (SMAD) proteins that translocate to the nucleus and regulate transcription of targeted genes. TGF-beta signaling can also activate non-SMAD pathways, which contribute to additional cellular responses [210]. Activation of TGFß signaling and cooperation with the TNFα and WNT pathways contributes to increases in BCSC and OCSC populations, tumorigenicity, and chemoresistance through regulation of EMT [113,211,212,213,214,215,216]. By inducing the expression of TTG2, TGFß promotes EMT, spheroid formation, and metastasis in OC, while TTG2 knockdown decreases the number of cells with a CSC phenotype (CD44+ CD117+) [217]. Silencing of SNAIL, a known TGFß target, reverses stemness properties and inhibits tumor growth in OCSC [137]. Inhibition of TGFß has also been shown to be effective in restoring sensitivity to cisplatin in OCSC [218]. In addition, the paclitaxel-induced increase in TGFβ and IL8 in TNBC, which led to an increase in BCSC number, could be reversed by inhibition of the TGF-β type II receptor [219]. In both BC and OC, the TGFß signaling pathway influences TME, especially cancer-associated fibroblasts (CAF), to promote tumor growth. It transforms normal fibroblasts into CAF [220], which continue to produce extracellular matrix proteins that support the microenvironment niche for CSC growth [221]. In addition, platelet-derived TGFβ can activate the TGFβ/SMAD and NF-κB signaling pathways, which can stimulate cancer cells to metastasize, while inhibition of TGFβ can reduce the lung metastasis of BC [222]. Treatment of OC cell line SKOV-3 with SB525334, an inhibitor of TGFβRI, showed promising results in targeting CSC with high ALDH expression. The self-renewal ability of CSC and in vitro tumorigenicity were reduced when treated with SB525334, which indicates the possibility of CSC-targeted treatments for OCSC and other CSC with high ALDH expression, such as BCSC [223].
The NF-κB signaling pathway plays a crucial role in cell functions and processes such as inflammation, angiogenesis, differentiation, stemness, and metastasis in BC and OC [224,225]. The activity of NF-κB is regulated by the inhibitor of κB (IκB) proteins. These IκB proteins act as inhibitors, preventing NF-κB from entering the nucleus. The IκB kinase (IKK) complex is responsible for regulating the IκB. When specific signals, such as pro-inflammatory cytokines, are recognized, the IKK complex is activated. This activation leads to the degradation of IκBs and the subsequent release of NF-κB. As a result, NF-κB translocates to the nucleus and controls the transcription of targeted genes [226]. Constitutive activation of the NF-κB pathway is observed in BCSC and OCSC. Activation of NF-κB in TNBC correlates with an increase in the CD24−/CD44+/EpCAM+ fraction and ALDH+ fraction of BCSC [227], while inhibition of NF-κB decreases CD44 expression, invasiveness, and proliferation of TNBC cells [228]. Similarly, NF-κB was found to be constitutively active in CD44+ OCSC and to promote chemoresistance, spheroid formation, and self-renewal/proliferation of OCSC [229]. Inhibition of NF-κB restored the response to platinum therapy in OCSC [230] and specifically decreased the expression of CD44 and genes involved in OCSC self-renewal/proliferation [231]. Together with the JAK2/STAT signaling, NF-κB seems to be crucial for OCSC self-differentiation into endothelial cells and promotion of angiogenesis [232]. Moreover, NF-κB signaling is involved in the interaction of OCSC with the TME. Namely, OCSC release cytokines to promote M2 macrophages’ recruitment and support of the cancer stem niche [79,130,204,209]. Inhibition of the NF-κB signaling pathway with a dominant-negative version of IkBα in OC reduced the number of CD44+ OCSC and the expression of the stemness genes [233]. In BC, inhibitors of NF-κB signaling (parthenolide, pyrrolidinedithiocarbamate, and its analog diethyldithiocarbamate) inhibited MCF7 mammosphere growth, suggesting its preferential targeting of BCSC [234].
The Hippo signaling pathway regulates a wide range of biological processes, including cell survival, differentiation, cell proliferation, and tissue homeostasis [235]. The Hippo pathway involves a kinase cascade comprising mammalian Ste20-like kinases 1/2 (MST1/2) and large tumor suppressor 1/2 (LATS1/2), along with downstream effectors, namely, the transcriptional coactivators Yes-associated protein (YAP) and the transcriptional coactivator with PDZ-binding motif (TAZ). Various cellular factors such as cell density, cell polarity, soluble factors, stress signals, and mechanical cues activate the pathway [236]. YAP, the major target of the Hippo pathway, is a known oncogene that, when suppressed, restores sensitivity to platinum-based therapy in OC and leads to decreased stemness properties in BC and OC [237,238,239]. The self-renewal and chemoresistance of BCSC and OCSC depend on the activity of the YAP and Hippo downstream coactivator TEAD [239,240]. In BC, hyperactivated YAP leads to the formation of BCSC, while TAZ is responsible for maintaining stem-like properties in CD44+/CD24− BCSC [241]. Detachment of cells from the extracellular matrix (ECM) leads to YAP/TAZ inhibition, resulting in anoikis, while dysregulation of HIPPO/YAP/TAZ leads to anoikis resistance and EMT [241]. RICH1, a Rho GTPase-activating protein, increases the sensitivity of BC to chemotherapeutics and inhibits stem-cell-like properties by inhibiting nuclear translocation of YAP/TAZ. This emphasizes the possibility of CSC-targeted therapies through manipulation of the HIPPO signaling pathway [242]. Indeed, verteporfin has been shown to be a suppressor of the YAP–TEAD complex with anticancer properties. OC cell lines treated with verteporfin showed decreased in vitro migration and reduced tumor growth in xenograft mice [243].
The nuclear factor erythroid 2-related factor 2 (NRF2) signaling pathway is often associated with the enrichment of BCSC and OCSC, leading to therapy resistance in BC and OC. NRF2 is a transcriptional factor that regulates the expression of more than 250 genes. It is repressed in a complex with Kelch-like ECH-associated protein 1 (KEAP1) and subjected to ubiquitination and proteasomal degradation. However, NRF2 regulation/activation can include other repressors or competitors. Oxidative stress activates this pathway by blocking the binding of NRF2 with KEAP1, which fosters its translocation to the nucleus and activation of target genes with antioxidant-responsive elements (ARE) in their promoter [244]. Radioresistance and chemoresistance in BC are linked to the augmentation of ALDH+CD44+ BCSC [245], but mainly to ALDH+ BCSC [71]. Similarly, in OC, chemoresistance is linked to OCSC expressing high levels of ALDH1 and not CD24, CD117, or CD133 [246]. Stemness properties, such as drug resistance, colony/sphere formation, tumor growth, and high stemness marker expression, of both BCSC and OCSC are regulated by the p62/NRF2 axis. The molecular mechanism involves the activation of NRF2 through the p62-associated pathway [247], regulated by ALDH1A1 in OC [248] and CD44 in BC [249]. Furthermore, resistance to radiotherapy in BC is associated with activation of the NRF2 pathway, the enrichment of ALDH+ BCSC, and the promotion of EMT. The activation of the NRF2 signaling pathway in BC can be mediated by reactive oxygen species (ROS) [250] or can occur via the silencing of KEAP1 by miR200a [251]. GSK-3β and BTB Domain And CNC Homolog 1 (BACH1) seem not to be involved in the radiation-induced activation of NRF2 [251]. ML385-mediated inhibition of NRF2 has been found to enhance the sensitivity of BCSC to ionizing radiation, while the activation of NRF2 through sulforaphane diminishes this effect [250]. Furthermore, the inhibition of NRF2 in ALDH+ OC cells and CD44+ BC cells has demonstrated a reduction in CSC properties, including chemoresistance, tumor growth, and spheroid formation [233].
These findings highlight the significance of targeting pathways involved in CSC biology as a potential therapeutic strategy to overcome therapy resistance in breast and ovarian cancers.

5. BCSC- and OCSC-Microenvironment Communication

The microenvironment of both BC and OC includes several cell types, including stromal cells, adipocytes, carcinoma-associated mesenchymal stem cells (CA-MSC), and immune cells (Figure 2). All these cell types create a communication network where many signaling molecules are released and maintain a specific local microenvironment. Single-cell RNA-sequencing analysis of OC revealed several major clusters within the tumor: epithelial, mesenchyme, macrophage, differentiation, T cell, B cell, and endothelium clusters. Within the epithelial cluster, a specific carcinoembryonic cluster was identified, PEG10+, which showed high similarities with embryonic cells, and is associated with carcinogenesis [252].

5.1. The Connection between Adipocyte Populations and BCSC and OCSC

Obesity is a known risk factor for many cancer types, including OC and BC [253,254]. A recent single-cell and spatial transcriptomic meta-analysis of human white adipose tissue revealed the presence of over 60 distinct cell populations within four major cell types: adipocytes (~20%), fibroblast and adipogenic progenitors (~40%), vascular cells (~15%), and immune cells (~20%) [255]. Adipocytes, which are common to both OC and BC, contribute greatly to the carcinogenesis. High visceral fat-to-muscle ratio is a predictor of worse overall survival for OC patients [256]. Adipocytes secrete adipokines, growth factors, and hormones which are supportive for tumor growth. For example, resistin induces stemness and EMT of EOC cells, and triggers angiogenesis through VEGF and MMP-2 [257] Additionally, adipocytes via leptin signaling increase the expression of stem markers such as SOX2 and NOTCH2 and contribute to the enrichment of BCSC [258]. Adipocytes also induce lipid metabolism in cancer cells, and co-culturing cancer cells with adipocytes leads to an increase in β-oxidation, ROS, and lipid peroxidation [259,260]. Additionally, they induce nitrogen oxide (NO) synthesis in OC cells, which then use arginine from the TME and secrete citrulline, further inducing adipogenesis [261]. OC cells, on the other hand, also affect the behavior of adipocytes. WNT signaling components secreted from OC cells can induce defattening of adipocytes and their conversion into adipocyte-derived fibroblasts (ADF), which demonstrate CAF-like characteristics. This morphological change is followed by changes in reduced expression of adipose markers and an increase in fibroblast markers, creating a subpopulation of cells at the invasive front of the tumor. These cells can migrate to the tumor center and contribute to tumor invasiveness and migratory capacity [262]. Even more, there are breast adipose tissue-derived mesenchymal stromal/stem cells (bASC), which can de-differentiate into different CAF and stimulate proliferation, migration, chemoresistance, and stemness of BC cells [263,264]. ADF cocultured with OC cell lines induce their proliferation and migratory ability, and induce their fatty acid uptake and lipid accumulation [265,266]. However, there is also evidence of an inhibitory effect of OC on adipocyte differentiation through the ECM protein secreted protein acidic and rich in cysteine (SPARC), which is responsible for maintaining tissue homeostasis [267].
Adipose-derived stem cells (ADSC) exhibit features of mesenchymal stromal cells, and can differentiate into various cell types: adipocytes, osteoblasts, chondrocytes, or myocytes. ADSC-conditioned medium increases migration and invasion of OC cell lines and xenografts, and promotes EMT through the TGF-β pathway [268,269]. ADSC also influence the sphere formation and tumor initiation of BC, along with increased expression of stem markers ALDH1A1 and ABCG2 [270]. Conversely, exosomes from OC and BC cells can direct the differentiation of ADSC into a myofibroblast-like phenotype [271,272]. To understand the role of adipocytes in tumor development and progression, it has to be noted that white adipose tissue (WAT) is an endocrine organ that also has a metabolic function [273,274]. WAT secretes inflammatory cytokines such as IL1, IL6, IL10 and TNFα, but also TGFβ, monocyte chemoattractive protein-1 (MCP-1), angiogenic proteins such as C-X-C motif chemokine 5 (CXCL5) and VEGF, but also adipokines such as leptin, resistin, and adiponectin (reviewed in [273]). These cyto/chemo/adipokines play an important role in determining the fate of tumors in their vicinity. IL6, a pro-inflammatory cytokine secreted locally and systematically by adipocytes, is significant for tumor development, as high plasma levels correlate with poorer overall survival of BC patients [262,275]. The secretion of IL6 is caused by tumor stimulation of adipocytes, as shown by co-cultivation of murine adipocytes with human BC cell line ZR75.1 [262]. In response to IL6, CD44+CD27− BC cells specifically activate the JAK2/STAT3 pathway, whereas NF-κB signaling is not as specific for these basal-like BC cells [276]. Yet, in HER2+ BC, IL6 can promote stemness and metastases via NF-κB/STAT3 activation [277]. In OC, IL6 can activate the JAK/STAT3 signaling pathway, leading to tumor growth, metastasis, and EMT [278]. In addition, IL6 triggers NOTCH3-dependent upregulation of genes promoting malignant features of HR+ MCF7 spheroids [279]. TNFα, also a pro-inflammatory cytokine, can trigger the stemness phenotype and drug resistance of basal-like BC cells through activation of the KLF5/Ephrin type-A receptor 2 (EphA2) axis [280]. Furthermore, cancer-associated adipocytes (CAA) can trigger increased expression of pro-inflammatory cytokines IL6 and IL8 in co-cultures with TNBC cells MDA-MB-231 [281]. TNFα also influences the expression of CD44 in OC through the activation of JNK, where it increases CD44 expression; this activation of JNK results in a more aggressive phenotype [282]. TNFα production in OC leads to increases in IL6, VEGF, C-C motif ligand 2 (CCL2), and C-X-C motif chemokine ligand 12 (CXCL12), and lower levels of TNF-α lead to a reduction in tumor growth [283].

5.2. The Effect of Immune Cells on BCSC and OCSC

The tumor-promoting microenvironment is generated by stimulating angiogenesis (activation of PI3K pathway via VEGF and EGF), regulating adhesion (changes in composition of integrins), and inflammation (pro-inflammatory cytokines IL6 and IL8 and increased neutrophil to lymphocyte ratio). Bioactive lipids such as lysophosphatidic acid (LPA) and arachidonic acid (AA) are elevated in malignant ascites and contribute to immunosuppression [284]. Many immune cell subtypes are found accumulated within the ascites, including M2 polarized TAM, myeloid-derived suppressor cells (MDSC), CD4+ and CD8+ T cells, Tregs, DC2 dendritic cells, and cytokine-producing natural killer (NK) cells, while cytotoxic NK cells are reduced [55]. Similar trends can be seen in BC metastasis, where the microenvironment plays a crucial role in the spread of the tumor, and many immune cells promote immunosuppression and tumor progression, including M2 macrophages, neutrophils, CD8+, CD4+, and Tregs [285,286]. Extracellular vesicles (EV) mediate the crosstalk between tumor cells and their microenvironment. EV can trigger immunosuppression by arresting T cells, inducing TAM polarization into M2 subtype, and inducing IL6. Additionally, EV can induce transformation of host stromal cells into CAF [287]. In BC, EV promote lung metastasis [288], increase migration and invasion of BC cells, and enhance EMT [289], while inhibition of EV leads to decreased tumor growth and lung metastasis [290].
Classically activated macrophages, also known as M1, are pro-inflammatory, while M2 macrophages are further divided into different subgroups (M2a, M2b, M2c, and M2d); furthermore, they have immunosuppressive properties along with tumor-promoting properties, so they are often called tumor-associated macrophages (TAM) [291]. TAM are one of the most prevalent types of immune cells found in BC and affect BCSC properties. BCSC interacting with macrophages initiates their tumor-promoting function, and these TAM produce high levels of IL6, which in turn initiates activation of the STAT3 signaling pathway and consequent enrichment of BCSC [292]. TAM also interact with OCSC and have been shown to correlate with poor overall patient survival, chemoresistance, and promotion of metastasis in OC [293]. OCSC can influence M2 macrophages and promote immunosuppression, while at the same time M2 macrophages can promote stemness of OCSC [139].
One of the most important components of the immune system in relation to cancer cells is the population of cytotoxic CD8+ T cells, which are heavily involved in BCSC and OCSC function. Since the function of cytotoxic CD8+ T cells is to kill abnormal cells, they are of great importance in the regulation of cancer growth. At the onset of cancer growth, CD8+ cells are responsible for killing cancer cells, and their presence in cancer tissue correlates positively with better overall survival of BC patients [294]. However, when tumors advance, the role of CD8+ T cells changes—they can no longer destroy tumor cells and instead promote the growth of the tumor rather than its eradication. BCSC, OCSC, and stromal cells can secrete large quantities of TGF-ß, a well-known CD8+ cell inhibitor [295,296]. Other than TGF-ß, BCSC can express high levels of Programmed cell death ligand 1 (PD-L1). This is especially important because CD8+ T cells express inhibitory receptor Programmed death-1 (PD-1), and through their interaction, BCSC along with OCSC can inactivate CD8+ T cells, further improving the chances for tumor development [297,298]. TNBC cells, which exhibit high levels of PD-L1, form more mammospheres than cells with a lower expression of PD-L1 [299]. Furthermore, the high expression of PD-L1 is positively correlated with the expression of other stem markers in BCSC, such as CD44 and ALDH [299]. Interaction between MCF7 cells and CD8+ T cells that have been treated with Concanamycin A, so they are unable to lysate tumor cells, has been shown to increase the population of CD44+/CD24− cells with stem properties [300]. Even though the levels of CD8+ T cells correlate with better survival of patients with OC, this effect seems to be dependent on the presence of CD4+ tumor-infiltrating T cells (TIL), where CD25+FOXP3+ regulatory T cells mitigate the effect of CD8+ T cells [301]. This polarization of CD4+ cells plays a part in tumor formation and growth. CD4+ T helper cells play an important role in cancer inhibition and destruction, while regulatory T cells have been shown to promote cancer. The expression of SOX2 in BCSC recruits the regulatory T cells through secretion of chemokine (C-C motif) ligand 1 (CCL-1), where Tregs further promote CSC properties [302].

5.3. Cancer-Associated Fibroblasts and Mesenchymal Stem Cells

CAF are generally found in close physical relation to a tumor, and they are characterized by the expression of fibroblast activation protein (FAP) and actin smooth muscle (αSMA) [303]. BCSC express high levels of SHH, a ligand of the HH signaling pathway that can in turn activate the HH signaling pathway in CAF via paracrine activation, which in turn increase stem properties of BCSC [304]. CAF are involved in ECM remodeling, regulation of immune cells, and promotion of stem characteristics of BCSC and OCSC [305,306]. ECM remodeling by replacing collagen type 4 with collagen type 1 and 3 [307] or by MMP influences angiogenesis and metastasis [305]. CAF secrete chemokines and cytokines that regulate regulatory T cells (Treg)-mediated immunosuppression, cytotoxic T cell localization, and macrophage differentiation into TAM [305]. CAF can induce OCSC through IL8 signaling and activation of the NOTCH signaling pathway [306]. OC cells cocultured with CAF show increased chemoresistance and increased number of OCSC. An increase in OCSC is obtained through WNT signaling, and its inhibition reversed the effect of CAF on OCSC [308]. Through para- and autocrine interaction, CAF can influence OC angiogenesis, metastasis, and proliferation mediated by various signaling pathways, such as AKT/mTOR, TGF-β, and NF-κB [309]. These data indicate the important role of CAF in the progression of BC and OC through its interaction with CSC.
CA-MSC are another crucial component of the OC TME. They are defined by the expression of CD44, CD73, and CD90 markers, and can differentiate into adipose tissue, cartilage, or bone in vitro [310]. CA-MSC originate from local abdominal tissue and are induced by hypoxia [311]. Hypoxia can induce a quiescent state but also increase proliferation, invasion, and sphere-formation capabilities, as well as increased stem-like properties of OC cells [175,312]. Hypoxia can also influence the expression of BCSC markers, such as CD133, where low-oxygen status leads to an increase in CD133 expression [76]. One of the proposed mechanisms is the hypoxia-induced upregulation of NOTCH signaling, leading to the upregulation of SOX2 and ALDH [175]. Another mechanism proposes the accumulation of HIF-1α and HIF-2α in OC cells upon ammonia treatment [313]. Hypoxia also induces the expression of EMT hallmark proteins such as expression of SLUG and SNAIL in BC cells [314]. Sirtuin 1 (SIRT1) is a downstream target of HIF-1α and has been proposed as a potential therapeutic target for OC [315], while SIRT1 increases chemoresistance and metastasis, and its overexpression is connected to BC growth [316,317].

6. Cancer Immunogenicity and Evasion of Immunosurveillance of OC and BC

The immune system plays a crucial role in shaping the development and progression of cancer, and according to the “cancer immunoediting” hypothesis it comprises three phases: elimination, equilibrium, and escape. During the elimination phase, the immune system (innate and adaptive) detects and eliminates tumor cells that display foreign antigens on their surface. Yet, some tumor cells develop mechanisms to evade or suppress immune responses, leading to the equilibrium phase. In the equilibrium phase, the immune system (mainly T helper type 1 (Th1) cells, cytotoxic T cells, and cytokines of type 1 immunity (IL12, IL2, interferon gamma (IFN-γ)) keeps tumor cells dormant or growing slowly. The escape phase occurs when tumor cells develop genetic instability and evade immune detection, resulting in rapid growth and metastasis. It is when the tumor becomes clinically visible. This phase is aided by a proangiogenic microenvironment and the suppression of the immune response. This includes Treg cells, myeloid-derived suppressor cells, type 2 macrophages, IL10, TGF-ß, VEGF, 2,3-dioxygenase (IDO), arginase-1, and cyclooxygenase-2 (COX-2) as well as the overexpression of pro-survival proteins and inefficient presentation of tumor-associated antigens [318].
While BC was firstly perceived as an immunogenically “cold” tumor, OC is considered an immunogenic tumor, as it expresses so-called tumor-associated antigens (TAA) that can be recognized by the immune system. These antigens are produced by the tumor cell, phagocytosed, and processed by the dendritic cells, which then expose them on the major histocompatibility complex (MHC) class molecules on their surface. These are recognized by the T cells and trigger the immune response of the host. TAA identified in OC include HER2/neu, CA125, FR-α, CA153, HE-4, and others [319]. Today, BC is also considered an immunogenic tumor due to its highly dynamic immune heterogeneity. Indeed, immune cells are present in the tumor and TME but are exposed to highly immunosuppressive environment. TAA identified in BC are HER2, Mucin 1 (MUC1), carcinoembryonic antigen (CEA), NY-ESO-1, melanoma-associated antigens (MAGE), brachyury, cMET (MET receptor tyrosine kinase), and mesothelin [320]. HER2 is a tyrosine kinase receptor of the EGF family overexpressed in 15–30% of invasive BC and 20–30% of OC. Its overexpression correlates with worse survival of BC and OC patients and more aggressive disease [321,322]. Interestingly, the same HER2/neu sequence is recognized by HLA-A2-restricted tumor-specific cytotoxic T lymphocytes found in both BC and OC [323]. This suggests that targeting this sequence with immunotherapy could potentially be effective in both types of cancer.
NK cells act cytotoxically on the cells that lose MHC class molecules. In OC, CD24+ cell populations isolated from OC cell lines were more susceptible to NK cell cytotoxicity as they show loss of MHC class I molecules on their surface, and upregulation of NKG2D receptor, which is responsible for initiation of NK killing mechanism. On the other hand, CD24+ population showed increased resistance to cisplatin and doxorubicin compared to CD24− population [324]. In fact, a study comparing seven OC cell lines according to their level of differentiation and CD44 expression concluded that poorly differentiated lines, with high CD44 and low MHC class I expression, are effectively eliminated by NK cells, and this effectiveness drops with the increasing differentiation level and MHC class I expression [325]. However, the ascitic fluid of advanced OC suppresses the effect of NK cells [326]. In BC, NK cells have been shown to reduce tumor growth in vivo [327] and specifically kill CD44+CD24low/- BCSC from an HR+ cell line when activated with IL2 and IL15 [328]. Conversely, analysis of radioresistant TNBC showed enrichment in CD44+CD24low/- BCSC subpopulations that evade the cytotoxic activity of NK cells. Although NK cells invade the tumor enriched with CD44+CD24low/- BCSC, their activity is impaired due to altered expression of ligands on BCSC. Specifically, the expression of HLA-E, the ligand for the inhibitory NKG2A receptor, is increased and the expression of MICA/B, the ligands for the activating NKG2D receptor, is decreased. Cleavage by ADAM10 [329] or downregulation by the oncogenic miR20a [330] contribute to decrease in MICA/B expression and subsequent deficiency in NKG2D recognition. Moreover, the accumulation of MDSC that negatively regulate NK cell activation and function contributes to the evasion of CD44+CD24low/- BCSC from NK cells and BC progression as well [329]. In an early phase clinical trial, the infusion of activated allogeneic NK cells in BC and OC patients resulted in transient donor chimerism, but there was no notable increase in the number of transfused NK cells [331].
In both, OC and BC, noncanonical activation of the TGFβ pathway in CD8+ T cells upregulates CD103 and induces secretion of CXCL13. These CXCL13+CD103+CD8+ TIL subpopulations promote migration of B cells to the tumor and the formation of tertiary lymphoid structures (TLS) and are associated with better prognosis [332]. In addition, a large-scale meta-analysis identifying the immunogenic cell death (ICD)-metagene expression signature in BC, OC, and lung cancer according to patients’ better prognoses reveals a highly cancer-type-specific prognostic impact, albeit with some similarities between BC and OC. High expression of the BC-specific ICD-derived metagene signature (TNF/CXCR3/P2RX7/CASP1/NLRP3/IL1B/LY96/CD4+/CD8+A/CD8+B/PRF1/IFNG/IL17A/IL17RA) is associated with prolonged overall survival (OS) in BC and OC patients but not in lung cancer patients. Similarly, high expression of the OC-specific ICD-derived metagene signature (CALR/PIK3CA/TNF/IFNA/IFNB1/CXCR3/P2RX7/CASP1/IL1B/TLR4/CD4+/PRF1/IFNG/IL17A/IL17RA) is associated with prolonged OS in OC patients and to some extent with prolonged OS in BC patients, whereas high expression of the lung-cancer-specific ICD-derived metagene signature is associated with prolonged OS only in lung cancer patients. This suggests that immune or inflammatory responses may operate in a more unified manner in BC and OC [333].
OC tissues express PD-L1 in approximately 50% of cases, and its expression correlates with high CD8 expression and moderate CD4 expression. Interestingly, the expression of PD-L1 colocalizes with the stem cell markers CD44 and LGR5 [334]. In BC, the expression of PD-L1 is three-fold higher in BCSC than in more differentiated cancer cells [335] and correlates with immune evasion [336]. Moreover, PD-L1 can be found in various immune cells, including macrophages, CD4+, FOXP3+, and CD8+ T cells within the BC TME, and higher levels are associated with better prognosis in TNBC [337]. In OC, a higher percentage of PD-L1 positive cells was found in the OC infiltrating cells compared to peripheral blood, and the highest in peritoneal fluid [338]. However, a meta-analysis of PD-L1 expression in OC revealed that PD-L1 does not have prognostic value in OC [339], in contrast to BC where a similar meta-analysis identified high PD-L1 expression as a negative prognostic factor [340]. However, PD-L1 upregulation has been associated with the development of carboplatin resistance in OC [341]. CD24, one of the OCSC markers, is expressed at higher levels than PD-L1 in OC, and interacts with Siglec-10 expressed on TAM, leading to anti-phagocytic activity of TAM and immune evasion. The same interaction has also been demonstrated for TNBC, and the authors propose anti-CD24 immunotherapy as a mechanism to promote immune response in OC and TNBC patients [342].
Cancer progression, metastasis, recurrence, and resistance occurs due to the BCSC and OCSC escape from immune surveillance [343,344]. They achieve immune evasion through several mechanisms. In OC, activation of the NF-κB signaling pathway directs the macrophages towards M2 polarization, which is associated with anti-inflammatory and immunosuppressive processes [209]. M2 macrophages express CD39 and CD73, which are responsible for the conversion of ATP to adenosine, which additionally pushes the macrophages towards the M2 phenotype, and suppresses CD4+ T, CD8+ T, and NK cells, thus generating a self-amplifying mechanism for immune evasion [345]. CD163+ TAM are more frequent in the ascites of OC patients compared to the peripheral blood of the same patients, and exosomes derived from these cells promote adhesion and migration of EOC cells [346]. Interaction of BCSC with macrophages initiates their tumor-promoting function, and these TAM produce high levels of IL6 which in turn initiates activation of the STAT3 signaling pathway and consequent enrichment of BCSC. These BCSC are also enriched in SOX2, NANOG, Stem cells antigen-1 (SCA-1), and OCT4 [292]. CSC can produce chemokines, such as CCL2, CCL3, CCL5, and CCL8, to recruit monocytes, thus influencing monocyte migration [347].
Regulatory T cells CD4+CD25+CD3+ are abundant in the tumor mass and the malignant ascites of OC patients with later stage disease, and they suppress the production of IFN-γ and IL2 and subsequent T cell activation [348]. Aging contributes to the reduced function of the immune system, as aged CD4+ TIL and aged CD20+ B cells are less active than young ones [349]. In fact, ascites-derived T cells show poor proliferation after stimulation with αCD3/28 beads and are unresponsive to IL2. The components responsible for this effect were shown to be the lipids within the ascites [350]. Polarization of CD4+ T cells also plays a part in tumor formation and growth. CD4+ T helpers play an important role in cancer inhibition and destruction, while regulatory T cells have been shown to promote cancer. Expression of SOX2 in BCSC recruits the CD4+ regulatory T cells through secretion of CCL1, where Tregs further promote cancer cell stem properties [302].

