Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jun 1.
Published in final edited form as: Pharmacol Ther. 2020 Mar 18;210:107524. doi: 10.1016/j.pharmthera.2020.107524

The tubal epigenome – an emerging target for ovarian cancer

Hunter D Reavis 1,2,3, Ronny Drapkin 1,2,4
PMCID: PMC7237289  NIHMSID: NIHMS1577264  PMID: 32197795

Abstract

Ovarian cancer is the most lethal gynecologic malignancy in the United States. The mortality of this disease is primarily attributed to challenges in early detection and therapeutic resistance. Recent studies indicate that the majority of high-grade serous ovarian carcinomas (HGSCs) originate from aberrant fallopian tube epithelial (FTE) cells. This shift in thinking about ovarian cancer pathogenesis has been met with an effort to identify the early genetic and epigenetic changes that underlie the transformation of normal FTE cells and prompt them to migrate and colonize the ovary, ultimately giving rise to aggressive HGSC. While identification of these early changes is important for biomarker discovery, the emergence of epigenetic alterations in FTE chromatin may also provide new opportunities for early detection, prevention, and therapeutic intervention. Here we provide a comprehensive overview of the current knowledge regarding early epigenetic reprogramming that precedes HGSC tumor development, the way that these alterations affect both intrinsic and extrinsic tumor properties, and how the epigenome may be targeted to thwart HGSC tumorigenesis.

Keywords: ovarian cancer, fallopian tube epithelium, STIC, H2Bub1, epigenetics

1. Introduction

Ovarian cancer is the most lethal gynecological malignancy in the United States. The term ‘ovarian cancer’ itself is a broad umbrella term describing a collection of heterogeneous neoplasms affecting the ovary. These neoplasms include germ-cell, sex cord-stromal, and epithelial tumors (Crum, Nucci, Haefner, & Peters)(Figure 1).

Figure 1.

Figure 1.

Ovarian cancer statistics. Ovarian cancer is a heterogeneous disease with epithelial tumors accounting for the majority of cases. The major histologic subtypes of epithelial ovarian carcinoma include serous, clear cell, mucinous and endometrioid tumors. High-grade serous carcinomas are the most common and are typically diagnosed after they have spread beyond the confines of the fallopian tube and ovary.

Most ovarian tumors are epithelial in nature, comprising approximately 90% of ovarian cancer diagnoses, with only 20% of these patients diagnosed at early disease stages ("Cancer Statistics Center: Ovarian Cancer," 2019). Women with invasive epithelial carcinomas are often symptomatic, but the early manifestations of the disease are non-specific and not pathognomonic for ovarian cancer. The lack of specific symptomatology accounts, in part, for the low rates of early detection, and the typical diagnosis at an advanced metastatic stage. The American Cancer Society estimates the 5-year survival rate for patients with metastatic epithelial ovarian cancer (EOC) is less than 30%. This poor survival rate yields a daunting death-to-case ratio, especially when compared with other female reproductive cancer types (International Agency for Research on Cancer, World Health Organization, Stewart, & Wild). With such a striking threat to women’s health, the pressing need to better understand disease pathogenesis, improve methods for early detection, and develop durable treatment options remains.

EOCs are stratified into four major histologic subtypes: clear cell, endometrioid, mucinous, and serous carcinomas (Figure 1). Both clear cell and endometrioid carcinomas are often associated with endometriosis, whereas the origin of mucinous ovarian cancers has been recently attributed to non-gynecological precursors (Cheasley, et al., 2019; Elias, et al., 2018; Lheureux, Gourley, Vergote, & Oza, 2019; Vaughan, et al., 2011).

Of the four subtypes of EOC, the majority are serous carcinomas that are further subdivided into high-grade or low-grade, depending on tumor dissemination and morphological characteristics. Low-grade serous carcinomas classified as ‘Type I’ carcinomas, are typically early stage tumors localized to the ovary and have more favorable clinical outcomes, accounting for only 10% of serous carcinoma deaths. High-grade serous ovarian carcinomas (Type II) are diagnosed at advanced stages and the highly proliferative nature of these disseminated tumors makes early diagnosis extremely challenging. Overall, high-grade serous carcinomas (HGSCs) are the most common form of epithelial ovarian carcinomas, comprising 75% of all EOC diagnoses (Karst & Drapkin, 2010; Kurman & Shih Ie, 2016; Lheureux, Gourley, et al., 2019).

The current standard-of-care for HGSC is cytoreductive surgery and treatment with a combination of taxanes and platinum-based chemotherapies. While patients tend to initially respond to this treatment regimen, many patients recur and develop chemoresistant tumors. Without a targeted treatment option, patients ultimately succumb to the disease (Agarwal & Kaye, 2003).

From a genomic standpoint, HGSC is primarily a copy number disease with few recurrent somatic mutations. In fact, the genomic landscape of HGSC displays more copy number alterations than any other tumor type studied to date and the only known recurrent somatic mutation in HGSC is in TP53, exhibited in nearly 100% of tumors (Cancer Genome Atlas Research, 2011; Vang, et al., 2016). Analyses of copy number alterations in HGSC revealed many common loci of chromosomal amplification or deletion. Moreover, the aneuploidy exhibited in these cells ultimately arises from a high degree of chromosomal and genomic instability associated with HGSC tumor cells. In fact, gene breakage events have been shown to inactivate tumor suppressor genes (TSGs), including RB1, NF1, RAD51B, and PTEN (Patch, et al., 2015). Approximately 50% of HGSCs are deficient in homologous recombination (HR) repair, with the most common defect being mutations in the BRCA1 or BRCA2 TSGs. Importantly, it is estimated that 15-20% of HGSC patients exhibit germline mutations in BRCA1/2 that may be detected prior to cancer development through germline testing (Alsop, et al., 2012; Domchek & Robson, 2019). Identification of defects in the HR pathway has been essential for screening germline mutations in high-risk women as well as for guiding therapies targeting acquired BRCA1/2 somatic mutations in HGSCs. These HR defects portend a vulnerability to DNA damaging agents, a heavily investigated therapeutic avenue; targeted therapeutics, particularly poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitors, have been widely utilized in the clinic to take advantage of HR-deficient ovarian cancers (Ledermann, 2019; Lheureux, Gourley, et al., 2019; Mirza, Pignata, & Ledermann, 2018). PARP inhibitors, of which there are now four FDA approved compounds, trap PARP molecules on DNA lesions and encourage double strand break repair via HR. In the absence of functional BRCA1/2, repair cannot be achieved, and the accumulated DNA damage forces the cells to undergo apoptosis. Much like the chemotherapeutic resistance exhibited by platinum and taxane treated tumors, acquired and de novo PARP inhibitor resistances challenge the efficacy of this alternative targeted therapy. With this in mind, many groups have set out to identify mechanisms of PARP inhibitor resensitization utilizing various techniques including whole genome CRISPR screens and microRNA classification (Choi, et al., 2016; Meghani, et al., 2018; Pettitt, et al., 2018; Zimmermann, et al., 2018). Identification of such vulnerabilities has prompted optimization of PARP inhibitor efficiency by either treating upfront with standard-of-care chemotherapy regimens or in combination with other drugs targeting the DNA repair pathway including ATR and CHK1 inhibitors (H. Kim, et al., 2017; Lheureux, Braunstein, & Oza, 2019; Parmar, et al., 2019; Yazinski, et al., 2017). While classic alterations like BRCA1/2 mutations currently identify patients that might respond to targeted therapy, the point at which these somatic aberrations are acquired during early tumor development is less well studied. Understanding the genesis of these alterations would provide an opportunity for risk-reduction and chemoprevention by targeting malignant cells before full tumor onset.

