Abstract
Ovarian cancer is the most lethal gynecologic malignancy and the fifth leading cause of cancer-related death in women. Although outcomes have improved in recent years, there remains an unmet clinical need to understand the early pathogenesis of ovarian cancer in order to identify new diagnostic approaches and agents of chemoprevention and chemotherapy. While high grade serous ovarian cancer (HGSOC), the most abundant histotype, was initially thought to arise from the ovarian surface epithelium, there is an increasing body of evidence suggesting that HGSOC originates in the fallopian tube. With this new understanding of cell of origin, understanding of disease development requires analysis with a novel perspective. Currently, factors that drive the initiation and migration of dysplastic tubal epithelial cells from the fallopian tube to the ovary are not yet fully defined. These factors include common mutations to fallopian tube epithelial cells, as well as factors originating from both the fallopian tube and ovary which are capable of inducing transformation and dissemination in said cells. Here, we review these changes, their causative agents, and various potential means of intervention.
Keywords: carcinogenesis, chemoprevention, chemotherapy, oviduct, STIC, TP53
1. INTRODUCTION
Ovarian cancer is the fifth leading cause of cancer-related death in women. While outcomes have improved in recent years, the current five-year survival rate remains a dismal 47% [1]. This is largely attributed to the late stage of presentation, as the majority of ovarian cancer patients are diagnosed with metastatic disease, at which point the five-year survival rate can be as low as 17% [2]. As such, there remains an urgent need to clarify the early pathogenesis of ovarian cancers in hopes of identifying new diagnostic techniques, as well as effective means of prevention and treatment.
Clinically, ovarian cancers are segregated into five predominant histotypes, the most common being high-grade serous ovarian cancer (HGSOC). HGSOC carries a particularly poor prognosis and is responsible for 70–80% of ovarian cancer deaths [3]. Though HGSOC was initially thought to arise from the ovarian surface epithelium, there is now clear evidence that HGSOC can also originate in the fallopian tube epithelium [4–12]. This is significant, as the cell of origin alters screening modalities, imaging practices, surgical intervention, and target-based drug discovery. Ultimately, identifying the cells of origin will better enable detection of pre-invasive disease and intervention before these cells disseminate and invade the ovary.
Recent evidence supports that many HGSOCs originate in the epithelial cells at the distal tips of the fimbriae of the fallopian tube, known as the oviduct in non-human species. For instance, approximately 40–60% of HGSOC patients harbor additional lesions in the fallopian tube [5–7]. As these lesions typically share identical TP53 mutations with their HGSOC counterparts, they likely represent the clonal origin of HGSOC [8, 9]. Additionally, salpingectomy is associated with a 35% reduction in ovarian cancer risk while tubal ligation reduces risk by 13% [10, 11]. In a study comparing HGSOC specimens to normal epithelial tissues from several other organs with cDNA microarrays, HGSOC mRNA expression patterns closely resembled that of normal fallopian tube epithelial cells yet differed from other sites including the ovary [12]. This was paralleled by additional studies using RNA sequencing, further confirming the high degree of similarity between HGSOC and the fallopian tube epithelium [9].
Hence, while it is now understood that HGSOC can originate from fallopian tube epithelial cells, how these cells initially transform and disseminate to colonize the ovary remains unclear. Understanding the local cues that direct these processes may present a new opportunity for intervention. In this review, we will survey the known local cues that mediate both physiologic and pathologic crosstalk between the ovary and fallopian tube and discuss potential means of intervention.
The ovary releases follicles and secretes associated hormones into and onto the fallopian tube in the form of follicular fluid, which contains a variety of factors now believed to mobilize dysplastic fallopian tube epithelial cells, potentially recruiting them to the ovary via chemotactic gradient. Here, we describe the initial genetic insults common to ovarian cancer patients, as well as the contents of fallopian tube and ovarian secretions and follicular fluid that are reported to direct these abnormal cells to the ovary where they establish overt tumors (Figure 1).
Figure 1. Signals of Interest in Ovarian Cancer Development.
Serous tubal intraepithelial carcinomas (STICs) and other precursor lesions of ovarian cancer exist throughout the fallopian tubes and are regularly exposed to several potentially oncogenic factors. These factors stem from secretions, from fallopian tube and ovarian epithelium, and follicular fluid that is released upon follicle rupture during the reproductive cycle. They can be protective or transformative, and stimulate proliferation or metastasis of fallopian tube epithelial cells.
2. INITIATION OF CANCER IN THE FALLOPIAN TUBE EPITHELIUM
One of the earliest potential HGSOC precursor lesions, dubbed secretory cell outgrowths (SCOUTs), consist of at least 30 secretory fallopian tube epithelial cells and are located all throughout the fallopian tube [13]. SCOUTs are almost entirely devoid of PAX2, a transcription factor normally expressed in fallopian tube epithelial cells [13–16]. Interestingly, while SCOUTs are more frequently found in the fallopian tubes of patients with HGSOC, their exact relationship to HGSOC has yet to be determined [17]. The Cancer Genome Atlas and a follow up analysis have reported that 96–100% of HGSOC carry TP53 mutations [18]. As TP53 is a tumor suppressor, alteration to the gene via mutation or deletion could enable unchecked cell growth, reduced DNA repair, and eventual development of cancer. The most accepted preneoplastic lesion identified in relation to HGSOC is the p53 signature, consisting of benign secretory cells exhibiting robust p53 staining (or a total lack of staining), positive ϒH2AX staining, and lack of Ki-67 staining [19]. Serous tubal intraepithelial carcinomas or STICs, share the same characteristics of DNA damage and p53 mutations as p53 signatures, but are composed of malignant cells featuring a high mitotic index and disorganized growth [19]. The prevailing theory is that p53 signatures progress to STICs, which are then believed to ultimately develop into HGSOC [8, 19–21]. Several chemical signals have been suggested to induce transformation in fallopian tube epithelial cells and subsequently play a role in HGSOC initiation.
2.1. FOLLICULAR FLUID
While the association of decreased ovulation with decreased risk of developing ovarian cancer is well-established epidemiologically, it remains unexplained biologically. During ovulation, ovarian follicles release fluid which washes the surrounding tissues including the fallopian tube. Secreted factors found in follicular fluid may be involved in the initiation or development of ovarian cancer. While some components of follicular fluid and their effects on neighboring tissues are discussed below, other components such as follicle stimulating hormone, anti-Mullerian hormone, luteinizing hormone, and androgens, are covered in depth by Emori and Drapkin [22].
Ovulation may instigate ovarian cancer development due to the release of follicular fluid on fallopian tube epithelia. When fallopian tube epithelial cells were treated with follicular fluid, they demonstrated a 1.7-fold increase in phospho-ϒH2AX, a known DNA double-stranded break response [23]. Additionally, compared to negative controls, 5% of the follicular fluid treated cells stained positive for TP53, suggesting that follicular fluid may induce TP53 stabilization [23]. This is noteworthy due to the common presentation of TP53 mutations in ovarian cancer precursor lesions. Interestingly, human fallopian tube epithelial cells express activation-induced cytidine deaminase (AID), which creates DNA mutations by converting a C:G base pair to a T:A base pair [24]. When treated with follicular fluid, fallopian tube epithelial cells demonstrated a significant induction of AID, an increase in double-strand DNA breaks, and a reduction in DNA methylation [24]. Similarly, when fallopian tube epithelial cells were exposed to follicular fluid, an increase in expression of APOBEC cytidine deaminases, which also cause DNA mutations, was reported [25]. Transient overexpression of a particular APOBEC cytidine deaminase, APOBEC3a, was able to induce double-strand DNA breaks in fallopian tube epithelial cells [25]. Collectively, these DNA damaging responses to follicular fluid support its potential role as a mutagen.
Reactive oxygen species, either contained within or produced as a response to follicular fluid, have also been suggested as instigating factors in the transformation of fallopian tube epithelial cells. In a model of ovulation, where tissue was collected 12 hours post injection with human chorionic gonadotropin, murine oviductal epithelial cells demonstrated increased levels of phospho-ϒH2AX, suggesting an increase in DNA damage [26]. Pro-inflammatory macrophages, which are known to produce reactive oxygen species were detected after ovulation [26]. Furthermore, follicular fluid treatment of fallopian tube secretory epithelial cells induced an increase in intracellular reactive oxygen species [27]. Treatment of cells harboring a p53 mutation with excess reactive oxygen species led to an increase in miR-182 expression which allowed cells to bypass senescence [27]. Together these data suggest that the release of reactive oxygen species-containing follicular fluid during ovulation may directly induce DNA damage in tubal epithelial cells or recruit pro-inflammatory immune cells.
In addition to studying the effects of follicular fluid as a whole, its specific components have also been investigated. Transferrin, an iron transport protein, has been identified as a component in follicular fluid that may induce DNA damage. Treatment of fallopian tube epithelial cells with transferrin increased expression of phospho-ϒH2AX and formation of double-strand breaks while knockdown of the transferrin receptor or treatment with small interfering RNA abrogated this effect [28]. Ferryl hemoglobin, found in ovulatory follicular and peritoneal fluids, rescued p53-deficient oviduct epithelial cells from lethal reactive oxygen species by consuming these species extracellularly and reducing NADPH oxidase-mediated cell death [29]. However, while the cells exposed to high levels of reactive oxygen species were able to evade apoptosis due to the protection by ferryl hemoglobin, they still harbored double-stranded DNA breaks suggesting that their rescue could allow these damaged cells to transform [29]. Similarly, fallopian tube epithelial cells treated with non-transferrin bound iron displayed increased cell number, migration, and DNA damage relative to untreated cells [30]. Iron treated cells also demonstrated increased expression of oncogenic markers such as β-catenin, Myc, and ecotropic virus integration site 1 [30].