7. Experimental Models for Distinguishing CSC Populations

One of the most common problems with studying CSC has been the selection of a method for the isolation and characterization of CSC. Throughout the years, several types of models and methods were used in order to distinguish CSC populations, all with their advantages and limitations.
The use of culture models in cancer research provides a valuable tool for investigating various aspects of tumor biology, including drug response, drug resistance, TME, and stem-cell-like properties. While the predominantly used 2D cultures provide a simple and high-throughput method in this field, they lack physiological relevance due to the altered 3D architecture, cell-to-cell interactions, and behavior. Three-dimensional models, by increasing complexity, include spheroid cultures of tumor cells, spheroid co-cultures of two or more cell types, and organoids developed from patient material, as the most complex system. These models more accurately mimic the complex architecture and microenvironment of tumors, cell heterogeneity, and the presence of CSC, but their use can present challenges such as difficult imaging, higher cost, and a lack of standardization. Several reviews already discussed the advantages and disadvantages of using 3D versus 2D cultures in cancer research. They listed different 3D culture models, including spheroids grown on ultra-low attachment plates or in spinner flasks, synthetic or natural hydrogel models and 3D-printed scaffolds, microfluidic organ-on-a-chip models, and cell line or patient-derived xenografts, and discussed their applications and limitations [351,352,353,354,355,356,357,358,359,360]. The ability of 3D models to mimic the architecture and microenvironment of tumors enables the maintenance of undifferentiated states and cancer stem-cell-like properties, providing insight into the role of CSC in cancer initiation, progression, and resistance to treatment.
Studies have shown that 3D models exhibit increased resistance to chemotherapeutics compared to 2D cultures, and they can demonstrate the efficacy of drugs in eradicating CSC populations and reducing spheroid size. An increased resistance to chemotherapeutics was shown on spheroid models [361,362,363,364,365,366,367,368,369,370,371], scaffold-based models [372,373,374,375,376,377,378,379,380], and organs-on-a-chip [381,382,383] when compared to 2D cell cultures. Numerous studies have reported the use of various models in BC and OC research showing increased levels of cancer stem-like markers. For example, cultivating the BC cell lines in natural 3D collagen scaffolds promoted EMT and resulted in increased tumorigenicity and BCSC populations with a CD44+/CD24− phenotype and markers OCT4, SOX2, SOX4, JAG1, and CD49f [384]. Spheroids grown on hydrogel-based 3D scaffolds maintained a drug-resistant phenotype (CD44+/CD24−/ALDH1+) and exhibited an increased drug resistance [374]. Cultivating BC cell lines on a polycaprolactone (PCL) scaffolds in 3D culture conditions resulted in an elevated proportion of BCSC populations (CD44+/CD24−) [385], an upregulation of stem cell markers (OCT3/4, SOX2, SOX4, and CD49f) and increased invasive capability [386], and an increased metastatic potential [387], accompanied by a significant increase in the resistance to chemotherapy [388]. Omentum is one of the preferred sites of OC metastases, so 3D omentum-inspired hydrogel and a four-cell-culture 3D model using primary mesothelial cells, fibroblasts, adipocyte cells, and high-grade OC cell lines were developed to study OC metastasis and drug response of patient-derived OC cells [389,390]. In addition, single-cell-derived metastatic OC spheroids (sMOCS) from ascites showed key features of cancer stemness of the original metastasis only in 3D and not in a 2D model [391]. Recently, a more sophisticated dynamical 3D model, mimicking hydrodynamic forces OC cells experience in the peritoneal cavity, was developed [392].
Xenograft models have also been successfully used in BC and OC research, and both BCSC and OCSC were shown to accelerate the tumor growth in mice [393,394]. Patient-derived xenografts (PDX) have been used in therapy efficiency and chemoresistance studies and were shown to be a good model for observing the changes in CD44+/CD24− BCSC populations and ALDH1+ OCSC populations [104,179,394,395]. PDX models have been shown to exhibit clinical and molecular characteristics of primary tumors, and can provide valuable information on tumor growth, metastasis, drug efficacy, and even prognosis both in breast [396,397,398] and ovarian cancer [399]. PDX models identified the hilum region of the mouse ovary, the transitional/junction area between ovarian surface epithelium (OSE), mesothelium, and tubal epithelium, as a previously unrecognized stem cell niche of the OSE [400].
Furthermore, decellularizing primary BC tissues led to the development of patient-derived scaffolds (PDS), which, when recellularized with BC cell lines, led to the development of stem-cell-like properties and a gene-expression profile similar to xenograft cultures [401]. In follow-up studies, the same group reported a change in BCSC markers NANOG, POU5F1, and ABCG2, and increased resistance to chemotherapeutics (5-fluorouracil, doxorubicin, and paclitaxel) [402] and endocrine therapies [403] in cells grown in PDS compared to 2D cultures, as well as an upregulation of CSC markers and a change of gene expression linked to prognostic features of the original cancer [404].
Regarding the patient material, the models most often used are the paraffin-embedded tissue slides and fresh or fresh frozen tissue samples (including ascites aspirates, biopsies, or solid tumors excised during surgery). They are used for the detection of gene and/or protein expression and mutation detection. For example, immunohistochemical staining and/or CSC-markers-based cell sorting revealed the abundance and CSC distribution between normal and cancer tissue at different stages and clinical outcomes [405,406,407,408].
Fresh tissues can be used to establish organoids/tumoroids in vitro, or for propagation in mice in PDX models [409].

8. CSC-Targeted Therapies

The heterogeneity of OC and BC is largely attributed to the presence of a subset of CSC, which have the ability to differentiate into multiple cancer cell types under various stimuli. Aggressive subtypes typically have a higher proportion of CSC, which can drive tumor development, progression, and resistance to therapy.
While primary tumors treated with specific therapy may initially respond well, they often become resistant and relapse when treated again with the same compound. This resistance to therapy is partially mediated by CSC, which can evade initial treatment by persisting in a quiescent, low proliferative state, and then re-activating and repopulating the tumor mass after therapy, often adopting a more resistant phenotype. CSC are intrinsically more resistant due to their active DNA repair, increased expression of ABC transporters, ALDH1, pro-survival BCL-2 protein family, and activation of signaling pathways such as MYC, PI3K/AKT, WNT, NOTCH, HH, NF-κB, and others [410,411,412]. However, chemotherapy can also have unintended consequences. While it can kill cancer cells, dying cancer cells can release IL8, which activates self-renewal and regeneration of CSC, potentially leading to the recurrence of cancer cells, as observed in BC [413]. Although OC is initially sensitive to chemotherapy, a high recurrence rate is observed due to the development of chemoresistance, which is thought to be mediated by OCSC [82].
Therefore, there is a great need for appropriate treatment strategies that are effective against CSC and improve the outcome of cancer therapy. Several approaches have been proposed to target CSC, including the use of small molecules, antibodies, and immune-based therapies. Therapeutic approaches for targeting CSC heavily depend on the identification and isolation of CSC using cell-surface markers. However, targeting these markers poses a challenge due to their overlap with embryonic and adult stem cells, which can potentially affect normal adult stem cells and impair the process of normal tissue regeneration [414]. Furthermore, the shared signaling pathways and transcription factors between CSC and normal stem cells present an additional challenge [415]. Moreover, the scarcity of CSC within tumors adds to the difficulty of isolating and identifying them [180]. Another promising approach in cancer treatment is the use of combination therapies, in which drugs targeting different signaling pathways and cell types are used simultaneously to improve treatment efficacy. Some of the treatment strategies commonly used for BC and OC are listed in Table 2.
Specific biomarkers of BCSC and OCSC are being utilized in treatment strategies. Section 3 of the mentioned source suggests that anti-CD44 or anti-EpCAM antibodies could be employed for treating BCSC and OCSC. Notably, catumaxomab, an anti-EpCAM antibody, received approval from the European Commission in 2009 for the treatment of EpCAM-overexpressing tumors such as BC and OC [443,444]. The combination of catumaxomab and activated T cells shows promising potential as a powerful therapeutic approach for combating chemoresistant TNBC cells that express EpCAM [445]. Another approach involves the use of an anti-CD133 antibody, which has shown promise in OC and BC. A fusion protein comprising an anti-single-chain variable fragment (scFv) peptide sequence that targets the extracellular domain of human CD133 (anti-CD133scFV) and deimmunized PE38KDEL (dCD133KDEL) demonstrated reduced growth of the NIH:OVCAR5 OC cell line and inhibited tumor progression in xenografts [446]. In a recent review, Tume et al. explored the potential of anti-CD133 for BCSC treatment. While this treatment approach offers certain advantages, the authors also highlighted several challenges, suggesting that combining conventional treatments with anti-CD133 may be a promising strategy [447]. In a glioblastoma PDX model, the CD133-specific chimeric antigen receptor T cell (CAR-T) known as CART133 demonstrated remarkable anti-tumor efficacy. Notably, it did not exhibit any harmful effects on normal CD133+ hematopoietic stem cells in humanized CD34+ mice. These findings suggest that CART133 may hold promise for targeting other treatment-resistant CD133+ tumors [448].
A subset of OC and BC cases caused by mutations in BRCA1 and BRCA2 genes are sensitive to Poly (ADP-ribose) polymerase (PARP) inhibitors. Although PARP inhibition shows encouraging results even in platinum-resistant patients, resistance to PARP inhibitors can develop [449]. This resistance may be due in part to PARP inhibition inducing the CD133+ and CD117+ cell populations [450]. In BC, which are generally more resistant to PARP inhibitors, the combination of PARP inhibitor Niraparib and cyclin-dependent kinase inhibitor dinaciclib reduced EMT and cancer stem-like cell phenotypes [451]. The downregulation of RAD51, which is thought to mediate the resistance to PARP inhibitors, sensitized BCSC to PARP inhibition and resulted in reduced tumor growth [452].
ALM201, a 23-residue peptide derived from the N-terminal region of the FK506-binding protein like (FKBPL), is a promising candidate for targeting OCSC, as it specifically binds to the CD44 receptor and exhibits anti-angiogenic and anti-CSC activity. In several OC cell lines, ALM201 reduced tumorsphere-forming efficiency, but its efficacy was not consistent in vivo in a xenograft model due to differences in the microenvironment [427]. Another peptide derivative of FKBPL, AD-01, showed promising activity in BC, reducing ESA+/CD44+/CD24− and ALDH+ cell numbers, self-renewal ability, and expression of OCT4, NANOG, and SOX2 in BC cell lines and xenografts [453].
As the MEK1/2-ERK1/2 pathway is active in the majority of HGSOC cases, inhibition of this pathway was also tested as a potential therapeutic approach. MEK1/2 inhibitor trametinib showed an inhibitory effect on HGSOC proliferation and induction G0/G1 cell cycle arrest in these cells. However, it was also shown to promote stemness, as higher percentages of CD133+ cells were detected after trametinib treatment, as well as an increase in gene expression of stemness markers SOX2, NANOG, OCT4, and ALDH1A in vitro and in vivo. Therefore, the authors stress the possibility of treatment failure and/or resistance due to these mechanisms [454].
In a preclinical study, the PI3K inhibitor XL147 was used in combination with trastuzumab to treat trastuzumab-resistant BC. The results showed that the combination therapy reduced cancer cell proliferation and increased apoptosis. The treatment also decreased mammosphere formation, inhibited tumor growth, and reduced ALDH activity. This suggests that the addition of a PI3K inhibitor to trastuzumab treatment could potentially improve therapeutic outcomes for trastuzumab-resistant BC patients [455].
Repurposing of existing therapeutics has yielded some promising candidates for targeting CSC, including metformin, an antidiabetic drug; salinomycin, an antimicrobial agent commonly used in agriculture; and calcium channel blockers. Metformin has been shown to have the potential to inhibit both OCSC and BCSC, with low concentrations selectively targeting CD44+ CD117+ OCSC while not affecting OC cells, resulting in the inhibition of OCSC growth in vitro and in vivo [163,456]. In BC, treatment with metformin alone has been shown to decrease the CD44+/CD24− population of BCSC and reduce their ability to form mammospheres. When combined with doxorubicin, a synergistic effect was more effective in the reduction of tumor mass and prevention of relapse than either drug alone [3]. However, metformin-induced activation of AMPK has shown a context-dependent effect and a survival-promoting role in dormant ER+ breast cancer cells, the subpopulation of cancer cells responsible for therapy resistance and cancer recurrence [457].
Salinomycin has also been shown to be a selective inhibitor of BCSC and OCSC. It induces apoptosis in OCSC and selectively kills BCSC while reducing tumor growth and EMT [416,417,458]. However, its disadvantage is poor solubility in water and poor bioavailability, which can be remedied by conjugation with CD133-targeted nanoparticles [459]. Calcium channel blockers (manidipine, lacidipine, benidipine, and lomerizine) have been identified as good candidates because they inhibit AKT and ERK signaling, decrease stemness, and promote OCSC apoptosis [460]. Similarly, a calcium channel blocker (amlodipine) inhibits the growth of BC cell lines, leads to downregulation of p-ERK1/2, and inhibits colony formation [461].
Natural products are often explored as potential new therapeutics for cancer treatment. Shikonin (SHK), for example, induced apoptosis and inhibited migration, invasion, and xenograft tumor growth, as well as the expression of CSC-related markers (ALDH1, OCT4, SOX2, and NANOG) in OC [462]. A very similar effect was described for BC, where it was shown that the downregulation of CSC markers was modulated by the inhibition of STAT3, FAK, and SRC [463].
In recent years, the need for more precise and efficient therapy of CSC has increased—CSC vaccines for OC (targeting CD117+CD44+ OCSC) were able to activate immune responses to autologous tumor antigens, which in turn reduced OCSC, prolonged survival, and reduced tumor growth in mice [464]. This is consistent with evidence showing that CSC vaccines with high ROR1 expression effectively activate the immune response against OC [465,466]. Vaccines against BCSC are also being studied—vaccines containing human BCSC-lysates have been shown to prolong the lives of mice with BC. Other types of vaccines have also been found to target BCSC and reduce tumor growth (reviewed in [467]).
Overall, a better understanding of CSC biology and their interactions with the TME is needed to develop effective therapies that can improve cancer treatment outcomes.

9. Conclusions

Breast and ovarian cancers are among the most common and deadly cancers affecting women worldwide. BC and OC show marked intertumor heterogeneity, which subdivides them into various subtypes, and even intratumor heterogeneity. Although BC and OC arise from different tissues, they share approximately 50% of the most frequently mutated genes. Integrated molecular analysis revealed major differences between BC subtypes and many similarities between HGSOC and basal-like TNBC, suggesting a related etiology [38]. Even if the primary tumors initially respond well, recurrence is common in both BC and OC. Current research highlights the importance of CSC in tumorigenesis, as CSC play an important role in cancer initiation, progression, survival, metastasis, and chemoresistance. They can be distinguished by certain CSC-related markers. Herein, we highlighted the commonalities of BSCS and OCSC in terms of CSC marker expression, pathway activation, immune evasion, and microenvironment remodeling, and how this knowledge could be used for CSC-targeted therapy. Further advances in experimental models and the discovery of potential additional CSC-related markers or combinations thereof that are responsible for more aggressive phenotypes will enable the enrichment of CSC and a more targeted approach to study complex CSC biology. Therefore, to improve patient outcomes, further research is essential to unravel the complex role of CSC in BC and OC. Understanding the underlying mechanisms of CSC in BC and OC will be crucial for developing new treatment strategies.