2. Origins of High-Grade Serous Carcinomas (HGSCs)

For many years, it was generally accepted that HGSCs were the product of genetic changes in the ovarian surface epithelium itself (Fathalla, 1971). However, more recent analysis of surrounding tissues revealed that this may not be the case for the majority of HGSCs. Shortly after the discovery of the BRCA1/2 genes in the mid-1990s, at-risk patients were offered risk-reducing surgery in the form of salpingo-oophorectomy for ovarian cancer prevention. As early as 2000-2001, studies emerged showing that these prophylactic specimens occasionally harbored abnormal cells in the fallopian tube rather than the ovary (Piek, et al., 2001; Zweemer, et al., 2000) (Figure 2). An understanding of these tubal lesions emerged with the development of the SEE-FIM (Sectioning and Extensively Examining the FIMbriated end) protocol to comprehensively examine the fallopian tube (Kindelberger, et al., 2007; Y. Lee, et al., 2007; Medeiros, et al., 2006). In otherwise healthy BRCA1/2 mutation carriers, the SEE-FIM protocol showed that approximately 5% of these women harbored an occult precursor to HGSC in the FTE, termed Serous Tubal Intraepithelial Carcinoma (STIC) (Medeiros, et al., 2006; Mingels, et al., 2012; Shaw, Rouzbahman, Pizer, Pintilie, & Begley, 2009; Wethington, et al., 2013). The subsequent evaluation of the fallopian tubes in patients with HGSC showed that up to 60% harbor precursor lesions in the fallopian tube, suggesting that preneoplastic events in the fallopian tube are the progenitors of HGSC in the majority of HGSC patients, regardless of BRCA1/2 mutation status (Chen, et al., 2017; Kindelberger, et al., 2007; Kroeger & Drapkin, 2017). In the setting of advanced disease, HGSC cells can colonize the FT and appear as a STIC but in reality are metastatic deposits (Chen, et al., 2017; Eckert, et al., 2016; Kroeger & Drapkin, 2017; Labidi-Galy, et al., 2017; McDaniel, et al., 2015). Altogether, this histopathological evidence ultimately prompted a change in the HGSC origin hypothesis to suggest that early aberrations in the epithelium of fallopian tube fimbriae may result in HGSC tumorigenesis.

Figure 2.

Figure 2.

Schematic for evolution of HGSC from malignant FTE precursor cells. Mutations in TP53 are a very early event in the pathogenesis of HGSC, occurring in benign-appearing secretory cells. These preneoplastic lesions are referred to as ‘p53 signatures.’ Acquisition of a neoplastic phenotype and proliferative capacity results in the development of serous tubal intraepithelial carcinoma (STIC). The question mark represents the unknown molecular changes associated with this transition. Invasion of the basement membrane and localized dissemination to the ovary and/or peritoneal cavity heralds the development of invasive HGSC and the associated clinical scenario.

One of the main markers of the link between the fallopian tube and ovarian tumors is the early loss of the universal marker of HGSC, TP53. Mutations in this TSG likely determines the FTE cell fate, as it becomes vulnerable to further genomic instability that will eventually give rise to STIC and the primary tumor. This concept of early TP53 loss of function was originally dubbed a ‘p53 signature’ when first observed as high p53 immunohistochemical staining of tubal secretory epithelial cells in BRCA mutant carrier patients (Y. Lee, et al., 2007). Following the mutation of TP53 in the fallopian tube, cells undergo malignant transformation to form dysplastic STIC lesions in the FT. Whether all TP53 mutations are capable of perpetuating subsequent aberrations is unclear (Tuna, et al., 2019). However, not all HGSCs have an identifiable STIC lesion in the FT (Chen, et al., 2017; Kroeger & Drapkin, 2017). While some animal studies suggest that HGSC can arise from the ovarian surface epithelium, we currently lack human data supporting the presence of precursors in the OSE (Flesken-Nikitin, et al., 2013; Zhang, et al., 2019). In addition to technical difficulties in identifying precursor lesions, which include sampling error, inter-observer variability, and consumption of the precursor by advanced disease (Carlson, et al., 2010; Mahe, et al., 2013; Rabban, Garg, Crawford, Chen, & Zaloudek, 2014; Vang, et al., 2012; Visvanathan, et al., 2011), a more recent concept suggests that TP53 mutant cells have undergone ‘precursor escape,’ whereby benign-appearing but TP53 mutated FT cells are shed to the ovary or peritoneum and undergo transformation at a later time (Soong, Kolin, Teschan, & Crum, 2018). In this scenario, transformation occurs at a non-tubal site but the cell of origin is still a TP53 mutated FT cell. Douglas Levine and colleagues recently tested the ‘precursor escape’ hypothesis by profiling HGSC with and without associated STIC lesions using copy number alterations and transcriptomics for RNA and microRNA expression. They argued that if HGSCs without STICs have a non-tubal origin then the expression profiles of those HGSCs should differ from those with a tubal origin. Analyses of the data from copy number alterations, messenger RNA sequencing and microRNA profiling failed to identify any significant differences between HGSCs with or without precursor STIC lesions (Ducie, et al., 2017). To infer the most probable site of origin, they calculated the similarity between HGSC and normal fallopian tube, normal ovary and normal peritoneum through molecular barcodes, and found that the most likely site of origin for all cases is indeed the distal fallopian tube. This is further supported by DNA methylome analyses showing that the methylomes of HGSC and FTE are significantly and consistently more highly conserved than are HGSC and OSE (Klinkebiel, Zhang, Akers, Odunsi, & Karpf, 2016).

Next generation DNA sequencing studies have provided further support for the tubal hypothesis. Using either macrodissection or laser-capture microdissection to precisely isolate the epithelial compartment of tubal precursor lesions, HGSC, and metastases to the ovary, omentum, and other structures in the peritoneum, whole-exome sequencing was combined with analyses for copy number variation and phylogeny to show that STICs are the precursor to HGSCs (Eckert, et al., 2016; Labidi-Galy, et al., 2017; McDaniel, et al., 2015; R. C. Wu, et al., 2019). Two studies included p53 signatures in their analyses and showed that mutations in TP53 are the earliest and only recurring event in this disease (Labidi-Galy 2017; Wu et al 2019). However, each patient also exhibited distinct mutations within the p53 signatures and STIC lesions. Loss of heterozygosity and copy number alterations, the hallmark of HGSC, were also detected in precursor lesions but to a lesser degree than the associated HGSC, indicating their earlier occurrence in the tumor evolutionary timeline. Importantly, both of these studies used mathematical modeling to estimate the time between the development of the earliest neoplastic clones in the FT and the development of ovarian and other metastatic lesions. They similarly concluded that the average time between a STIC and an ovarian tumor is 6-7 years. However, in patients with metastatic lesions, the time between the ovarian carcinoma and development of metastases is rapid, typically less than 2 years. Therefore, the window of opportunity for future studies aimed at prevention or early detection is between the emergence of a STIC and its colonization of the ovary.

Spontaneous HGSC induction in genetically engineered mouse models (GEMMs) has provided experimental support for tubal transformation in HGSC. With a Cre-recombinase system, it has become possible to knockout TSGs under the control of tissue specific promoters like Pax8 or Ovgp1. While mice lack the fallopian tube structures of the human female reproductive system, knockout of TSGs such as TP53, BRCA1/2, and/or PTEN in the most similar structure, the oviduct, gives rise to both STICs and serous ovarian carcinomas (J. Kim, et al., 2012; Morin & Weeraratna, 2016; Perets, et al., 2013; Sherman-Baust, et al., 2014; Stuckelberger & Drapkin, 2018; R. Wu, et al., 2016; Zhai, et al., 2017; Zhang, et al., 2019). Importantly, these GEMMs recapitulate the copy number alterations seen in human HGSCs (McCool, et al., 2019; Perets, et al., 2013).

3. Fallopian Tube Epithelial (FTE) Precursors and the Epigenome

In addition to recent studies defining the genetic evolution of HGSC from the FTE, mounting data suggests that epigenomic dysregulation also contributes to HGSC pathogenesis (Figure 3). Alterations in DNA methylation (particularly changes in CpG island methylation upstream of promoters), histone modifications (primarily methylation and acetylation marks previously associated with repression or activation of nearby transcription, respectively), and chromatin remodeling genes (mutations in writers, readers, and erasers of the post-translational modifications) ultimately regulate transcriptional activity and chromatin accessibility of the genome, and have thus garnered interest in understanding how epigenetic aberrations drive tumorigenesis (Bennett & Licht, 2018; Jones, Issa, & Baylin, 2016; Sen, Shah, Nativio, & Berger, 2016; Valencia & Kadoch, 2019).

Figure 3.

Figure 3.

Regulation of chromatin dynamics by epigenetic remodelers and the effects of chromatin accessibility on tumorigenic gene transcription. On the left, chromatin compaction can be regulated by removal of histone acetyl marks by deacetylating enzymes (HDACs), deposition of histone methyl marks by histone methyltransferases (KMTs), E3 ligase-mediated histone monoubiquitination, or direct methylation of DNA by DNA methyltransferases (DNMTs). Negative regulation of these processes, shown on the right, leads to chromatin relaxation, mediated by addition of acetyl marks by histone acetyltransferases (HATs), removal of monoubiquitin by deubiquitinases (DUBs), or removal of histone methyl marks by lysine demethylases (KDMs).