Insulin-like growth factor 2 (IGF2) is a peptide hormone found in follicular fluid [31]. When fallopian tube epithelial cells were treated with follicular fluid, anchorage independent growth increased, an effect abrogated by inhibition of the IGF receptor [31]. Interestingly, a clinical study noted that IGF2 expression was higher in ovarian tumor tissues than those of healthy ovaries [32]. Higher expression of IGF2 was also associated with decreased overall survival in patients [32]. A separate study observing the effect of follicular fluid on fallopian tube epithelial cells noted rapid upregulation of the inflammatory cytokine interleukin-8 (IL-8) [23]. Beyond its canonical roles in inflammation, IL-8 has also been implicated in a variety of additional cell processes in ovarian tissue, namely increased proliferation. Furthermore, Transforming Growth Factor β (TGFβ) has also been identified as a component of follicular fluid for which fallopian tube epithelial cells express receptors. One study observing the effect of TGFβ on oviductal epithelium demonstrated a reduction in PAX2 expression, which could potentiate precursor lesions through loss of tumor suppressive action [33]. These data suggest that while some components of follicular fluid could contribute to the initiation of ovarian cancer, others could play a role in tumor maintenance by promoting proliferation.
As previously mentioned, decreased ovulation is associated with decreased risk of developing ovarian cancer. In concordance, epidemiologic data also suggests that the use of oral contraception reduces ovarian cancer risk since its components, progestin with or without estrogen, block ovulation [34]. Intrauterine devices (IUDs), another popular mechanism of contraception, prevent pregnancy through both the inflammatory response to a foreign body, which is toxic to sperm and ova, as well as local effects specific to the type of IUD [35]. For example, levonorgestrel IUDs release progestins which can prevent ovulation and thicken cervical mucus to prevent fertilization and implantation [35]. Interestingly, a retrospective review described an association between IUD use and decreased risk of developing ovarian cancer [36]. However, as this study was retrospective, the specific type of IUD was unavailable for subset analysis so the resulting association warrants further investigation in order to frame its relation to ovulation.
2.2. ESTROGEN
The local role of estrogen on the fallopian tube primarily involves facilitating the movement and maturation of eggs, sperm, and embryo rolling [37]. However, exposure to estrogens, including the natural hormone estradiol, is considered a risk factor for development of ovarian cancer [38–40]. Estrogen induced changes include genetic mutations, altered gene expression, as well as downstream effects of estrogen-induced oxidants like DNA strand breaks and adduct formation [41, 42]. Interestingly, while treatment of murine ovarian epithelial cells with estradiol did not result in cellular proliferation and migration, RNA sequencing of these cells revealed increased expression of proliferation and anti-apoptosis transcripts [43]. Furthermore, early lesions in the fallopian tube composed of PAX2 deficient cells have been shown to have increased estrogen receptor [44]. Clinically, women taking estrogen only hormone replacement therapy have increased risk of developing ovarian cancer compared with those who have never taken estrogen only therapy or estrogen-progesterone combination therapy [38, 39]. Together, this data suggests that estrogens may be involved in the development of ovarian cancer.
Hormonal therapy has shown efficacy in certain cancers. In the treatment of ovarian cancer, various clinical studies analyzing both antiestrogens and inhibitors of aromatase, which converts testosterone to estradiol, have demonstrated modest efficacy [45]. This is supported by a separate analysis of 53 different trials of endocrine therapies which demonstrated a clinical benefit rate of 41%, with an increased effect in estrogen receptor positive tumors [46]. Additionally, several clinical trials have shown that tamoxifen provides a response rate of 15% in platinum-resistant ovarian cancer which has led to its use as a second-line treatment since its toxicity profile is more favorable than that of other chemotherapy agents [47]. However, studies of endocrine therapies require more robust analysis pertaining to subtype sensitivity and optimal treatment settings.
2.3. PROGESTERONE
Progesterone, like estrogen, is a steroid hormone component of follicular fluid which is necessary for proper follicle development and ovulation. Progesterone is believed to mainly have a protective effect against ovarian cancer. In studies of patients with ovarian cancer, increased progesterone receptor expression was associated with improved overall and disease-free survival [48, 49]. This is further supported by the observation that women taking combined estrogen-progestin hormone replacement therapy or oral contraceptives have decreased relative risk of ovarian cancer when compared with non-users [50–52]. Additionally, pregnancy is associated with decreased risk of ovarian cancer, an observation likely due to higher levels of progesterone during pregnancy [53].
3. FACTORS OF FALLOPIAN TUBE ORIGIN AND THEIR ROLE IN DISEASE
The change in paradigm regarding cell of origin creates an opportunity to study disease onset and progression from an angle that had previously been neglected. For instance, as recent research has found, there are several factors that originate in the fallopian tube that both serve as a marker of origin for HGSOC and to influence disease progression.
3.1. CA-125
Serum cancer antigen 125 (CA-125), a glycoprotein also known as MUC16, is expressed in epithelial cells in normal tissues like the genitourinary tract and overexpressed in some epithelial cancers [54]. MUC16 expression in epithelial ovarian cancers aligns with that of fallopian tube epithelium, suggesting the latter as the cell of origin [55]. CA-125 is currently utilized as a surrogate biomarker in clinical practice to gauge response to treatment and detect recurrence [56]. However, CA-125 is not used presently for ovarian cancer screening.
In addition to its use as a surrogate biomarker, CA-125/MUC16 appear to contribute to disease development. For example, knockdown of MUC16 in ovarian cancer cells resulted in increased synapses between these and natural killer cells, and increased lysis of ovarian cancer cells [57]. In vivo, mice with ovarian tumors featuring MUC16-knockdown displayed increased survival compared to controls [58]. These data suggest that CA-125/MUC16 are capable of suppressing the innate anti-tumor immune system response. Another suggested role for CA-125 is in metastasis, since stable expression of MUC16 increased motility and invasiveness [59]. CA-125 was also associated with decreased expression of E-cadherin and increased expression of N-cadherin and vimentin suggesting its effects on metastasis and epithelial to mesenchymal transition (EMT) [59]. Additionally, down-regulation of MUC16 led to increased susceptibility of ovarian cancer cells to cisplatin [60]. While similar effects were not noticed in response to paclitaxel, this data supports the role of MUC16 in genotoxic chemotherapy resistance [60].
Several anti-CA-125/MUC16 therapies have been investigated. One such potential treatment aimed to target MUC16-mesothelin interactions via a fusion of human antibody Fc region to the binding domain of mesothelin, but it remains to be examined in a clinical setting [61]. In patients with recurrent ovarian cancer, co-treatment of chemotherapy and oregovomab, a murine monoclonal antibody directed against CA-125, resulted in immune responses to CA-125 and improved time to progression and survival [62]. Another strategy utilizing DMUC5754A, composed of an anti-MUC16 antibody conjugated to a microtubule-disrupting agent, demonstrated anti-tumor activity in patients with MUC16 expressing tumors with an acceptable safety profile in a Phase I clinical trial [63].
3.2. PROLACTIN
Prolactin, most commonly referenced as a neuroendocrine hormone secreted from the anterior pituitary, is also produced in many peripheral tissues including the brain, mammary glands, placenta, amnion, decidua, and uterus [64]. Similarly, prolactin receptors are found throughout the body [64]. Interestingly, both human ovaries and fallopian tube epithelia produce prolactin and express prolactin receptors indicating that prolactin may act as a hormone and auto- or paracrine regulatory factor [65, 66].
A clinical study noted that compared with pancreatic, lung, and breast cancers, prolactin levels were highest in patients with endometrial and ovarian cancers [67]. In a study of 230 patients with ovarian cancer, 120 of which had serous histology, increased prolactin was associated with an increased risk of ovarian cancer in the subgroup of patients with a BMI > 25kg/m2 [68]. Recently, a prolactin receptor targeted imaging platform has demonstrated more specific and sensitive detection of ovarian cancer when compared with currently utilized contrast agents like GdDTPA [69].
The clinical data suggesting prolactin could be involved in ovarian cancer aligns with in vitro data. In a murine spontaneous model of fallopian tube-derived ovarian cancer, a prolactin-like gene (Prl2c2) was the most abundant transcript and silencing it reduced tumor burden [70]. In the ovarian cancer cell line OV-90, prolactin receptor antagonists decreased cell number and migration [71]. In both the transformed human fallopian tube epithelium cell line FT33-TAg-Myc and the ovarian cancer cell line OVCAR8, treatment with prolactin induced proliferation through the receptor [66]. When mice were injected with OVCAR3 cells that had prolactin receptors deleted, no tumor growth was noted [66]. Moreover, incubating OVCAR3 cells with prolactin prior to treatment with cisplatin protected the cells from drug-induced apoptosis [67]. Collectively, these data indicate that since prolactin is normally secreted from the fallopian tube epithelia, it likely acts in an autocrine manner to induce clonal proliferation of tumorigenic cells and potentiate chemoresistance.
For this reason, prolactin and prolactin receptor antagonists are attractive treatment modalities. Of note, when orthotopic models of human ovarian cancer were treated with the prolactin antagonist G129R, the result was an inhibition of tumor growth and increase in autophagy [72]. However, little research has been conducted regarding the prolactin signaling pathway and its potential for therapeutic use in ovarian cancer and much remains to be concluded.
3.3. PAX8
While paired box transcription factor 8 (PAX8) is normally expressed in the fallopian tube epithelium, it is not usually expressed in the ovarian surface epithelium [73]. However, PAX8 is almost universally expressed in HGSOC tumors [73]. These data serve as support for PAX8 as a marker of cell origin, demonstrating that HGSOC can stem from fallopian tube epithelial cells.