Author Contributions

Conceptualization, L.M.; writing—original draft preparation I.L., M.K., M.M., N.P., A.Č.G., M.S. and L.M.; writing—review and editing, I.L., M.K., A.Č.G., M.S. and L.M.; visualization M.K., I.L., M.S. and L.M.; supervision, L.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ferlay, J.; Ervik, M.; Lam, F.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today. Available online: https://gco.iarc.fr/today (accessed on 21 March 2023).
  2. Reid, B.M.; Permuth, J.B.; Sellers, T.A. Epidemiology of ovarian cancer: A review. Cancer Biol. Med. 2017, 14, 9–32. [Google Scholar] [CrossRef] [Green Version]
  3. Rebbeck, T.R.; Mitra, N.; Wan, F.; Sinilnikova, O.M.; Healey, S.; McGuffog, L.; Mazoyer, S.; Chenevix-Trench, G.; Easton, D.F.; Antoniou, A.C.; et al. Association of type and location of BRCA1 and BRCA2 mutations with risk of breast and ovarian cancer. JAMA 2015, 313, 1347–1361. [Google Scholar] [CrossRef] [Green Version]
  4. Fu, X.; Tan, W.; Song, Q.; Pei, H.; Li, J. BRCA1 and Breast Cancer: Molecular Mechanisms and Therapeutic Strategies. Front. Cell Dev. Biol. 2022, 10, 813457. [Google Scholar] [CrossRef]
  5. Eliade, M.; Skrzypski, J.; Baurand, A.; Jacquot, C.; Bertolone, G.; Loustalot, C.; Coutant, C.; Guy, F.; Fumoleau, P.; Duffourd, Y.; et al. The transfer of multigene panel testing for hereditary breast and ovarian cancer to healthcare: What are the implications for the management of patients and families? Oncotarget 2017, 8, 1957–1971. [Google Scholar] [CrossRef] [Green Version]
  6. Kuchenbaecker, K.B.; Hopper, J.L.; Barnes, D.R.; Phillips, K.-A.; Mooij, T.M.; Roos-Blom, M.-J.; Jervis, S.; van Leeuwen, F.E.; Milne, R.L.; Andrieu, N.; et al. Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 2017, 317, 2402–2416. [Google Scholar] [CrossRef] [Green Version]
  7. Chen, J.; Bae, E.; Zhang, L.; Hughes, K.; Parmigiani, G.; Braun, D.; Rebbeck, T.R. Penetrance of Breast and Ovarian Cancer in Women Who Carry a BRCA1/2 Mutation and Do Not Use Risk-Reducing Salpingo-Oophorectomy: An Updated Meta-Analysis. JNCI cancer Spectr. 2020, 4, pkaa029. [Google Scholar] [CrossRef]
  8. Hartmann, L.C.; Schaid, D.J.; Woods, J.E.; Crotty, T.P.; Myers, J.L.; Arnold, P.G.; Petty, P.M.; Sellers, T.A.; Johnson, J.L.; McDonnell, S.K.; et al. Efficacy of bilateral prophylactic mastectomy in women with a family history of breast cancer. N. Engl. J. Med. 1999, 340, 77–84. [Google Scholar] [CrossRef] [Green Version]
  9. Domchek, S.M.; Friebel, T.M.; Singer, C.F.; Evans, D.G.; Lynch, H.T.; Isaacs, C.; Garber, J.E.; Neuhausen, S.L.; Matloff, E.; Eeles, R.; et al. Association of risk-reducing surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and mortality. JAMA 2010, 304, 967–975. [Google Scholar] [CrossRef] [Green Version]
  10. Rebbeck, T.R.; Friebel, T.; Lynch, H.T.; Neuhausen, S.L.; van ’t Veer, L.; Garber, J.E.; Evans, G.R.; Narod, S.A.; Isaacs, C.; Matloff, E.; et al. Bilateral prophylactic mastectomy reduces breast cancer risk in BRCA1 and BRCA2 mutation carriers: The PROSE Study Group. J. Clin. Oncol. 2004, 22, 1055–1062. [Google Scholar] [CrossRef] [Green Version]
  11. Choi, Y.-H.; Terry, M.B.; Daly, M.B.; MacInnis, R.J.; Hopper, J.L.; Colonna, S.; Buys, S.S.; Andrulis, I.L.; John, E.M.; Kurian, A.W.; et al. Association of Risk-Reducing Salpingo-Oophorectomy with Breast Cancer Risk in Women with BRCA1 and BRCA2 Pathogenic Variants. JAMA Oncol. 2021, 7, 585–592. [Google Scholar] [CrossRef]
  12. Perri, T.; Levin, G.; Naor-Revel, S.; Eliassi-Revivo, P.; Lifshitz, D.; Friedman, E.; Korach, J. Risk-reducing salpingo-oophorectomy and breast cancer incidence among Jewish BRCA1/BRCA2-mutation carriers-an Israeli matched-pair study. Int. J. Gynaecol. Obstet. 2022, 157, 431–436. [Google Scholar] [CrossRef]
  13. Conduit, C.; Milne, R.L.; Friedlander, M.L.; Phillips, K.-A. Bilateral Salpingo-oophorectomy and Breast Cancer Risk for BRCA1 and BRCA2 Mutation Carriers: Assessing the Evidence. Cancer Prev. Res. 2021, 14, 983–994. [Google Scholar] [CrossRef] [PubMed]
  14. Michaelson-Cohen, R.; Gabizon–Peretz, S.; Armon, S.; Srebnik-Moshe, N.; Mor, P.; Tomer, A.; Levy-Lahad, E.; Paluch-Shimon, S. Breast cancer risk and hormone replacement therapy among BRCA carriers after risk-reducing salpingo-oophorectomy. Eur. J. Cancer 2021, 148, 95–102. [Google Scholar] [CrossRef] [PubMed]
  15. Prat, J. New insights into ovarian cancer pathology. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2012, 23 (Suppl. S1), x111–x117. [Google Scholar] [CrossRef] [PubMed]
  16. Johnson, K.S.; Conant, E.F.; Soo, M.S. Molecular Subtypes of Breast Cancer: A Review for Breast Radiologists. J. Breast Imaging 2021, 3, 12–24. [Google Scholar] [CrossRef]
  17. Feng, Z.; Wen, H.; Bi, R.; Ju, X.; Chen, X.; Yang, W.; Wu, X. A clinically applicable molecular classification for high-grade serous ovarian cancer based on hormone receptor expression. Sci. Rep. 2016, 6, 25408. [Google Scholar] [CrossRef] [Green Version]
  18. Lafourcade, A.; His, M.; Baglietto, L.; Boutron-Ruault, M.C.; Dossus, L.; Rondeau, V. Factors associated with breast cancer recurrences or mortality and dynamic prediction of death using history of cancer recurrences: The French E3N cohort. BMC Cancer 2018, 18, 171. [Google Scholar] [CrossRef]
  19. Garzon, S.; Laganà, A.S.; Casarin, J.; Raffaelli, R.; Cromi, A.; Franchi, M.; Barra, F.; Alkatout, I.; Ferrero, S.; Ghezzi, F. Secondary and tertiary ovarian cancer recurrence: What is the best management? Gland Surg. 2020, 9, 1118–1129. [Google Scholar] [CrossRef]
  20. Courtney, D.; Davey, M.G.; Moloney, B.M.; Barry, M.K.; Sweeney, K.; McLaughlin, R.P.; Malone, C.M.; Lowery, A.J.; Kerin, M.J. Breast cancer recurrence: Factors impacting occurrence and survival. Ir. J. Med. Sci. 2022, 191, 2501–2510. [Google Scholar] [CrossRef]
  21. Pedersen, R.N.; Esen, B.Ö.; Mellemkjær, L.; Christiansen, P.; Ejlertsen, B.; Lash, T.L.; Nørgaard, M.; Cronin-Fenton, D. The Incidence of Breast Cancer Recurrence 10-32 Years After Primary Diagnosis. JNCI J. Natl. Cancer Inst. 2022, 114, 391–399. [Google Scholar] [CrossRef]
  22. Colombo, N.; Lorusso, D.; Scollo, P. Impact of Recurrence of Ovarian Cancer on Quality of Life and Outlook for the Future. Int. J. Gynecol. Cancer 2017, 27, 1134–1140. [Google Scholar] [CrossRef] [Green Version]
  23. Bishop, A.J.; Ensor, J.; Moulder, S.L.; Shaitelman, S.F.; Edson, M.A.; Whitman, G.J.; Bishnoi, S.; Hoffman, K.E.; Stauder, M.C.; Valero, V.; et al. Prognosis for patients with metastatic breast cancer who achieve a no-evidence-of-disease status after systemic or local therapy. Cancer 2015, 121, 4324–4332. [Google Scholar] [CrossRef] [Green Version]
  24. Bilyk, O.; Coatham, M.; Jewer, M.; Postovit, L.-M. Epithelial-to-Mesenchymal Transition in the Female Reproductive Tract: From Normal Functioning to Disease Pathology. Front. Oncol. 2017, 7, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yamulla, R.J.; Nalubola, S.; Flesken-Nikitin, A.; Nikitin, A.Y.; Schimenti, J.C. Most Commonly Mutated Genes in High-Grade Serous Ovarian Carcinoma Are Nonessential for Ovarian Surface Epithelial Stem Cell Transformation. Cell Rep. 2020, 32, 108086. [Google Scholar] [CrossRef]
  26. Deng, K.; Yang, C.; Tan, Q.; Song, W.; Lu, M.; Zhao, W.; Lou, G.; Li, Z.; Li, K.; Hou, Y. Sites of distant metastases and overall survival in ovarian cancer: A study of 1481 patients. Gynecol. Oncol. 2018, 150, 460–465. [Google Scholar] [CrossRef]
  27. Tan, D.S.P.; Agarwal, R.; Kaye, S.B. Mechanisms of transcoelomic metastasis in ovarian cancer. Lancet Oncol. 2006, 7, 925–934. [Google Scholar] [CrossRef] [PubMed]
  28. Tjhay, F.; Motohara, T.; Tayama, S.; Narantuya, D.; Fujimoto, K.; Guo, J.; Sakaguchi, I.; Honda, R.; Tashiro, H.; Katabuchi, H. CD44 variant 6 is correlated with peritoneal dissemination and poor prognosis in patients with advanced epithelial ovarian cancer. Cancer Sci. 2015, 106, 1421–1428. [Google Scholar] [CrossRef] [Green Version]
  29. Al Habyan, S.; Kalos, C.; Szymborski, J.; McCaffrey, L. Multicellular detachment generates metastatic spheroids during intra-abdominal dissemination in epithelial ovarian cancer. Oncogene 2018, 37, 5127–5135. [Google Scholar] [CrossRef]
  30. Yeung, T.-L.; Leung, C.S.; Yip, K.-P.; Au Yeung, C.L.; Wong, S.T.C.; Mok, S.C. Cellular and molecular processes in ovarian cancer metastasis. A Review in the Theme: Cell and Molecular Processes in Cancer Metastasis. Am. J. Physiol. Cell Physiol. 2015, 309, C444–C456. [Google Scholar] [CrossRef] [Green Version]
  31. Di, J.; Duiveman-de Boer, T.; Zusterzeel, P.L.M.; Figdor, C.G.; Massuger, L.F.A.G.; Torensma, R. The stem cell markers Oct4A, Nanog and c-Myc are expressed in ascites cells and tumor tissue of ovarian cancer patients. Cell. Oncol. 2013, 36, 363–374. [Google Scholar] [CrossRef] [PubMed]
  32. Latifi, A.; Luwor, R.B.; Bilandzic, M.; Nazaretian, S.; Stenvers, K.; Pyman, J.; Zhu, H.; Thompson, E.W.; Quinn, M.A.; Findlay, J.K.; et al. Isolation and characterization of tumor cells from the ascites of ovarian cancer patients: Molecular phenotype of chemoresistant ovarian tumors. PLoS ONE 2012, 7, e46858. [Google Scholar] [CrossRef] [Green Version]
  33. Bregenzer, M.E.; Horst, E.N.; Mehta, P.; Novak, C.M.; Repetto, T.; Mehta, G. The Role of Cancer Stem Cells and Mechanical Forces in Ovarian Cancer Metastasis. Cancers 2019, 11, 1008. [Google Scholar] [CrossRef] [Green Version]
  34. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Proteomics. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
  35. Rakha, E.; Toss, M.; Quinn, C. Specific cell differentiation in breast cancer: A basis for histological classification. J. Clin. Pathol. 2022, 75, 76–84. [Google Scholar] [CrossRef]
  36. Feng, Y.; Spezia, M.; Huang, S.; Yuan, C.; Zeng, Z.; Zhang, L.; Ji, X.; Liu, W.; Huang, B.; Luo, W.; et al. Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis. 2018, 5, 77–106. [Google Scholar] [CrossRef]
  37. Ng, C.K.Y.; Bidard, F.-C.; Piscuoglio, S.; Geyer, F.C.; Lim, R.S.; de Bruijn, I.; Shen, R.; Pareja, F.; Berman, S.H.; Wang, L.; et al. Genetic Heterogeneity in Therapy-Naïve Synchronous Primary Breast Cancers and Their Metastases. Clin. Cancer Res. 2017, 23, 4402–4415. [Google Scholar] [CrossRef] [Green Version]
  38. Cancer Genome Atlas Network Comprehensive molecular portraits of human breast tumours. Nature 2012, 490, 61–70. [CrossRef] [Green Version]
  39. Martins, F.C.; de Santiago, I.; Trinh, A.; Xian, J.; Guo, A.; Sayal, K.; Jimenez-Linan, M.; Deen, S.; Driver, K.; Mack, M.; et al. Combined image and genomic analysis of high-grade serous ovarian cancer reveals PTEN loss as a common driver event and prognostic classifier. Genome Biol. 2014, 15, 526. [Google Scholar] [CrossRef] [Green Version]
  40. Martins, F.C.; Couturier, D.-L.; Paterson, A.; Karnezis, A.N.; Chow, C.; Nazeran, T.M.; Odunsi, A.; Gentry-Maharaj, A.; Vrvilo, A.; Hein, A.; et al. Clinical and pathological associations of PTEN expression in ovarian cancer: A multicentre study from the Ovarian Tumour Tissue Analysis Consortium. Br. J. Cancer 2020, 123, 793–802. [Google Scholar] [CrossRef]
  41. Li, W.; Gu, X.; Liu, C.; Shi, Y.; Wang, P.; Zhang, N.; Wu, R.; Leng, L.; Xie, B.; Song, C.; et al. A synergetic effect of BARD1 mutations on tumorigenesis. Nat. Commun. 2021, 12, 1243. [Google Scholar] [CrossRef]
  42. Tegally, H.; Kensler, K.H.; Mungloo-Dilmohamud, Z.; Ghoorah, A.W.; Rebbeck, T.R.; Baichoo, S. Discovering novel driver mutations from pan-cancer analysis of mutational and gene expression profiles. PLoS ONE 2020, 15, e0242780. [Google Scholar] [CrossRef] [PubMed]
  43. Dick, J.E. Breast cancer stem cells revealed. Proc. Natl. Acad. Sci. USA 2003, 100, 3547–3549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chhabra, S.N.; Booth, B.W. Asymmetric cell division of mammary stem cells. Cell Div. 2021, 16, 5. [Google Scholar] [CrossRef] [PubMed]
  45. Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [Green Version]
  46. Xu, H.; Tian, Y.; Yuan, X.; Wu, H.; Liu, Q.; Pestell, R.G.; Wu, K. The role of CD44 in epithelial-mesenchymal transition and cancer development. Onco. Targets. Ther. 2015, 8, 3783–3792. [Google Scholar] [CrossRef] [Green Version]
  47. Al-Othman, N.; Alhendi, A.; Ihbaisha, M.; Barahmeh, M.; Alqaraleh, M.; Al-Momany, B.Z. Role of CD44 in breast cancer. Breast Dis. 2020, 39, 1–13. [Google Scholar] [CrossRef]
  48. Lim, S.-C.; Oh, S.-H. The role of CD24 in various human epithelial neoplasias. Pathol. Res. Pract. 2005, 201, 479–486. [Google Scholar] [CrossRef]
  49. Kristiansen, G.; Winzer, K.J.; Mayordomo, E.; Bellach, J.; Schlüns, K.; Denkert, C.; Dahl, E.; Pilarsky, C.; Altevogt, P.; Guski, H.; et al. CD24 Expression Is a New Prognostic Marker in Breast Cancer. Clin. Cancer Res. 2003, 9, 4906–4913. [Google Scholar]
  50. Kristiansen, G.; Denkert, C.; Schlüns, K.; Dahl, E.; Pilarsky, C.; Hauptmann, S. CD24 is expressed in ovarian cancer and is a new independent prognostic marker of patient survival. Am. J. Pathol. 2002, 161, 1215–1221. [Google Scholar] [CrossRef] [Green Version]
  51. Park, S.Y.; Lee, H.E.; Li, H.; Shipitsin, M.; Gelman, R.; Polyak, K. Heterogeneity for stem cell-related markers according to tumor subtype and histologic stage in breast cancer. Clin. Cancer Res. 2010, 16, 876–887. [Google Scholar] [CrossRef] [Green Version]
  52. Gao, M.-Q.; Choi, Y.-P.; Kang, S.; Youn, J.H.; Cho, N.-H. CD24+ cells from hierarchically organized ovarian cancer are enriched in cancer stem cells. Oncogene 2010, 29, 2672–2680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Surowiak, P.; Materna, V.; Kaplenko, I.; Spaczyński, M.; Dietel, M.; Kristiansen, G.; Lage, H.; Zabel, M. Unfavorable prognostic value of CD24 expression in sections from primary and relapsed ovarian cancer tissue. Int. J. Gynecol. Cancer 2006, 16, 515–521. [Google Scholar] [CrossRef]
  54. Choi, Y.-L.; Kim, S.-H.; Shin, Y.K.; Hong, Y.-C.; Lee, S.-J.; Kang, S.Y.; Ahn, G. Cytoplasmic CD24 expression in advanced ovarian serous borderline tumors. Gynecol. Oncol. 2005, 97, 379–386. [Google Scholar] [CrossRef]
  55. Gao, Y.; Foster, R.; Yang, X.; Feng, Y.; Shen, J.K.; Mankin, H.J.; Hornicek, F.J.; Amiji, M.M.; Duan, Z. Up-regulation of CD44 in the development of metastasis, recurrence and drug resistance of ovarian cancer. Oncotarget 2015, 6, 9313–9326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ponta, H.; Sherman, L.; Herrlich, P.A. CD44: From adhesion molecules to signalling regulators. Nat. Rev. Mol. Cell Biol. 2003, 4, 33–45. [Google Scholar] [CrossRef]
  57. Uchino, M.; Kojima, H.; Wada, K.; Imada, M.; Onoda, F.; Satofuka, H.; Utsugi, T.; Murakami, Y. Nuclear β-catenin and CD44 upregulation characterize invasive cell populations in non-aggressive MCF-7 breast cancer cells. BMC Cancer 2010, 10, 414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Börjesson, P.K.E.; Postema, E.J.; Roos, J.C.; Colnot, D.R.; Marres, H.A.M.; van Schie, M.H.; Stehle, G.; de Bree, R.; Snow, G.B.; Oyen, W.J.G.; et al. Phase I therapy study with (186)Re-labeled humanized monoclonal antibody BIWA 4 (bivatuzumab) in patients with head and neck squamous cell carcinoma. Clin. Cancer Res. 2003, 9, 3961S–3972S. [Google Scholar] [PubMed]
  59. Meng, E.; Long, B.; Sullivan, P.; McClellan, S.; Finan, M.A.; Reed, E.; Shevde, L.; Rocconi, R.P. CD44+/CD24− ovarian cancer cells demonstrate cancer stem cell properties and correlate to survival. Clin. Exp. Metastasis 2012, 29, 939–948. [Google Scholar] [CrossRef] [PubMed]
  60. Li, X.; Lewis, M.T.; Huang, J.; Gutierrez, C.; Osborne, C.K.; Wu, M.F.; Hilsenbeck, S.G.; Pavlick, A.; Zhang, X.; Chamness, G.C.; et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J. Natl. Cancer Inst. 2008, 100, 672–679. [Google Scholar] [CrossRef]
  61. Roy, R.; Willan, P.; Clarke, R.; Farnie, G. Differentiation therapy: Targeting breast cancer stem cells to reduce resistance to radiotherapy and chemotherapy. Breast Cancer Res. 2010, 12, O5. [Google Scholar] [CrossRef] [Green Version]
  62. Ricardo, S.; Vieira, A.F.; Gerhard, R.; Leitão, D.; Pinto, R.; Cameselle-Teijeiro, J.F.; Milanezi, F.; Schmitt, F.; Paredes, J. Breast cancer stem cell markers CD44, CD24 and ALDH1: Expression distribution within intrinsic molecular subtype. J. Clin. Pathol. 2011, 64, 937–944. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, Z.; Wang, Q.; Wang, Q.; Wang, Y.; Chen, J. Prognostic Significance of CD24 and CD44 in Breast Cancer: A Meta-Analysis. Int. J. Biol. Markers 2017, 32, 75–82. [Google Scholar] [CrossRef] [PubMed]
  64. Friedl, P.; Alexander, S. Cancer invasion and the microenvironment: Plasticity and reciprocity. Cell 2011, 147, 992–1009. [Google Scholar] [CrossRef] [Green Version]
  65. Sheridan, C.; Kishimoto, H.; Fuchs, R.K.; Mehrotra, S.; Bhat-Nakshatri, P.; Turner, C.H.; Goulet, R.; Badve, S.; Nakshatri, H. CD44+/CD24- breast cancer cells exhibit enhanced invasive properties: An early step necessary for metastasis. Breast Cancer Res. 2006, 8, R59. [Google Scholar] [CrossRef] [Green Version]
  66. Li, W.; Ma, H.; Zhang, J.; Zhu, L.; Wang, C.; Yang, Y. Unraveling the roles of CD44/CD24 and ALDH1 as cancer stem cell markers in tumorigenesis and metastasis. Sci. Rep. 2017, 7, 13856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Yang, C.; Cao, M.; Liu, Y.; He, Y.; Du, Y.; Zhang, G.; Gao, F. Inducible formation of leader cells driven by CD44 switching gives rise to collective invasion and metastases in luminal breast carcinomas. Oncogene 2019, 38, 7113–7132. [Google Scholar] [CrossRef]
  68. Zanoni, M.; Bravaccini, S.; Fabbri, F.; Arienti, C. Emerging Roles of Aldehyde Dehydrogenase Isoforms in Anti-cancer Therapy Resistance. Front. Med. 2022, 9, 795762. [Google Scholar] [CrossRef]
  69. Deng, S.; Yang, X.; Lassus, H.; Liang, S.; Kaur, S.; Ye, Q.; Li, C.; Wang, L.-P.; Roby, K.F.; Orsulic, S.; et al. Distinct expression levels and patterns of stem cell marker, aldehyde dehydrogenase isoform 1 (ALDH1), in human epithelial cancers. PLoS ONE 2010, 5, e10277. [Google Scholar] [CrossRef]
  70. Landen, C.N.; Goodman, B.; Katre, A.A.; Steg, A.D.; Nick, A.M.; Stone, R.L.; Miller, L.D.; Mejia, P.V.; Jennings, N.B.; Gershenson, D.M.; et al. Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer. Mol. Cancer Ther. 2010, 9, 3186–3199. [Google Scholar] [CrossRef] [Green Version]
  71. Tanei, T.; Morimoto, K.; Shimazu, K.; Kim, S.J.; Tanji, Y.; Taguchi, T.; Tamaki, Y.; Noguchi, S. Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin-based chemotherapy for breast cancers. Clin. Cancer Res. 2009, 15, 4234–4241. [Google Scholar] [CrossRef] [Green Version]
  72. Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 Is a Marker of Normal and Malignant Human Mammary Stem Cells and a Predictor of Poor Clinical Outcome. Cell Stem Cell 2007, 1, 555–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Corbeil, D.; Röper, K.; Fargeas, C.A.; Joester, A.; Huttner, W.B. Prominin: A Story of Cholesterol, Plasma Membrane Protrusions and Human Pathology. Traffic 2001, 2, 82–91. [Google Scholar] [CrossRef] [Green Version]
  74. Anderson, L.H.; Boulanger, C.A.; Smith, G.H.; Carmeliet, P.; Watson, C.J. Stem cell marker prominin-1 regulates branching morphogenesis, but not regenerative capacity, in the mammary gland. Dev. Dyn. 2011, 240, 674–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Wright, M.H.; Calcagno, A.M.; Salcido, C.D.; Carlson, M.D.; Ambudkar, S.V.; Varticovski, L. Brca1 breast tumors contain distinct CD44+/CD24-and CD133+cells with cancer stem cell characteristics. Breast Cancer Res. 2008, 10, R10. [Google Scholar] [CrossRef] [Green Version]
  76. Brugnoli, F.; Grassilli, S.; Al-Qassab, Y.; Capitani, S.; Bertagnolo, V. CD133 in Breast Cancer Cells: More than a Stem Cell Marker. J. Oncol. 2019, 2019, 7512632. [Google Scholar] [CrossRef] [Green Version]
  77. Liu, T.J.; Sun, B.C.; Zhao, X.L.; Zhao, X.M.; Sun, T.; Gu, Q.; Yao, Z.; Dong, X.Y.; Zhao, N.; Liu, N. CD133+ cells with cancer stem cell characteristics associates with vasculogenic mimicry in triple-negative breast cancer. Oncogene 2013, 32, 544–553. [Google Scholar] [CrossRef] [Green Version]
  78. Ferrandina, G.; Bonanno, G.; Pierelli, L.; Perillo, A.; Procoli, A.; Mariotti, A.; Corallo, M.; Martinelli, E.; Rutella, S.; Paglia, A.; et al. Expression of CD133-1 and CD133-2 in ovarian cancer. Int. J. Gynecol. Cancer 2008, 18, 506–514. [Google Scholar] [CrossRef]
  79. Long, H.; Xie, R.; Xiang, T.; Zhao, Z.; Lin, S.; Liang, Z.; Chen, Z.; Zhu, B. Autocrine CCL5 signaling promotes invasion and migration of CD133+ ovarian cancer stem-like cells via NF-κB-mediated MMP-9 upregulation. Stem Cells 2012, 30, 2309–2319. [Google Scholar] [CrossRef]
  80. Mal, A.; Bukhari, A.B.; Singh, R.K.; Kapoor, A.; Barai, A.; Deshpande, I.; Wadasadawala, T.; Ray, P.; Sen, S.; De, A. EpCAM-Mediated Cellular Plasticity Promotes Radiation Resistance and Metastasis in Breast Cancer. Front. Cell Dev. Biol. 2020, 8, 597673. [Google Scholar] [CrossRef]
  81. Hiraga, T.; Ito, S.; Nakamura, H. EpCAM expression in breast cancer cells is associated with enhanced bone metastasis formation. Int. J. Cancer 2016, 138, 1698–1708. [Google Scholar] [CrossRef] [Green Version]
  82. Walters Haygood, C.L.; Arend, R.C.; Straughn, J.M.; Buchsbaum, D.J. Ovarian cancer stem cells: Can targeted therapy lead to improved progression-free survival? World J. Stem Cells 2014, 6, 441–447. [Google Scholar] [CrossRef]
  83. Zheng, J.; Zhao, S.; Yu, X.; Huang, S.; Liu, H.Y. Simultaneous targeting of CD44 and EpCAM with a bispecific aptamer effectively inhibits intraperitoneal ovarian cancer growth. Theranostics 2017, 7, 1373–1388. [Google Scholar] [CrossRef]
  84. Münz, M.; Murr, A.; Kvesic, M.; Rau, D.; Mangold, S.; Pflanz, S.; Lumsden, J.; Volkland, J.; Fagerberg, J.; Riethmüller, G.; et al. Side-by-side analysis of five clinically tested anti-EpCAM monoclonal antibodies. Cancer Cell Int. 2010, 10, 44. [Google Scholar] [CrossRef] [Green Version]