While particularly interesting in the setting of HGSC development from early FT precursors, this paradigm of epigenetic dysregulation driving tumorigenesis has been previously established in other disease states (Tsai & Baylin, 2011). For example, mutations in genes regulating DNA methylation, histone methylation, and histone acetylation are common in prostate tumors (Yegnasubramanian, De Marzo, & Nelson, 2019). These regulatory genes include epigenetic ‘writers’ that deposit the respective post-translational modification (PTM), ‘readers’ that catalyze downstream signaling in the presence of the PTM, and ‘erasers’ that return the site to its unmodified state. The crosstalk between epigenetic regulatory components ultimately allows for dynamic regulation of cellular signaling by manipulating transcription and localized condensation and relaxation of the chromatin. In the scope of prostate cancer, one of the most well-studied epigenetic phenomena is DNA hypermethylation where the balance between DNA methyltransferases and demethyltransferases becomes disrupted. This disruption leads to an increased frequency of methyl marks near promoter regions of important TSGs such as GSTP1, RASSF1A, and APC (Yegnasubramanian, et al., 2019). Accumulation of methyl marks at promoter CpG islands blocks transcription factor binding and has been associated with transcriptional repression of TSGs. Loss of tumor suppression enables the outgrowth of cancer precursor cells and prompts tumorigenesis. Understanding when these events occur during tumorigenesis and which key regulators become dysfunctional provides researchers with a new approach to therapeutics.

DNA methylation

The rise of detection methods for covalent DNA modification patterns has encouraged investigation of epigenomic similarities between FTE precursors and HGSC. DNA methylome analysis of patient samples from tumor tissue, FTE, and ovarian surface epithelium revealed that dominant changes in the HGSC epigenome most directly correspond to methylation patterns in the matched FTE (Klinkebiel, et al., 2016). In a larger-scale analysis of DNA methylation signatures, Pisanic et al. showed that there are indeed detectable hypermethylation loci at TUBB6, IRX2, and c17orf64 promoters that reliably differentiate malignant FTE from benign tissue (Pisanic, et al., 2018). Further analysis of the HGSC methylome has begun to identify how early FT epigenetic reprogramming ultimately drives changes that initiate characteristic genomic instability in BRCA1/2 mutant tumors (Bartlett, et al., 2016). Whether these altered or aberrant patterns harbor clinical utility for early detection or as biomarkers to strengthen risk-assessment panels is under investigation.

In addition to BRCA1/2 mutations, HR deficiency in HGSC patients has also been associated with BRCA1 promoter region hypermethylation in approximately 10% of cases (Baldwin, et al., 2000; Esteller, et al., 2000; Swisher, et al., 2017). In support of this idea, it has been shown that BRCA1 promoter hypermethylation can be detected in serum DNA of stage I HGSC patients (Ibanez de Caceres, et al., 2004). Much like BRCA1/2 mutant tumors, those with BRCA1 promoter hypermethylation are targetable with PARP inhibitor treatment. Conversely, BRCA1 promoter hypomethylation has been identified as a mechanism of resistance in PARPi-treated patients (Ter Brugge, et al., 2016). To support this idea, a number of studies have shown that DNA hypomethylation is a predominant feature in cancers, including HGSC (Ehrlich, 2002; Widschwendter, et al., 2004; Woloszynska-Read, et al., 2008). Significantly, a 2018 study has provided strong evidence that DNA hypomethylation, or more specifically loss of 5-hmC, may serve as a diagnostic and prognostic biomarker for HGSC (Tucker, et al., 2018). These findings of early detectable alterations in the epigenome are important not only for diagnostic and therapeutic purposes, but also to aid in understanding the preliminary events affecting downstream gene expression patterns seen in HGSC. While these studies identify readily detectable epigenetic alterations specific to HGSC, the search for the molecular mechanisms that are driving these early DNA methylation changes is still ongoing.

Based on the notion that promoter methylation transcriptionally inactivates TSGs early in tumorigenesis, DNA methyltransferases (DNMTs) have been targeted in the clinic. Originally approved for use in myelodysplastic syndrome, 5-azacytidine (AZA) and 5-aza-2’-deoxycytidine (decitabine) are DNMT inhibitors (DNMTi) that have shown promise in resensitization of platinum resistant ovarian cancers (Matei, et al., 2012). It is believed that this reduced promoter methylation corresponds to re-expression of genes controlling DNA damage repair and immune response and therefore inhibits the cancer cells ability to evade death in the presence of standard chemotherapy (Fang, et al., 2018; Stone, et al., 2017; Travers, et al., 2019; Wang, et al., 2015). Given that epigenetic remodeling events like DNA demethylation can enhance anti-tumor response to chemotherapy along with the fact that these events precede tumorigenesis, it is clear that epigenetic instability is a crucial vulnerability in ovarian cancer. However, due to the global nature of DNA methylation and the few catalytically active DNMTs in the human proteome, there is a concern that inhibition of these enzymes may be harmful in a monotherapy setting. While hypomethylation upstream of TSGs may inhibit the rise of cancer cells, consequent hypomethylation upstream of oncogenes may alternatively advance tumorigenesis.

Histone modifications

The human genome is composed of DNA wrapped tightly around octameric histone proteins to form nucleosomes of compact chromatin; changes in addition or removal of covalent PTMs on histones can change the polarity of neighboring chromatin and cause compaction or relaxation to regulate gene transcription. By this logic, understanding early epigenetic histone modifications that precede genetic aberrations but stimulate the development of HGSC is critical. These epigenetic modifications encompass a myriad of alterations including but not limited to, methylation, acetylation, phosphorylation, lactylation, and ubiquitination (Audia & Campbell, 2016; Dawson & Kouzarides, 2012; Zhao & Shilatifard, 2019).

In the context of HGSC development, we recently reported that there is a marked decrease in histone H2B monoubiquitination (H2Bub1) between wild-type FTE and associated STIC lesions, followed by its eventual absence in the primary HGSC (Hooda, et al., 2019). A mechanism for loss of H2Bub1 was originally reported in HeLa cells where it was demonstrated that knockdown of the E3 ubiquitin ligase, RNF20, is sufficient to suppress H2B monoubiquitination and selectively attenuate expression of TSGs such as TP53 (Shema, et al., 2008). Knocking down RNF20 in FT secretory cells by RNA interference leads to increased migration, clonogenicity, and anchorage-independent growth of the cells (Hooda et al. 2019). These malignant phenotypes are in part caused by an increase in immune cytokine secretion, including interleukin-6, and an increase in cell adhesion programs. Given that loss of H2Bub1 occurs in over 70% of HGSC cases, these findings emphasize the importance of epigenetic maintenance, especially H2Bub1, in ovarian cancer progression (Dickson, et al., 2016; Hooda, et al., 2019).

While RNF20 is believed to be the canonical E3 ligase that deposits ubiquitin on H2B at lysine 120, recent studies have proposed that BRCA1 may also be an important non-canonical E3 ligase responsible for the regulation of H2Bub1 (Dickson, et al., 2016). Identifying epigenetic regulators that are commonly altered in HGSC provides new therapeutic opportunities to overcome the challenges of treating advanced disease. Alternatively, targeting ubiquitin-ligase/de-ubiquitinase pathways involved in early epigenetic reprogramming of FTE may provide a new avenue for therapeutic intervention. For example, approximately 53% of HGSC patients exhibit copy number loss of the E3 ligase RNF20 (Cerami, et al., 2012; Gao, et al., 2013; Hooda, et al., 2019). While, it may be difficult to reverse the heterozygous loss of the E3 ligase, a more tangible therapeutic goal may instead be targeting of de-ubiquitinating enzymes (DUBs) whose amplification in HGSC may be further driving H2Bub1 loss during tumorigenesis. While the DUBs predominantly responsible for this phenomenon in FTE to HGSC remain unidentified, the prospect of DUB inhibition shows promise in other cancer contexts (Cheng, et al., 2019; Harrigan, Jacq, Martin, & Jackson, 2018; Poondla, Chandrasekaran, Kim, & Ramakrishna, 2019).