In addition, PAX8 also plays a role in disease development. For example, knockdown of PAX8 expression in a human ovarian cancer cell line resulted in reduction of proliferation, migration, and invasion, as well as tumorigenesis in an in vivo model [74]. Similarly, knockdown of PAX8 through shRNA was able to reduce viability and induce apoptosis of a majority of tested ovarian cancer cell lines [75]. Another study observing the effect of PAX8 expression in ovarian surface epithelial cells demonstrated increased proliferation and migration, as well as upregulation of various factors associated with EMT [76]. Interestingly, while knockdown of PAX8 in several human HGSOC cell lines decreased proliferation and increased apoptosis, knockdown in oviductal epithelium had little to no effect on proliferation, migration, or apoptosis [76]. This suggests an opportunity to target PAX8 pharmacologically in order to reduce the viability of transformed cells without affecting normal tissues.
While PAX8 expression may not be a predictive biomarker as it does not associate with tumor stage or disease outcome, its high prevalence in HGSOC cases makes its inhibition a suitable candidate for therapeutic intervention. One such possible intervention is thiostrepton, an antibiotic that inhibits translation by binding the prokaryotic ribosome, which was able to reduce PAX8 and inhibit tumor progression in vivo [77]. Another investigation of anti-PAX8 therapy centers around class I histone deacetylase inhibitors (HDACis). Panobinostat (a pan-HDACi) and romidespin (a class I HDACi) were able to eradicate PAX8 protein and PAX8-positive tumor cells, as well as inhibit migration through disruption of PAX8 transcription and downstream regulation [78]. Further in vivo analysis showed that treatment with HDACis resulted in decreased tumor burden and metastatic dissemination while co-treatment with HDACis and platinum chemotherapy indicated less tumor progression than single therapy treatment [78]. Taken together, these data suggest PAX8 may be both a marker of cell of origin and a worthwhile therapeutic target for HGSOC.
4. DISSEMINATION OF FALLOPIAN TUBE CELLS TO THE OVARY
Fallopian tube dysplastic cells likely exhibit organotropism for the ovary since the majority of HGSOC patients are diagnosed with large ovarian masses. To this point, tail vein injection of four separate human ovarian cancer cell lines led to a total of 66% of mice developing ovarian metastases while injection of four non-ovarian cancer cell lines led to no ovarian metastases [79]. This suggests that ovarian cancer cells have a particular proclivity for spread to the ovary. Since a murine model showed superovulation increased the colonization of the ovary following xenograft of ovarian tumor initiating cells, this predisposition correlates with ovulation [80]. Additionally, when tumorigenic fallopian tube epithelial cells were injected in murine bursa, all mice developed ovarian tumors and were sacrificed due to tumor burden by day 90, while no mice receiving intraperitoneal injections developed tumors for the duration of the experiment [81]. In transgenic mouse model which spontaneously develops ovarian cancer, removal of the ovaries reduced peritoneal metastases, suggesting that ovaries are required for such progression [82]. While these data support the tendency for fallopian tube cells to migrate to the ovary preferentially, which signaling molecules contribute to this dissemination are yet unclear. However, several potential factors have been explored and are discussed in the following sections.
4.1. WNT Signaling
Recent evidence appears to implicate WNT signaling in the recruitment of abnormal fallopian tube epithelial cells to the ovary. WNT4 appears to be significantly amplified in murine models of oviductal oncogenesis, as do select WNT receptors Frz2, 4, 5, and 8 [70]. Interestingly, a single nucleotide polymorphism in the WNT4 gene appears to have a significant association with the incidence of ovarian carcinoma [83]. A genome wide association study examining susceptibility loci for high-grade epithelial ovarian cancer noted that amplification of the WNT4 locus is associated with increased incidence of disease [84]. RNA sequencing of a murine oviductal cell model lacking PTEN and characterized as high grade oviductal carcinoma displayed significant upregulation of WNT4 and several other genes associated with non-canonical WNT signaling [85]. This upregulation was similarly validated in a transgenic model with conditional PTEN knockout, and was required for tumor cell migration and colonization of the ovary in a β-Catenin-independent manner [86]. These results were confirmed in human tumor microarrays and ovarian cancer cells lines, which substantiate a pro-migratory role for WNT4 in ovarian cancer [86]. Interestingly, this study also demonstrated that WNT4 is strongly expressed by the fallopian tube, implicating its use as a marker of cell origin as well [86]. Taken together, these data suggest that the abundant WNT4 in the fallopian tube may have key roles in stimulating the migration and invasion of abnormal cells, thereby aiding the initiation of HGSOC. However, while a few treatment methods involving canonical WNT signaling are currently under investigation, therapeutic agents involving non-canonical signaling remain unstudied.
4.2. SDF-1
Stromal cell-derived factor 1 (SDF-1) and its receptor, CXCR4, play essential roles in trafficking haemato- and lymphopoietic cells as well as stem and progenitor cells [87]. SDF-1 and CXCR4 are also expressed in ovarian cell lines and ovarian tumors, and SDF-1 is a known component of follicular fluid [88]. Notably, in patients with ovarian cancer, an increased expression of SDF-1 is associated with reduced overall survival while an increased expression of CXCR4 is associated with reduced progression-free survival and overall survival [88, 89]. The CXCR4 and SDF-1 interaction has been shown to stimulate invasion and proliferation in human ovarian cancer cell lines. CXCR4 upregulation is also associated with disease progression and metastasis in human ovarian cancer specimens [90]. Ovarian tumor initiating cells treated with a CXCR4 antagonist demonstrated a significant reduction in migration [80]. The activation of ERK induced by the SDF-1 and CXCR4 interaction was abrogated when cells were treated with the same antagonist [80]. Altogether, these data suggest that SDF-1 and CXCR4 potentiate metastasis of ovarian cancer both from the fallopian tube to the ovary as well as from the ovary to the peritoneum.
As such, inhibition of the SDF-1/CXCR4 signaling axis poses an interesting opportunity for development of therapeutic agents. For example, treatment of ovarian cancer cells with AMD3100, a selective CXCR4 antagonist, resulted in promotion of apoptosis and inhibition of proliferation, migration, and invasion [91]. In addition, mifepristone, an FDA approved chemical abortifacient, was able to suppress SDF-1 induced migration, invasion, and adhesion of ovarian cancer cells [92]. Furthermore, pretreatment with mifepristone and co-treatment with mifepristone and SDF-1 led to decreased tumor weight when compared to treatment with SDF-1 alone [92]. However, more research is needed to determine if these methods would be sustainable treatments for ovarian cancer.
4.3. ACTIVIN A
Activin A, a classic ovarian hormone belonging to the TGFβ superfamily, is composed of two inhibin βA subunits, and signals through type I and type II activin receptors. In murine oviductal cells, treatment with activin A resulted in induction of both EMT as well as increased migration [93]. Similar effects were noted in ovarian cancer cells with the additional observation that treatment with activin A inhibitors decreased migration [93]. According to clinical data in Oncomine, serous tumors had higher levels of activin A receptors and lower levels of activin A signaling inhibitors when compared with normal ovarian samples [93]. Additionally, higher expression of both the activin A type II receptor and subunit was associated with decreased disease-free survival in patients [93]. Together, these data suggest that ovarian activin A is capable of inducing migration in oviductal cells and contributes to disease progression. However, a recent phase I clinical trial studying the efficacy of the activin A inhibitor, STM 434, demonstrated no significant antitumor effect and resulted in several adverse events [94].
4.4. NOREPINEPHRINE
Norepinephrine is a stress hormone secreted by adrenal glands and stored in various tissues throughout the body. In a co-culture model of malignant human fallopian tube epithelial cells and murine ovaries, imaging mass spectrometry showed that tumorigenic murine oviductal epithelial cells, but not their normal counterparts, stimulated norepinephrine secretion from the ovary [95]. Norepinephrine induced invasion in these cells as well, suggesting it may be one of the factors driving the primary metastasis of ovarian cancer from the fallopian tube to the ovary [95]. Interestingly, treatment of both fallopian tube and ovarian epithelial cells with norepinephrine resulted in the down-regulation of several genes involved in cell death, potentially allowing initial malignant cells to evade apoptosis [96].
As of late, norepinephrine has also been of interest at the clinical level. It is well known that despite optimal first-line treatment (surgery and chemotherapy), around 70–80% of patients with ovarian cancer will relapse with recurrent disease that is resistant to first-line chemotherapeutic agents [97]. While this trend has not been entirely explained, norepinephrine has been shown to stimulate DUSP1 expression which reduced platinum agent induced apoptosis in ovarian cancer cells suggesting norepinephrine may mediate chemoresistance [98]. Additionally, in a study of patients with epithelial ovarian cancer, use of beta-adrenergic receptor blockers was associated with a 54% reduced chance of mortality compared with non-users [99]. While similar studies have inspired clinical trials observing the effect beta-blockers may have on ovarian cancer patients without hypertension, little has been done to investigate the effect of beta-blockers as a means of chemoprevention in high risk patients [100]. Altogether, these data suggest that norepinephrine may significantly mediate cancer progression and as such warrants further therapeutic consideration.