  85. Schmidt, M.; Scheulen, M.E.; Dittrich, C.; Obrist, P.; Marschner, N.; Dirix, L.; Schmidt, M.; Rüttinger, D.; Schuler, M.; Reinhardt, C.; et al. An open-label, randomized phase II study of adecatumumab, a fully human anti-EpCAM antibody, as monotherapy in patients with metastatic breast cancer. Ann. Oncol. 2010, 21, 275–282. [Google Scholar] [CrossRef]
  86. Seimetz, D.; Lindhofer, H.; Bokemeyer, C. Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3) as a targeted cancer immunotherapy. Cancer Treat. Rev. 2010, 36, 458–467. [Google Scholar] [CrossRef]
  87. Szotek, P.P.; Pieretti-Vanmarcke, R.; Masiakos, P.T.; Dinulescu, D.M.; Connolly, D.; Foster, R.; Dombkowski, D.; Preffer, F.; MacLaughlin, D.T.; Donahoe, P.K. Ovarian cancer side population defines cells with stem cell-like characteristics and Mullerian Inhibiting Substance responsiveness. Proc. Natl. Acad. Sci. USA 2006, 103, 11154–11159. [Google Scholar] [CrossRef] [Green Version]
  88. Moserle, L.; Indraccolo, S.; Ghisi, M.; Frasson, C.; Fortunato, E.; Canevari, S.; Miotti, S.; Tosello, V.; Zamarchi, R.; Corradin, A.; et al. The Side Population of Ovarian Cancer Cells Is a Primary Target of IFN-α Antitumor Effects. Cancer Res. 2008, 68, 5658–5668. [Google Scholar] [CrossRef] [Green Version]
  89. Hu, L.; McArthur, C.; Jaffe, R.B. Ovarian cancer stem-like side-population cells are tumourigenic and chemoresistant. Br. J. Cancer 2010, 102, 1276–1283. [Google Scholar] [CrossRef] [Green Version]
  90. Leccia, F.; Del Vecchio, L.; Mariotti, E.; Di Noto, R.; Morel, A.P.; Puisieux, A.; Salvatore, F.; Ansieau, S. ABCG2, a novel antigen to sort luminal progenitors of BRCA1- breast cancer cells. Mol. Cancer 2014, 13, 213. [Google Scholar] [CrossRef] [Green Version]
  91. Honeth, G.; Bendahl, P.O.; Ringnér, M.; Saal, L.H.; Gruvberger-Saal, S.K.; Lövgren, K.; Grabau, D.; Fernö, M.; Borg, Å.; Hegardt, C. The CD44+/CD24 phenotype is enriched in basal-like breast tumors. Breast Cancer Res. 2008, 10, R53. [Google Scholar] [CrossRef] [Green Version]
  92. Croker, A.K.; Goodale, D.; Chu, J.; Postenka, C.; Hedley, B.D.; Hess, D.A.; Allan, A.L. High aldehyde dehydrogenase and expression of cancer stem cell markers selects for breast cancer cells with enhanced malignant and metastatic ability. J. Cell. Mol. Med. 2009, 13, 2236–2252. [Google Scholar] [CrossRef] [Green Version]
  93. Kryczek, I.; Liu, S.; Roh, M.; Vatan, L.; Szeliga, W.; Wei, S.; Banerjee, M.; Mao, Y.; Kotarski, J.; Wicha, M.S.; et al. Expression of aldehyde dehydrogenase and CD133 defines ovarian cancer stem cells. Int. J. Cancer 2012, 130, 29–39. [Google Scholar] [CrossRef] [Green Version]
  94. Siu, M.K.Y.; Wong, E.S.Y.; Kong, D.S.H.; Chan, H.Y.; Jiang, L.; Wong, O.G.W.; Lam, E.W.-F.; Chan, K.K.L.; Ngan, H.Y.S.; Le, X.-F.; et al. Stem cell transcription factor NANOG controls cell migration and invasion via dysregulation of E-cadherin and FoxJ1 and contributes to adverse clinical outcome in ovarian cancers. Oncogene 2013, 32, 3500–3509. [Google Scholar] [CrossRef] [Green Version]
  95. Han, J.; Zhang, F.; Yu, M.; Zhao, P.; Ji, W.; Zhang, H.; Wu, B.; Wang, Y.; Niu, R. RNA interference-mediated silencing of NANOG reduces cell proliferation and induces G0/G1 cell cycle arrest in breast cancer cells. Cancer Lett. 2012, 321, 80–88. [Google Scholar] [CrossRef]
  96. Liu, K.; Xie, F.; Gao, A.; Zhang, R.; Zhang, L.; Xiao, Z.; Hu, Q.; Huang, W.; Huang, Q.; Lin, B.; et al. SOX2 regulates multiple malignant processes of breast cancer development through the SOX2/miR-181a-5p, miR-30e-5p/TUSC3 axis. Mol. Cancer 2017, 16, 62. [Google Scholar] [CrossRef] [Green Version]
  97. Leis, O.; Eguiara, A.; Lopez-Arribillaga, E.; Alberdi, M.J.; Hernandez-Garcia, S.; Elorriaga, K.; Pandiella, A.; Rezola, R.; Martin, A.G. Sox2 expression in breast tumours and activation in breast cancer stem cells. Oncogene 2012, 31, 1354–1365. [Google Scholar] [CrossRef] [Green Version]
  98. Wen, Y.; Hou, Y.; Huang, Z.; Cai, J.; Wang, Z. SOX2 is required to maintain cancer stem cells in ovarian cancer. Cancer Sci. 2017, 108, 719–731. [Google Scholar] [CrossRef] [Green Version]
  99. Mohiuddin, I.S.; Wei, S.J.; Kang, M.H. Role of OCT4 in cancer stem-like cells and chemotherapy resistance. Biochim. Biophys. Acta—Mol. Basis Dis. 2020, 1866, 165432. [Google Scholar] [CrossRef]
  100. Yan, H.C.; Fang, L.S.; Xu, J.; Qiu, Y.Y.; Lin, X.M.; Huang, H.X.; Han, Q.Y. The identification of the biological characteristics of human ovarian cancer stem cells. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 3497–3503. [Google Scholar]
  101. Zhang, S.; Balch, C.; Chan, M.W.W.; Lai, H.-C.; Matei, D.; Schilder, J.M.M.; Yan, P.S.S.; Huang, T.H.-M.H.-M.; Nephew, K.P.P. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008, 68, 4311–4320. [Google Scholar] [CrossRef] [Green Version]
  102. Boesch, M.; Zeimet, A.G.; Reimer, D.; Schmidt, S.; Gastl, G.; Parson, W.; Spoeck, F.; Hatina, J.; Wolf, D.; Sopper, S. The side population of ovarian cancer cells defines a heterogeneous compartment exhibiting stem cell characteristics. Oncotarget 2014, 5, 7027–7039. [Google Scholar] [CrossRef] [Green Version]
  103. Zhang, Q.; Han, Z.; Zhu, Y.; Chen, J.; Li, W. The Role and Specific Mechanism of OCT4 in Cancer Stem Cells: A Review. Int. J. Stem Cells 2020, 13, 312. [Google Scholar] [CrossRef]
  104. Zhang, S.; Zhang, H.; Ghia, E.M.; Huang, J.; Wu, L.; Zhang, J.; Lam, S.; Lei, Y.; He, J.; Cui, B.; et al. Inhibition of chemotherapy resistant breast cancer stem cells by a ROR1 specific antibody. Proc. Natl. Acad. Sci. USA 2019, 116, 1370–1377. [Google Scholar] [CrossRef] [Green Version]
  105. Henry, C.; Llamosas, E.; Knipprath-Meszaros, A.; Schoetzau, A.; Obermann, E.; Fuenfschilling, M.; Caduff, R.; Fink, D.; Hacker, N.; Ward, R.; et al. Targeting the ROR1 and ROR2 receptors in epithelial ovarian cancer inhibits cell migration and invasion. Oncotarget 2015, 6, 40310–40326. [Google Scholar] [CrossRef] [Green Version]
  106. Zou, H.; Luo, J.; Guo, Y.; Liu, Y.; Wang, Y.; Deng, L.; Li, P. RNA-binding protein complex LIN28/MSI2 enhances cancer stem cell-like properties by modulating Hippo-YAP1 signaling and independently of Let-7. Oncogene 2022, 41, 1657–1672. [Google Scholar] [CrossRef]
  107. Peng, S.; Maihle, N.J.; Huang, Y. Pluripotency factors Lin28 and Oct4 identify a sub-population of stem cell-like cells in ovarian cancer. Oncogene 2010, 29, 2153–2159. [Google Scholar] [CrossRef] [Green Version]
  108. Lu, H.; Clauser, K.R.; Tam, W.L.; Fröse, J.; Ye, X.; Eaton, E.N.; Reinhardt, F.; Donnenberg, V.S.; Bhargava, R.; Carr, S.A.; et al. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat. Cell Biol. 2014, 16, 1105–1117. [Google Scholar] [CrossRef] [Green Version]
  109. Connor, E.V.; Saygin, C.; Braley, C.; Wiechert, A.C.; Karunanithi, S.; Crean-Tate, K.; Abdul-Karim, F.W.; Michener, C.M.; Rose, P.G.; Lathia, J.D.; et al. Thy-1 predicts poor prognosis and is associated with self-renewal in ovarian cancer. J. Ovarian Res. 2019, 12, 112. [Google Scholar] [CrossRef] [Green Version]
  110. Yang, L.; Tang, H.; Kong, Y.; Xie, X.; Chen, J.; Song, C.; Liu, X.; Ye, F.; Li, N.; Wang, N.; et al. LGR5 Promotes Breast Cancer Progression and Maintains Stem-Like Cells Through Activation of Wnt/β-Catenin Signaling. Stem Cells 2015, 33, 2913–2924. [Google Scholar] [CrossRef]
  111. Kim, H.; Lee, D.H.; Park, E.; Myung, J.K.; Park, J.H.; Kim, D.I.; Kim, S.I.; Lee, M.; Kim, Y.; Park, C.M.; et al. Differential epithelial and stromal LGR5 expression in ovarian carcinogenesis. Sci. Rep. 2022, 12, 11200. [Google Scholar] [CrossRef]
  112. Liu, W.; Zhang, J.; Gan, X.; Shen, F.; Yang, X.; Du, N.; Xia, D.; Liu, L.; Qiao, L.; Pan, J.; et al. LGR5 promotes epithelial ovarian cancer proliferation, metastasis, and epithelial–mesenchymal transition through the Notch1 signaling pathway. Cancer Med. 2018, 7, 3132–3142. [Google Scholar] [CrossRef]
  113. Lo, P.K.; Kanojia, D.; Liu, X.; Singh, U.P.; Berger, F.G.; Wang, Q.; Chen, H. CD49f and CD61 identify Her2/neu-induced mammary tumor-initiating cells that are potentially derived from luminal progenitors and maintained by the integrin-TGFΒ signaling. Oncogene 2012, 31, 2614–2626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Wiechert, A.; Saygin, C.; Thiagarajan, P.S.; Rao, V.S.; Hale, J.S.; Gupta, N.; Hitomi, M.; Nagaraj, A.B.; DiFeo, A.; Lathia, J.D.; et al. Cisplatin induces stemness in ovarian cancer. Oncotarget 2016, 7, 30511–30522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Moein, S.; Tenen, D.G.; Amabile, G.; Chai, L. SALL4: An Intriguing Therapeutic Target in Cancer Treatment. Cells 2022, 11, 2601. [Google Scholar] [CrossRef] [PubMed]
  116. Sharbatoghli, M.; Shamshiripour, P.; Fattahi, F.; Kalantari, E.; Habibi Shams, Z.; Panahi, M.; Totonchi, M.; Asadi-Lari, Z.; Madjd, Z.; Saeednejad Zanjani, L. Co-expression of cancer stem cell markers, SALL4/ALDH1A1, is associated with tumor aggressiveness and poor survival in patients with serous ovarian carcinoma. J. Ovarian Res. 2022, 15, 17. [Google Scholar] [CrossRef] [PubMed]
  117. Srinivasan, M.; Bharali, D.J.; Sudha, T.; Khedr, M.; Guest, I.; Sell, S.; Glinsky, G.V.; Mousa, S.A. Downregulation of Bmi1 in breast cancer stem cells suppresses tumor growth and proliferation. Oncotarget 2017, 8, 38731–38742. [Google Scholar] [CrossRef] [Green Version]
  118. Zhao, Q.; Qian, Q.; Cao, D.; Yang, J.; Gui, T.; Shen, K. Role of BMI1 in epithelial ovarian cancer: Investigated via the CRISPR/Cas9 system and RNA sequencing. J. Ovarian Res. 2018, 11, 31. [Google Scholar] [CrossRef]
  119. Grange, C.; Lanzardo, S.; Cavallo, F.; Camussi, G.; Bussolati, B. SCA-1 identifies the tumor-initiating cells in mammary tumors of BALB-neuT trangenic mice. Neoplasia 2008, 10, 1433–1443. [Google Scholar] [CrossRef] [Green Version]
  120. Liu, L.; Yin, B.; Yi, Z.; Liu, X.J.; Hu, Z.Q.; Gao, W.C.; Yu, H.W.; Li, Q.Q. Breast cancer stem cells characterized by CD70 expression preferentially metastasize to the lungs. Breast Cancer 2018, 25, 706–716. [Google Scholar] [CrossRef]
  121. Vassilopoulos, A.; Chisholm, C.; Lahusen, T.; Zheng, H.; Deng, C.-X. A critical role of CD29 and CD49f in mediating metastasis for cancer-initiating cells isolated from a Brca1-associated mouse model of breast cancer. Oncogene 2014, 33, 5477–5482. [Google Scholar] [CrossRef]
  122. Yu, F.; Li, J.; Chen, H.; Fu, J.; Ray, S.; Huang, S.; Zheng, H.; Ai, W. Kruppel-like factor 4 (KLF4) is required for maintenance of breast cancer stem cells and for cell migration and invasion. Oncogene 2011, 30, 2161–2172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. 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. [Google Scholar] [CrossRef] [PubMed]
  124. Kim, D.K.; Ham, M.H.; Lee, S.Y.; Shin, M.J.; Kim, Y.E.; Song, P.; Suh, D.-S.; Kim, J.H. CD166 promotes the cancer stem-like properties of primary epithelial ovarian cancer cells. BMB Rep. 2020, 53, 622–627. [Google Scholar] [CrossRef] [PubMed]
  125. Mitsui, H.; Shibata, K.; Suzuki, S.; Umezu, T.; Mizuno, M.; Kajiyama, H.; Kikkawa, F. Functional interaction between peritoneal mesothelial cells and stem cells of ovarian yolk sac tumor (SC-OYST) in peritoneal dissemination. Gynecol. Oncol. 2012, 124, 303–310. [Google Scholar] [CrossRef]
  126. Eyre, R.; Harvey, I.; Stemke-Hale, K.; Lennard, T.W.J.; Tyson-Capper, A.; Meeson, A.P. Reversing paclitaxel resistance in ovarian cancer cells via inhibition of the ABCB1 expressing side population. Tumour Biol. 2014, 35, 9879–9892. [Google Scholar] [CrossRef]
  127. Coffman, L.; Mooney, C.; Lim, J.; Bai, S.; Silva, I.; Gong, Y.; Yang, K.; Buckanovich, R.J. Endothelin receptor-A is required for the recruitment of antitumor T cells and modulates chemotherapy induction of cancer stem cells. Cancer Biol. Ther. 2013, 14, 184–192. [Google Scholar] [CrossRef] [Green Version]
  128. Kong, X.; Wang, X.; Xu, W.; Behera, S.; Hellermann, G.; Kumar, A.; Lockey, R.F.; Mohapatra, S.; Mohapatra, S.S. Natriuretic peptide receptor a as a novel anticancer target. Cancer Res. 2008, 68, 249–256. [Google Scholar] [CrossRef] [Green Version]
  129. Fan, Q.; Zhang, W.; Emerson, R.E.; Xu, Y. ZIP4 Is a Novel Cancer Stem Cell Marker in High-Grade Serous Ovarian Cancer. Cancers 2020, 12, 3692. [Google Scholar] [CrossRef]
  130. Xiang, T.; Long, H.; He, L.; Han, X.; Lin, K.; Liang, Z.; Zhuo, W.; Xie, R.; Zhu, B. Interleukin-17 produced by tumor microenvironment promotes self-renewal of CD133+ cancer stem-like cells in ovarian cancer. Oncogene 2015, 34, 165–176. [Google Scholar] [CrossRef]
  131. Wei, X.; Dombkowski, D.; Meirelles, K.; Pieretti-Vanmarcke, R.; Szotek, P.P.; Chang, H.L.; Preffer, F.I.; Mueller, P.R.; Teixeira, J.; MacLaughlin, D.T.; et al. Mullerian inhibiting substance preferentially inhibits stem/progenitors in human ovarian cancer cell lines compared with chemotherapeutics. Proc. Natl. Acad. Sci. USA 2010, 107, 18874–18879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. d’Adhemar, C.J.; Spillane, C.D.; Gallagher, M.F.; Bates, M.; Costello, K.M.; Barry-O’Crowley, J.; Haley, K.; Kernan, N.; Murphy, C.; Smyth, P.C.; et al. The MyD88+ phenotype is an adverse prognostic factor in epithelial ovarian cancer. PLoS ONE 2014, 9, e100816. [Google Scholar] [CrossRef] [Green Version]
  133. Zhang, L.; Ma, R.; Gao, M.; Zhao, Y.; Lv, X.; Zhu, W.; Han, L.; Su, P.; Fan, Y.; Yan, Y.; et al. SNORA72 Activates the Notch1/c-Myc Pathway to Promote Stemness Transformation of Ovarian Cancer Cells. Front. Cell Dev. Biol. 2020, 8, 583087. [Google Scholar] [CrossRef] [PubMed]
  134. Zeng, Y.A.; Nusse, R. Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture. Cell Stem Cell 2010, 6, 568–577. [Google Scholar] [CrossRef] [Green Version]
  135. Ng, A.; Tan, S.; Singh, G.; Rizk, P.; Swathi, Y.; Tan, T.Z.; Huang, R.Y.-J.; Leushacke, M.; Barker, N. Lgr5 marks stem/progenitor cells in ovary and tubal epithelia. Nat. Cell Biol. 2014, 16, 745–757. [Google Scholar] [CrossRef]
  136. Logan, C.Y.; Nusse, R. The Wnt signaling pathway in development and disease. Annu. Rev. Cell Dev. Biol. 2004, 20, 781–810. [Google Scholar] [CrossRef] [Green Version]
  137. Hojo, N.; Huisken, A.L.; Wang, H.; Chirshev, E.; Kim, N.S.; Nguyen, S.M.; Campos, H.; Glackin, C.A.; Ioffe, Y.J.; Unternaehrer, J.J. Snail knockdown reverses stemness and inhibits tumour growth in ovarian cancer. Sci. Rep. 2018, 8, 8704. [Google Scholar] [CrossRef] [Green Version]
  138. Chau, W.K.; Ip, C.K.; Mak, A.S.C.; Lai, H.-C.; Wong, A.S.T. c-Kit mediates chemoresistance and tumor-initiating capacity of ovarian cancer cells through activation of Wnt/β-catenin-ATP-binding cassette G2 signaling. Oncogene 2013, 32, 2767–2781. [Google Scholar] [CrossRef] [Green Version]
  139. Raghavan, S.; Mehta, P.; Xie, Y.; Lei, Y.L.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Lin, S.Y.; Xia, W.; Wang, J.C.; Kwong, K.Y.; Spohn, B.; Wen, Y.; Pestell, R.G.; Hung, M.C. β-catenin, a novel prognostic marker for breast cancer: Its roles in cyclin D1 expression and cancer progression. Proc. Natl. Acad. Sci. USA 2000, 97, 4262–4266. [Google Scholar] [CrossRef] [Green Version]
  141. Monteiro, J.; Gaspar, C.; Richer, W.; Franken, P.F.; Sacchetti, A.; Joosten, R.; Idali, A.; Brandao, J.; Decraene, C.; Fodde, R. Cancer stemness in Wnt-driven mammary tumorigenesis. Carcinogenesis 2014, 35, 2–13. [Google Scholar] [CrossRef] [Green Version]
  142. Wu, Y.; Ginther, C.; Kim, J.; Mosher, N.; Chung, S.; Slamon, D.; Vadgama, J.V. Expression of Wnt3 activates Wnt/β-catenin pathway and promotes EMT-like phenotype in trastuzumab-resistant HER2-overexpressing breast cancer cells. Mol. Cancer Res. 2012, 10, 1597–1606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Jang, G.B.; Kim, J.Y.; Cho, S.D.; Park, K.S.; Jung, J.Y.; Lee, H.Y.; Hong, I.S.; Nam, J.S. Blockade of Wnt/β-catenin signaling suppresses breast cancer metastasis by inhibiting CSC-like phenotype. Sci. Rep. 2015, 5, 12465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Shan, N.L.; Shin, Y.; Yang, G.; Furmanski, P.; Suh, N. Breast Cancer Stem Cells: A Review of Their Characteristics and The Agents That Affect Them. Mol. Carcinog. 2021, 60, 73. [Google Scholar] [CrossRef]
  145. Xu, J.; Prosperi, J.R.; Choudhury, N.; Olopade, O.I.; Goss, K.H. β-Catenin Is Required for the Tumorigenic Behavior of Triple-Negative Breast Cancer Cells. PLoS ONE 2015, 10, e0117097. [Google Scholar] [CrossRef] [Green Version]
  146. Yan, Y.; Liu, F.; Han, L.; Zhao, L.; Chen, J.; Olopade, O.I.; He, M.; Wei, M. HIF-2α promotes conversion to a stem cell phenotype and induces chemoresistance in breast cancer cells by activating Wnt and Notch pathways. J. Exp. Clin. Cancer Res. 2018, 37, 256. [Google Scholar] [CrossRef] [Green Version]
  147. Rascio, F.; Spadaccino, F.; Rocchetti, M.T.; Castellano, G.; Stallone, G.; Netti, G.S.; Ranieri, E. The Pathogenic Role of PI3K/AKT Pathway in Cancer Onset and Drug Resistance: An Updated Review. Cancers 2021, 13, 3949. [Google Scholar] [CrossRef]
  148. Stemke-Hale, K.; Gonzalez-Angulo, A.M.; Lluch, A.; Neve, R.M.; Kuo, W.L.; Davies, M.; Carey, M.; Hu, Z.; Guan, Y.; Sahin, A.; et al. An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res. 2008, 68, 6084–6091. [Google Scholar] [CrossRef] [Green Version]
  149. Guo, T.; Dong, X.; Xie, S.; Zhang, L.; Zeng, P.; Zhang, L. Cellular Mechanism of Gene Mutations and Potential Therapeutic Targets in Ovarian Cancer. Cancer Manag. Res. 2021, 13, 3081–3100. [Google Scholar] [CrossRef]
  150. Li, H.; Prever, L.; Hirsch, E.; Gulluni, F. Targeting PI3K/AKT/mTOR Signaling Pathway in Breast Cancer. Cancers (Basel). 2021, 13, 3517. [Google Scholar] [CrossRef]
  151. Kinross, K.M.; Montgomery, K.G.; Kleinschmidt, M.; Waring, P.; Ivetac, I.; Tikoo, A.; Saad, M.; Hare, L.; Roh, V.; Mantamadiotis, T.; et al. An activating Pik3ca mutation coupled with Pten loss is sufficient to initiate ovarian tumorigenesis in mice. J. Clin. Invest. 2012, 122, 553–557. [Google Scholar] [CrossRef]
  152. du Rusquec, P.; Blonz, C.; Frenel, J.S.; Campone, M. Targeting the PI3K/Akt/mTOR pathway in estrogen-receptor positive HER2 negative advanced breast cancer. Ther. Adv. Med. Oncol. 2020, 12, 1758835920940939. [Google Scholar] [CrossRef]
  153. Li, H.X.; Zeng, J.F.; Shen, K. PI3K/AKT/mTOR signaling pathway as a therapeutic target for ovarian cancer. Arch. Gynecol. Obstet. 2014, 290, 1067–1078. [Google Scholar] [CrossRef]
  154. Miricescu, D.; Totan, A.; Stanescu-Spinu, I.I.; Badoiu, S.C.; Stefani, C.; Greabu, M. PI3K/AKT/mTOR Signaling Pathway in Breast Cancer: From Molecular Landscape to Clinical Aspects. Int. J. Mol. Sci. 2021, 22, 173. [Google Scholar] [CrossRef] [PubMed]
  155. 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. [Google Scholar] [CrossRef] [PubMed]
  156. Shayesteh, L.; Lu, Y.; Kuo, W.L.; Baldocchi, R.; Godfrey, T.; Collins, C.; Pinkel, D.; Powell, B.; Mills, G.B.; Gray, J.W. PlK3CA is implicated as an oncogene in ovarian cancer. Nat. Genet. 1999, 21, 99–102. [Google Scholar] [CrossRef]
  157. Madsen, R.R.; Erickson, E.C.; Rueda, O.M.; Robin, X.; Caldas, C.; Toker, A.; Semple, R.K.; Vanhaesebroeck, B. Positive correlation between transcriptomic stemness and PI3K/AKT/mTOR signaling scores in breast cancer, and a counterintuitive relationship with PIK3CA genotype. PLoS Genet. 2021, 17, e1009876. [Google Scholar] [CrossRef]
  158. Zhou, J.; Wulfkuhle, J.; Zhang, H.; Gu, P.; Yang, Y.; Deng, J.; Margolick, J.B.; Liotta, L.A.; Petricoin, E.; Zhang, Y. Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc. Natl. Acad. Sci. USA 2007, 104, 16158–16163. [Google Scholar] [CrossRef] [Green Version]
  159. Russo, A.; Colina, J.A.; Moy, J.; Baligod, S.; Czarnecki, A.A.; Varughese, P.; Lantvit, D.D.; Dean, M.J.; Burdette, J.E. Silencing PTEN in the fallopian tube promotes enrichment of cancer stem cell-like function through loss of PAX2. Cell Death Dis. 2021, 12, 375. [Google Scholar] [CrossRef]
  160. Rivas, S.; Gómez-Oro, C.; Antón, I.M.; Wandosell, F. Role of Akt isoforms controlling cancer stem cell survival, phenotype and self-renewal. Biomedicines 2018, 6, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Douville, J.; Beaulieu, R.; Balicki, D. ALDH1 as a Functional Marker of Cancer Stem and Progenitor Cells. Stem Cells Dev. 2009, 18, 17–26. [Google Scholar] [CrossRef] [PubMed]
  162. Lee, H.; Park, H.J.; Park, C.S.; Oh, E.T.; Choi, B.H.; Williams, B.; Lee, C.K.; Song, C.W. Response of breast cancer cells and cancer stem cells to metformin and hyperthermia alone or combined. PLoS ONE 2014, 9, e87979. [Google Scholar] [CrossRef] [PubMed]
  163. Zhang, R.; Zhang, P.; Wang, H.; Hou, D.; Li, W.; Xiao, G.; Li, C. Inhibitory effects of metformin at low concentration on epithelial-mesenchymal transition of CD44(+)CD117(+) ovarian cancer stem cells. Stem Cell Res. Ther. 2015, 6, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Wang, Y.; Luo, X.; Wu, N.; Liao, Q.; Wang, J. SRC-3/TRAF4 facilitates ovarian cancer development by activating the PI3K/AKT signaling pathway. Med. Oncol. 2023, 40, 76. [Google Scholar] [CrossRef]
  165. Gu, Y.; Gao, H.; Zhang, H.; John, A.; Zhu, X.; Shivaram, S.; Yu, J.; Weinshilboum, R.M.; Wang, L. TRAF4 hyperactivates HER2 signaling and contributes to Trastuzumab resistance in HER2-positive breast cancer. Oncogene 2022, 41, 4119–4129. [Google Scholar] [CrossRef]
  166. Li, L.; Deng, C.-X.; Chen, Q. SRC-3, a Steroid Receptor Coactivator: Implication in Cancer. Int. J. Mol. Sci. 2021, 22, 4760. [Google Scholar] [CrossRef]
  167. Jiang, N.; Hu, Y.; Wang, M.; Zhao, Z.; Li, M. The Notch Signaling Pathway Contributes to Angiogenesis and Tumor Immunity in Breast Cancer. Breast Cancer Targets Ther. 2022, 14, 291–309. [Google Scholar] [CrossRef]