The rationale for targeting ubiquitin-specific histone modifications in HGSC stems from the recent success in targeting other chromatin remodelers. In ovarian clear cell carcinoma (OCCC), common changes in chromatin remodelers such as the overexpression of the histone methyltransferase, EZH2 exist. EZH2 has been most thoroughly investigated as a target in large B-cell lymphomas, and the success of small molecule inhibitors against EZH2 in this context has spurred interest in the ovarian cancer field. In addition to EZH2 overexpression, mutations in the ARID1A component of the SWI/SNF chromatin remodeling complex have been reported in almost 60% of OCCC patients (Bitler, et al., 2015). A screen of ARID1A-mutant OCCCs revealed significant single agent sensitivity to EZH2 inhibitors and these compounds can be exploited for in vitro and in vivo synthetic lethality in this setting. With such a large percentage of OCCC patients exhibiting mutations in chromatin remodeling proteins, strategic targeting of EZH2 can inhibit histone methylation events that silence TSGs which are important for OCCC progression. Similarly, the potential for use of EZH2 inhibitors has been investigated in high-grade tumors. Karakashev et al. investigated markers of EZH2 inhibitor sensitivity in HGSC and found that a subset of tumors marked by CARM1 overexpression also benefit from EZH2i monotherapy (Karakashev, et al., 2018). CARM1 actively silences EZH2 target genes and the introduction of EZH2 inhibition in these settings synergizes to suppress pro-tumorigenic effectors. Altogether, these results are striking in that they identify subpopulations of patients that may show particular benefit from epigenetic therapies.

As previously mentioned, one of the main concerns with the use of DNA methylation and histone modification-targeted therapeutics is “broad reprogramming” (Jones, et al., 2016). For example, most small molecule inhibitors targeting acetylation pathways have been aimed at histone deacetylating enzymes (HDACs); the goal has been to maintain anti-tumorigenic gene expression by preventing removal of chromatin-relaxing acetyl marks (H. J. Kim & Bae, 2011). However, due to the genome-wide nature of histone acetylation, monotherapy with HDAC inhibitors has seen little success. For this reason, focused efforts to identify drug combinations and specific patient populations that may benefit from HDAC inhibition continue (Batty, Malouf, & Issa, 2009). Bitler et al. found that ARID1A-mutant OCCC patients not only benefit from EZH2 inhibition but also HDAC6 inhibition (Bitler, et al., 2017). While these small molecule inhibitors have been heavily investigated in the clinic, it remains unclear how to best use them in the setting of HGSC. By identifying biomarkers for epigenetic vulnerabilities and honing in on localized regulation of the epigenome, off-target effects can be minimized, which will ultimately improve patient quality of life.

While the traditional approach for development of targeted therapies in HGSC has revolved around genetic defects, the emergence of early epigenetic changes in precursor cells provides an opportunity for new investigation. Early changes in chromatin dynamics suggest that prophylactic intervention could prevent high-risk precursors from progressing, and inhibit the milieu of pro-tumorigenic pathways regulated by this epigenetic reprogramming.

4. Role of the Early Epigenome in Sculpting the Tumor Microenvironment

With an established role for epigenetic reprogramming in epithelial precursor cells, there is an effort to identify how these changes may be functionally promoting tumorigenesis. It is known that there are discrete differences in DNA methylation and histone ubiquitination patterns in HGSC development, and it is of interest to identify other relevant alterations that nurture a pro-tumorigenic environment. Epigenetic changes that affect chromatin structure ultimately dictate accessibility of the genome necessary to preserve cellular homeostasis. When chromatin conformation becomes more euchromatic in regions rich in oncogenes, or more heterochromatic in regions rich in tumor suppressors, cell intrinsic and extrinsic properties are altered in favor of a pro-tumor microenvironment. A 2018 pan-cancer study reported a striking correlation between accessible chromatin and RNA expression, showing that euchromatic regions surrounding transcriptional start sites for oncogenes such as BCL2 and SRC are associated with increased expression levels in a variety of tissue types (Corces, et al., 2018). Selective preparation or ‘poising’ of chromatin through epigenetic dysregulation may affect downstream gene expression that can promote hallmarks of cancer including, but not limited to, migration and tumor-promoting inflammation (Hanahan & Weinberg, 2011).

The epithelial-to-mesenchymal transition (EMT) describes the shift of epithelial precursor cells to a more migratory and invasive phenotype. Post-translational histone modifications have recently been implicated in this pro-tumorigenic change from the ‘default’ epithelial program (Tam & Weinberg, 2013). EZH2 and SUZ12 are two components of the polycomb repressive complex 2 (PRC2)that have been identified as important players in promoting H3K27me3 methylation of promoters upstream of classic epithelial program genes including E-cadherin. Enhanced H3K27me3 methylation at these promoter sites effectively represses transcription of epithelial programs and may therefore be promoting the EMT seen in epithelial ovarian carcinomas. While H3K27me3 is considered to be a transcriptionally repressive post-translational modification, other methyl marks like H3K4me3 are considered to be transcriptionally activating. Silencing of epithelial gene programs may indeed drive the EMT in epithelial carcinomas, but enhanced transcriptional activity upstream of mesenchymal genes may bolster this effect. In a 2013 study, Chaffer et al. showed that the introduction of growth factors to poised chromatin containing both H3K27me3 and H3K4me3 marks at ZEB1 promoters shift transcription of this pro-mesenchymal gene toward a more active state by promoting preferential presence of the euchromatic H3K4me3 mark in breast cancer cells (Chaffer, et al., 2013). Altogether these data suggest an important role for epigenetic reprogramming and its modulation of tumorigenic properties.

Several recent studies have also begun to highlight the role of epigenetic alterations in cell autonomous promotion of an inflammatory microenvironment. As previously mentioned, HGSC development has been linked to the gradual loss of H2Bub1 (Figure 4). While the loss of this mark in early precursor cells affects several pro-tumorigenic pathways, one of particular interest includes the effect on chronic inflammation. Heterozygous knockout of the Rnf20 E3 ligase in a mouse model led to decreased H2Bub1 levels and secretion of proinflammatory cytokines, including interleukin-6 (IL-6), by nuclear factor κB (NF-κB). Concordantly, RNF20+/− mice are predisposed to acute and chronic colonic inflammation and inflammation-associated colorectal cancer, with excessive myeloid-derived suppressor cells (MDSCs) that may quench anti-tumoral T cell activity (Tarcic, et al., 2016). Similarly, in vitro analysis of FT cell lines with reduced H2Bub1 revealed increased IL-6 that enhanced migration of FT cells lacking H2Bub1. Increased secretion of this cytokine may favorably prime the fallopian tube microenvironment to give rise to both STIC lesions as well as eventual HGSCs (Hooda, et al., 2019). IL-6 mediates pancreatic cancer metastasis to the liver by encouraging pro-metastatic niche formation (J. W. Lee, et al., 2019). Therefore, it is likely that IL-6 secretion by transformed FTE cells similarly prepares the ovarian microenvironment for invasion (Reavis & Drapkin, 2019). Together, these findings highlight the importance of H2Bub1 in HGSC and other early epigenetic reprogramming events in pro-metastatic niche formation and warrant translational investigation for prophylactic inhibition of these events.

Figure 4.

Figure 4.

Morphology and immunohistochemical staining demonstrating gradual loss of H2Bub1 throughout HGSC development (A). H2Bub1 is dynamically regulated by positive effectors like the E3 ligase RNF20 and negative deubiquitinating effectors (B). Loss of H2Bub1 can have profound effects on tumor cell biology leading to increased migration, cell adhesion and inflammatory signals dominating in the absence of H2Bub1 (C).

5. Summary

While The Cancer Genome Atlas and other genomic studies have cataloged the landscape of ovarian cancer, there is an emerging appreciation that alterations in the epigenome also play a role in tumor development and progress. Matched DNA methylation signatures between HGSC patient FTE and the primary tumor have confirmed not only that FT cells are likely HGSC progenitors, but also that detrimental epigenetic changes occur early during tumorigenesis. DNA methylation can impair DNA damage repair by suppressing BRCA1 transcription, and the loss of H2Bub1 can trigger both cell migration and a proinflammatory tumor microenvironment. Identifying the key enzymes involved in these pathogenic processes can aid in the development of novel biomarkers for early detection as well as potential therapeutics that would help prevent ovarian cancer before it begins.