5. GENETIC ALTERATIONS AND THE TUMOR MICROENVIRONMENT
Evidence suggests that mutations observed in HGSOC are often reflected in their fallopian tube lesion precursors [21]. For example, a study examining ovarian tumors and STIC lesions from individual patients determined that most had identical mutations in several genes including TP53, PTEN, BRCA1, and BRCA2. As such, in the context of the fallopian tube initiation model, it is likely that these mutations originated from tubal cells early in tumorigenesis. Given the established roles for these genes in cell proliferation and motility, it is also likely that cells from STIC lesions harboring select mutations may have aberrant responses to the paracrine factors described in this review article.
cBioportal was used to examine patient data from The Cancer Genome Atlas provisional dataset of high grade serous ovarian tumors along with the literature, which implicated several pathways that are likely to alter paracrine or autocrine responses to the ovarian microenvironment [101, 102]. Mutations affecting BRCA1, BRCA2, and PALB2 are common to ovarian cancer (Table 1), with BRCA genes being the most common genetic risk factor for HGSOC. These genes are known for their roles in the homologous recombination DNA repair pathway. Given the known DNA damaging effects of several components of the follicular fluid such as iron, AID, and various reactive oxygen species, it is important to consider that FTE cells deficient in the homologous recombination pathway might incur more damage or repair damage more slowly. Mutations to TP53 also modify cellular responses to DNA damage and are present at 96–100% of HGSOC. As previously mentioned, fallopian tube derived high-grade serous ovarian carcinomas carry early TP53 mutations. This is noteworthy because expression of mutant p53 in murine oviductal cells dramatically increased expression of the progesterone receptor [103]. Treatment of p53-null murine oviductal epithelial and p53-defective human fallopian tube epithelial cells with progesterone induced necroptosis [104]. In a Trp53−/− murine model, similar necroptotic effects were observed upon treatment with progesterone while inhibition of the progesterone receptor led to double-strand break accumulation [104]. Activation of the necroptosis pathway could explain the protective effect of progesterone against ovarian cancer. Interestingly, tumors harboring TP53 mutations display increased invasive potential in responses to norepinephrine [95]. Cyclin E1 (CCNE1) is amplified in 12.97% of HGSOC patients (Table 1) and has been found in early fallopian tube lesions, which is noteworthy considering constitutive CCNE1 expression has been shown to induce chromosomal instability [105]. For instance, CCNE1 amplification is associated with increased expression of ϒH2AX foci suggesting that these cells may also accumulate damage in response to follicular fluid differently than wild-type fallopian tube epithelial cells.
Table 1.
The frequency of genomic alterations in key signaling genes in the TCGA Firehose Legacy ovarian serous cystadenocarcinoma cohort
Gene | Total Alterations | Mutation | Amplification | Deletion | mRNA Upregulation | mRNA Downregulation | Multiple Alterations |
---|---|---|---|---|---|---|---|
TP53 | 88.29% | 87.03% | - | 0.32% | - | - | 0.95% |
PIK3CA | 35.44% | 0.63% | 18.35% | - | 6.65% | - | 9.81% |
NF1 | 26.58% | 1.9% | - | 0.95% | 0.63% | 14.24% | 8.86% |
CCNE1 | 26.27% | - | 12.97% | - | 2.22% | 2.85% | 8.23% |
SMAD2 | 20.33% | - | 1.1% | 1.1% | 1.65% | 12.64% | 3.85% |
PTEN | 17.41% | 0.32% | 0.63% | 0.63% | 2.85% | 7.91% | 5.06% |
BRCA2 | 11.71% | 3.48% | 1.9% | 0.95% | 3.48% | 1.27% | 0.63% |
PALB2 | 11.08% | 1.27% | 0.63% | - | 0.95% | 8.23% | - |
BRCA1 | 8.23% | 3.8% | 0.32% | 1.58% | 0.95% | 0.95% | 0.63% |
cBioportal was utilized to analyze ovarian serous cystadenocarcinoma patient data from The Cancer Genome Analysis Firehose Legacy patient data set (N = 606) according to the original references [101, 102]. Protocols for sequencing analysis of TCGA dataset can be located here: https://tcga-data.nci.nih.gov/tcga/. Genetic analyses were conducted using only samples with mutation data (N = 316).
In addition to the 87% of patients harboring TP53 mutations, mutations affecting PI3K signaling were also prevalent (Table 1). 35% of patients had either mRNA and/or copy number amplification of PIK3CA, while 17.4% had loss of PTEN. As both genes have been linked to pathologic activation of PI3K signals, dysplastic tubal cells harboring these mutations are likely to have abnormal responses to signals which exert their effects through the PI3K pathway. Current models appear to substantiate this notion, as loss of PTEN results in increased WNT4 signaling and a highly invasive phenotype [86]. Further, in the PAX8-Cre transgenic murine models of oviductal derived HGSOC with loss of PTEN, mutant TP53, and either mutant BRCA1 or BRCA2, these tumors formed rapidly and when animals were ovariectomized, they failed to disseminate into the peritoneal space [82]. Ovarian cancer patients also displayed frequent loss of NF1, a negative regulator of RAS signaling, which may also have significant effects on cellular responses involving downstream MAPK signaling networks including PI3K (Table 1). As activin A, estrogen, prolactin, and SDF1 are dependent on RAS/PI3K signaling to exert their pro-migratory functions, the contributions of these mutations to tumor cell recruitment warrant further study. Interestingly, we also observed frequent mRNA downregulation of SMAD2, a critical mediator of activin and TGFβ signaling (Table 1), both of which are known mediators of the tumor microenvironment contributing to several hallmark features of tumorigenesis including proliferation, migration, epithelial to mesenchymal transition, and immune evasion [106, 107]. As such, should loss of SMAD2 occur early in precursor lesions, this may have substantial ramifications in responses to TGFβ family ligands in the follicular fluid or ovarian secretions.
In summary, genomic alteration to these and many genes in dysplastic fallopian tube epithelial cells may lead to extensive signal reprogramming, subsequently affecting the interactions of said cells with paracrine signals from the surrounding fallopian tissue or ovary. Though potentially important to our understanding of HGSOC pathogenesis, how such mutations alter the interaction of tubal cells with the local tumor microenvironment is largely unstudied. Hence, further examination is necessary to better evaluate the early effects of these and other mutations, particularly regarding the design of early intervention strategies.
6. CONCLUSION
Several factors in both fallopian tube and ovarian secretions along with those contained in follicular fluid may be responsible for the transformation and dissemination of dysplastic fallopian tube epithelial cells to the ovary aiding in the establishment of ovarian cancer. These factors range in effect which include transformation, proliferation, and migration. While some of these factors are always present in the tissues, others are acquired during malignancy. A combination of mutations common to ovarian cancers can ultimately lead to pathway modifications capable of disease development (Figure 2). For instance, one of the earliest mutations found in SCOUT lesions is a loss of PAX2, which leads to increased susceptibility to estrogen signaling. Since estrogen signaling can then stimulate cellular damage and resistance of apoptosis, this loss of PAX2 can allow for perpetuation of damaged cells. Other common mutations found in early lesions include alterations to p53 and loss of PTEN. Lesions featuring these genotypic alterations are known to have increased WNT4, which results in increased capacity for metastasis and invasion. Additionally, mutation of p53 may alter the response of fallopian tube cells to progesterone to induce necroptosis. The surrounding norepinephrine secreted from ovarian tissue is capable of stimulating migration of these cells as well. In addition to the aforementioned mutations and impactful factors, follicular fluid is released into the environment surrounding both the fallopian tube and ovary on a monthly basis from the age of puberty (10–14 years) until menopause (45–55 years). Ultimately, a combination of events such as the ones described can transform benign fallopian tube epithelial cells into lesions capable of further progression, metastasis, and invasion of the ovary allowing for the formation of overt tumors. Importantly, while many of the components mentioned in this review have provided potential points of therapeutic intervention, further research on these various signals is required in order to fully elucidate their role in the initiation and progression of ovarian cancer in order to develop effective chemotherapeutic agents.
Figure 2. Changes in Ovarian-Fallopian Tube Communication from Normal Fallopian Tube Epithelium to Intraepithelial Carcinoma.
Before the occurrence of any transformational events, normal fallopian tube epithelium secretes several factors that likely play a role in disease development. If the epithelium forms a SCOUT lesion, a serous outgrowth typically PAX2 null, it then expresses increased estrogen receptor leading to increased estrogen signaling. If cells undergo loss of PTEN, they express higher levels of WNT4 and are susceptible to the proliferative and metastatic effects of norepinephrine. Mutation of p53 leads to the development of a signature lesion. In p53 null signature lesion cells, progesterone (P4) can protectively induce necroptosis. Combinations of these mutation and factor-induced transformations promote development of serous tubal intraepithelial carcinomas (STICs). STICs are considered malignant and are capable of dissemination to and invasion of the ovary.
HIGHLIGHTS.
Ovarian cancer is the most lethal gynecologic malignancy
Fallopian tube epithelium is a source for high grade ovarian cancer
New analysis of transformation and metastasis can improve diagnosis and treatment
ACKNOWLEDGEMENTS
This work is supported by grant CA240301 and startup funds provided by the University of Illinois Urbana-Champaign. We would also like to acknowledge BioRender for enabling the creation of the figures featured in this work.
Abbreviations:
- HGSOC
High grade serous ovarian cancer
- SCOUTs
secretory cell outgrowths
- STICs
serous tubal intraepithelial carcinomas
- TGFβ
Transforming Growth Factor β
- AID
activation-induced cytidine deaminase
- IGF2
insulin-like growth factor 2
- IL-8
interleukin 8
- IUD
intrauterine device
- CA-125
serum cancer antigen 125
- EMT
epithelial to mesenchymal transition
- TCGA
The Cancer Genome Atlas
- PAX8
paired box transcription factor 8
- HDACis
histone deacetylase inhibitors
- SDF1
stromal cell-derived factor 1
- P4
progesterone
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
CONFLICT OF INTEREST STATEMENT
The authors have no conflicts to disclose.