  168. Lobry, C.; Oh, P.; Mansour, M.R.; Look, A.T.; Aifantis, I. Notch signaling: Switching an oncogene to a tumor suppressor. Blood 2014, 123, 2451–2459. [Google Scholar] [CrossRef] [Green Version]
  169. Hopfer, O.; Zwahlen, D.; Fey, M.F.; Aebi, S. The Notch pathway in ovarian carcinomas and adenomas. Br. J. Cancer 2005, 93, 709–718. [Google Scholar] [CrossRef] [Green Version]
  170. Giuli, M.V.; Mancusi, A.; Giuliani, E.; Screpanti, I.; Checquolo, S. Notch signaling in female cancers: A multifaceted node to overcome drug resistance. Cancer drug Resist. 2021, 4, 805–836. [Google Scholar] [CrossRef]
  171. Liu, Z.; Zhu, Y.; Li, F.; Xie, Y. GATA1-regulated JAG1 promotes ovarian cancer progression by activating Notch signal pathway. Protoplasma 2020, 257, 901–910. [Google Scholar] [CrossRef]
  172. Kim, M.J.; Kim, A.-R.; Jeong, J.-Y.; Kim, K.; Kim, T.-H.; Lee, C.; Chung, K.; Ko, Y.-H.; An, H.-J. Correlation of ALDH1 and Notch3 Expression: Clinical implication in Ovarian Carcinomas. J. Cancer 2017, 8, 3331–3342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Edwards, A.; Brennan, K. Notch Signalling in Breast Development and Cancer. Front. cell Dev. Biol. 2021, 9, 692173. [Google Scholar] [CrossRef] [PubMed]
  174. Ibragimova, M.; Tsyganov, M.; Litviakov, N. Tumour Stem Cells in Breast Cancer. Int. J. Mol. Sci. 2022, 23, 5058. [Google Scholar] [CrossRef]
  175. Seo, E.J.; Kim, D.K.; Jang, I.H.; Choi, E.J.; Shin, S.H.; Lee, S.I.; Kwon, S.-M.; Kim, K.-H.; Suh, D.-S.; Kim, J.H. Hypoxia-NOTCH1-SOX2 signaling is important for maintaining cancer stem cells in ovarian cancer. Oncotarget 2016, 7, 55624–55638. [Google Scholar] [CrossRef] [Green Version]
  176. Weijzen, S.; Rizzo, P.; Braid, M.; Vaishnav, R.; Jonkheer, S.M.; Zlobin, A.; Osborne, B.A.; Gottipati, S.; Aster, J.C.; Hahn, W.C.; et al. Activation of Notch-1 signaling maintains the neoplastic phenotype in human Ras-transformed cells. Nat. Med. 2002, 8, 979–986. [Google Scholar] [CrossRef] [PubMed]
  177. Nigam, A. Breast Cancer Stem Cells, Pathways and Therapeutic Perspectives 2011. Indian J. Surg. 2013, 75, 170–180. [Google Scholar] [CrossRef] [Green Version]
  178. McGowan, P.M.; Simedrea, C.; Ribot, E.J.; Foster, P.J.; Palmieri, D.; Steeg, P.S.; Allan, A.L.; Chambers, A.F. Notch1 inhibition alters the CD44 hi/CD24 lo population and reduces the formation of brain metastases from breast cancer. Mol. Cancer Res. 2011, 9, 834–844. [Google Scholar] [CrossRef] [Green Version]
  179. Qiu, M.; Peng, Q.; Jiang, I.; Carroll, C.; Han, G.; Rymer, I.; Lippincott, J.; Zachwieja, J.; Gajiwala, K.; Kraynov, E.; et al. Specific inhibition of Notch1 signaling enhances the antitumor efficacy of chemotherapy in triple negative breast cancer through reduction of cancer stem cells. Cancer Lett. 2013, 328, 261–270. [Google Scholar] [CrossRef]
  180. Ma, H.; Tian, T.; Cui, Z. Targeting ovarian cancer stem cells: A new way out. Stem Cell Res. Ther. 2023, 14, 28. [Google Scholar] [CrossRef]
  181. Grudzien, P.; Lo, S.; Albain, K.S.; Robinson, P.; Rajan, P.; Strack, P.R.; Golde, T.E.; Miele, L.; Foreman, K.E. Inhibition of notch signaling reduces the stem-like population of breast cancer cells and prevents mammosphere formation. Anticancer Res. 2010, 30, 3853–3867. [Google Scholar] [CrossRef]
  182. Muñoz-Galván, S.; Carnero, A. Targeting Cancer Stem Cells to Overcome Therapy Resistance in Ovarian Cancer. Cells 2020, 9, 1402. [Google Scholar] [CrossRef] [PubMed]
  183. Varjosalo, M.; Taipale, J. Hedgehog: Functions and mechanisms. Genes Dev. 2008, 22, 2454–2472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Cochrane, C.R.; Szczepny, A.; Watkins, D.N.; Cain, J.E. Hedgehog Signaling in the Maintenance of Cancer Stem Cells. Cancers 2015, 7, 1554–1585. [Google Scholar] [CrossRef]
  185. Sari, I.N.; Phi, L.T.H.; Jun, N.; Wijaya, Y.T.; Lee, S.; Kwon, H.Y. Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells. Cells 2018, 7, 208. [Google Scholar] [CrossRef] [Green Version]
  186. Pietrobono, S.; Gagliardi, S.; Stecca, B. Non-canonical Hedgehog Signaling Pathway in Cancer: Activation of GLI Transcription Factors Beyond Smoothened. Front. Genet. 2019, 10, 556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Liao, X.; Siu, M.K.Y.; Au, C.W.H.; Wong, E.S.Y.; Chan, H.Y.; Ip, P.P.C.; Ngan, H.Y.S.; Cheung, A.N.Y. Aberrant activation of hedgehog signaling pathway in ovarian cancers: Effect on prognosis, cell invasion and differentiation. Carcinogenesis 2009, 30, 131–140. [Google Scholar] [CrossRef]
  188. Clement, V.; Sanchez, P.; de Tribolet, N.; Radovanovic, I.; Ruiz i Altaba, A. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr. Biol. 2007, 17, 165–172. [Google Scholar] [CrossRef]
  189. Zhao, H.; Li, N.; Pang, Y.; Zhao, J.; Wu, X. Gli affects the stemness and prognosis of epithelial ovarian cancer via homeobox protein NANOG. Mol. Med. Rep. 2021, 23, 128. [Google Scholar] [CrossRef]
  190. Ray, A.; Meng, E.; Reed, E.; Shevde, L.A.; Rocconi, R.P. Hedgehog signaling pathway regulates the growth of ovarian cancer spheroid forming cells. Int. J. Oncol. 2011, 39, 797–804. [Google Scholar] [CrossRef]
  191. Liu, S.; Dontu, G.; Mantle, I.D.; Patel, S.; Ahn, N.S.; Jackson, K.W.; Suri, P.; Wicha, M.S. Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res. 2006, 66, 6063–6071. [Google Scholar] [CrossRef] [Green Version]
  192. He, M.; Fu, Y.; Yan, Y.; Xiao, Q.; Wu, H.; Yao, W.; Zhaov, H.; Zhao, L.; Jiang, Q.; Yu, Z.; et al. The Hedgehog signalling pathway mediates drug response of MCF-7 mammosphere cells in breast cancer patients. Clin. Sci. 2015, 129, 809–822. [Google Scholar] [CrossRef] [PubMed]
  193. Riaz, S.K.; Khan, J.S.; Shah, S.T.A.; Wang, F.; Ye, L.; Jiang, W.G.; Malik, M.F.A. Involvement of hedgehog pathway in early onset, aggressive molecular subtypes and metastatic potential of breast cancer. Cell Commun. Signal. 2018, 16, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Han, B.; Qu, Y.; Jin, Y.; Yu, Y.; Deng, N.; Wawrowsky, K.; Zhang, X.; Li, N.; Bose, S.; Wang, Q.; et al. FOXC1 Activates Smoothened-Independent Hedgehog Signaling in Basal-like Breast Cancer. Cell Rep. 2015, 13, 1046–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Chen, Q.; Gao, G.; Luo, S. Hedgehog signaling pathway and ovarian cancer. Chin. J. Cancer Res. 2013, 25, 346–353. [Google Scholar] [CrossRef] [PubMed]
  196. Chen, Q.; Xu, R.; Zeng, C.; Lu, Q.; Huang, D.; Shi, C.; Zhang, W.; Deng, L.; Yan, R.; Rao, H.; et al. Down-Regulation of Gli Transcription Factor Leads to the Inhibition of Migration and Invasion of Ovarian Cancer Cells via Integrin β4-Mediated FAK Signaling. PLoS ONE 2014, 9, e88386. [Google Scholar] [CrossRef] [Green Version]
  197. Bhateja, P.; Cherian, M.; Majumder, S.; Ramaswamy, B. The Hedgehog Signaling Pathway: A Viable Target in Breast Cancer? Cancers 2019, 11, 1126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Levy, D.E.; Darnell, J.E. Stats: Transcriptional control and biological impact. Nat. Rev. Mol. Cell Biol. 2002, 3, 651–662. [Google Scholar] [CrossRef]
  199. Chung, S.S.; Aroh, C.; Vadgama, J.V. Constitutive activation of STAT3 signaling regulates hTERT and promotes stem cell-like traits in human breast cancer cells. PLoS ONE 2013, 8, e83971. [Google Scholar] [CrossRef]
  200. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
  201. Abubaker, K.; Luwor, R.B.; Zhu, H.; McNally, O.; Quinn, M.A.; Burns, C.J.; Thompson, E.W.; Findlay, J.K.; Ahmed, N. Inhibition of the JAK2/STAT3 pathway in ovarian cancer results in the loss of cancer stem cell-like characteristics and a reduced tumor burden. BMC Cancer 2014, 14, 317. [Google Scholar] [CrossRef] [Green Version]
  202. Burgos-Ojeda, D.; Wu, R.; McLean, K.; Chen, Y.-C.; Talpaz, M.; Yoon, E.; Cho, K.R.; Buckanovich, R.J. CD24+ Ovarian Cancer Cells Are Enriched for Cancer-Initiating Cells and Dependent on JAK2 Signaling for Growth and Metastasis. Mol. Cancer Ther. 2015, 14, 1717–1727. [Google Scholar] [CrossRef] [Green Version]
  203. Ruan, Z.; Yang, X.; Cheng, W. OCT4 accelerates tumorigenesis through activating JAK/STAT signaling in ovarian cancer side population cells. Cancer Manag. Res. 2019, 11, 389–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Wang, D.; Xiang, T.; Zhao, Z.; Lin, K.; Yin, P.; Jiang, L.; Liang, Z.; Zhu, B. Autocrine interleukin-23 promotes self-renewal of CD133+ ovarian cancer stem-like cells. Oncotarget 2016, 7, 76006–76020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. To, S.Q.; Dmello, R.S.; Richards, A.K.; Ernst, M.; Chand, A.L. STAT3 Signaling in Breast Cancer: Multicellular Actions and Therapeutic Potential. Cancers 2022, 14, 429. [Google Scholar] [CrossRef]
  206. Wei, W.; Tweardy, D.J.; Zhang, M.; Zhang, X.; Landua, J.; Petrovic, I.; Bu, W.; Roarty, K.; Hilsenbeck, S.G.; Rosen, J.M.; et al. STAT3 Signaling Is Activated Preferentially in Tumor-Initiating Cells in Claudin-Low Models of Human Breast Cancer. Stem Cells 2014, 32, 2571–2582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Lin, L.; Hutzen, B.; Lee, H.F.; Peng, Z.; Wang, W.; Zhao, C.; Lin, H.J.; Sun, D.; Li, P.K.; Li, C.; et al. Evaluation of STAT3 signaling in ALDH+ and ALDH+/CD44+/CD24- subpopulations of breast cancer cells. PLoS ONE 2013, 8, e82821. [Google Scholar] [CrossRef] [Green Version]
  208. Weng, Y.-S.; Tseng, H.-Y.; Chen, Y.-A.; Shen, P.-C.; Al Haq, A.T.; Chen, L.-M.; Tung, Y.-C.; Hsu, H.-L. MCT-1/miR-34a/IL-6/IL-6R signaling axis promotes EMT progression, cancer stemness and M2 macrophage polarization in triple-negative breast cancer. Mol. Cancer 2019, 18, 42. [Google Scholar] [CrossRef] [Green Version]
  209. Deng, X.; Zhang, P.; Liang, T.; Deng, S.; Chen, X.; Zhu, L. Ovarian cancer stem cells induce the M2 polarization of macrophages through the PPARγ and NF-κB pathways. Int. J. Mol. Med. 2015, 36, 449–454. [Google Scholar] [CrossRef] [Green Version]
  210. Tzavlaki, K.; Moustakas, A. TGF-β Signaling. Biomolecules 2020, 10, 487. [Google Scholar] [CrossRef] [Green Version]
  211. Bruna, A.; Greenwood, W.; Le Quesne, J.; Teschendorff, A.; Miranda-Saavedra, D.; Rueda, O.M.; Sandoval, J.L.; Vidakovic, A.T.; Saadi, A.; Pharoah, P.; et al. TGFβ induces the formation of tumour-initiating cells in claudin low breast cancer. Nat. Commun. 2012, 3, 1055. [Google Scholar] [CrossRef]
  212. Rafehi, S.; Ramos Valdes, Y.; Bertrand, M.; McGee, J.; Préfontaine, M.; Sugimoto, A.; DiMattia, G.E.; Shepherd, T.G. TGFβ signaling regulates epithelial-mesenchymal plasticity in ovarian cancer ascites-derived spheroids. Endocr. Relat. Cancer 2016, 23, 147–159. [Google Scholar] [CrossRef] [Green Version]
  213. Asiedu, M.K.; Ingle, J.N.; Behrens, M.D.; Radisky, D.C.; Knutson, K.L. TGFβ/TNFα-Mediated Epithelial–Mesenchymal Transition Generates Breast Cancer Stem Cells with a Claudin-Low Phenotype. Cancer Res. 2011, 71, 4707–4719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Bellomo, C.; Caja, L.; Moustakas, A. Transforming growth factor β as regulator of cancer stemness and metastasis. Br. J. Cancer 2016, 115, 761–769. [Google Scholar] [CrossRef] [Green Version]
  215. Xu, X.; Zhang, L.; He, X.; Zhang, P.; Sun, C.; Xu, X.; Lu, Y.; Li, F. TGF-β plays a vital role in triple-negative breast cancer (TNBC) drug-resistance through regulating stemness, EMT and apoptosis. Biochem. Biophys. Res. Commun. 2018, 502, 160–165. [Google Scholar] [CrossRef] [PubMed]
  216. Chihara, Y.; Shimoda, M.; Hori, A.; Ohara, A.; Naoi, Y.; Ikeda, J.; Kagara, N.; Tanei, T.; Shimomura, A.; Shimazu, K.; et al. A small-molecule inhibitor of SMAD3 attenuates resistance to anti-HER2 drugs in HER2-positive breast cancer cells. Breast Cancer Res. Treat. 2017, 166, 55–68. [Google Scholar] [CrossRef] [PubMed]
  217. Cao, L.; Shao, M.; Schilder, J.; Guise, T.; Mohammad, K.S.; Matei, D. Tissue transglutaminase links TGF-β, epithelial to mesenchymal transition and a stem cell phenotype in ovarian cancer. Oncogene 2012, 31, 2521–2534. [Google Scholar] [CrossRef] [Green Version]
  218. Wang, C.-W.; Lee, B.-H.; Tai, C.-J. The inhibition of cordycepin on cancer stemness in TGF-beta induced chemo-resistant ovarian cancer cell. Oncotarget 2017, 8, 111912–111921. [Google Scholar] [CrossRef] [Green Version]
  219. Bhola, N.E.; Balko, J.M.; Dugger, T.C.; Kuba, M.G.; Sánchez, V.; Sanders, M.; Stanford, J.; Cook, R.S.; Arteaga, C.L. TGF-β inhibition enhances chemotherapy action against triple-negative breast cancer. J. Clin. Investig. 2013, 123, 1348–1358. [Google Scholar] [CrossRef]
  220. Guido, C.; Whitaker-Menezes, D.; Capparelli, C.; Balliet, R.; Lin, Z.; Pestell, R.G.; Howell, A.; Aquila, S.; Andò, S.; Martinez-Outschoorn, U.; et al. Metabolic reprogramming of cancer-associated fibroblasts by TGF-β drives tumor growth: Connecting TGF-β signaling with “Warburg-like” cancer metabolism and L-lactate production. Cell Cycle 2012, 11, 3019–3035. [Google Scholar] [CrossRef] [Green Version]
  221. Yeung, T.-L.; Leung, C.S.; Wong, K.-K.; Samimi, G.; Thompson, M.S.; Liu, J.; Zaid, T.M.; Ghosh, S.; Birrer, M.J.; Mok, S.C. TGF-β modulates ovarian cancer invasion by upregulating CAF-derived versican in the tumor microenvironment. Cancer Res. 2013, 73, 5016–5028. [Google Scholar] [CrossRef] [Green Version]
  222. Labelle, M.; Begum, S.; Hynes, R.O. Direct Signaling between Platelets and Cancer Cells Induces an Epithelial-Mesenchymal-Like Transition and Promotes Metastasis. Cancer Cell 2011, 20, 576–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Wen, H.; Qian, M.; He, J.; Li, M.; Yu, Q.; Leng, Z. Inhibiting of self-renewal, migration and invasion of ovarian cancer stem cells by blocking TGF-β pathway. PLoS ONE 2020, 15, e0230230. [Google Scholar] [CrossRef] [PubMed]
  224. Vazquez-Santillan, K.; Melendez-Zajgla, J.; Jimenez-Hernandez, L.E.; Gaytan-Cervantes, J.; Munõz-Galindo, L.; Pinã-Sanchez, P.; Martinez-Ruiz, G.; Torres, J.; Garcia-Lopez, P.; Gonzalez-Torres, C.; et al. NF-kappaΒ-inducing kinase regulates stem cell phenotype in breast cancer. Sci. Reports 2016, 6, 37340. [Google Scholar] [CrossRef] [Green Version]
  225. Sarkar, F.H.; Li, Y.; Wang, Z.; Kong, D. NF-kappaB signaling pathway and its therapeutic implications in human diseases. Int. Rev. Immunol. 2008, 27, 293–319. [Google Scholar] [CrossRef]
  226. Rinkenbaugh, A.L.; Baldwin, A.S. The NF-κB Pathway and Cancer Stem Cells. Cells 2016, 5, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Yamamoto, M.; Taguchi, Y.; Ito-Kureha, T.; Semba, K.; Yamaguchi, N.; Inoue, J.I. NF-κB non-cell-autonomously regulates cancer stem cell populations in the basal-like breast cancer subtype. Nat. Commun. 2013, 4, 2299. [Google Scholar] [CrossRef] [Green Version]
  228. Smith, S.M.; Lyu, Y.L.; Cai, L. NF-κB affects proliferation and invasiveness of breast cancer cells by regulating CD44 expression. PLoS ONE 2014, 9, e106966. [Google Scholar] [CrossRef] [Green Version]
  229. Alvero, A.B.; Chen, R.; Fu, H.-H.; Montagna, M.; Schwartz, P.E.; Rutherford, T.; Silasi, D.-A.; Steffensen, K.D.; Waldstrom, M.; Visintin, I.; et al. Molecular phenotyping of human ovarian cancer stem cells unravel the mechanisms for repair and chemo-resistance. Cell Cycle 2009, 8, 158–166. [Google Scholar] [CrossRef] [Green Version]
  230. House, C.D.; Jordan, E.; Hernandez, L.; Ozaki, M.; James, J.M.; Kim, M.; Kruhlak, M.J.; Batchelor, E.; Elloumi, F.; Cam, M.C.; et al. NFκB Promotes Ovarian Tumorigenesis via Classical Pathways That Support Proliferative Cancer Cells and Alternative Pathways That Support ALDH+ Cancer Stem-like Cells. Cancer Res. 2017, 77, 6927–6940. [Google Scholar] [CrossRef] [Green Version]
  231. Gonzalez-Torres, C.; Gaytan-Cervantes, J.; Vazquez-Santillan, K.; Mandujano-Tinoco, E.A.; Ceballos-Cancino, G.; Garcia-Venzor, A.; Zampedri, C.; Sanchez-Maldonado, P.; Mojica-Espinosa, R.; Jimenez-Hernandez, L.E.; et al. NF-κB Participates in the Stem Cell Phenotype of Ovarian Cancer Cells. Arch. Med. Res. 2017, 48, 343–351. [Google Scholar] [CrossRef]
  232. Alvero, A.B.; Fu, H.-H.; Holmberg, J.; Visintin, I.; Mor, L.; Marquina, C.C.; Oidtman, J.; Silasi, D.-A.; Mor, G. Stem-like ovarian cancer cells can serve as tumor vascular progenitors. Stem Cells 2009, 27, 2405–2413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Hallis, S.P.; Kim, S.K.; Lee, J.-H.; Kwak, M.-K. Association of NRF2 with HIF-2α-induced cancer stem cell phenotypes in chronic hypoxic condition. Redox Biol. 2023, 60, 102632. [Google Scholar] [CrossRef] [PubMed]
  234. Zhou, J.; Zhang, H.; Gu, P.; Bai, J.; Margolick, J.B.; Zhang, Y. NF-κB pathway inhibitors preferentially inhibit breast cancer stem-like cells. Breast Cancer Res. Treat. 2008, 111, 419–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Calses, P.C.; Crawford, J.J.; Lill, J.R.; Dey, A. Hippo Pathway in Cancer: Aberrant Regulation and Therapeutic Opportunities. Trends in cancer 2019, 5, 297–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Fu, M.; Hu, Y.; Lan, T.; Guan, K.-L.; Luo, T.; Luo, M. The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct. Target. Ther. 2022, 7, 376. [Google Scholar] [CrossRef]
  237. Hall, C.A.; Wang, R.; Miao, J.; Oliva, E.; Shen, X.; Wheeler, T.; Hilsenbeck, S.G.; Orsulic, S.; Goode, S. Hippo pathway effector Yap is an ovarian cancer oncogene. Cancer Res. 2010, 70, 8517–8525. [Google Scholar] [CrossRef] [Green Version]
  238. Muñoz-Galván, S.; Felipe-Abrio, B.; Verdugo-Sivianes, E.M.; Perez, M.; Jiménez-García, M.P.; Suarez-Martinez, E.; Estevez-Garcia, P.; Carnero, A. Downregulation of MYPT1 increases tumor resistance in ovarian cancer by targeting the Hippo pathway and increasing the stemness. Mol. Cancer 2020, 19, 7. [Google Scholar] [CrossRef] [Green Version]
  239. Quinn, H.M.; Vogel, R.; Popp, O.; Mertins, P.; Lan, L.; Messerschmidt, C.; Landshammer, A.; Lisek, K.; Château-Joubert, S.; Marangoni, E.; et al. YAP and β-Catenin Cooperate to Drive Oncogenesis in Basal Breast Cancer. Cancer Res. 2021, 81, 2116–2127. [Google Scholar] [CrossRef] [PubMed]
  240. Xia, Y.; Zhang, Y.-L.; Yu, C.; Chang, T.; Fan, H.-Y. YAP/TEAD co-activator regulated pluripotency and chemoresistance in ovarian cancer initiated cells. PLoS ONE 2014, 9, e109575. [Google Scholar] [CrossRef]
  241. Park, J.H.; Shin, J.E.; Park, H.W. The Role of Hippo Pathway in Cancer Stem Cell Biology. Mol. Cells 2018, 41, 83–92. [Google Scholar] [CrossRef]
  242. Tian, Q.; Gao, H.; Zhou, Y.; Zhu, L.; Yang, J.; Wang, B.; Liu, P.; Yang, J. RICH1 inhibits breast cancer stem cell traits through activating kinases cascade of Hippo signaling by competing with Merlin for binding to Amot-p80. Cell Death Dis. 2022, 13, 71. [Google Scholar] [CrossRef] [PubMed]
  243. Li, Z.; Feng, J.; Gou, J.; Jia, J.; Yi, T.; Cui, T. Verteporfin, a suppressor of YAP–TEAD complex, presents promising antitumor properties on ovarian cancer. Onco. Targets. Ther. 2016, 9, 5371–5381. [Google Scholar] [CrossRef] [Green Version]
  244. Jaganjac, M.; Milkovic, L.; Sunjic, S.B.; Zarkovic, N. The NRF2, Thioredoxin, and Glutathione System in Tumorigenesis and Anticancer Therapies. Antioxidants 2020, 9, 1151. [Google Scholar] [CrossRef] [PubMed]
  245. Croker, A.K.; Allan, A.L. Inhibition of aldehyde dehydrogenase (ALDH) activity reduces chemotherapy and radiation resistance of stem-like ALDHhiCD44+ human breast cancer cells. Breast Cancer Res. Treat. 2012, 133, 75–87. [Google Scholar] [CrossRef]
  246. Ayub, T.H.; Keyver-Paik, M.-D.; Debald, M.; Rostamzadeh, B.; Thiesler, T.; Schröder, L.; Barchet, W.; Abramian, A.; Kaiser, C.; Kristiansen, G.; et al. Accumulation of ALDH1-positive cells after neoadjuvant chemotherapy predicts treatment resistance and prognosticates poor outcome in ovarian cancer. Oncotarget 2015, 6, 16437–16448. [Google Scholar] [CrossRef] [Green Version]
  247. Ryoo, I.-G.; Choi, B.-H.; Kwak, M.-K. Activation of NRF2 by p62 and proteasome reduction in sphere-forming breast carcinoma cells. Oncotarget 2015, 6, 8167–8184. [Google Scholar] [CrossRef] [Green Version]
  248. Kim, D.; Choi, B.-H.; Ryoo, I.-G.; Kwak, M.-K. High NRF2 level mediates cancer stem cell-like properties of aldehyde dehydrogenase (ALDH)-high ovarian cancer cells: Inhibitory role of all-trans retinoic acid in ALDH/NRF2 signaling. Cell Death Dis. 2018, 9, 896. [Google Scholar] [CrossRef] [Green Version]
  249. Ryoo, I.; Choi, B.; Ku, S.-K.; Kwak, M.-K. High CD44 expression mediates p62-associated NFE2L2/NRF2 activation in breast cancer stem cell-like cells: Implications for cancer stem cell resistance. Redox Biol. 2018, 17, 246–258. [Google Scholar] [CrossRef]
  250. Qin, S.; He, X.; Lin, H.; Schulte, B.A.; Zhao, M.; Tew, K.D.; Wang, G.Y. Nrf2 inhibition sensitizes breast cancer stem cells to ionizing radiation via suppressing DNA repair. Free Radic. Biol. Med. 2021, 169, 238–247. [Google Scholar] [CrossRef]
  251. Kamble, D.; Mahajan, M.; Dhat, R.; Sitasawad, S. Keap1-Nrf2 Pathway Regulates ALDH and Contributes to Radioresistance in Breast Cancer Stem Cells. Cells 2021, 10, 83. [Google Scholar] [CrossRef]
  252. Zhao, H.; Gao, Y.; Miao, J.; Chen, S.; Li, J.; Li, Z.; Yin, C.; Yue, W. Single-cell RNA-seq highlights a specific carcinoembryonic cluster in ovarian cancer. Cell Death Dis. 2021, 12, 1082. [Google Scholar] [CrossRef] [PubMed]
  253. Patel, A.V.; Patel, K.S.; Teras, L.R. Excess body fatness and cancer risk: A summary of the epidemiologic evidence. Surg. Obes. Relat. Dis. 2023, 19, 742–745. [Google Scholar] [CrossRef] [PubMed]
  254. Quail, D.F.; Dannenberg, A.J. The obese adipose tissue microenvironment in cancer development and progression. Nat. Rev. Endocrinol. 2019, 15, 139–154. [Google Scholar] [CrossRef] [PubMed]
  255. Massier, L.; Jalkanen, J.; Elmastas, M.; Zhong, J.; Wang, T.; Nono Nankam, P.A.; Frendo-Cumbo, S.; Bäckdahl, J.; Subramanian, N.; Sekine, T.; et al. An integrated single cell and spatial transcriptomic map of human white adipose tissue. Nat. Commun. 2023, 14, 1438. [Google Scholar] [CrossRef] [PubMed]