Acknowledgements

The authors thank members of the Drapkin lab for fruitful discussions. We thank Drs. Rugang Zhang, Patrice Morin, Paola Vermeer, and Peter Deraska for suggestions and critical reading of the manuscript. This work was supported by the National Cancer Institute P50 CA228991, the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation, The Honorable Tina Brozman ‘Tina’s Wish’ Foundation, The Claneil Foundation, and the Shooting for a Cure Foundation.

Abbreviations

BRCA

breast cancer susceptibility gene

EOC

epithelial ovarian cancer

FTE

fallopian tube epithelium

HGSC

high-grade serous ovarian carcinoma

HR

homologous recombination

OCCC

ovarian clear cell carcinoma

PARP

poly (ADP-ribose) polymerase

STIC

serous tubal intraepithelial carcinoma

TSG

tumor suppressor gene

Footnotes

Conflicts of Interest

R. Drapkin is a consultant/advisory board member for Repare Therapeutics, Mersana Therapeutics, and Siamab Therapeutics. No other authors declare potential conflicts of interest.

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.

Bibliography

  1. Agarwal R, & Kaye SB (2003). Ovarian cancer: strategies for overcoming resistance to chemotherapy. Nat Rev Cancer, 3, 502–516. [DOI] [PubMed] [Google Scholar]
  2. Alsop K, Fereday S, Meldrum C, deFazio A, Emmanuel C, George J, Dobrovic A, Birrer MJ, Webb PM, Stewart C, Friedlander M, Fox S, Bowtell D, & Mitchell G (2012). BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J Clin Oncol, 30, 2654–2663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Audia JE, & Campbell RM (2016). Histone Modifications and Cancer. Cold Spring Harb Perspect Biol, 8, a019521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Baldwin RL, Nemeth E, Tran H, Shvartsman H, Cass I, Narod S, & Karlan BY (2000). BRCA1 promoter region hypermethylation in ovarian carcinoma: a population-based study. Cancer Res, 60, 5329–5333. [PubMed] [Google Scholar]
  5. Bartlett TE, Chindera K, McDermott J, Breeze CE, Cooke WR, Jones A, Reisel D, Karegodar ST, Arora R, Beck S, Menon U, Dubeau L, & Widschwendter M (2016). Epigenetic reprogramming of fallopian tube fimbriae in BRCA mutation carriers defines early ovarian cancer evolution. Nat Commun, 7, 11620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Batty N, Malouf GG, & Issa JP (2009). Histone deacetylase inhibitors as anti-neoplastic agents. Cancer Lett, 280, 192–200. [DOI] [PubMed] [Google Scholar]
  7. Bennett RL, & Licht JD (2018). Targeting Epigenetics in Cancer. Annu Rev Pharmacol Toxicol, 58, 187–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bitler BG, Aird KM, Garipov A, Li H, Amatangelo M, Kossenkov AV, Schultz DC, Liu Q, Shih Ie M, Conejo-Garcia JR, Speicher DW, & Zhang R (2015). Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med, 21, 231–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bitler BG, Wu S, Park PH, Hai Y, Aird KM, Wang Y, Zhai Y, Kossenkov AV, Vara-Ailor A, Rauscher FJ III, Zou W, Speicher DW, Huntsman DG, Conejo-Garcia JR, Cho KR, Christianson DW, & Zhang R (2017). ARID1A-mutated ovarian cancers depend on HDAC6 activity. Nat Cell Biol, 19, 962–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Cancer Genome Atlas Research, N. (2011). Integrated genomic analyses of ovarian carcinoma. Nature, 474, 609–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Cancer Statistics Center: Ovarian Cancer. In. (2019). American Cancer Society. [Google Scholar]
  12. Carlson JW, Jarboe EA, Kindelberger D, Nucci MR, Hirsch MS, & Crum CP (2010). Serous tubal intraepithelial carcinoma: diagnostic reproducibility and its implications. Int J Gynecol Pathol, 29, 310–314. [DOI] [PubMed] [Google Scholar]
  13. 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 (2012). The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov, 2, 401–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Chaffer CL, Marjanovic ND, Lee T, Bell G, Kleer CG, Reinhardt F, D'Alessio AC, Young RA, & Weinberg RA (2013). Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell, 154, 61–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cheasley D, Wakefield MJ, Ryland GL, Allan PE, Alsop K, Amarasinghe KC, Ananda S, Anglesio MS, Au-Yeung G, Bohm M, Bowtell DDL, Brand A, Chenevix-Trench G, Christie M, Chiew YE, Churchman M, DeFazio A, Demeo R, Dudley R, Fairweather N, Fedele CG, Fereday S, Fox SB, Gilks CB, Gourley C, Hacker NF, Hadley AM, Hendley J, Ho GY, Hughes S, Hunstman DG, Hunter SM, Jobling TW, Kalli KR, Kaufmann SH, Kennedy CJ, Kobel M, Le Page C, Li J, Lupat R, McNally OM, McAlpine JN, Mes-Masson AM, Mileshkin L, Provencher DM, Pyman J, Rahimi K, Rowley SM, Salazar C, Samimi G, Saunders H, Semple T, Sharma R, Sharpe AJ, Stephens AN, Thio N, Torres MC, Traficante N, Xing Z, Zethoven M, Antill YC, Scott CL, Campbell IG, & Gorringe KL (2019). The molecular origin and taxonomy of mucinous ovarian carcinoma. Nat Commun, 10, 3935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chen F, Gaitskell K, Garcia MJ, Albukhari A, Tsaltas J, & Ahmed AA (2017). Serous tubal intraepithelial carcinomas associated with high-grade serous ovarian carcinomas: a systematic review. BJOG, 124, 872–878. [DOI] [PubMed] [Google Scholar]
  17. Cheng J, Guo J, North BJ, Wang B, Cui CP, Li H, Tao K, Zhang L, & Wei W (2019). Functional analysis of deubiquitylating enzymes in tumorigenesis and development. Biochim Biophys Acta Rev Cancer, 1872, 188312. [DOI] [PubMed] [Google Scholar]
  18. Choi YE, Meghani K, Brault ME, Leclerc L, He YJ, Day TA, Elias KM, Drapkin R, Weinstock DM, Dao F, Shih KK, Matulonis U, Levine DA, Konstantinopoulos PA, & Chowdhury D (2016). Platinum and PARP Inhibitor Resistance Due to Overexpression of MicroRNA-622 in BRCA1-Mutant Ovarian Cancer. Cell Rep, 14, 429–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Corces MR, Granja JM, Shams S, Louie BH, Seoane JA, Zhou W, Silva TC, Groeneveld C, Wong CK, Cho SW, Satpathy AT, Mumbach MR, Hoadley KA, Robertson AG, Sheffield NC, Felau I, Castro MAA, Berman BP, Staudt LM, Zenklusen JC, Laird PW, Curtis C, Cancer Genome Atlas Analysis, N., Greenleaf WJ, & Chang HY (2018). The chromatin accessibility landscape of primary human cancers. Science, 362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Crum CP, Nucci MR, Haefner HK, & Peters WA Diagnostic gynecologic and obstetric pathology. In (Third edition ed., pp. 1 online resource (pages)). [Google Scholar]
  21. Dawson MA, & Kouzarides T (2012). Cancer epigenetics: from mechanism to therapy. Cell, 150, 12–27. [DOI] [PubMed] [Google Scholar]
  22. Dickson KA, Cole AJ, Gill AJ, Clarkson A, Gard GB, Chou A, Kennedy CJ, Henderson BR, Australian Ovarian Cancer, S., Fereday S, Traficante N, Alsop K, Bowtell DD, deFazio A, Clifton-Bligh R, & Marsh DJ (2016). The RING finger domain E3 ubiquitin ligases BRCA1 and the RNF20/RNF40 complex in global loss of the chromatin mark histone H2B monoubiquitination (H2Bub1) in cell line models and primary high-grade serous ovarian cancer. Hum Mol Genet, 25, 5460–5471. [DOI] [PubMed] [Google Scholar]