REFERENCES
- [1].Siegel RL, Miller KD, Jemal A, Cancer statistics, 2018, CA Cancer J Clin, 68 (2018) 7–30. [DOI] [PubMed] [Google Scholar]
- [2].N. A Howlader N, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA (eds)., SEER Cancer Statistics Review, National Cancer Institute; Bethesda, MD, 1975–2016, based on November 2018 SEER data submission, posted to the SEER web site, April 2019. [Google Scholar]
- [3].Bowtell DD, Bohm S, Ahmed AA, Aspuria PJ, Bast RC Jr., Beral V, Berek JS, Birrer MJ, Blagden S, Bookman MA, Brenton JD, Chiappinelli KB, Martins FC, Coukos G, Drapkin R, Edmondson R, Fotopoulou C, Gabra H, Galon J, Gourley C, Heong V, Huntsman DG, Iwanicki M, Karlan BY, Kaye A, Lengyel E, Levine DA, Lu KH, McNeish IA, Menon U, Narod SA, Nelson BH, Nephew KP, Pharoah P, Powell DJ Jr., Ramos P, Romero IL, Scott CL, Sood AK, Stronach EA, Balkwill FR, Rethinking ovarian cancer II: reducing mortality from high-grade serous ovarian cancer, Nat Rev Cancer, 15 (2015) 668–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Kurman RJ, Shih Ie M, The Dualistic Model of Ovarian Carcinogenesis: Revisited, Revised, and Expanded, Am J Pathol, 186 (2016) 733–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Karnezis AN, Cho KR, Gilks CB, Pearce CL, Huntsman DG, The disparate origins of ovarian cancers: pathogenesis and prevention strategies, Nat Rev Cancer, 17 (2017) 65–74. [DOI] [PubMed] [Google Scholar]
- [6].Kindelberger DW, Lee Y, Miron A, Hirsch MS, Feltmate C, Medeiros F, Callahan MJ, Garner EO, Gordon RW, Birch C, Berkowitz RS, Muto MG, Crum CP, Intraepithelial carcinoma of the fimbria and pelvic serous carcinoma: Evidence for a causal relationship, Am J Surg Pathol, 31 (2007) 161–169. [DOI] [PubMed] [Google Scholar]
- [7].Perets R, Drapkin R, It’s Totally Tubular….Riding The New Wave of Ovarian Cancer Research, Cancer Res, 76 (2016) 10–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Lee Y, Miron A, Drapkin R, Nucci MR, Medeiros F, Saleemuddin A, Garber J, Birch C, Mou H, Gordon RW, Cramer DW, McKeon FD, Crum CP, A candidate precursor to serous carcinoma that originates in the distal fallopian tube, J Pathol, 211 (2007) 26–35. [DOI] [PubMed] [Google Scholar]
- [9].Ducie J, Dao F, Considine M, Olvera N, Shaw PA, Kurman RJ, Shih IM, Soslow RA, Cope L, Levine DA, Molecular analysis of high-grade serous ovarian carcinoma with and without associated serous tubal intra-epithelial carcinoma, Nat Commun, 8 (2017) 990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Falconer H, Yin L, Gronberg H, Altman D, Ovarian cancer risk after salpingectomy: a nationwide population-based study, J Natl Cancer Inst, 107 (2015). [DOI] [PubMed] [Google Scholar]
- [11].Madsen C, Baandrup L, Dehlendorff C, Kjaer SK, Tubal ligation and salpingectomy and the risk of epithelial ovarian cancer and borderline ovarian tumors: a nationwide case-control study, Acta Obstet Gynecol Scand, 94 (2015) 86–94. [DOI] [PubMed] [Google Scholar]
- [12].Marquez RT, Baggerly KA, Patterson AP, Liu J, Broaddus R, Frumovitz M, Atkinson EN, Smith DI, Hartmann L, Fishman D, Berchuck A, Whitaker R, Gershenson DM, Mills GB, Bast RC Jr., Lu KH, Patterns of gene expression in different histotypes of epithelial ovarian cancer correlate with those in normal fallopian tube, endometrium, and colon, Clin Cancer Res, 11 (2005) 6116–6126. [DOI] [PubMed] [Google Scholar]
- [13].Chen EY, Mehra K, Mehrad M, Ning G, Miron A, Mutter GL, Monte N, Quade BJ, McKeon FD, Yassin Y, Xian W, Crum CP, Secretory cell outgrowth, PAX2 and serous carcinogenesis in the Fallopian tube, J Pathol, 222 (2010) 110–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Song H, Kwan SY, Izaguirre DI, Zu Z, Tsang YT, Tung CS, King ER, Mok SC, Gershenson DM, Wong KK, PAX2 Expression in Ovarian Cancer, Int J Mol Sci, 14 (2013) 6090–6105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Roh MH, Yassin Y, Miron A, Mehra KK, Mehrad M, Monte NM, Mutter GL, Nucci MR, Ning G, McKeon FD, Hirsch MS, Wa X, Crum CP, High-grade fimbrial-ovarian carcinomas are unified by altered p53, PTEN and PAX2 expression, Mod Pathol, 23 (2010) 1316–1324. [DOI] [PubMed] [Google Scholar]
- [16].Al-Hujaily EM, Tang Y, Yao DS, Carmona E, Garson K, Vanderhyden BC, Divergent Roles of PAX2 in the Etiology and Progression of Ovarian Cancer, Cancer Prev Res (Phila), 8 (2015) 1163–1173. [DOI] [PubMed] [Google Scholar]
- [17].Quick CM, Ning G, Bijron J, Laury A, Wei TS, Chen EY, Vargas SO, Betensky RA, McKeon FD, Xian W, Crum CP, PAX2-null secretory cell outgrowths in the oviduct and their relationship to pelvic serous cancer, Mod Pathol, 25 (2012) 449–455. [DOI] [PubMed] [Google Scholar]
- [18].Vang R, Levine DA, Soslow RA, Zaloudek C, Shih Ie M, Kurman RJ, Molecular Alterations of TP53 are a Defining Feature of Ovarian High-Grade Serous Carcinoma: A Rereview of Cases Lacking TP53 Mutations in The Cancer Genome Atlas Ovarian Study, Int J Gynecol Pathol, 35 (2016) 48–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Jarboe E, Folkins A, Nucci MR, Kindelberger D, Drapkin R, Miron A, Lee Y, Crum CP, Serous carcinogenesis in the fallopian tube: a descriptive classification, Int J Gynecol Pathol, 27 (2008) 1–9. [DOI] [PubMed] [Google Scholar]
- [20].Li HX, Lu ZH, Shen K, Cheng WJ, Malpica A, Zhang J, Wei JJ, Zhang ZH, Liu J, Advances in serous tubal intraepithelial carcinoma: correlation with high grade serous carcinoma and ovarian carcinogenesis, Int J Clin Exp Pathol, 7 (2014) 848–857. [PMC free article] [PubMed] [Google Scholar]
- [21].Labidi-Galy SI, Papp E, Hallberg D, Niknafs N, Adleff V, Noe M, Bhattacharya R, Novak M, Jones S, Phallen J, Hruban CA, Hirsch MS, Lin DI, Schwartz L, Maire CL, Tille JC, Bowden M, Ayhan A, Wood LD, Scharpf RB, Kurman R, Wang TL, Shih IM, Karchin R, Drapkin R, Velculescu VE, High grade serous ovarian carcinomas originate in the fallopian tube, Nat Commun, 8 (2017) 1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Emori MM, Drapkin R, The hormonal composition of follicular fluid and its implications for ovarian cancer pathogenesis, Reprod Biol Endocrinol, 12 (2014) 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Bahar-Shany K, Brand H, Sapoznik S, Jacob-Hirsch J, Yung Y, Korach J, Perri T, Cohen Y, Hourvitz A, Levanon K, Exposure of fallopian tube epithelium to follicular fluid mimics carcinogenic changes in precursor lesions of serous papillary carcinoma, Gynecol Oncol, 132 (2014) 322–327. [DOI] [PubMed] [Google Scholar]
- [24].Sapoznik S, Bahar-Shany K, Brand H, Pinto Y, Gabay O, Glick-Saar E, Dor C, Zadok O, Barshack I, Zundelevich A, Gal-Yam EN, Yung Y, Hourvitz A, Korach J, Beiner M, Jacob J, Levanon EY, Barak M, Aviel-Ronen S, Levanon K, Activation-Induced Cytidine Deaminase Links Ovulation-Induced Inflammation and Serous Carcinogenesis, Neoplasia, 18 (2016) 90–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Brachova P, Alvarez NS, Van Voorhis BJ, Christenson LK, Cytidine deaminase Apobec3a induction in fallopian epithelium after exposure to follicular fluid, Gynecol Oncol, 145 (2017) 577–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].King SM, Hilliard TS, Wu LY, Jaffe RC, Fazleabas AT, Burdette JE, The impact of ovulation on fallopian tube epithelial cells: evaluating three hypotheses connecting ovulation and serous ovarian cancer, Endocr Relat Cancer, 18 (2011) 627–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Liu Y, Qiang W, Xu X, Dong R, Karst AM, Liu Z, Kong B, Drapkin RI, Wei JJ, Role of miR-182 in response to oxidative stress in the cell fate of human fallopian tube epithelial cells, Oncotarget, 6 (2015) 38983–38998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Shigeta S, Toyoshima M, Kitatani K, Ishibashi M, Usui T, Yaegashi N, Transferrin facilitates the formation of DNA double-strand breaks via transferrin receptor 1: the possible involvement of transferrin in carcinogenesis of high-grade serous ovarian cancer, Oncogene, 35 (2016) 3577–3586. [DOI] [PubMed] [Google Scholar]
- [29].Huang HS, Hsu CF, Chu SC, Chen PC, Ding DC, Chang MY, Chu TY, Haemoglobin in pelvic fluid rescues Fallopian tube epithelial cells from reactive oxygen species stress and apoptosis, J Pathol, 240 (2016) 484–494. [DOI] [PubMed] [Google Scholar]
- [30].Rockfield S, Kee Y, Nanjundan M, Chronic iron exposure and c-Myc/H-ras-mediated transformation in fallopian tube cells alter the expression of EVI1, amplified at 3q26.2 in ovarian cancer, Oncogenesis, 8 (2019) 46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Hsu CF, Huang HS, Chen PC, Ding DC, Chu TY, IGF-axis confers transformation and regeneration of fallopian tube fimbria epithelium upon ovulation, EBioMedicine, 41 (2019) 597–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Dong Y, Li J, Han F, Chen H, Zhao X, Qin Q, Shi R, Liu J, High IGF2 expression is associated with poor clinical outcome in human ovarian cancer, Oncol Rep, 34 (2015) 936–942. [DOI] [PubMed] [Google Scholar]
- [33].Alwosaibai K, Abedini A, Al-Hujaily EM, Tang Y, Garson K, Collins O, Vanderhyden BC, PAX2 maintains the differentiation of mouse oviductal epithelium and inhibits the transition to a stem cell-like state, Oncotarget, 8 (2017) 76881–76897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].La Vecchia C, Ovarian cancer: epidemiology and risk factors, Eur J Cancer Prev, 26 (2017) 55–62. [DOI] [PubMed] [Google Scholar]
- [35].Madden T, Intrauterine contraception: Background and device types, 2019.