  256. Ham, S.; Choi, J.H.; Shin, S.G.; Lee, E.-J. High visceral fat-to-muscle ratio is an independent factor that predicts worse overall survival in patients with primary epithelial ovarian, fallopian tube, and peritoneal cancer. J. Ovarian Res. 2023, 16, 19. [Google Scholar] [CrossRef] [PubMed]
  257. Parafiniuk, K.; Skiba, W.; Pawłowska, A.; Suszczyk, D.; Maciejczyk, A.; Wertel, I. The Role of the Adipokine Resistin in the Pathogenesis and Progression of Epithelial Ovarian Cancer. Biomedicines 2022, 10, 920. [Google Scholar] [CrossRef]
  258. Brock, C.K.; Hebert, K.L.; Artiles, M.; Wright, M.K.; Cheng, T.; Windsor, G.O.; Nguyen, K.; Alzoubi, M.S.; Collins-Burow, B.M.; Martin, E.C.; et al. A Role for Adipocytes and Adipose Stem Cells in the Breast Tumor Microenvironment and Regenerative Medicine. Front. Physiol. 2021, 12, 751239. [Google Scholar] [CrossRef]
  259. Mukherjee, A.; Chiang, C.-Y.; Daifotis, H.A.; Nieman, K.M.; Fahrmann, J.F.; Lastra, R.R.; Romero, I.L.; Fiehn, O.; Lengyel, E. Adipocyte-Induced FABP4 Expression in Ovarian Cancer Cells Promotes Metastasis and Mediates Carboplatin Resistance. Cancer Res. 2020, 80, 1748–1761. [Google Scholar] [CrossRef] [Green Version]
  260. Nieman, K.M.; Kenny, H.A.; Penicka, C.V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M.R.; Romero, I.L.; Carey, M.S.; Mills, G.B.; Hotamisligil, G.S.; et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 2011, 17, 1498–1503. [Google Scholar] [CrossRef] [Green Version]
  261. Salimian Rizi, B.; Caneba, C.; Nowicka, A.; Nabiyar, A.W.; Liu, X.; Chen, K.; Klopp, A.; Nagrath, D. Nitric Oxide Mediates Metabolic Coupling of Omentum-Derived Adipose Stroma to Ovarian and Endometrial Cancer Cells. Cancer Res. 2015, 75, 456–471. [Google Scholar] [CrossRef] [Green Version]
  262. Bochet, L.; Lehuédé, C.; Dauvillier, S.; Wang, Y.Y.; Dirat, B.; Laurent, V.; Dray, C.; Guiet, R.; Maridonneau-Parini, I.; Le Gonidec, S.; et al. Adipocyte-Derived Fibroblasts Promote Tumor Progression and Contribute to the Desmoplastic Reaction in Breast Cancer. Cancer Res. 2013, 73, 5657–5668. [Google Scholar] [CrossRef] [Green Version]
  263. Ritter, A.; Kreis, N.-N.; Roth, S.; Friemel, A.; Safdar, B.K.; Hoock, S.C.; Wildner, J.M.; Allert, R.; Louwen, F.; Solbach, C.; et al. Cancer-educated mammary adipose tissue-derived stromal/stem cells in obesity and breast cancer: Spatial regulation and function. J. Exp. Clin. Cancer Res. 2023, 42, 35. [Google Scholar] [CrossRef] [PubMed]
  264. Ritter, A.; Kreis, N.-N.; Hoock, S.C.; Solbach, C.; Louwen, F.; Yuan, J. Adipose Tissue-Derived Mesenchymal Stromal/Stem Cells, Obesity and the Tumor Microenvironment of Breast Cancer. Cancers 2022, 14, 3908. [Google Scholar] [CrossRef] [PubMed]
  265. Iyoshi, S.; Yoshihara, M.; Nakamura, K.; Sugiyama, M.; Koya, Y.; Kitami, K.; Uno, K.; Mogi, K.; Tano, S.; Tomita, H.; et al. Pro-tumoral behavior of omental adipocyte-derived fibroblasts in tumor microenvironment at the metastatic site of ovarian cancer. Int. J. Cancer 2021, 149, 1961–1972. [Google Scholar] [CrossRef]
  266. Ladanyi, A.; Mukherjee, A.; Kenny, H.A.; Johnson, A.; Mitra, A.K.; Sundaresan, S.; Nieman, K.M.; Pascual, G.; Benitah, S.A.; Montag, A.; et al. Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene 2018, 37, 2285–2301. [Google Scholar] [CrossRef] [PubMed]
  267. John, B.; Naczki, C.; Patel, C.; Ghoneum, A.; Qasem, S.; Salih, Z.; Said, N. Regulation of the bi-directional cross-talk between ovarian cancer cells and adipocytes by SPARC. Oncogene 2019, 38, 4366–4383. [Google Scholar] [CrossRef] [Green Version]
  268. Liu, X.; Zhao, G.; Huo, X.; Wang, Y.; Tigyi, G.; Zhu, B.-M.; Yue, J.; Zhang, W. Adipose-Derived Stem Cells Facilitate Ovarian Tumor Growth and Metastasis by Promoting Epithelial to Mesenchymal Transition Through Activating the TGF-β Pathway. Front. Oncol. 2021, 11, 756011. [Google Scholar] [CrossRef]
  269. Nowicka, A.; Marini, F.C.; Solley, T.N.; Elizondo, P.B.; Zhang, Y.; Sharp, H.J.; Broaddus, R.; Kolonin, M.; Mok, S.C.; Thompson, M.S.; et al. Human omental-derived adipose stem cells increase ovarian cancer proliferation, migration, and chemoresistance. PLoS ONE 2013, 8, e81859. [Google Scholar] [CrossRef]
  270. Wei, H.-J.; Zeng, R.; Lu, J.-H.; Lai, W.-F.T.; Chen, W.-H.; Liu, H.-Y.; Chang, Y.-T.; Deng, W.-P. Adipose-derived stem cells promote tumor initiation and accelerate tumor growth by interleukin-6 production. Oncotarget 2015, 6, 7713–7726. [Google Scholar] [CrossRef] [Green Version]
  271. Cho, J.A.; Park, H.; Lim, E.H.; Kim, K.H.; Choi, J.S.; Lee, J.H.; Shin, J.W.; Lee, K.W. Exosomes from ovarian cancer cells induce adipose tissue-derived mesenchymal stem cells to acquire the physical and functional characteristics of tumor-supporting myofibroblasts. Gynecol. Oncol. 2011, 123, 379–386. [Google Scholar] [CrossRef]
  272. Cho, J.A.; Park, H.; Lim, E.H.; Lee, K.W. Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Int. J. Oncol. 2012, 40, 130–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Divella, R.; De Luca, R.; Abbate, I.; Naglieri, E.; Daniele, A. Obesity and cancer: The role of adipose tissue and adipo-cytokines-induced chronic inflammation. J. Cancer 2016, 7, 2346–2359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Rausch, L.K.; Netzer, N.C.; Hoegel, J.; Pramsohler, S. The Linkage between Breast Cancer, Hypoxia, and Adipose Tissue. Front. Oncol. 2017, 7, 211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Bachelot, T.; Ray-Coquard, I.; Menetrier-Caux, C.; Rastkha, M.; Duc, A.; Blay, J.-Y. Prognostic value of serum levels of interleukin 6 and of serum and plasma levels of vascular endothelial growth factor in hormone-refractory metastatic breast cancer patients. Br. J. Cancer 2003, 88, 1721–1726. [Google Scholar] [CrossRef] [Green Version]
  276. Marotta, L.L.C.; Almendro, V.; Marusyk, A.; Shipitsin, M.; Schemme, J.; Walker, S.R.; Bloushtain-Qimron, N.; Kim, J.J.; Choudhury, S.A.; Maruyama, R.; et al. The JAK2/STAT3 signaling pathway is required for growth of CD44+CD24– stem cell–like breast cancer cells in human tumors. J. Clin. Investig. 2011, 121, 2723–2735. [Google Scholar] [CrossRef]
  277. Liu, S.; Lee, J.S.; Jie, C.; Park, M.H.; Iwakura, Y.; Patel, Y.; Soni, M.; Reisman, D.; Chen, H. HER2 Overexpression Triggers an IL1α Proinflammatory Circuit to Drive Tumorigenesis and Promote Chemotherapy Resistance. Cancer Res. 2018, 78, 2040–2051. [Google Scholar] [CrossRef] [Green Version]
  278. Browning, L.; Patel, M.; Bring Horvath, E.; Tawara, K.; Jorcyk, C.L. IL-6 and ovarian cancer: Inflammatory cytokines in promotion of metastasis. Cancer Manag. Res. 2018, 10, 6685–6693. [Google Scholar] [CrossRef] [Green Version]
  279. Sansone, P.; Storci, G.; Tavolari, S.; Guarnieri, T.; Giovannini, C.; Taffurelli, M.; Ceccarelli, C.; Santini, D.; Paterini, P.; Marcu, K.B.; et al. IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. J. Clin. Investig. 2007, 117, 3988–4002. [Google Scholar] [CrossRef]
  280. Zhao, P.; Sun, J.; Huang, X.; Zhang, X.; Liu, X.; Liu, R.; Du, G.; Gan, W.; Yang, C.; Tang, Y.; et al. Targeting the KLF5-EphA2 axis can restrain cancer stemness and overcome chemoresistance in basal-like breast cancer. Int. J. Biol. Sci. 2023, 19, 1861–1874. [Google Scholar] [CrossRef]
  281. Nickel, A.; Blücher, C.; Al Kadri, O.; Schwagarus, N.; Müller, S.; Schaab, M.; Thiery, J.; Burkhardt, R.; Stadler, S.C. Adipocytes induce distinct gene expression profiles in mammary tumor cells and enhance inflammatory signaling in invasive breast cancer cells. Sci. Rep. 2018, 8, 9482. [Google Scholar] [CrossRef] [Green Version]
  282. Muthukumaran, N.; Miletti-González, K.E.; Ravindranath, A.K.; Rodríguez-Rodríguez, L. Tumor Necrosis Factor-α Differentially Modulates CD44 Expression in Ovarian Cancer Cells. Mol. Cancer Res. 2006, 4, 511–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Kulbe, H.; Thompson, R.; Wilson, J.L.; Robinson, S.; Hagemann, T.; Fatah, R.; Gould, D.; Ayhan, A.; Balkwill, F. The Inflammatory Cytokine Tumor Necrosis Factor-α Generates an Autocrine Tumor-Promoting Network in Epithelial Ovarian Cancer Cells. Cancer Res. 2007, 67, 585–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Rickard, B.P.; Conrad, C.; Sorrin, A.J.; Ruhi, M.K.; Reader, J.C.; Huang, S.A.; Franco, W.; Scarcelli, G.; Polacheck, W.J.; Roque, D.M.; et al. Malignant Ascites in Ovarian Cancer: Cellular, Acellular, and Biophysical Determinants of Molecular Characteristics and Therapy Response. Cancers 2021, 13, 4318. [Google Scholar] [CrossRef] [PubMed]
  285. Salemme, V.; Centonze, G.; Cavallo, F.; Defilippi, P.; Conti, L. The Crosstalk Between Tumor Cells and the Immune Microenvironment in Breast Cancer: Implications for Immunotherapy. Front. Oncol. 2021, 11, 610303. [Google Scholar] [CrossRef] [PubMed]
  286. Li, J.J.; Tsang, J.Y.; Tse, G.M. Tumor Microenvironment in Breast Cancer-Updates on Therapeutic Implications and Pathologic Assessment. Cancers 2021, 13, 4233. [Google Scholar] [CrossRef] [PubMed]
  287. Feng, W.; Dean, D.C.; Hornicek, F.J.; Shi, H.; Duan, Z. Exosomes promote pre-metastatic niche formation in ovarian cancer. Mol. Cancer 2019, 18, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. González-Callejo, P.; Gener, P.; Díaz-Riascos, Z.V.; Conti, S.; Cámara-Sánchez, P.; Riera, R.; Mancilla, S.; García-Gabilondo, M.; Peg, V.; Arango, D.; et al. Extracellular vesicles secreted by triple-negative breast cancer stem cells trigger premetastatic niche remodeling and metastatic growth in the lungs. Int. J. Cancer 2023, 152, 2153–2165. [Google Scholar] [CrossRef]
  289. Kogure, A.; Yoshioka, Y.; Ochiya, T. Extracellular vesicles in cancer metastasis: Potential as therapeutic targets and materials. Int. J. Mol. Sci. 2020, 21, 4463. [Google Scholar] [CrossRef]
  290. Bobrie, A.; Krumeich, S.; Reyal, F.; Recchi, C.; Moita, L.F.; Seabra, M.C.; Ostrowski, M.; Théry, C. Rab27a Supports Exosome-Dependent and -Independent Mechanisms That Modify the Tumor Microenvironment and Can Promote Tumor Progression. Cancer Res. 2012, 72, 4920–4930. [Google Scholar] [CrossRef] [Green Version]
  291. Fico, F.; Santamaria-Martínez, A. The Tumor Microenvironment as a Driving Force of Breast Cancer Stem Cell Plasticity. Cancers 2020, 12, 3863. [Google Scholar] [CrossRef]
  292. Radharani, N.N.V.; Yadav, A.S.; Nimma, R.; Kumar, T.V.S.; Bulbule, A.; Chanukuppa, V.; Kumar, D.; Patnaik, S.; Rapole, S.; Kundu, G.C. Tumor-associated macrophage derived IL-6 enriches cancer stem cell population and promotes breast tumor progression via Stat-3 pathway. Cancer Cell Int. 2022, 22, 122. [Google Scholar] [CrossRef]
  293. El-Arabey, A.A.; Alkhalil, S.S.; Al-Shouli, S.T.; Awadalla, M.E.; Alhamdi, H.W.; Almanaa, T.N.; Mohamed, S.S.E.M.; Abdalla, M. Revisiting macrophages in ovarian cancer microenvironment: Development, function and interaction. Med. Oncol. 2023, 40, 142. [Google Scholar] [CrossRef] [PubMed]
  294. Ali, H.R.; Provenzano, E.; Dawson, S.J.; Blows, F.M.; Liu, B.; Shah, M.; Earl, H.M.; Poole, C.J.; Hiller, L.; Dunn, J.A.; et al. Association between CD8+ T-cell infiltration and breast cancer survival in 12 439 patients. Ann. Oncol. 2014, 25, 1536–1543. [Google Scholar] [CrossRef] [PubMed]
  295. Shipitsin, M.; Campbell, L.L.; Argani, P.; Weremowicz, S.; Bloushtain-Qimron, N.; Yao, J.; Nikolskaya, T.; Serebryiskaya, T.; Beroukhim, R.; Hu, M.; et al. Molecular Definition of Breast Tumor Heterogeneity. Cancer Cell 2007, 11, 259–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Wilczyński, J.R.; Wilczyński, M.; Paradowska, E. Cancer Stem Cells in Ovarian Cancer—A Source of Tumor Success and a Challenging Target for Novel Therapies. Int. J. Mol. Sci. 2022, 23, 2496. [Google Scholar] [CrossRef] [PubMed]
  297. Wu, Y.; Chen, M.; Wu, P.; Chen, C.; Xu, Z.P.; Gu, W. Increased PD-L1 expression in breast and colon cancer stem cells. Clin. Exp. Pharmacol. Physiol. 2017, 44, 602–604. [Google Scholar] [CrossRef]
  298. Matsuzaki, J.; Gnjatic, S.; Mhawech-Fauceglia, P.; Beck, A.; Miller, A.; Tsuji, T.; Eppolito, C.; Qian, F.; Lele, S.; Shrikant, P.; et al. Tumor-infiltrating NY-ESO-1–specific CD8 + T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 7875–7880. [Google Scholar] [CrossRef] [Green Version]
  299. Castagnoli, L.; Cancila, V.; Cordoba-Romero, S.L.; Faraci, S.; Talarico, G.; Belmonte, B.; Iorio, M.V.; Milani, M.; Volpari, T.; Chiodoni, C.; et al. WNT signaling modulates PD-L1 expression in the stem cell compartment of triple-negative breast cancer. Oncogene 2019, 38, 4047–4060. [Google Scholar] [CrossRef] [Green Version]
  300. Stein, R.G.; Ebert, S.; Schlahsa, L.; Scholz, C.J.; Braun, M.; Hauck, P.; Horn, E.; Monoranu, C.M.; Thiemann, V.J.; Wustrow, M.P.; et al. Cognate nonlytic interactions between CD8+ T cells and breast cancer cells induce cancer stem cell-like properties. Cancer Res. 2019, 79, 1507–1519. [Google Scholar] [CrossRef] [Green Version]
  301. Sato, E.; Olson, S.H.; Ahn, J.; Bundy, B.; Nishikawa, H.; Qian, F.; Jungbluth, A.A.; Frosina, D.; Gnjatic, S.; Ambrosone, C.; et al. Intraepithelial CD8 + tumor-infiltrating lymphocytes and a high CD8 +/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl. Acad. Sci. USA 2005, 102, 18538–18543. [Google Scholar] [CrossRef] [Green Version]
  302. Xu, Y.; Dong, X.; Qi, P.; Ye, Y.; Shen, W.; Leng, L.; Wang, L.; Li, X.; Luo, X.; Chen, Y.; et al. Sox2 communicates with Tregs through CCL1 to promote the stemness property of breast cancer cells. Stem Cells 2017, 35, 2351. [Google Scholar] [CrossRef] [Green Version]
  303. Loh, J.J.; Ma, S. The Role of Cancer-Associated Fibroblast as a Dynamic Player in Mediating Cancer Stemness in the Tumor Microenvironment. Front. Cell Dev. Biol. 2021, 9, 727640. [Google Scholar] [CrossRef] [PubMed]
  304. Valenti, G.; Quinn, H.M.; Heynen, G.J.J.E.; Lan, L.; Holland, J.D.; Vogel, R.; Wulf-Goldenberg, A.; Birchmeier, W. Cancer stem cells regulate cancer-associated fibroblasts via activation of hedgehog signaling in mammary gland tumors. Cancer Res. 2017, 77, 2134–2147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Elwakeel, E.; Weigert, A. Breast Cancer CAFs: Spectrum of Phenotypes and Promising Targeting Avenues. Int. J. Mol. Sci. 2021, 22, 11636. [Google Scholar] [CrossRef] [PubMed]
  306. Ji, Z.; Tian, W.; Gao, W.; Zang, R.; Wang, H.; Yang, G. Cancer-Associated Fibroblast-Derived Interleukin-8 Promotes Ovarian Cancer Cell Stemness and Malignancy Through the Notch3-Mediated Signaling. Front. Cell Dev. Biol. 2021, 9, 684505. [Google Scholar] [CrossRef]
  307. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef]
  308. Fang, Y.; El Aziz, M.A.A.; Tiwari, K.; Mitra, A.K. Abstract 4685: Cancer associated fibroblasts promote ovarian cancer chemoresistance by inducing cancer stem cells through Wnt signaling. Cancer Res. 2019, 79, 4685. [Google Scholar] [CrossRef]
  309. Sommerfeld, L.; Finkernagel, F.; Jansen, J.M.; Wagner, U.; Nist, A.; Stiewe, T.; Müller-Brüsselbach, S.; Sokol, A.M.; Graumann, J.; Reinartz, S.; et al. The multicellular signalling network of ovarian cancer metastases. Clin. Transl. Med. 2021, 11, e633. [Google Scholar] [CrossRef]
  310. McLean, K.; Gong, Y.; Choi, Y.; Deng, N.; Yang, K.; Bai, S.; Cabrera, L.; Keller, E.; McCauley, L.; Cho, K.R.; et al. Human ovarian carcinoma–associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered {BMP} production. J. Clin. Investig. 2011, 121, 3206–3219. [Google Scholar] [CrossRef] [Green Version]
  311. Coffman, L.G.; Pearson, A.T.; Frisbie, L.G.; Freeman, Z.; Christie, E.; Bowtell, D.D.; Buckanovich, R.J. Ovarian Carcinoma-Associated Mesenchymal Stem Cells Arise from Tissue-Specific Normal Stroma. Stem Cells 2019, 37, 257–269. [Google Scholar] [CrossRef] [Green Version]
  312. Liang, D.; Ma, Y.; Liu, J.; Trope, C.G.; Holm, R.; Nesland, J.M.; Suo, Z. The hypoxic microenvironment upgrades stem-like properties of ovarian cancer cells. BMC Cancer 2012, 12, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  313. Kitajima, S.; Lee, K.L.; Hikasa, H.; Sun, W.; Huang, R.Y.-J.; Yang, H.; Matsunaga, S.; Yamaguchi, T.; Araki, M.; Kato, H.; et al. Hypoxia-inducible factor-1α promotes cell survival during ammonia stress response in ovarian cancer stem-like cells. Oncotarget 2017, 8, 114481–114494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Chen, J.; Imanaka, N.; Chen, J.; Griffin, J.D. Hypoxia potentiates Notch signaling in breast cancer leading to decreased E-cadherin expression and increased cell migration and invasion. Br. J. Cancer 2010, 102, 351–360. [Google Scholar] [CrossRef] [PubMed]
  315. Qin, J.; Liu, Y.; Lu, Y.; Liu, M.; Li, M.; Li, J.; Wu, L. Hypoxia-inducible factor 1 alpha promotes cancer stem cells-like properties in human ovarian cancer cells by upregulating SIRT1 expression. Sci. Rep. 2017, 7, 10592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  316. Jin, X.; Wei, Y.; Xu, F.; Zhao, M.; Dai, K.; Shen, R.; Yang, S.; Zhang, N. SIRT1 promotes formation of breast cancer through modulating Akt activity. J. Cancer 2018, 9, 2012–2023. [Google Scholar] [CrossRef] [Green Version]
  317. Shi, L.; Tang, X.; Qian, M.; Liu, Z.; Meng, F.; Fu, L.; Wang, Z.; Zhu, W.G.; Huang, J.D.; Zhou, Z.; et al. A SIRT1-centered circuitry regulates breast cancer stemness and metastasis. Oncogene 2018, 37, 6299–6315. [Google Scholar] [CrossRef] [Green Version]
  318. Lasek, W. Cancer immunoediting hypothesis: History, clinical implications and controversies. Cent. J. Immunol. 2022, 47, 168–174. [Google Scholar] [CrossRef]
  319. Ding, J.; Zhang, Y.; Che, Y. Ovarian cancer stem cells: Critical roles in anti-tumor immunity. Front. Genet. 2022, 13, 998220. [Google Scholar] [CrossRef]
  320. Gatti-Mays, M.E.; Balko, J.M.; Gameiro, S.R.; Bear, H.D.; Prabhakaran, S.; Fukui, J.; Disis, M.L.; Nanda, R.; Gulley, J.L.; Kalinsky, K.; et al. If we build it they will come: Targeting the immune response to breast cancer. NPJ Breast Cancer 2019, 5, 37. [Google Scholar] [CrossRef] [Green Version]
  321. Luo, H.; Xu, X.; Ye, M.; Sheng, B.; Zhu, X. The prognostic value of HER2 in ovarian cancer: A meta-analysis of observational studies. PLoS ONE 2018, 13, e0191972. [Google Scholar] [CrossRef] [Green Version]
  322. Iqbal, N.; Iqbal, N. Human Epidermal Growth Factor Receptor 2 (HER2) in Cancers: Overexpression and Therapeutic Implications. Mol. Biol. Int. 2014, 2014, 852748. [Google Scholar] [CrossRef] [PubMed]
  323. Peoples, G.E.; Goedegebuure, P.S.; Smith, R.; Linehan, D.C.; Yoshino, I.; Eberlein, T.J. Breast and ovarian cancer-specific cytotoxic T lymphocytes recognize the same HER2/neu-derived peptide. Proc. Natl. Acad. Sci. USA 1995, 92, 432–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  324. Koh, J.; Lee, S.; Park, H.; Lee, H.J.; Cho, N.H.; Kim, J. Susceptibility of CD24+ ovarian cancer cells to anti-cancer drugs and natural killer cells. Biochem. Biophys. Res. Commun. 2012, 427, 373–378. [Google Scholar] [CrossRef] [PubMed]
  325. Chovatiya, N.; Kaur, K.; Huerta-Yepez, S.; Chen, P.-C.; Neal, A.; DiBernardo, G.; Gumrukcu, S.; Memarzadeh, S.; Jewett, A. Inability of ovarian cancers to upregulate their MHC-class I surface expression marks their aggressiveness and increased susceptibility to NK cell-mediated cytotoxicity. Cancer Immunol. Immunother. 2022, 71, 2929–2941. [Google Scholar] [CrossRef]
  326. Uppendahl, L.D.; Dahl, C.M.; Miller, J.S.; Felices, M.; Geller, M.A. Natural Killer Cell-Based Immunotherapy in Gynecologic Malignancy: A Review. Front. Immunol. 2017, 8, 1825. [Google Scholar] [CrossRef] [Green Version]
  327. Dewan, M.Z.; Terunuma, H.; Takada, M.; Tanaka, Y.; Abe, H.; Sata, T.; Toi, M.; Yamamoto, N. Role of natural killer cells in hormone-independent rapid tumor formation and spontaneous metastasis of breast cancer cells in vivo. Breast Cancer Res. Treat. 2007, 104, 267–275. [Google Scholar] [CrossRef]
  328. Yin, T.; Wang, G.; He, S.; Liu, Q.; Sun, J.; Wang, Y. Human cancer cells with stem cell-like phenotype exhibit enhanced sensitivity to the cytotoxicity of IL-2 and IL-15 activated natural killer cells. Cell. Immunol. 2016, 300, 41–45. [Google Scholar] [CrossRef]
  329. Jin, H.; Kim, H.J. NK Cells Lose Their Cytotoxicity Function against Cancer Stem Cell-Rich Radiotherapy-Resistant Breast Cancer Cell Populations. Int. J. Mol. Sci. 2021, 22, 9639. [Google Scholar] [CrossRef]
  330. Wang, B.; Wang, Q.; Wang, Z.; Jiang, J.; Yu, S.-C.; Ping, Y.-F.; Yang, J.; Xu, S.-L.; Ye, X.-Z.; Xu, C.; et al. Metastatic Consequences of Immune Escape from NK Cell Cytotoxicity by Human Breast Cancer Stem Cells. Cancer Res. 2014, 74, 5746–5757. [Google Scholar] [CrossRef] [Green Version]
  331. Geller, M.A.; Cooley, S.; Judson, P.L.; Ghebre, R.; Carson, L.F.; Argenta, P.A.; Jonson, A.L.; Panoskaltsis-Mortari, A.; Curtsinger, J.; McKenna, D.; et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy 2011, 13, 98–107. [Google Scholar] [CrossRef] [Green Version]
  332. Workel, H.H.; Lubbers, J.M.; Arnold, R.; Prins, T.M.; van der Vlies, P.; de Lange, K.; Bosse, T.; van Gool, I.C.; Eggink, F.A.; Wouters, M.C.A.; et al. A Transcriptionally Distinct CXCL13+CD103+CD8+ T-cell Population Is Associated with B-cell Recruitment and Neoantigen Load in Human Cancer. Cancer Immunol. Res. 2019, 7, 784–796. [Google Scholar] [CrossRef] [Green Version]
  333. Garg, A.D.; De Ruysscher, D.; Agostinis, P. Immunological metagene signatures derived from immunogenic cancer cell death associate with improved survival of patients with lung, breast or ovarian malignancies: A large-scale meta-analysis. Oncoimmunology 2016, 5, e1069938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Alwosaibai, K.; Aalmri, S.; Mashhour, M.; Ghandorah, S.; Alshangiti, A.; Azam, F.; Selwi, W.; Gharaibeh, L.; Alatawi, Y.; Alruwaii, Z.; et al. PD-L1 is highly expressed in ovarian cancer and associated with cancer stem cells populations expressing CD44 and other stem cell markers. BMC Cancer 2023, 23, 13. [Google Scholar] [CrossRef]