  23. Domchek SM, & Robson ME (2019). Update on Genetic Testing in Gynecologic Cancer. J Clin Oncol, 37, 2501–2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Ducie J, Dao F, Considine M, Olvera N, Shaw PA, Kurman RJ, Shih IM, Soslow RA, Cope L, & Levine DA (2017). Molecular analysis of high-grade serous ovarian carcinoma with and without associated serous tubal intra-epithelial carcinoma. Nat Commun, 8, 990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Eckert MA, Pan S, Hernandez KM, Loth RM, Andrade J, Volchenboum SL, Faber P, Montag A, Lastra R, Peter ME, Yamada SD, & Lengyel E (2016). Genomics of Ovarian Cancer Progression Reveals Diverse Metastatic Trajectories Including Intraepithelial Metastasis to the Fallopian Tube. Cancer Discov, 6, 1342–1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Ehrlich M (2002). DNA methylation in cancer: too much, but also too little. Oncogene, 21, 5400–5413. [DOI] [PubMed] [Google Scholar]
  27. Elias KM, Tsantoulis P, Tille JC, Vitonis A, Doyle LA, Hornick JL, Kaya G, Barnes L, Cramer DW, Puppa G, Stuckelberger S, Hooda J, Dietrich PY, Goggins M, Kerr CL, Birrer M, Hirsch MS, Drapkin R, & Labidi-Galy SI (2018). Primordial germ cells as a potential shared cell of origin for mucinous cystic neoplasms of the pancreas and mucinous ovarian tumors. J Pathol, 246, 459–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Esteller M, Silva JM, Dominguez G, Bonilla F, Matias-Guiu X, Lerma E, Bussaglia E, Prat J, Harkes IC, Repasky EA, Gabrielson E, Schutte M, Baylin SB, & Herman JG (2000). Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst, 92, 564–569. [DOI] [PubMed] [Google Scholar]
  29. Fang F, Cardenas H, Huang H, Jiang G, Perkins SM, Zhang C, Keer HN, Liu Y, Nephew KP, & Matei D (2018). Genomic and Epigenomic Signatures in Ovarian Cancer Associated with Resensitization to Platinum Drugs. Cancer Res, 78, 631–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Fathalla MF (1971). Incessant ovulation--a factor in ovarian neoplasia? Lancet, 2, 163. [DOI] [PubMed] [Google Scholar]
  31. Flesken-Nikitin A, Hwang CI, Cheng CY, Michurina TV, Enikolopov G, & Nikitin AY (2013). Ovarian surface epithelium at the junction area contains a cancer-prone stem cell niche. Nature, 495, 241–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. 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 (2013). Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal, 6, pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Hanahan D, & Weinberg RA (2011). Hallmarks of cancer: the next generation. Cell, 144, 646–674. [DOI] [PubMed] [Google Scholar]
  34. Harrigan JA, Jacq X, Martin NM, & Jackson SP (2018). Deubiquitylating enzymes and drug discovery: emerging opportunities. Nat Rev Drug Discov, 17, 57–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Hooda J, Novak M, Salomon MP, Matsuba C, Ramos RI, MacDuffie E, Song M, Hirsch MS, Lester J, Parkash V, Karlan BY, Oren M, Hoon DS, & Drapkin R (2019). Early Loss of Histone H2B Monoubiquitylation Alters Chromatin Accessibility and Activates Key Immune Pathways That Facilitate Progression of Ovarian Cancer. Cancer Res, 79, 760–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Ibanez de Caceres I, Battagli C, Esteller M, Herman JG, Dulaimi E, Edelson MI, Bergman C, Ehya H, Eisenberg BL, & Cairns P (2004). Tumor cell-specific BRCA1 and RASSF1A hypermethylation in serum, plasma, and peritoneal fluid from ovarian cancer patients. Cancer Res, 64, 6476–6481. [DOI] [PubMed] [Google Scholar]
  37. International Agency for Research on Cancer, World Health Organization, Stewart BW, & Wild C World cancer report 2014. [Google Scholar]
  38. Jones PA, Issa JP, & Baylin S (2016). Targeting the cancer epigenome for therapy. Nat Rev Genet, 17, 630–641. [DOI] [PubMed] [Google Scholar]
  39. Karakashev S, Zhu H, Wu S, Yokoyama Y, Bitler BG, Park PH, Lee JH, Kossenkov AV, Gaonkar KS, Yan H, Drapkin R, Conejo-Garcia JR, Speicher DW, Ordog T, & Zhang R (2018). CARM1-expressing ovarian cancer depends on the histone methyltransferase EZH2 activity. Nat Commun, 9, 631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Karst AM, & Drapkin R (2010). Ovarian cancer pathogenesis: a model in evolution. J Oncol, 2010, 932371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Kim H, George E, Ragland R, Rafail S, Zhang R, Krepler C, Morgan M, Herlyn M, Brown E, & Simpkins F (2017). Targeting the ATR/CHK1 Axis with PARP Inhibition Results in Tumor Regression in BRCA-Mutant Ovarian Cancer Models. Clin Cancer Res, 23, 3097–3108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Kim HJ, & Bae SC (2011). Histone deacetylase inhibitors: molecular mechanisms of action and clinical trials as anti-cancer drugs. Am J Transl Res, 3, 166–179. [PMC free article] [PubMed] [Google Scholar]
  43. Kim J, Coffey DM, Creighton CJ, Yu Z, Hawkins SM, & Matzuk MM (2012). High-grade serous ovarian cancer arises from fallopian tube in a mouse model. Proc Natl Acad Sci U S A, 109, 3921–3926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. 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 (2007). Intraepithelial carcinoma of the fimbria and pelvic serous carcinoma: Evidence for a causal relationship. Am J Surg Pathol, 31, 161–169. [DOI] [PubMed] [Google Scholar]
  45. Klinkebiel D, Zhang W, Akers SN, Odunsi K, & Karpf AR (2016). DNA Methylome Analyses Implicate Fallopian Tube Epithelia as the Origin for High-Grade Serous Ovarian Cancer. Mol Cancer Res, 14, 787–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Kroeger PT Jr., & Drapkin R (2017). Pathogenesis and heterogeneity of ovarian cancer. Curr Opin Obstet Gynecol, 29, 26–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Kurman RJ, & Shih Ie M (2016). The Dualistic Model of Ovarian Carcinogenesis: Revisited, Revised, and Expanded. Am J Pathol, 186, 733–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. 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 (2017). High grade serous ovarian carcinomas originate in the fallopian tube. Nat Commun, 8, 1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Ledermann JA (2019). Extending the scope of PARP inhibitors in ovarian cancer. Lancet Oncol, 20, 470–472. [DOI] [PubMed] [Google Scholar]
  50. Lee JW, Stone ML, Porrett PM, Thomas SK, Komar CA, Li JH, Delman D, Graham K, Gladney WL, Hua X, Black TA, Chien AL, Majmundar KS, Thompson JC, Yee SS, O'Hara MH, Aggarwal C, Xin D, Shaked A, Gao M, Liu D, Borad MJ, Ramanathan RK, Carpenter EL, Ji A, de Beer MC, de Beer FC, Webb NR, & Beatty GL (2019). Hepatocytes direct the formation of a pro-metastatic niche in the liver. Nature, 567, 249–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. 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 (2007). A candidate precursor to serous carcinoma that originates in the distal fallopian tube. J Pathol, 211, 26–35. [DOI] [PubMed] [Google Scholar]
  52. Lheureux S, Braunstein M, & Oza AM (2019). Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA Cancer J Clin, 69, 280–304. [DOI] [PubMed] [Google Scholar]
  53. Lheureux S, Gourley C, Vergote I, & Oza AM (2019). Epithelial ovarian cancer. Lancet, 393, 1240–1253. [DOI] [PubMed] [Google Scholar]
  54. Mahe E, Tang S, Deb P, Sur M, Lytwyn A, & Daya D (2013). Do deeper sections increase the frequency of detection of serous tubal intraepithelial carcinoma (STIC) in the "sectioning and extensively examining the FIMbriated end" (SEE-FIM) protocol? Int J Gynecol Pathol, 32, 353–357. [DOI] [PubMed] [Google Scholar]