- [36].Wheeler LJ, Desanto K, Teal SB, Sheeder J, Guntupalli SR, Intrauterine Device Use and Ovarian Cancer Risk: A Systematic Review and Meta-analysis, Obstet Gynecol, 134 (2019) 791–800. [DOI] [PubMed] [Google Scholar]
- [37].Menezo Y, Guerin P, The mammalian oviduct: biochemistry and physiology, Eur J Obstet Gynecol Reprod Biol, 73 (1997) 99–104. [DOI] [PubMed] [Google Scholar]
- [38].Rodriguez C, Patel AV, Calle EE, Jacob EJ, Thun MJ, Estrogen replacement therapy and ovarian cancer mortality in a large prospective study of US women, JAMA, 285 (2001) 1460–1465. [DOI] [PubMed] [Google Scholar]
- [39].Lacey JV Jr., Mink PJ, Lubin JH, Sherman ME, Troisi R, Hartge P, Schatzkin A, Schairer C, Menopausal hormone replacement therapy and risk of ovarian cancer, JAMA, 288 (2002) 334–341. [DOI] [PubMed] [Google Scholar]
- [40].Ho SM, Estrogen, progesterone and epithelial ovarian cancer, Reprod Biol Endocrinol, 1 (2003) 73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Liehr JG, Is estradiol a genotoxic mutagenic carcinogen?, Endocr Rev, 21 (2000) 40–54. [DOI] [PubMed] [Google Scholar]
- [42].Cavalieri E, Frenkel K, Liehr JG, Rogan E, Roy D, Estrogens as endogenous genotoxic agents--DNA adducts and mutations, J Natl Cancer Inst Monogr, (2000) 75–93. [DOI] [PubMed] [Google Scholar]
- [43].Moyle-Heyrman G, Schipma MJ, Dean M, Davis DA, Burdette JE, Genome-wide transcriptional regulation of estrogen receptor targets in fallopian tube cells and the role of selective estrogen receptor modulators, J Ovarian Res, 9 (2016) 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Colina J, Varughese P, Karthikeyan S, Salvi A, Modi D, Burdette JE, Reduced PAX2 expression in murine fallopian tube cells enhances estrogen receptor signaling, Carcinogenesis, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Langdon SP, Gourley C, Gabra H, Stanley B, Endocrine therapy in epithelial ovarian cancer, Expert Rev Anticancer Ther, 17 (2017) 109–117. [DOI] [PubMed] [Google Scholar]
- [46].Paleari L, DeCensi A, Endocrine therapy in ovarian cancer: where do we stand?, Curr Opin Obstet Gynecol, 30 (2018) 17–22. [DOI] [PubMed] [Google Scholar]
- [47].Markman M, Bookman MA, Second-line treatment of ovarian cancer, Oncologist, 5 (2000) 26–35. [DOI] [PubMed] [Google Scholar]
- [48].Sieh W, Kobel M, Longacre TA, Bowtell DD, deFazio A, Goodman MT, Hogdall E, Deen S, Wentzensen N, Moysich KB, Brenton JD, Clarke BA, Menon U, Gilks CB, Kim A, Madore J, Fereday S, George J, Galletta L, Lurie G, Wilkens LR, Carney ME, Thompson PJ, Matsuno RK, Kjaer SK, Jensen A, Hogdall C, Kalli KR, Fridley BL, Keeney GL, Vierkant RA, Cunningham JM, Brinton LA, Yang HP, Sherman ME, Garcia-Closas M, Lissowska J, Odunsi K, Morrison C, Lele S, Bshara W, Sucheston L, Jimenez-Linan M, Driver K, Alsop J, Mack M, McGuire V, Rothstein JH, Rosen BP, Bernardini MQ, Mackay H, Oza A, Wozniak EL, Benjamin E, Gentry-Maharaj A, Gayther SA, Tinker AV, Prentice LM, Chow C, Anglesio MS, Johnatty SE, Chenevix-Trench G, Whittemore AS, Pharoah PD, Goode EL, Huntsman DG, Ramus SJ, Hormone-receptor expression and ovarian cancer survival: an Ovarian Tumor Tissue Analysis consortium study, Lancet Oncol, 14 (2013) 853–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Luo H, Li S, Zhao M, Sheng B, Zhu H, Zhu X, Prognostic value of progesterone receptor expression in ovarian cancer: a meta-analysis, Oncotarget, 8 (2017) 36845–36856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Faber MT, Jensen A, Frederiksen K, Glud E, Hogdall E, Hogdall C, Blaakaer J, Kjaer SK, Oral contraceptive use and impact of cumulative intake of estrogen and progestin on risk of ovarian cancer, Cancer Causes Control, 24 (2013) 2197–2206. [DOI] [PubMed] [Google Scholar]
- [51].Beral V, Million Women Study C, Bull D, Green J, Reeves G, Ovarian cancer and hormone replacement therapy in the Million Women Study, Lancet, 369 (2007) 1703–1710. [DOI] [PubMed] [Google Scholar]
- [52].Hankinson SE, Colditz GA, Hunter DJ, Spencer TL, Rosner B, Stampfer MJ, A quantitative assessment of oral contraceptive use and risk of ovarian cancer, Obstet Gynecol, 80 (1992) 708–714. [PubMed] [Google Scholar]
- [53].Adami HO, Hsieh CC, Lambe M, Trichopoulos D, Leon D, Persson I, Ekbom A, Janson PO, Parity, age at first childbirth, and risk of ovarian cancer, Lancet, 344 (1994) 1250–1254. [DOI] [PubMed] [Google Scholar]
- [54].Haridas D, Ponnusamy MP, Chugh S, Lakshmanan I, Seshacharyulu P, Batra SK, MUC16: molecular analysis and its functional implications in benign and malignant conditions, FASEB J, 28 (2014) 4183–4199. [DOI] [PubMed] [Google Scholar]
- [55].O’Shannessy DJ, Jackson SM, Twine NC, Hoffman BE, Dezso Z, Agoulnik SI, Somers EB, Gene expression analyses support fallopian tube epithelium as the cell of origin of epithelial ovarian cancer, Int J Mol Sci, 14 (2013) 13687–13703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Lowry KP, Lee SI, Imaging and Screening of Ovarian Cancer, Radiol Clin North Am, 55 (2017) 1251–1259. [DOI] [PubMed] [Google Scholar]
- [57].Gubbels JA, Felder M, Horibata S, Belisle JA, Kapur A, Holden H, Petrie S, Migneault M, Rancourt C, Connor JP, Patankar MS, MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells, Mol Cancer, 9 (2010) 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Felder M, Kapur A, Rakhmilevich AL, Qu X, Sondel PM, Gillies SD, Connor J, Patankar MS, MUC16 suppresses human and murine innate immune responses, Gynecol Oncol, 152 (2019) 618–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Theriault C, Pinard M, Comamala M, Migneault M, Beaudin J, Matte I, Boivin M, Piche A, Rancourt C, MUC16 (CA125) regulates epithelial ovarian cancer cell growth, tumorigenesis and metastasis, Gynecol Oncol, 121 (2011) 434–443. [DOI] [PubMed] [Google Scholar]
- [60].Boivin M, Lane D, Piche A, Rancourt C, CA125 (MUC16) tumor antigen selectively modulates the sensitivity of ovarian cancer cells to genotoxic drug-induced apoptosis, Gynecol Oncol, 115 (2009) 407–413. [DOI] [PubMed] [Google Scholar]
- [61].Xiang X, Feng M, Felder M, Connor JP, Man YG, Patankar MS, Ho M, HN125: A Novel Immunoadhesin Targeting MUC16 with Potential for Cancer Therapy, J Cancer, 2 (2011) 280–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Gordon AN, Schultes BC, Gallion H, Edwards R, Whiteside TL, Cermak JM, Nicodemus CF, CA125- and tumor-specific T-cell responses correlate with prolonged survival in oregovomab-treated recurrent ovarian cancer patients, Gynecol Oncol, 94 (2004) 340–351. [DOI] [PubMed] [Google Scholar]
- [63].Liu JF, Moore KN, Birrer MJ, Berlin S, Matulonis UA, Infante JR, Wolpin B, Poon KA, Firestein R, Xu J, Kahn R, Wang Y, Wood K, Darbonne WC, Lackner MR, Kelley SK, Lu X, Choi YJ, Maslyar D, Humke EW, Burris HA, Phase I study of safety and pharmacokinetics of the anti-MUC16 antibody-drug conjugate DMUC5754A in patients with platinum-resistant ovarian cancer or unresectable pancreatic cancer, Ann Oncol, 27 (2016) 2124–2130. [DOI] [PubMed] [Google Scholar]
- [64].Freeman ME, Kanyicska B, Lerant A, Nagy G, Prolactin: structure, function, and regulation of secretion, Physiol Rev, 80 (2000) 1523–1631. [DOI] [PubMed] [Google Scholar]
- [65].Schwarzler P, Untergasser G, Hermann M, Dirnhofer S, Abendstein B, Berger P, Prolactin gene expression and prolactin protein in premenopausal and postmenopausal human ovaries, Fertil Steril, 68 (1997) 696–701. [DOI] [PubMed] [Google Scholar]