  335. Mansour, F.A.; Al-Mazrou, A.; Al-Mohanna, F.; Al-Alwan, M.; Ghebeh, H. PD-L1 is overexpressed on breast cancer stem cells through notch3/mTOR axis. Oncoimmunology 2020, 9, 1729299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  336. Darvin, P.; Sasidharan Nair, V.; Elkord, E. PD-L1 Expression in Human Breast Cancer Stem Cells Is Epigenetically Regulated through Posttranslational Histone Modifications. J. Oncol. 2019, 2019, 3958908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  337. Sobral-Leite, M.; Van de Vijver, K.; Michaut, M.; van der Linden, R.; Hooijer, G.K.J.; Horlings, H.M.; Severson, T.M.; Mulligan, A.M.; Weerasooriya, N.; Sanders, J.; et al. Assessment of PD-L1 expression across breast cancer molecular subtypes, in relation to mutation rate, BRCA1-like status, tumor-infiltrating immune cells and survival. Oncoimmunology 2018, 7, e1509820. [Google Scholar] [CrossRef] [Green Version]
  338. Pawłowska, A.; Kwiatkowska, A.; Suszczyk, D.; Chudzik, A.; Tarkowski, R.; Barczyński, B.; Kotarski, J.; Wertel, I. Clinical and Prognostic Value of Antigen-Presenting Cells with PD-L1/PD-L2 Expression in Ovarian Cancer Patients. Int. J. Mol. Sci. 2021, 22, 11563. [Google Scholar] [CrossRef]
  339. Wang, L. Prognostic effect of programmed death-ligand 1 (PD-L1) in ovarian cancer: A systematic review, meta-analysis and bioinformatics study. J. Ovarian Res. 2019, 12, 37. [Google Scholar] [CrossRef] [Green Version]
  340. Li, X.; Li, M.; Lian, Z.; Zhu, H.; Kong, L.; Wang, P.; Yu, J. Prognostic Role of Programmed Death Ligand-1 Expression in Breast Cancer: A Systematic Review and Meta-Analysis. Target. Oncol. 2016, 11, 753–761. [Google Scholar] [CrossRef]
  341. Jang, Y.S.; Kim, T.W.; Ryu, J.S.; Kong, H.J.; Jang, S.H.; Nam, G.H.; Kim, J.H.; Jeon, S. Upregulation of programmed death ligand-1 in tumor-associated macrophages affects chemotherapeutic response in ovarian cancer cells. PLoS ONE 2023, 18, e0277285. [Google Scholar] [CrossRef]
  342. Barkal, A.A.; Brewer, R.E.; Markovic, M.; Kowarsky, M.; Barkal, S.A.; Zaro, B.W.; Krishnan, V.; Hatakeyama, J.; Dorigo, O.; Barkal, L.J.; et al. CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature 2019, 572, 392–396. [Google Scholar] [CrossRef] [PubMed]
  343. Rizzo, S.; Hersey, J.M.; Mellor, P.; Dai, W.; Santos-Silva, A.; Liber, D.; Luk, L.; Titley, I.; Carden, C.P.; Box, G.; et al. Ovarian Cancer Stem Cell–Like Side Populations Are Enriched Following Chemotherapy and Overexpress EZH2. Mol. Cancer Ther. 2011, 10, 325–335. [Google Scholar] [CrossRef] [Green Version]
  344. Steg, A.D.; Bevis, K.S.; Katre, A.A.; Ziebarth, A.; Dobbin, Z.C.; Alvarez, R.D.; Zhang, K.; Conner, M.; Landen, C.N. Stem cell pathways contribute to clinical chemoresistance in ovarian cancer. Clin. Cancer Res. 2012, 18, 869–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Montalbán Del Barrio, I.; Penski, C.; Schlahsa, L.; Stein, R.G.; Diessner, J.; Wöckel, A.; Dietl, J.; Lutz, M.B.; Mittelbronn, M.; Wischhusen, J.; et al. Adenosine-generating ovarian cancer cells attract myeloid cells which differentiate into adenosine-generating tumor associated macrophages—A self-amplifying, CD39- and CD73-dependent mechanism for tumor immune escape. J. Immunother. Cancer 2016, 4, 49. [Google Scholar] [CrossRef] [Green Version]
  346. Zhang, X.; Wang, J.; Liu, N.; Wu, W.; Li, H.; Chen, J.; Guo, X. Molecular mechanism of CD163+ tumor-associated macrophage (TAM)-derived exosome-induced cisplatin resistance in ovarian cancer ascites. Ann. Transl. Med. 2022, 10, 1014. [Google Scholar] [CrossRef] [PubMed]
  347. Allavena, P.; Digifico, E.; Belgiovine, C. Macrophages and cancer stem cells: A malevolent alliance. Mol. Med. 2021, 27, 121. [Google Scholar] [CrossRef]
  348. Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef] [PubMed]
  349. Harper, E.; Sheedy, E.; Stack, M. With Great Age Comes Great Metastatic Ability: Ovarian Cancer and the Appeal of the Aging Peritoneal Microenvironment. Cancers 2018, 10, 230. [Google Scholar] [CrossRef] [Green Version]
  350. Wefers, C.; Duiveman-de Boer, T.; Zusterzeel, P.; Massuger, L.; Fuchs, D.; Torensma, R.; Wheelock, C.; de Vries, I. Different Lipid Regulation in Ovarian Cancer: Inhibition of the Immune System. Int. J. Mol. Sci. 2018, 19, 273. [Google Scholar] [CrossRef] [Green Version]
  351. Holen, I.; Speirs, V.; Morrissey, B.; Blyth, K. In vivo models in breast cancer research: Progress, challenges and future directions. Dis. Model. Mech. 2017, 10, 359–371. [Google Scholar] [CrossRef] [Green Version]
  352. Sztankovics, D.; Moldvai, D.; Petővári, G.; Gelencsér, R.; Krencz, I.; Raffay, R.; Dankó, T.; Sebestyén, A. 3D bioprinting and the revolution in experimental cancer model systems-A review of developing new models and experiences with in vitro 3D bioprinted breast cancer tissue-mimetic structures. Pathol. Oncol. Res. 2023, 29, 1610996. [Google Scholar] [CrossRef] [PubMed]
  353. Huerta-Reyes, M.; Aguilar-Rojas, A. Three-dimensional models to study breast cancer (Review). Int. J. Oncol. 2021, 58, 331–343. [Google Scholar] [CrossRef] [PubMed]
  354. Fröhlich, E. The Variety of 3D Breast Cancer Models for the Study of Tumor Physiology and Drug Screening. Int. J. Mol. Sci. 2023, 24, 7116. [Google Scholar] [CrossRef] [PubMed]
  355. Salinas-Vera, Y.M.; Valdés, J.; Pérez-Navarro, Y.; Mandujano-Lazaro, G.; Marchat, L.A.; Ramos-Payán, R.; Nuñez-Olvera, S.I.; Pérez-Plascencia, C.; López-Camarillo, C. Three-Dimensional 3D Culture Models in Gynecological and Breast Cancer Research. Front. Oncol. 2022, 12, 826113. [Google Scholar] [CrossRef] [PubMed]
  356. Fisher, M.F.; Rao, S.S. Three-dimensional culture models to study drug resistance in breast cancer. Biotechnol. Bioeng. 2020, 117, 2262–2278. [Google Scholar] [CrossRef]
  357. Polonio-Alcalá, E.; Rabionet, M.; Ruiz-Martínez, S.; Ciurana, J.; Puig, T. Three-Dimensional Manufactured Supports for Breast Cancer Stem Cell Population Characterization. Curr. Drug Targets 2018, 20, 839–851. [Google Scholar] [CrossRef]
  358. Balachander, G.M.; Kotcherlakota, R.; Nayak, B.; Kedaria, D.; Rangarajan, A.; Chatterjee, K. 3D Tumor Models for Breast Cancer: Whither We Are and What We Need. ACS Biomater. Sci. Eng. 2021, 7, 3470–3486. [Google Scholar] [CrossRef]
  359. Murayama, T.; Gotoh, N. Patient-Derived Xenograft Models of Breast Cancer and Their Application. Cells 2019, 8, 621. [Google Scholar] [CrossRef] [Green Version]
  360. Xu, F.; Burg, K.J.L. Three-dimensional polymeric systems for cancer cell studies. Cytotechnology 2007, 54, 135–143. [Google Scholar] [CrossRef] [Green Version]
  361. Doublier, S.; Belisario, D.C.; Polimeni, M.; Annaratone, L.; Riganti, C.; Allia, E.; Ghigo, D.; Bosia, A.; Sapino, A. HIF-1 activation induces doxorubicin resistance in MCF7 3-D spheroids via P-glycoprotein expression: A potential model of the chemo-resistance of invasive micropapillary carcinoma of the breast. BMC Cancer 2012, 12, 4. [Google Scholar] [CrossRef] [Green Version]
  362. Dubois, C.; Dufour, R.; Daumar, P.; Aubel, C.; Szczepaniak, C.; Blavignac, C.; Mounetou, E.; Penault-Llorca, F.; Bamdad, M. Development and cytotoxic response of two proliferative MDA-MB-231 and non-proliferative SUM1315 three-dimensional cell culture models of triple-negative basal-like breast cancer cell lines. Oncotarget 2017, 8, 95316–95331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  363. Ham, S.L.; Thakuri, P.S.; Plaster, M.; Li, J.; Luker, K.E.; Luker, G.D.; Tavana, H. Three-dimensional tumor model mimics stromal—Breast cancer cells signaling. Oncotarget 2018, 9, 249–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  364. Brancato, V.; Gioiella, F.; Imparato, G.; Guarnieri, D.; Urciuolo, F.; Netti, P.A. 3D breast cancer microtissue reveals the role of tumor microenvironment on the transport and efficacy of free-doxorubicin in vitro. Acta Biomater. 2018, 75, 200–212. [Google Scholar] [CrossRef] [PubMed]
  365. Lee, S.Y.; Jeong, E.-K.; Jeon, H.M.; Kim, C.H.; Kang, H.S. Implication of necrosis-linked p53 aggregation in acquired apoptotic resistance to 5-FU in MCF-7 multicellular tumour spheroids. Oncol. Rep. 2010, 24, 73–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  366. Rashidi, M.R.W.; Mehta, P.; Bregenzer, M.; Raghavan, S.; Fleck, E.M.; Horst, E.N.; Harissa, Z.; Ravikumar, V.; Brady, S.; Bild, A.; et al. Engineered 3D Model of Cancer Stem Cell Enrichment and Chemoresistance. Neoplasia 2019, 21, 822–836. [Google Scholar] [CrossRef]
  367. Imamura, Y.; Mukohara, T.; Shimono, Y.; Funakoshi, Y.; Chayahara, N.; Toyoda, M.; Kiyota, N.; Takao, S.; Kono, S.; Nakatsura, T.; et al. Comparison of 2D- and 3D-culture models as drug-testing platforms in breast cancer. Oncol. Rep. 2015, 33, 1837–1843. [Google Scholar] [CrossRef] [Green Version]
  368. Lovitt, C.J.; Shelper, T.B.; Avery, V.M. Evaluation of chemotherapeutics in a three-dimensional breast cancer model. J. Cancer Res. Clin. Oncol. 2015, 141, 951–959. [Google Scholar] [CrossRef]
  369. Lovitt, C.J.; Shelper, T.B.; Avery, V.M. Doxorubicin resistance in breast cancer cells is mediated by extracellular matrix proteins. BMC Cancer 2018, 18, 41. [Google Scholar] [CrossRef] [Green Version]
  370. Breslin, S.; O’Driscoll, L.; Breslin, S.; O’Driscoll, L. The relevance of using 3D cell cultures, in addition to 2D monolayer cultures, when evaluating breast cancer drug sensitivity and resistance. Oncotarget 2016, 7, 45745–45756. [Google Scholar] [CrossRef] [Green Version]
  371. dit Faute, M.A.; Laurent, L.; Ploton, D.; Poupon, M.-F.; Jardillier, J.-C.; Bobichon, H. Distinctive alterations of invasiveness, drug resistance and cell-cell organization in 3D-cultures of MCF-7, a human breast cancer cell line, and its multidrug resistant variant. Clin. Exp. Metastasis 2002, 19, 161–167. [Google Scholar] [CrossRef]
  372. Gomes, L.R.; Rocha, C.R.R.; Martins, D.J.; Fiore, A.P.Z.P.; Kinker, G.S.; Bruni-Cardoso, A.; Menck, C.F.M. ATR mediates cisplatin resistance in 3D-cultured breast cancer cells via translesion DNA synthesis modulation. Cell Death Dis. 2019, 10, 459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  373. Gangadhara, S.; Smith, C.; Barrett-Lee, P.; Hiscox, S. 3D culture of Her2+ breast cancer cells promotes AKT to MAPK switching and a loss of therapeutic response. BMC Cancer 2016, 16, 345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  374. Hong, S.; Song, J.M. 3D bioprinted drug-resistant breast cancer spheroids for quantitative in situ evaluation of drug resistance. Acta Biomater. 2022, 138, 228–239. [Google Scholar] [CrossRef] [PubMed]
  375. Rezakhani, L.; Rahmati, S.; Ghasemi, S.; Alizadeh, M.; Alizadeh, A. A comparative study of the effects of crab derived exosomes and doxorubicin in 2 & 3-dimensional in vivo models of breast cancer. Chem. Phys. Lipids 2022, 243, 105179. [Google Scholar] [CrossRef]
  376. Wang, Y.; Shi, W.; Kuss, M.; Mirza, S.; Qi, D.; Krasnoslobodtsev, A.; Zeng, J.; Band, H.; Band, V.; Duan, B. 3D Bioprinting of Breast Cancer Models for Drug Resistance Study. ACS Biomater. Sci. Eng. 2018, 4, 4401–4411. [Google Scholar] [CrossRef]
  377. Horning, J.L.; Sahoo, S.K.; Vijayaraghavalu, S.; Dimitrijevic, S.; Vasir, J.K.; Jain, T.K.; Panda, A.K.; Labhasetwar, V. 3-D tumor model for in vitro evaluation of anticancer drugs. Mol. Pharm. 2008, 5, 849–862. [Google Scholar] [CrossRef]
  378. Gong, X.; Lin, C.; Cheng, J.; Su, J.; Zhao, H.; Liu, T.; Wen, X.; Zhao, P. Generation of Multicellular Tumor Spheroids with Microwell-Based Agarose Scaffolds for Drug Testing. PLoS ONE 2015, 10, e0130348. [Google Scholar] [CrossRef] [Green Version]
  379. Baker, A.E.G.; Tam, R.Y.; Shoichet, M.S. Independently Tuning the Biochemical and Mechanical Properties of 3D Hyaluronan-Based Hydrogels with Oxime and Diels–Alder Chemistry to Culture Breast Cancer Spheroids. Biomacromolecules 2017, 18, 4373–4384. [Google Scholar] [CrossRef]
  380. Arya, A.D.; Hallur, P.M.; Karkisaval, A.G.; Gudipati, A.; Rajendiran, S.; Dhavale, V.; Ramachandran, B.; Jayaprakash, A.; Gundiah, N.; Chaubey, A. Gelatin Methacrylate Hydrogels as Biomimetic Three-Dimensional Matrixes for Modeling Breast Cancer Invasion and Chemoresponse in Vitro. ACS Appl. Mater. Interfaces 2016, 8, 22005–22017. [Google Scholar] [CrossRef]
  381. Shin, K.; Klosterhoff, B.S.; Han, B. Characterization of Cell-Type-Specific Drug Transport and Resistance of Breast Cancers Using Tumor-Microenvironment-on-Chip. Mol. Pharm. 2016, 13, 2214–2223. [Google Scholar] [CrossRef] [Green Version]
  382. Ozcelikkale, A.; Shin, K.; Noe-Kim, V.; Elzey, B.D.; Dong, Z.; Zhang, J.T.; Kim, K.; Kwon, I.C.; Park, K.; Han, B. Differential response to doxorubicin in breast cancer subtypes simulated by a microfluidic tumor model. J. Control. Release 2017, 266, 129–139. [Google Scholar] [CrossRef] [PubMed]
  383. Wang, S.; Mao, S.; Li, M.; Li, H.F.; Lin, J.M. Near-physiological microenvironment simulation on chip to evaluate drug resistance of different loci in tumour mass. Talanta 2019, 191, 67–73. [Google Scholar] [CrossRef] [PubMed]
  384. Chen, L.; Xiao, Z.; Meng, Y.; Zhao, Y.; Han, J.; Su, G.; Chen, B.; Dai, J. The enhancement of cancer stem cell properties of MCF-7 cells in 3D collagen scaffolds for modeling of cancer and anti-cancer drugs. Biomaterials 2012, 33, 1437–1444. [Google Scholar] [CrossRef] [PubMed]
  385. Palomeras, S.; Rabionet, M.; Ferrer, I.; Sarrats, A.; Garcia-Romeu, M.; Puig, T.; Ciurana, J. Breast Cancer Stem Cell Culture and Enrichment Using Poly(ε-Caprolactone) Scaffolds. Molecules 2016, 21, 537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  386. Feng, S.; Duan, X.; Lo, P.-K.; Liu, S.; Liu, X.; Chen, H.; Wang, Q. Expansion of breast cancer stem cells with fibrous scaffolds. Integr. Biol. 2013, 5, 768–777. [Google Scholar] [CrossRef] [Green Version]
  387. Balachander, G.M.; Balaji, S.A.; Rangarajan, A.; Chatterjee, K. Enhanced Metastatic Potential in a 3D Tissue Scaffold toward a Comprehensive in Vitro Model for Breast Cancer Metastasis. ACS Appl. Mater. Interfaces 2015, 7, 27810–27822. [Google Scholar] [CrossRef]
  388. Sims-Mourtada, J.; Niamat, R.A.; Samuel, S.; Eskridge, C.; Kmiec, E.B. Enrichment of breast cancer stem-like cells by growth on electrospun polycaprolactone-chitosan nanofiber scaffolds. Int. J. Nanomed. 2014, 9, 995–1003. [Google Scholar] [CrossRef] [Green Version]
  389. Malacrida, B.; Pearce, O.M.T.; Balkwill, F.R. Building in vitro 3D human multicellular models of high-grade serous ovarian cancer. STAR Protoc. 2022, 3, 101086. [Google Scholar] [CrossRef]
  390. Brooks, E.A.; Gencoglu, M.F.; Corbett, D.C.; Stevens, K.R.; Peyton, S.R. An omentum-inspired 3D PEG hydrogel for identifying ECM-drivers of drug resistant ovarian cancer. APL Bioeng. 2019, 3, 026106. [Google Scholar] [CrossRef] [Green Version]
  391. Velletri, T.; Villa, C.E.; Cilli, D.; Barzaghi, B.; Lo Riso, P.; Lupia, M.; Luongo, R.; López-Tobón, A.; De Simone, M.; Bonnal, R.J.P.; et al. Single cell-derived spheroids capture the self-renewing subpopulations of metastatic ovarian cancer. Cell Death Differ. 2022, 29, 614–626. [Google Scholar] [CrossRef]
  392. Li, S.-S.; Ip, C.K.M.; Tang, M.Y.H.; Sy, S.K.H.; Yung, S.; Chan, T.-M.; Yang, M.; Shum, H.C.; Wong, A.S.T. Modeling Ovarian Cancer Multicellular Spheroid Behavior in a Dynamic 3D Peritoneal Microdevice. J. Vis. Exp. 2017, 120, e55337. [Google Scholar] [CrossRef] [Green Version]
  393. Liu, S.; Ginestier, C.; Ou, S.J.; Clouthier, S.G.; Patel, S.H.; Monville, F.; Korkaya, H.; Heath, A.; Dutcher, J.; Kleer, C.G.; et al. Breast cancer stem cells are regulated by mesenchymal stem cells through cytokine networks. Cancer Res. 2011, 71, 614–624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  394. Wang, Y.; Cardenas, H.; Fang, F.; Condello, S.; Taverna, P.; Segar, M.; Liu, Y.; Nephew, K.P.; Matei, D. Epigenetic targeting of ovarian cancer stem cells. Cancer Res. 2014, 74, 4922–4936. [Google Scholar] [CrossRef] [Green Version]
  395. Hu, Y.; Yagüe, E.; Zhao, J.; Wang, L.; Bai, J.; Yang, Q.; Pan, T.; Zhao, H.; Liu, J.; Zhang, J. Sabutoclax, pan-active BCL-2 protein family antagonist, overcomes drug resistance and eliminates cancer stem cells in breast cancer. Cancer Lett. 2018, 423, 47–59. [Google Scholar] [CrossRef]
  396. DeRose, Y.S.; Wang, G.; Lin, Y.-C.; Bernard, P.S.; Buys, S.S.; Ebbert, M.T.W.; Factor, R.; Matsen, C.; Milash, B.A.; Nelson, E.; et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat. Med. 2011, 17, 1514–1520. [Google Scholar] [CrossRef] [PubMed]
  397. Dobrolecki, L.E.; Airhart, S.D.; Alferez, D.G.; Aparicio, S.; Behbod, F.; Bentires-Alj, M.; Brisken, C.; Bult, C.J.; Cai, S.; Clarke, R.B.; et al. Patient-derived xenograft (PDX) models in basic and translational breast cancer research. Cancer Metastasis Rev. 2016, 35, 547–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  398. Liu, H.; Patel, M.R.; Prescher, J.A.; Patsialou, A.; Qian, D.; Lin, J.; Wen, S.; Chang, Y.-F.; Bachmann, M.H.; Shimono, Y.; et al. Cancer stem cells from human breast tumors are involved in spontaneous metastases in orthotopic mouse models. Proc. Natl. Acad. Sci. USA 2010, 107, 18115–18120. [Google Scholar] [CrossRef] [Green Version]
  399. Weroha, S.J.; Becker, M.A.; Enderica-Gonzalez, S.; Harrington, S.C.; Oberg, A.L.; Maurer, M.J.; Perkins, S.E.; AlHilli, M.; Butler, K.A.; McKinstry, S.; et al. Tumorgrafts as in vivo surrogates for women with ovarian cancer. Clin. Cancer Res. 2014, 20, 1288–1297. [Google Scholar] [CrossRef] [Green Version]
  400. Flesken-Nikitin, A.; Hwang, C.-I.; Cheng, C.-Y.; Michurina, T.V.; Enikolopov, G.; Nikitin, A.Y. Ovarian surface epithelium at the junction area contains cancer-prone stem cell niche. Nature 2013, 495, 241–245. [Google Scholar] [CrossRef] [Green Version]
  401. Landberg, G.; Fitzpatrick, P.; Isakson, P.; Jonasson, E.; Karlsson, J.; Larsson, E.; Svanström, A.; Rafnsdottir, S.; Persson, E.; Gustafsson, A.; et al. Patient-derived scaffolds uncover breast cancer promoting properties of the microenvironment. Biomaterials 2020, 235, 119705. [Google Scholar] [CrossRef]
  402. Leiva, M.C.; Garre, E.; Gustafsson, A.; Svanström, A.; Bogestål, Y.; Håkansson, J.; Ståhlberg, A.; Landberg, G. Breast cancer patient-derived scaffolds as a tool to monitor chemotherapy responses in human tumor microenvironments. J. Cell. Physiol. 2021, 236, 4709. [Google Scholar] [CrossRef] [PubMed]
  403. Gustafsson, A.; Garre, E.; Leiva, M.C.; Salerno, S.; Ståhlberg, A.; Landberg, G. Patient-derived scaffolds as a drug-testing platform for endocrine therapies in breast cancer. Sci. Rep. 2021, 11, 13334. [Google Scholar] [CrossRef] [PubMed]
  404. Garre, E.; Gustafsson, A.; Leiva, M.C.; Håkansson, J.; Ståhlberg, A.; Kovács, A.; Landberg, G. Breast Cancer Patient-Derived Scaffolds Can Expose Unique Individual Cancer Progressing Properties of the Cancer Microenvironment Associated with Clinical Characteristics. Cancers 2022, 14, 2172. [Google Scholar] [CrossRef]
  405. Omole, E.B.; Aijaz, I.; Ellegate, J.; Isenhart, E.; Desouki, M.M.; Mastri, M.; Humphrey, K.; Dougherty, E.M.; Rosario, S.R.; Nastiuk, K.L.; et al. Combined BRCA2 and MAGEC3 Expression Predict Outcome in Advanced Ovarian Cancers. Cancers 2022, 14, 4724. [Google Scholar] [CrossRef] [PubMed]
  406. Parte, S.C.; Batra, S.K.; Kakar, S.S. Characterization of stem cell and cancer stem cell populations in ovary and ovarian tumors. J. Ovarian Res. 2018, 11, 69. [Google Scholar] [CrossRef]
  407. Ali, H.R.; Dawson, S.-J.; Blows, F.M.; Provenzano, E.; Pharoah, P.D.; Caldas, C. Cancer stem cell markers in breast cancer: Pathological, clinical and prognostic significance. Breast Cancer Res. 2011, 13, R118. [Google Scholar] [CrossRef] [Green Version]
  408. Abraham, B.K.; Fritz, P.; McClellan, M.; Hauptvogel, P.; Athelogou, M.; Brauch, H. Prevalence of CD44+/CD24-/low cells in breast cancer may not be associated with clinical outcome but may favor distant metastasis. Clin. Cancer Res. 2005, 11, 1154–1159. [Google Scholar] [CrossRef]
  409. Cheng, L.; Ramesh, A.V.; Flesken-Nikitin, A.; Choi, J.; Nikitin, A.Y. Mouse models for cancer stem cell research. Toxicol. Pathol. 2010, 38, 62–71. [Google Scholar] [CrossRef]
  410. Abdullah, L.N.; Chow, E.K.-H. Mechanisms of chemoresistance in cancer stem cells. Clin. Transl. Med. 2013, 2, 3. [Google Scholar] [CrossRef] [Green Version]
  411. Zhang, S.; Cui, B.; Lai, H.; Liu, G.; Ghia, E.M.; Widhopf, G.F.; Zhang, Z.; Wu, C.C.N.; Chen, L.; Wu, R.; et al. Ovarian cancer stem cells express ROR1, which can be targeted for anti-cancer-stem-cell therapy. Proc. Natl. Acad. Sci. USA 2014, 111, 17266–17271. [Google Scholar] [CrossRef] [Green Version]
  412. Konrad, C.V.; Murali, R.; Varghese, B.A.; Nair, R. The role of cancer stem cells in tumor heterogeneity and resistance to therapy. Can. J. Physiol. Pharmacol. 2017, 95, 1–15. [Google Scholar] [CrossRef] [Green Version]
  413. Mao, X.-D.; Wei, X.; Xu, T.; Li, T.-P.; Liu, K.-S. Research progress in breast cancer stem cells: Characterization and future perspectives. Am. J. Cancer Res. 2022, 12, 3208–3222. [Google Scholar] [PubMed]
  414. Kim, W.-T.; Ryu, C.J. Cancer stem cell surface markers on normal stem cells. BMB Rep. 2017, 50, 285–298. [Google Scholar] [CrossRef] [Green Version]
  415. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target. Ther. 2020, 5, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  416. Palomeras, S.; Ruiz-Martínez, S.; Puig, T. Targeting Breast Cancer Stem Cells to Overcome Treatment Resistance. Molecules 2018, 23, 2193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  417. Zou, M.; Yin, X.; Zhou, X.; Niu, X.; Wang, Y.; Su, M. Salinomycin-Loaded High-Density Lipoprotein Exerts Promising Anti-Ovarian Cancer Effects by Inhibiting Epithelial–Mesenchymal Transition. Int. J. Nanomed. 2022, 17, 4059–4071. [Google Scholar] [CrossRef] [PubMed]