  55. Matei D, Fang F, Shen C, Schilder J, Arnold A, Zeng Y, Berry WA, Huang T, & Nephew KP (2012). Epigenetic resensitization to platinum in ovarian cancer. Cancer Res, 72, 2197–2205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. McCool K, Freeman ZT, Zhai Y, Wu R, Hu K, Liu CJ, Tomlins SA, Fearon ER, Magnuson B, Kuick R, & Cho KR (2019). Murine Oviductal High-Grade Serous Carcinomas Mirror the Genomic Alterations, Gene Expression Profiles, and Immune Microenvironment of Their Human Counterparts. Cancer Res. [DOI] [PMC free article] [PubMed]
  57. McDaniel AS, Stall JN, Hovelson DH, Cani AK, Liu CJ, Tomlins SA, & Cho KR (2015). Next-Generation Sequencing of Tubal Intraepithelial Carcinomas. JAMA Oncol, 1, 1128–1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Medeiros F, Muto MG, Lee Y, Elvin JA, Callahan MJ, Feltmate C, Garber JE, Cramer DW, & Crum CP (2006). The tubal fimbria is a preferred site for early adenocarcinoma in women with familial ovarian cancer syndrome. Am J Surg Pathol, 30, 230–236. [DOI] [PubMed] [Google Scholar]
  59. Meghani K, Fuchs W, Detappe A, Drane P, Gogola E, Rottenberg S, Jonkers J, Matulonis U, Swisher EM, Konstantinopoulos PA, & Chowdhury D (2018). Multifaceted Impact of MicroRNA 493-5p on Genome-Stabilizing Pathways Induces Platinum and PARP Inhibitor Resistance in BRCA2-Mutated Carcinomas. Cell Rep, 23, 100–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Mingels MJ, Roelofsen T, van der Laak JA, de Hullu JA, van Ham MA, Massuger LF, Bulten J, & Bol M (2012). Tubal epithelial lesions in salpingo-oophorectomy specimens of BRCA-mutation carriers and controls. Gynecol Oncol, 127, 88–93. [DOI] [PubMed] [Google Scholar]
  61. Mirza MR, Pignata S, & Ledermann JA (2018). Latest clinical evidence and further development of PARP inhibitors in ovarian cancer. Ann Oncol, 29, 1366–1376. [DOI] [PubMed] [Google Scholar]
  62. Morin PJ, & Weeraratna AT (2016). Genetically-defined ovarian cancer mouse models. J Pathol, 238, 180–184. [DOI] [PubMed] [Google Scholar]
  63. Parmar K, Kochupurakkal BS, Lazaro JB, Wang ZC, Palakurthi S, Kirschmeier PT, Yang C, Sambel LA, Farkkila A, Reznichenko E, Reavis HD, Dunn CE, Zou L, Do KT, Konstantinopoulos PA, Matulonis UA, Liu JF, D'Andrea AD, & Shapiro GI (2019). The CHK1 Inhibitor Prexasertib Exhibits Monotherapy Activity in High-Grade Serous Ovarian Cancer Models and Sensitizes to PARP Inhibition. Clin Cancer Res, 25, 6127–6140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Patch AM, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, Nones K, Cowin P, Alsop K, Bailey PJ, Kassahn KS, Newell F, Quinn MC, Kazakoff S, Quek K, Wilhelm-Benartzi C, Curry E, Leong HS, Australian Ovarian Cancer Study, G., Hamilton A, Mileshkin L, Au-Yeung G, Kennedy C, Hung J, Chiew YE, Harnett P, Friedlander M, Quinn M, Pyman J, Cordner S, O'Brien P, Leditschke J, Young G, Strachan K, Waring P, Azar W, Mitchell C, Traficante N, Hendley J, Thorne H, Shackleton M, Miller DK, Arnau GM, Tothill RW, Holloway TP, Semple T, Harliwong I, Nourse C, Nourbakhsh E, Manning S, Idrisoglu S, Bruxner TJ, Christ AN, Poudel B, Holmes O, Anderson M, Leonard C, Lonie A, Hall N, Wood S, Taylor DF, Xu Q, Fink JL, Waddell N, Drapkin R, Stronach E, Gabra H, Brown R, Jewell A, Nagaraj SH, Markham E, Wilson PJ, Ellul J, McNally O, Doyle MA, Vedururu R, Stewart C, Lengyel E, Pearson JV, Waddell N, deFazio A, Grimmond SM, & Bowtell DD (2015). Whole-genome characterization of chemoresistant ovarian cancer. Nature, 521, 489–494. [DOI] [PubMed] [Google Scholar]
  65. 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 (2013). Transformation of the fallopian tube secretory epithelium leads to high-grade serous ovarian cancer in Brca;Tp53;Pten models. Cancer Cell, 24, 751–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Pettitt SJ, Krastev DB, Brandsma I, Drean A, Song F, Aleksandrov R, Harrell MI, Menon M, Brough R, Campbell J, Frankum J, Ranes M, Pemberton HN, Rafiq R, Fenwick K, Swain A, Guettler S, Lee JM, Swisher EM, Stoynov S, Yusa K, Ashworth A, & Lord CJ (2018). Genome-wide and high-density CRISPR-Cas9 screens identify point mutations in PARP1 causing PARP inhibitor resistance. Nat Commun, 9, 1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Piek JM, van Diest PJ, Zweemer RP, Jansen JW, Poort-Keesom RJ, Menko FH, Gille JJ, Jongsma AP, Pals G, Kenemans P, & Verheijen RH (2001). Dysplastic changes in prophylactically removed Fallopian tubes of women predisposed to developing ovarian cancer. J Pathol, 195, 451–456. [DOI] [PubMed] [Google Scholar]
  68. Pisanic TR 2nd, Cope LM, Lin SF, Yen TT, Athamanolap P, Asaka R, Nakayama K, Fader AN, Wang TH, Shih IM, & Wang TL (2018). Methylomic Analysis of Ovarian Cancers Identifies Tumor-Specific Alterations Readily Detectable in Early Precursor Lesions. Clin Cancer Res, 24, 6536–6547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Poondla N, Chandrasekaran AP, Kim KS, & Ramakrishna S (2019). Deubiquitinating enzymes as cancer biomarkers: new therapeutic opportunities? BMB Rep, 52, 181–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Rabban JT, Garg K, Crawford B, Chen LM, & Zaloudek CJ (2014). Early detection of high-grade tubal serous carcinoma in women at low risk for hereditary breast and ovarian cancer syndrome by systematic examination of fallopian tubes incidentally removed during benign surgery. Am J Surg Pathol, 38, 729–742. [DOI] [PubMed] [Google Scholar]
  71. Reavis H, & Drapkin R (2019). H2Bub1: Guardian of chromatin accessibility in ovarian cancer. Oncoscience, 6, 349–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Sen P, Shah PP, Nativio R, & Berger SL (2016). Epigenetic Mechanisms of Longevity and Aging. Cell, 166, 822–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Shaw PA, Rouzbahman M, Pizer ES, Pintilie M, & Begley H (2009). Candidate serous cancer precursors in fallopian tube epithelium of BRCA1/2 mutation carriers. Mod Pathol, 22, 1133–1138. [DOI] [PubMed] [Google Scholar]
  74. Shema E, Tirosh I, Aylon Y, Huang J, Ye C, Moskovits N, Raver-Shapira N, Minsky N, Pirngruber J, Tarcic G, Hublarova P, Moyal L, Gana-Weisz M, Shiloh Y, Yarden Y, Johnsen SA, Vojtesek B, Berger SL, & Oren M (2008). The histone H2B-specific ubiquitin ligase RNF20/hBRE1 acts as a putative tumor suppressor through selective regulation of gene expression. Genes Dev, 22, 2664–2676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Sherman-Baust CA, Kuhn E, Valle BL, Shih Ie M, Kurman RJ, Wang TL, Amano T, Ko MS, Miyoshi I, Araki Y, Lehrmann E, Zhang Y, Becker KG, & Morin PJ (2014). A genetically engineered ovarian cancer mouse model based on fallopian tube transformation mimics human high-grade serous carcinoma development. J Pathol, 233, 228–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Soong TR, Kolin DL, Teschan NJ, & Crum CP (2018). Back to the Future? The Fallopian Tube, Precursor Escape and a Dualistic Model of High-Grade Serous Carcinogenesis. Cancers (Basel), 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Stone ML, Chiappinelli KB, Li H, Murphy LM, Travers ME, Topper MJ, Mathios D, Lim M, Shih IM, Wang TL, Hung CF, Bhargava V, Wiehagen KR, Cowley GS, Bachman KE, Strick R, Strissel PL, Baylin SB, & Zahnow CA (2017). Epigenetic therapy activates type I interferon signaling in murine ovarian cancer to reduce immunosuppression and tumor burden. Proc Natl Acad Sci U S A, 114, E10981–E10990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Stuckelberger S, & Drapkin R (2018). Precious GEMMs: emergence of faithful models for ovarian cancer research. J Pathol, 245, 129–131. [DOI] [PubMed] [Google Scholar]