- [66].Karthikeyan S, Russo A, Dean M, Lantvit DD, Endsley M, Burdette JE, Prolactin signaling drives tumorigenesis in human high grade serous ovarian cancer cells and in a spontaneous fallopian tube derived model, Cancer Lett, 433 (2018) 221–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Levina VV, Nolen B, Su Y, Godwin AK, Fishman D, Liu J, Mor G, Maxwell LG, Herberman RB, Szczepanski MJ, Szajnik ME, Gorelik E, Lokshin AE, Biological significance of prolactin in gynecologic cancers, Cancer Res, 69 (2009) 5226–5233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Clendenen TV, Arslan AA, Lokshin AE, Liu M, Lundin E, Koenig KL, Berrino F, Hallmans G, Idahl A, Krogh V, Lukanova A, Marrangoni A, Muti P, Nolen BM, Ohlson N, Shore RE, Sieri S, Zeleniuch-Jacquotte A, Circulating prolactin levels and risk of epithelial ovarian cancer, Cancer Causes Control, 24 (2013) 741–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Sundaram KM, Zhang Y, Mitra AK, Kouadio JK, Gwin K, Kossiakoff AA, Roman BB, Lengyel E, Piccirilli JA, Prolactin Receptor-Mediated Internalization of Imaging Agents Detects Epithelial Ovarian Cancer with Enhanced Sensitivity and Specificity, Cancer Res, 77 (2017) 1684–1696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Endsley MP, Moyle-Heyrman G, Karthikeyan S, Lantvit DD, Davis DA, Wei JJ, Burdette JE, Spontaneous Transformation of Murine Oviductal Epithelial Cells: A Model System to Investigate the Onset of Fallopian-Derived Tumors, Front Oncol, 5 (2015) 154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Tan D, Chen KE, Khoo T, Walker AM, Prolactin increases survival and migration of ovarian cancer cells: importance of prolactin receptor type and therapeutic potential of S179D and G129R receptor antagonists, Cancer Lett, 310 (2011) 101–108. [DOI] [PubMed] [Google Scholar]
- [72].Wen Y, Zand B, Ozpolat B, Szczepanski MJ, Lu C, Yuca E, Carroll AR, Alpay N, Bartholomeusz C, Tekedereli I, Kang Y, Rupaimoole R, Pecot CV, Dalton HJ, Hernandez A, Lokshin A, Lutgendorf SK, Liu J, Hittelman WN, Chen WY, Lopez-Berestein G, Szajnik M, Ueno NT, Coleman RL, Sood AK, Antagonism of tumoral prolactin receptor promotes autophagy-related cell death, Cell Rep, 7 (2014) 488–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Ozcan A, Shen SS, Hamilton C, Anjana K, Coffey D, Krishnan B, Truong LD, PAX 8 expression in non-neoplastic tissues, primary tumors, and metastatic tumors: a comprehensive immunohistochemical study, Mod Pathol, 24 (2011) 751–764. [DOI] [PubMed] [Google Scholar]
- [74].Di Palma T, Lucci V, de Cristofaro T, Filippone MG, Zannini M, A role for PAX8 in the tumorigenic phenotype of ovarian cancer cells, BMC Cancer, 14 (2014) 292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Cheung HW, Cowley GS, Weir BA, Boehm JS, Rusin S, Scott JA, East A, Ali LD, Lizotte PH, Wong TC, Jiang G, Hsiao J, Mermel CH, Getz G, Barretina J, Gopal S, Tamayo P, Gould J, Tsherniak A, Stransky N, Luo B, Ren Y, Drapkin R, Bhatia SN, Mesirov JP, Garraway LA, Meyerson M, Lander ES, Root DE, Hahn WC, Systematic investigation of genetic vulnerabilities across cancer cell lines reveals lineage-specific dependencies in ovarian cancer, Proc Natl Acad Sci U S A, 108 (2011) 12372–12377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Rodgers LH, E O.h., Young AN, Burdette JE, Loss of PAX8 in high-grade serous ovarian cancer reduces cell survival despite unique modes of action in the fallopian tube and ovarian surface epithelium, Oncotarget, 7 (2016) 32785–32795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Hardy LR, Pergande MR, Esparza K, Heath KN, Onyuksel H, Cologna SM, Burdette JE, Proteomic analysis reveals a role for PAX8 in peritoneal colonization of high grade serous ovarian cancer that can be targeted with micelle encapsulated thiostrepton, Oncogene, 38 (2019) 6003–6016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Shi K, Yin X, Cai MC, Yan Y, Jia C, Ma P, Zhang S, Zhang Z, Gu Z, Zhang M, Di W, Zhuang G, PAX8 regulon in human ovarian cancer links lineage dependency with epigenetic vulnerability to HDAC inhibitors, Elife, 8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Coffman LG, Burgos-Ojeda D, Wu R, Cho K, Bai S, Buckanovich RJ, New models of hematogenous ovarian cancer metastasis demonstrate preferential spread to the ovary and a requirement for the ovary for abdominal dissemination, Transl Res, 175 (2016) 92–102 e102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Yang-Hartwich Y, Gurrea-Soteras M, Sumi N, Joo WD, Holmberg JC, Craveiro V, Alvero AB, Mor G, Ovulation and extra-ovarian origin of ovarian cancer, Sci Rep, 4 (2014) 6116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Dean M, Jin V, Russo A, Lantvit DD, Burdette JE, Exposure of the extracellular matrix and colonization of the ovary in metastasis of fallopian-tube-derived cancer, Carcinogenesis, 40 (2019) 41–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Perets R, Wyant GA, Muto KW, Bijron JG, Poole BB, Chin KT, Chen JY, Ohman AW, Stepule CD, Kwak S, Karst AM, Hirsch MS, Setlur SR, Crum CP, Dinulescu DM, Drapkin R, Transformation of the fallopian tube secretory epithelium leads to high-grade serous ovarian cancer in Brca;Tp53;Pten models, Cancer Cell, 24 (2013) 751–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Zhang J, Zhang P, Shen Y, Yang M, Zou H, Liu H, Relationship of WNT4 Gene with the Risk of Epithelial Ovarian Cancer: A Han Chinese Population-Based Association Study, Genet Test Mol Biomarkers, 22 (2018) 686–692. [DOI] [PubMed] [Google Scholar]
- [84].Kuchenbaecker KB, Ramus SJ, Tyrer J, Lee A, Shen HC, Beesley J, Lawrenson K, McGuffog L, Healey S, Lee JM, Spindler TJ, Lin YG, Pejovic T, Bean Y, Li Q, Coetzee S, Hazelett D, Miron A, Southey M, Terry MB, Goldgar DE, Buys SS, Janavicius R, Dorfling CM, van Rensburg EJ, Neuhausen SL, Ding YC, Hansen TV, Jonson L, Gerdes AM, Ejlertsen B, Barrowdale D, Dennis J, Benitez J, Osorio A, Garcia MJ, Komenaka I, Weitzel JN, Ganschow P, Peterlongo P, Bernard L, Viel A, Bonanni B, Peissel B, Manoukian S, Radice P, Papi L, Ottini L, Fostira F, Konstantopoulou I, Garber J, Frost D, Perkins J, Platte R, Ellis S, Embrace Godwin AK, Schmutzler RK, Meindl A, Engel C, Sutter C, Sinilnikova OM, Collaborators GS, Damiola F, Mazoyer S, Stoppa-Lyonnet D, Claes K, De Leeneer K, Kirk J, Rodriguez GC, Piedmonte M, O’Malley DM, de la Hoya M, Caldes T, Aittomaki K, Nevanlinna H, Collee JM, Rookus MA, Oosterwijk JC, Breast Cancer Family R, Tihomirova L, Tung N, Hamann U, Isaccs C, Tischkowitz M, Imyanitov EN, Caligo MA, Campbell IG, Hogervorst FB, Hebon Olah E, Diez O, Blanco I, Brunet J, Lazaro C, Pujana MA, Jakubowska A, Gronwald J, Lubinski J, Sukiennicki G, Barkardottir RB, Plante M, Simard J, Soucy P, Montagna M, Tognazzo S, Teixeira MR, Investigators KC, Pankratz VS, Wang X, Lindor N, Szabo CI, Kauff N, Vijai J, Aghajanian CA, Pfeiler G, Berger A, Singer CF, Tea MK, Phelan CM, Greene MH, Mai PL, Rennert G, Mulligan AM, Tchatchou S, Andrulis IL, Glendon G, Toland AE, Jensen UB, Kruse TA, Thomassen M, Bojesen A, Zidan J, Friedman E, Laitman Y, Soller M, Liljegren A, Arver B, Einbeigi Z, Stenmark-Askmalm M, Olopade OI, Nussbaum RL, Rebbeck TR, Nathanson KL, Domchek SM, Lu KH, Karlan BY, Walsh C, Lester J, S. Australian Cancer, G. Australian Ovarian Cancer Study Hein A, Ekici AB, Beckmann MW, Fasching PA, Lambrechts D, Van Nieuwenhuysen E, Vergote I, Lambrechts S, Dicks E, Doherty JA, Wicklund