  418. Hirsch, H.A.; Iliopoulos, D.; Tsichlis, P.N.; Struhl, K. Metformin Selectively Targets Cancer Stem Cells, and Acts Together with Chemotherapy to Block Tumor Growth and Prolong Remission. Cancer Res. 2009, 69, 7507–7511. [Google Scholar] [CrossRef] [Green Version]
  419. Liu, Q.; Hodge, J.; Wang, J.; Wang, Y.; Wang, L.; Singh, U.P.; Li, Y.; Yao, Y.; Wang, D.; Ai, W.; et al. Emodin reduces Breast Cancer Lung Metastasis by suppressing Macrophage-induced Breast Cancer Cell Epithelial-mesenchymal transition and Cancer Stem Cell formation. Theranostics 2020, 10, 8365–8381. [Google Scholar] [CrossRef]
  420. Long, H.; Chen, H.; Yan, J.; Cheng, H. Emodin exerts antitumor effects in ovarian cancer cell lines by preventing the development of cancer stem cells via epithelial mesenchymal transition. Oncol. Lett. 2022, 23, 95. [Google Scholar] [CrossRef]
  421. Shan, N.L.; Wahler, J.; Lee, H.J.; Bak, M.J.; Gupta, S.D.; Maehr, H.; Suh, N. Vitamin D compounds inhibit cancer stem-like cells and induce differentiation in triple negative breast cancer. J. Steroid Biochem. Mol. Biol. 2017, 173, 122–129. [Google Scholar] [CrossRef]
  422. Ji, M.; Liu, L.; Hou, Y.; Li, B. 1α,25-Dihydroxyvitamin D3 restrains stem cell-like properties of ovarian cancer cells by enhancing vitamin D receptor and suppressing CD44. Oncol. Rep. 2019, 41, 3393–3403. [Google Scholar] [CrossRef] [PubMed]
  423. Tsai, K.-J.; Tsai, H.-Y.; Tsai, C.-C.; Chen, T.-Y.; Hsieh, T.-H.; Chen, C.-L.; Mbuyisa, L.; Huang, Y.-B.; Lin, M.-W. Luteolin Inhibits Breast Cancer Stemness and Enhances Chemosensitivity through the Nrf2-Mediated Pathway. Molecules 2021, 26, 6452. [Google Scholar] [CrossRef] [PubMed]
  424. Cao, D.; Zhu, G.-Y.; Lu, Y.; Yang, A.; Chen, D.; Huang, H.-J.; Peng, S.-X.; Chen, L.-W.; Li, Y.-W. Luteolin suppresses epithelial-mesenchymal transition and migration of triple-negative breast cancer cells by inhibiting YAP/TAZ activity. Biomed. Pharmacother. 2020, 129, 110462. [Google Scholar] [CrossRef]
  425. Li, Y.; Hu, Y.; Yang, L.; Liu, J.; Cui, C.; Yang, M.; Zou, D.; Zhou, L.; Zhou, Q.; Ge, W.; et al. Luteolin directly binds to KDM4C and attenuates ovarian cancer stemness via epigenetic suppression of PPP2CA/YAP axis. Biomed. Pharmacother. 2023, 160, 114350. [Google Scholar] [CrossRef] [PubMed]
  426. McClements, L.; Annett, S.; Yakkundi, A.; O’Rourke, M.; Valentine, A.; Moustafa, N.; Alqudah, A.; Simões, B.M.; Furlong, F.; Short, A.; et al. FKBPL and its peptide derivatives inhibit endocrine therapy resistant cancer stem cells and breast cancer metastasis by downregulating DLL4 and Notch4. BMC Cancer 2019, 19, 351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  427. Annett, S.; Moore, G.; Short, A.; Marshall, A.; McCrudden, C.; Yakkundi, A.; Das, S.; McCluggage, W.G.; Nelson, L.; Harley, I.; et al. FKBPL-based peptide, ALM201, targets angiogenesis and cancer stem cells in ovarian cancer. Br. J. Cancer 2020, 122, 361–371. [Google Scholar] [CrossRef]
  428. Lacerda, L.; Reddy, J.P.; Liu, D.; Larson, R.; Li, L.; Masuda, H.; Brewer, T.; Debeb, B.G.; Xu, W.; Hortobágyi, G.N.; et al. Simvastatin Radiosensitizes Differentiated and Stem-Like Breast Cancer Cell Lines and Is Associated with Improved Local Control in Inflammatory Breast Cancer Patients Treated with Postmastectomy Radiation. Stem Cells Transl. Med. 2014, 3, 849–856. [Google Scholar] [CrossRef]
  429. Ehmsen, S.; Pedersen, M.H.; Wang, G.; Terp, M.G.; Arslanagic, A.; Hood, B.L.; Conrads, T.P.; Leth-Larsen, R.; Ditzel, H.J. Increased Cholesterol Biosynthesis Is a Key Characteristic of Breast Cancer Stem Cells Influencing Patient Outcome. Cell Rep. 2019, 27, 3927–3938.e6. [Google Scholar] [CrossRef] [Green Version]
  430. Kato, S.; Liberona, M.F.; Cerda-Infante, J.; Sánchez, M.; Henríquez, J.; Bizama, C.; Bravo, M.L.; Gonzalez, P.; Gejman, R.; Brañes, J.; et al. Simvastatin interferes with cancer ‘stem-cell’ plasticity reducing metastasis in ovarian cancer. Endocr. Relat. Cancer 2018, 25, 821–836. [Google Scholar] [CrossRef]
  431. Yan, Y.; Li, Z.; Xu, X.; Chen, C.; Wei, W.; Fan, M.; Chen, X.; Li, J.J.; Wang, Y.; Huang, J. All-trans retinoic acids induce differentiation and sensitize a radioresistant breast cancer cells to chemotherapy. BMC Complement. Altern. Med. 2016, 16, 113. [Google Scholar] [CrossRef] [Green Version]
  432. Schech, A.; Kazi, A.; Yu, S.; Shah, P.; Sabnis, G. Histone Deacetylase Inhibitor Entinostat Inhibits Tumor-Initiating Cells in Triple-Negative Breast Cancer Cells. Mol. Cancer Ther. 2015, 14, 1848–1857. [Google Scholar] [CrossRef] [Green Version]
  433. Wang, D.; Li, W.; Zhao, R.; Chen, L.; Liu, N.; Tian, Y.; Zhao, H.; Xie, M.; Lu, F.; Fang, Q.; et al. Stabilized Peptide HDAC Inhibitors Derived from HDAC1 Substrate H3K56 for the Treatment of Cancer Stem–Like Cells In Vivo. Cancer Res. 2019, 79, 1769–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  434. Choudhury, P.; Barua, A.; Roy, A.; Pattanayak, R.; Bhattacharyya, M.; Saha, P. Eugenol restricts Cancer Stem Cell population by degradation of β-catenin via N-terminal Ser37 phosphorylation-an in vivo and in vitro experimental evaluation. Chem. Biol. Interact. 2020, 316, 108938. [Google Scholar] [CrossRef] [PubMed]
  435. Islam, S.S.; Aboussekhra, A. Sequential combination of cisplatin with eugenol targets ovarian cancer stem cells through the Notch-Hes1 signalling pathway. J. Exp. Clin. Cancer Res. 2019, 38, 382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  436. Wu, Z.H.; Lin, C.; Liu, M.M.; Zhang, J.; Tao, Z.H.; Hu, X.C. Src inhibition can synergize with gemcitabine and reverse resistance in triple negative breast cancer cells via the AKT/c-jun pathway. PLoS ONE 2016, 11, e0169230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  437. Simpkins, F.; Jang, K.; Yoon, H.; Hew, K.E.; Kim, M.; Azzam, D.J.; Sun, J.; Zhao, D.; Ince, T.A.; Liu, W.; et al. Dual Src and MEK Inhibition Decreases Ovarian Cancer Growth and Targets Tumor Initiating Stem-Like Cells. Clin. Cancer Res. 2018, 24, 4874–4886. [Google Scholar] [CrossRef] [Green Version]
  438. Chen, L.; Yang, G.; Dong, H. Everolimus Reverses Palbociclib Resistance in ER+ Human Breast Cancer Cells by Inhibiting Phosphatidylinositol 3-Kinase(PI3K)/Akt/Mammalian Target of Rapamycin (mTOR) Pathway. Med. Sci. Monit. 2019, 25, 77–86. [Google Scholar] [CrossRef]
  439. Phan, N.L.-C.; Van Trinh, N.; Pham, P. Van Low concentrations of 5-aza-2’-deoxycytidine induce breast cancer stem cell differentiation by triggering tumor suppressor gene expression. Onco. Targets. Ther. 2016, 9, 49–59. [Google Scholar] [CrossRef] [Green Version]
  440. Ho, C.-M.; Lee, F.-K.; Yen, T.-L.; Huang, S.-H.; Cheng, W.-F. Everolimus combined with 5-aza-2-deoxycytidine generated potent anti-tumor effects on ovarian clear cell cancer stem-like/spheroid cells by inhibiting the COL6A3-AKT-mTOR pathway. Am. J. Cancer Res. 2022, 12, 1686–1706. [Google Scholar]
  441. Liu, P.; Kumar, I.S.; Brown, S.; Kannappan, V.; Tawari, P.E.; Tang, J.Z.; Jiang, W.; Armesilla, A.L.; Darling, J.L.; Wang, W. Disulfiram targets cancer stem-like cells and reverses resistance and cross-resistance in acquired paclitaxel-resistant triple-negative breast cancer cells. Br. J. Cancer 2013, 109, 1876–1885. [Google Scholar] [CrossRef] [Green Version]
  442. Guo, F.; Yang, Z.; Sehouli, J.; Kaufmann, A.M. Blockade of ALDH in Cisplatin-Resistant Ovarian Cancer Stem Cells In Vitro Synergistically Enhances Chemotherapy-Induced Cell Death. Curr. Oncol. 2022, 29, 2808–2822. [Google Scholar] [CrossRef] [PubMed]
  443. Spizzo, G.; Fong, D.; Wurm, M.; Ensinger, C.; Obrist, P.; Hofer, C.; Mazzoleni, G.; Gastl, G.; Went, P. EpCAM expression in primary tumour tissues and metastases: An immunohistochemical analysis. J. Clin. Pathol. 2011, 64, 415–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  444. Heiss, M.M.; Murawa, P.; Koralewski, P.; Kutarska, E.; Kolesnik, O.O.; Ivanchenko, V.V.; Dudnichenko, A.S.; Aleknaviciene, B.; Razbadauskas, A.; Gore, M.; et al. The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: Results of a prospective randomized phase II/III trial. Int. J. Cancer 2010, 127, 2209–2221. [Google Scholar] [CrossRef] [Green Version]
  445. Kubo, M.; Umebayashi, M.; Kurata, K.; Mori, H.; Kai, M.; Onishi, H.; Katano, M.; Nakamura, M.; Morisaki, T. Catumaxomab with Activated T-cells Efficiently Lyses Chemoresistant EpCAM-positive Triple-negative Breast Cancer Cell Lines. Anticancer Res. 2018, 38, 4273–4279. [Google Scholar] [CrossRef]
  446. Skubitz, A.P.N.; Taras, E.P.; Boylan, K.L.M.; Waldron, N.N.; Oh, S.; Panoskaltsis-Mortari, A.; Vallera, D.A. Targeting CD133 in an in vivo ovarian cancer model reduces ovarian cancer progression. Gynecol. Oncol. 2013, 130, 579–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  447. Tume, L.; Paco, K.; Ubidia-Incio, R.; Moya, J. CD133 in breast cancer cells and in breast cancer stem cells as another target for immunotherapy. Gac. Mex. Oncol. 2016, 15, 22–30. [Google Scholar] [CrossRef] [Green Version]
  448. Vora, P.; Venugopal, C.; Salim, S.K.; Tatari, N.; Bakhshinyan, D.; Singh, M.; Seyfrid, M.; Upreti, D.; Rentas, S.; Wong, N.; et al. The Rational Development of CD133-Targeting Immunotherapies for Glioblastoma. Cell Stem Cell 2020, 26, 832–844.e6. [Google Scholar] [CrossRef]
  449. Ahmed, N.; Kadife, E.; Raza, A.; Short, M.; Jubinsky, P.T.; 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. [Google Scholar] [CrossRef] [Green Version]
  450. Bellio, C.; DiGloria, C.; Foster, R.; James, K.; Konstantinopoulos, P.A.; Growdon, W.B.; Rueda, B.R. PARP Inhibition Induces Enrichment of DNA Repair–Proficient CD133 and CD117 Positive Ovarian Cancer Stem Cells. Mol. Cancer Res. 2019, 17, 431–445. [Google Scholar] [CrossRef] [Green Version]
  451. Zeniou, M.; Nguekeu-Zebaze, L.; Dantzer, F. Therapeutic considerations of PARP in stem cell biology: Relevance in cancer and beyond. Biochem. Pharmacol. 2019, 167, 107–115. [Google Scholar] [CrossRef]
  452. Liu, Y.; Burness, M.L.; Martin-Trevino, R.; Guy, J.; Bai, S.; Harouaka, R.; Brooks, M.D.; Shang, L.; Fox, A.; Luther, T.K.; et al. RAD51 Mediates Resistance of Cancer Stem Cells to PARP Inhibition in Triple-Negative Breast Cancer. Clin. Cancer Res. 2017, 23, 514–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  453. McClements, L.; Yakkundi, A.; Papaspyropoulos, A.; Harrison, H.; Ablett, M.P.; Jithesh, P.V.; McKeen, H.D.; Bennett, R.; Donley, C.; Kissenpfennig, A.; et al. Targeting Treatment-Resistant Breast Cancer Stem Cells with FKBPL and Its Peptide Derivative, AD-01, via the CD44 Pathway. Clin. Cancer Res. 2013, 19, 3881–3893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  454. Chesnokov, M.S.; Khan, I.; Park, Y.; Ezell, J.; Mehta, G.; Yousif, A.; Hong, L.J.; Buckanovich, R.J.; Takahashi, A.; Chefetz, I. The MEK1/2 Pathway as a Therapeutic Target in High-Grade Serous Ovarian Carcinoma. Cancers 2021, 13, 1369. [Google Scholar] [CrossRef] [PubMed]
  455. Chakrabarty, A.; Bhola, N.E.; Sutton, C.; Ghosh, R.; Kuba, M.G.; Dave, B.; Chang, J.C.; Arteaga, C.L. Trastuzumab-resistant cells rely on a HER2-PI3K-FoxO-survivin axis and are sensitive to PI3K inhibitors. Cancer Res. 2013, 73, 1190–1200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  456. Shank, J.J.; Yang, K.; Ghannam, J.; Cabrera, L.; Johnston, C.J.; Reynolds, R.K.; Buckanovich, R.J. Metformin targets ovarian cancer stem cells in vitro and in vivo. Gynecol. Oncol. 2012, 127, 390–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  457. Hampsch, R.A.; Wells, J.D.; Traphagen, N.A.; McCleery, C.F.; Fields, J.L.; Shee, K.; Dillon, L.M.; Pooler, D.B.; Lewis, L.D.; Demidenko, E.; et al. AMPK Activation by Metformin Promotes Survival of Dormant ERþ Breast Cancer Cells. Clin. Cancer Res. 2020, 26, 3707–3719. [Google Scholar] [CrossRef] [Green Version]
  458. Lee, H.G.; Shin, S.-J.J.; Chung, H.-W.W.; Kwon, S.-H.H.; Cha, S.-D.D.; Lee, J.-E.E.; Cho, C.-H.H. Salinomycin reduces stemness and induces apoptosis on human ovarian cancer stem cell. J. Gynecol. Oncol. 2017, 28, e14. [Google Scholar] [CrossRef] [Green Version]
  459. Mi, Y.; Huang, Y.; Deng, J. The enhanced delivery of salinomycin to CD133+ ovarian cancer stem cells through CD133 antibody conjugation with poly(lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles. Oncol. Lett. 2018, 15, 6611–6621. [Google Scholar] [CrossRef] [Green Version]
  460. Lee, H.; Kim, J.W.; Kim, D.K.; Choi, D.K.; Lee, S.; Yu, J.H.; Kwon, O.-B.; Lee, J.; Lee, D.-S.; Kim, J.H.; et al. Calcium Channels as Novel Therapeutic Targets for Ovarian Cancer Stem Cells. Int. J. Mol. Sci. 2020, 21, 2327. [Google Scholar] [CrossRef] [Green Version]
  461. Alqudah, M.; Al-Samman, R.; Azaizeh, M.; Alzoubi, K. Amlodipine inhibits proliferation, invasion, and colony formation of breast cancer cells. Biomed. Rep. 2022, 16, 50. [Google Scholar] [CrossRef]
  462. Chang, Y.-H.; Lin, Y.-J.; Huang, C.-Y.; Harnod, T.; Ding, D.-C. Shikonin impedes type 2 ovarian cancer progression via FasL/caspase-8 and mir-874-3p/XIAP axis and prohibits the properties of stemness. Am. J. Cancer Res. 2022, 12, 4584–4601. [Google Scholar] [PubMed]
  463. Thakur, R.; Trivedi, R.; Rastogi, N.; Singh, M.; Mishra, D.P. Inhibition of STAT3, FAK and Src mediated signaling reduces cancer stem cell load, tumorigenic potential and metastasis in breast cancer. Sci. Rep. 2015, 5, 10194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  464. Xu, H.; Zhao, F.; Wu, D.; Zhang, Y.; Bao, X.; Shi, F.; Cai, Y.; Dou, J. Eliciting effective tumor immunity against ovarian cancer by cancer stem cell vaccination. Biomed. Pharmacother. 2023, 161, 114547. [Google Scholar] [CrossRef] [PubMed]
  465. Wu, D.; Yu, X.; Wang, J.; Hui, X.; Zhang, Y.; Cai, Y.; Ren, M.; Guo, M.; Zhao, F.; Dou, J. Ovarian Cancer Stem Cells with High ROR1 Expression Serve as a New Prophylactic Vaccine for Ovarian Cancer. J. Immunol. Res. 2019, 2019, 9394615. [Google Scholar] [CrossRef] [Green Version]
  466. Wu, D.; Wang, J.; Cai, Y.; Ren, M.; Zhang, Y.; Shi, F.; Zhao, F.; He, X.; Pan, M.; Yan, C.; et al. Effect of targeted ovarian cancer immunotherapy using ovarian cancer stem cell vaccine. J. Ovarian Res. 2015, 8, 68. [Google Scholar] [CrossRef] [Green Version]
  467. Ruiu, R.; Di Lorenzo, A.; Cavallo, F.; Conti, L. Are Cancer Stem Cells a Suitable Target for Breast Cancer Immunotherapy? Front. Oncol. 2022, 12, 877384. [Google Scholar] [CrossRef]
Figure 1. Common oncogenic signaling pathways that have been implicated in the maintenance of BCSC and OCSC. Picture shows canonical types of activation of nine oncogenic pathways: WNT, PI3K/AKT/mTOR, NOTCH, HH-GLI, JAK/STAT, TGFβ, NF-κβ, Hippo, and NRF2 signaling pathways.
Figure 1. Common oncogenic signaling pathways that have been implicated in the maintenance of BCSC and OCSC. Picture shows canonical types of activation of nine oncogenic pathways: WNT, PI3K/AKT/mTOR, NOTCH, HH-GLI, JAK/STAT, TGFβ, NF-κβ, Hippo, and NRF2 signaling pathways.
Ijms 24 10683 g001
Figure 2. Tumor microenvironment of BC and OC. The most represented cell types are cancer-associated fibroblasts (CAF), adipocytes, immune cells, and carcinoma-associated mesenchymal stem cells (CA-MSC). All these cell types create a communication network and are accompanied by the secretion of different cytokines or chemokines which play an important role in determining the fate of the tumor.
Figure 2. Tumor microenvironment of BC and OC. The most represented cell types are cancer-associated fibroblasts (CAF), adipocytes, immune cells, and carcinoma-associated mesenchymal stem cells (CA-MSC). All these cell types create a communication network and are accompanied by the secretion of different cytokines or chemokines which play an important role in determining the fate of the tumor.
Ijms 24 10683 g002
Table 2. CSC-targeted strategies commonly used against BCSC and OCSC.
Table 2. CSC-targeted strategies commonly used against BCSC and OCSC.
CompoundMode of ActionExperimental ModelEffect
SalinomycinMembrane ionophore antibioticBC cell linesReduces BCSC proportion, and improves survival [416].
Salinomycin-HDLOC cell line (CD133+ population), normal ovarian epithelial line, xenograftsDownregulation of stemness markers (MYCN, NANOG, OCT4, SOX2) in vitro and in vivo, induction of apoptosis [417].
MetforminAntidiabetic agentBC cell lines (spheroids)Decreases CD44+/CD24−/population and mammospheres formation [418].
OC cell lines (spheroids), xenograftsInhibits the CD44+ CD117+ population, no effect on CD44+ ALDH+ population, inhibits EMT [163].
EmodinAnti-malarial and anti-allergic agentBC cell lines and mouse modelsSuppresses TGF-β1 production, reduces macrophage-induced EMT and BCSC formation, and reduces breast cancer lung metastasis [419].
OC cell lines, xenograftsInhibits the growth of OC cells in vitro and in vivo, reduces the number of CD44+/CD24− OCSC [420].
1α,25-dihydroxyvitamin D3VitaminBC cell linesInhibition of spheroid formation, downregulation of stemness markers (OCT4, CD44, NOTCH1, NOTCH2, NOTCH3) [421].
OC cell lines, xenograftsInhibition of spheroid formation, downregulation of stemness markers (OCT4, CD44, NANOG, SOX2, KLF4, ABCG2), delays onset of tumor formation [422].
LuteolinFlavonoid, antioxidantBC cell lines, xenograftsInhibition of stemness and chemoresistance via the NRF2-mediated pathway [423] and suppresses EMT and migration of TNBC cells by inhibition of Hippo/YAP pathway [424].
OC cell line, primary OC (CD133+ ALDH+ populations), xenograftsInhibition of stemness, inhibition of Hippo/YAP pathway, sensitization to paclitaxel and carboplatin [425].
ALM201Anti-angiogenic therapeutic peptideBC cell lines (spheroids),
xenografts
Inhibition of spheroid formation, decrease in in vivo lung metastasis, and sensitization to tamoxifen by downregulating NOTCH4 and DLL4 [426].
OC cell lines, xenograftsInhibition of spheroid formation, inhibition of CD44+ CD117+ population, induction of differentiation [427].
SimvastatinInhibition of cholesterol synthesisBC cell lines (spheroids),
PDX
Radiosensitizes mammospheres of inflammatory BC (IBC) and TNBC, yet radioprotects HR+ and HER2+ [428]; inhibition of spheroid formation in PDX-derived TNBC [429].
OC cell lines, primary cultures, xenograftsInhibition of spheroid formation, downregulation of CD44 and ALDH1A1, reduction of tumorigenesis and metastasis in vivo [430].
All-trans retinoic acid (ATRA)Vitamin A metaboliteBC cell lines,
xenografts
Induces differentiation, inhibits invasiveness and migration of BCSC, and sensitizes to chemotherapy [431].
OC cell lines (ALDH-high and ALDH-low),
xenografts
Inhibition of stemness properties, inhibition of p62/NRF2 axis, decreases in vivo tumor growth only in ALDH-high OCSC [248].
EntinostatHDAC inhibitorsBC cell lines, mouse modelReduces CD44+/CD24− cell population, ALDH-1 activity, BMI-1, NANOG, and OCT4. Reduces tumor formation at the primary site and lung metastasis [432].
16cyc-HxA, 16lin-HxA, and 16KAOC cell lines, xenograftsInduction of apoptosis in OCSC, reduction of tumor size in vivo [433].
EugenolInhibition of NOTCH pathwayBC cell lines, mouse modelDownregulation of stemness of secondary mammosphere, lower CD44+/CD24−/low population, stemness suppression [434].
OC cell lines, xenograftsReduction of chemotherapy-induced spheroid formation, inhibition of CD44+ and ALDH+ cells, increases tumor-free survival [435].
Saracatinib + gemcitabineInhibition of SRC + chemotherapeuticBC cell lines, mouse modelSynergistic effect, reverse drug resistance, inhibition tumor stemness, metastasis, and growth of BCSC (CD44+ OCT4+) [436].
Saracatinib + selumetinibSRC inhibitor + MEK inhibitorOC cell lines, primary cell lines, xenograftsSynergistic effect on cell cycle arrest, induction of apoptosis and autophagy, decrease in ALDH1+ population in vitro and in vivo [437].
EverolimusInhibitor of PI3K/Akt/mTOR signaling pathwayBC cell linesReverses Palbociclib resistance, decreases the expression of ALDH1 and NANOG, decreases cell migration, self-renewal, and EMT [438].
5-aza-2′-deoxycytidineInhibitor of DNA methylationBC cell linesDecreases the expression of CD44+/CD24−, activation of P53 expression, increases sensitivity to chemotherapy [439]
Everolimus + 5-aza-2-deoxycytidinemTOR inhibitor + inhibitor of DNA methylationOC cell lines, xenograftsSynergistic effect on OC tumorspheres (ALDH1+ CD44+) and tumor formation in vivo, induction of apoptosis [440].
Disulfiram + cisplatin + paclitaxelALDH inhibitor + chemotherapyBC cell linesDecreases ALDH activity, increases expression of SOX2 and NANOG. Disulfiram sensitizes BCSCs to chemotherapeutics [441].
OC cell linesSynergistic effect on OC cells, re-sensitization of cisplatin-resistant lines [442].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lučić, I.; Kurtović, M.; Mlinarić, M.; Piteša, N.; Čipak Gašparović, A.; Sabol, M.; Milković, L. Deciphering Common Traits of Breast and Ovarian Cancer Stem Cells and Possible Therapeutic Approaches. Int. J. Mol. Sci. 2023, 24, 10683. https://doi.org/10.3390/ijms241310683

AMA Style

Lučić I, Kurtović M, Mlinarić M, Piteša N, Čipak Gašparović A, Sabol M, Milković L. Deciphering Common Traits of Breast and Ovarian Cancer Stem Cells and Possible Therapeutic Approaches. International Journal of Molecular Sciences. 2023; 24(13):10683. https://doi.org/10.3390/ijms241310683

Chicago/Turabian Style

Lučić, Ivan, Matea Kurtović, Monika Mlinarić, Nikolina Piteša, Ana Čipak Gašparović, Maja Sabol, and Lidija Milković. 2023. "Deciphering Common Traits of Breast and Ovarian Cancer Stem Cells and Possible Therapeutic Approaches" International Journal of Molecular Sciences 24, no. 13: 10683. https://doi.org/10.3390/ijms241310683

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

Article Metrics

Back to TopTop