KG, Rossing MA, Rudolph A, Chang-Claude J, Wang-Gohrke S, Eilber U, Moysich KB, Odunsi K, Sucheston L, Lele S, Wilkens LR, Goodman MT, Thompson PJ, Shvetsov YB, Runnebaum IB, Durst M, Hillemanns P, Dork T, Antonenkova N, Bogdanova N, Leminen A, Pelttari LM, Butzow R, Modugno F, Kelley JL, Edwards RP, Ness RB, du Bois A, Heitz F, Schwaab I, Harter P, Matsuo K, Hosono S, Orsulic S, Jensen A, Kjaer SK, Hogdall E, Hasmad HN, Azmi MA, Teo SH, Woo YL, Fridley BL, Goode EL, Cunningham JM, Vierkant RA, Bruinsma F, Giles GG, Liang D, Hildebrandt MA, Wu X, Levine DA, Bisogna M, Berchuck A, Iversen ES, Schildkraut JM, Concannon P, Weber RP, Cramer DW, Terry KL, Poole EM, Tworoger SS, Bandera EV, Orlow I, Olson SH, Krakstad C, Salvesen HB, Tangen IL, Bjorge L, van Altena AM, Aben KK, Kiemeney LA, Massuger LF, Kellar M, Brooks-Wilson A, Kelemen LE, Cook LS, Le ND, Cybulski C, Yang H, Lissowska J, Brinton LA, Wentzensen N, Hogdall C, Lundvall L, Nedergaard L, Baker H, Song H, Eccles D, McNeish I, Paul J, Carty K, Siddiqui N, Glasspool R, Whittemore AS, Rothstein JH, McGuire V, Sieh W, Ji BT, Zheng W, Shu XO, Gao YT, Rosen B, Risch HA, McLaughlin JR, Narod SA, Monteiro AN, Chen A, Lin HY, Permuth-Wey J, Sellers TA, Tsai YY, Chen Z, Ziogas A, Anton-Culver H, Gentry-Maharaj A, Menon U, Harrington P, Lee AW, Wu AH, Pearce CL, Coetzee G, Pike MC, Dansonka-Mieszkowska A, Timorek A, Rzepecka IK, Kupryjanczyk J, Freedman M, Noushmehr H, Easton DF, Offit K, Couch FJ, Gayther S, Pharoah PP, Antoniou AC, Chenevix-Trench G, Consortium B of Investigators of Modifiers of, Brca, Identification of six new susceptibility loci for invasive epithelial ovarian cancer, Nat Genet, 47 (2015) 164–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Eddie SL, Quartuccio SM, E O.h., Moyle-Heyrman G, Lantvit DD, Wei JJ, Vanderhyden BC, Burdette JE, Tumorigenesis and peritoneal colonization from fallopian tube epithelium, Oncotarget, 6 (2015) 20500–20512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Russo A, Czarnecki AA, Dean M, Modi DA, Lantvit DD, Hardy L, Baligod S, Davis DA, Wei JJ, Burdette JE, PTEN loss in the fallopian tube induces hyperplasia and ovarian tumor formation, Oncogene, 37 (2018) 1976–1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Kucia M, Jankowski K, Reca R, Wysoczynski M, Bandura L, Allendorf DJ, Zhang J, Ratajczak J, Ratajczak MZ, CXCR4-SDF-1 signalling, locomotion, chemotaxis and adhesion, J Mol Histol, 35 (2004) 233–245. [DOI] [PubMed] [Google Scholar]
- [88].Popple A, Durrant LG, Spendlove I, Rolland P, Scott IV, Deen S, Ramage JM, The chemokine, CXCL12, is an independent predictor of poor survival in ovarian cancer, Br J Cancer, 106 (2012) 1306–1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Jiang YP, Wu XH, Shi B, Wu WX, Yin GR, Expression of chemokine CXCL12 and its receptor CXCR4 in human epithelial ovarian cancer: an independent prognostic factor for tumor progression, Gynecol Oncol, 103 (2006) 226–233. [DOI] [PubMed] [Google Scholar]
- [90].Barbolina MV, Kim M, Liu Y, Shepard J, Belmadani A, Miller RJ, Shea LD, Stack MS, Microenvironmental regulation of chemokine (C-X-C-motif) receptor 4 in ovarian carcinoma, Mol Cancer Res, 8 (2010) 653–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Liu Y, Ren CC, Yang L, Xu YM, Chen YN, Role of CXCL12-CXCR4 axis in ovarian cancer metastasis and CXCL12-CXCR4 blockade with AMD3100 suppresses tumor cell migration and invasion in vitro, J Cell Physiol, 234 (2019) 3897–3909. [DOI] [PubMed] [Google Scholar]
- [92].Zheng N, Chen J, Liu W, Liu J, Li T, Chen H, Wang J, Jia L, Mifepristone inhibits ovarian cancer metastasis by intervening in SDF-1/CXCR4 chemokine axis, Oncotarget, 8 (2017) 59123–59135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Dean M, Davis DA, Burdette JE, Activin A stimulates migration of the fallopian tube epithelium, an origin of high-grade serous ovarian cancer, through non-canonical signaling, Cancer Lett, 391 (2017) 114–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Tao JJ, Cangemi NA, Makker V, Cadoo KA, Liu JF, Rasco DW, Navarro WH, Haqq CM, Hyman DM, First-in-Human Phase I Study of the Activin A Inhibitor, STM 434, in Patients with Granulosa Cell Ovarian Cancer and Other Advanced Solid Tumors, Clin Cancer Res, 25 (2019) 5458–5465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Zink KE, Dean M, Burdette JE, Sanchez LM, Imaging Mass Spectrometry Reveals Crosstalk between the Fallopian Tube and the Ovary that Drives Primary Metastasis of Ovarian Cancer, ACS Cent Sci, 4 (2018) 1360–1370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Gjyshi A, Dash S, Cen L, Cheng CH, Zhang C, Yoder SJ, Teer JK, Armaiz-Pena GN, Monteiro ANA, Early transcriptional response of human ovarian and fallopian tube surface epithelial cells to norepinephrine, Sci Rep, 8 (2018) 8291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Matsuo K, Lin YG, Roman LD, Sood AK, Overcoming platinum resistance in ovarian carcinoma, Expert Opin Investig Drugs, 19 (2010) 1339–1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Kang Y, Nagaraja AS, Armaiz-Pena GN, Dorniak PL, Hu W, Rupaimoole R, Liu T, Gharpure KM, Previs RA, Hansen JM, Rodriguez-Aguayo C, Ivan C, Ram P, Sehgal V, Lopez-Berestein G, Lutgendorf SK, Cole SW, Sood AK, Adrenergic Stimulation of DUSP1 Impairs Chemotherapy Response in Ovarian Cancer, Clin Cancer Res, 22 (2016) 1713–1724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Diaz ES, Karlan BY, Li AJ, Impact of beta blockers on epithelial ovarian cancer survival, Gynecol Oncol, 127 (2012) 375–378. [DOI] [PubMed] [Google Scholar]
- [100].Propranolol Hydrochloride and Chemotherapy in Treating Patients With Ovarian, Primary Peritoneal, or Fallopian Tube Cancer, M.D. Anderson Cancer Center.
- [101].Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N, The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data, Cancer Discov, 2 (2012) 401–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, Cerami E, Sander C, Schultz N, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal, Sci Signal, 6 (2013) pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Quartuccio SM, Karthikeyan S, Eddie SL, Lantvit DD, E O.h., Modi DA, Wei JJ, Burdette JE, Mutant p53 expression in fallopian tube epithelium drives cell migration, Int J Cancer, 137 (2015) 1528–1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Wu NY, Huang HS, Chao TH, Chou HM, Fang C, Qin CZ, Lin CY, Chu TY, Zhou HH, Progesterone Prevents High-Grade Serous Ovarian Cancer by Inducing Necroptosis of p53-Defective Fallopian Tube Epithelial Cells, Cell Rep, 18 (2017) 2557–2565. [DOI] [PubMed] [Google Scholar]
- [105].Karst AM, Jones PM, Vena N, Ligon AH, Liu JF, Hirsch MS, Etemadmoghadam D, Bowtell DD, Drapkin R, Cyclin E1 deregulation occurs early in secretory cell transformation to promote formation of fallopian tube-derived high-grade serous ovarian cancers, Cancer Res, 74 (2014) 1141–1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Loomans HA, Andl CD, Intertwining of Activin A and TGFbeta Signaling: Dual Roles in Cancer Progression and Cancer Cell Invasion, Cancers (Basel), 7 (2014) 70–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Principe DR, Doll JA, Bauer J, Jung B, Munshi HG, Bartholin L, Pasche B, Lee C, Grippo PJ, TGF-beta: duality of function between tumor prevention and carcinogenesis, J Natl Cancer Inst, 106 (2014) djt369. [DOI] [PMC free article] [PubMed] [Google Scholar]