  79. Swisher EM, Lin KK, Oza AM, Scott CL, Giordano H, Sun J, Konecny GE, Coleman RL, Tinker AV, O'Malley DM, Kristeleit RS, Ma L, Bell-McGuinn KM, Brenton JD, Cragun JM, Oaknin A, Ray-Coquard I, Harrell MI, Mann E, Kaufmann SH, Floquet A, Leary A, Harding TC, Goble S, Maloney L, Isaacson J, Allen AR, Rolfe L, Yelensky R, Raponi M, & McNeish IA (2017). Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol, 18, 75–87. [DOI] [PubMed] [Google Scholar]
  80. Tam WL, & Weinberg RA (2013). The epigenetics of epithelial-mesenchymal plasticity in cancer. Nat Med, 19, 1438–1449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Tarcic O, Pateras IS, Cooks T, Shema E, Kanterman J, Ashkenazi H, Boocholez H, Hubert A, Rotkopf R, Baniyash M, Pikarsky E, Gorgoulis VG, & Oren M (2016). RNF20 Links Histone H2B Ubiquitylation with Inflammation and Inflammation-Associated Cancer. Cell Rep, 14, 1462–1476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Ter Brugge P, Kristel P, van der Burg E, Boon U, de Maaker M, Lips E, Mulder L, de Ruiter J, Moutinho C, Gevensleben H, Marangoni E, Majewski I, Jozwiak K, Kloosterman W, van Roosmalen M, Duran K, Hogervorst F, Turner N, Esteller M, Cuppen E, Wesseling J, & Jonkers J (2016). Mechanisms of Therapy Resistance in Patient-Derived Xenograft Models of BRCA1-Deficient Breast Cancer. J Natl Cancer Inst, 108. [DOI] [PubMed] [Google Scholar]
  83. Travers M, Brown SM, Dunworth M, Holbert CE, Wiehagen KR, Bachman KE, Foley JR, Stone ML, Baylin SB, Casero RA Jr., & Zahnow CA (2019). DFMO and 5-Azacytidine Increase M1 Macrophages in the Tumor Microenvironment of Murine Ovarian Cancer. Cancer Res, 79, 3445–3454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Tsai HC, & Baylin SB (2011). Cancer epigenetics: linking basic biology to clinical medicine. Cell Res, 21, 502–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Tucker DW, Getchell CR, McCarthy ET, Ohman AW, Sasamoto N, Xu S, Ko JY, Gupta M, Shafrir A, Medina JE, Lee JJ, MacDonald LA, Malik A, Hasselblatt KT, Li W, Zhang H, Kaplan SJ, Murphy GF, Hirsch MS, Liu JF, Matulonis UA, Terry KL, Lian CG, & Dinulescu DM (2018). Epigenetic Reprogramming Strategies to Reverse Global Loss of 5-Hydroxymethylcytosine, a Prognostic Factor for Poor Survival in High-grade Serous Ovarian Cancer. Clin Cancer Res, 24, 1389–1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Tuna M, Ju Z, Yoshihara K, Amos CI, Tanyi JL, & Mills GB (2019). Clinical relevance of TP53 hotspot mutations in high-grade serous ovarian cancers. Br J Cancer. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Valencia AM, & Kadoch C (2019). Chromatin regulatory mechanisms and therapeutic opportunities in cancer. Nat Cell Biol, 21, 152–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Vang R, Levine DA, Soslow RA, Zaloudek C, Shih Ie M, & Kurman RJ (2016). 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, 48–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Vang R, Visvanathan K, Gross A, Maambo E, Gupta M, Kuhn E, Li RF, Ronnett BM, Seidman JD, Yemelyanova A, Shih Ie M, Shaw PA, Soslow RA, & Kurman RJ (2012). Validation of an algorithm for the diagnosis of serous tubal intraepithelial carcinoma. Int J Gynecol Pathol, 31, 243–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Vaughan S, Coward JI, Bast RC Jr., Berchuck A, Berek JS, Brenton JD, Coukos G, Crum CC, Drapkin R, Etemadmoghadam D, Friedlander M, Gabra H, Kaye SB, Lord CJ, Lengyel E, Levine DA, McNeish IA, Menon U, Mills GB, Nephew KP, Oza AM, Sood AK, Stronach EA, Walczak H, Bowtell DD, & Balkwill FR (2011). Rethinking ovarian cancer: recommendations for improving outcomes. Nat Rev Cancer, 11, 719–725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Visvanathan K, Vang R, Shaw P, Gross A, Soslow R, Parkash V, Shih Ie M, & Kurman RJ (2011). Diagnosis of serous tubal intraepithelial carcinoma based on morphologic and immunohistochemical features: a reproducibility study. Am J Surg Pathol, 35, 1766–1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Wang L, Amoozgar Z, Huang J, Saleh MH, Xing D, Orsulic S, & Goldberg MS (2015). Decitabine Enhances Lymphocyte Migration and Function and Synergizes with CTLA-4 Blockade in a Murine Ovarian Cancer Model. Cancer Immunol Res, 3, 1030–1041. [DOI] [PubMed] [Google Scholar]
  93. Wethington SL, Park KJ, Soslow RA, Kauff ND, Brown CL, Dao F, Otegbeye E, Sonoda Y, Abu-Rustum NR, Barakat RR, Levine DA, & Gardner GJ (2013). Clinical outcome of isolated serous tubal intraepithelial carcinomas (STIC). Int J Gynecol Cancer, 23, 1603–1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Widschwendter M, Jiang G, Woods C, Muller HM, Fiegl H, Goebel G, Marth C, Muller-Holzner E,, Zeimet AG, Laird PW, & Ehrlich M (2004). DNA hypomethylation and ovarian cancer biology. Cancer Res, 64, 4472–4480. [DOI] [PubMed] [Google Scholar]
  95. Woloszynska-Read A, Mhawech-Fauceglia P, Yu J, Odunsi K, & Karpf AR (2008). Intertumor and intratumor NY-ESO-1 expression heterogeneity is associated with promoter-specific and global DNA methylation status in ovarian cancer. Clin Cancer Res, 14, 3283–3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Wu R, Zhai Y, Kuick R, Karnezis AN, Garcia P, Naseem A, Hu TC, Fearon ER, & Cho KR (2016). Impact of oviductal versus ovarian epithelial cell of origin on ovarian endometrioid carcinoma phenotype in the mouse. J Pathol, 240, 341–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Wu RC, Wang P, Lin SF, Zhang M, Song Q, Chu T, Wang BG, Kurman RJ, Vang R, Kinzler K, Tomasetti C, Jiao Y, Shih IM, & Wang TL (2019). Genomic landscape and evolutionary trajectories of ovarian cancer precursor lesions. J Pathol, 248, 41–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Yazinski SA, Comaills V, Buisson R, Genois MM, Nguyen HD, Ho CK, Todorova Kwan T, Morris R, Lauffer S, Nussenzweig A, Ramaswamy S, Benes CH, Haber DA, Maheswaran S, Birrer MJ, & Zou L (2017). ATR inhibition disrupts rewired homologous recombination and fork protection pathways in PARP inhibitor-resistant BRCA-deficient cancer cells. Genes Dev, 31, 318–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Yegnasubramanian S, De Marzo AM, & Nelson WG (2019). Prostate Cancer Epigenetics: From Basic Mechanisms to Clinical Implications. Cold Spring Harb Perspect Med, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Zhai Y, Wu R, Kuick R, Sessine MS, Schulman S, Green M, Fearon ER, & Cho KR (2017). High-grade serous carcinomas arise in the mouse oviduct via defects linked to the human disease. J Pathol, 243, 16–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Zhang S, Dolgalev I, Zhang T, Ran H, Levine DA, & Neel BG (2019). Both fallopian tube and ovarian surface epithelium are cells-of-origin for high-grade serous ovarian carcinoma. Nat Commun, 10, 5367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Zhao Z, & Shilatifard A (2019). Epigenetic modifications of histones in cancer. Genome Biol, 20, 245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Zimmermann M, Murina O, Reijns MAM, Agathanggelou A, Challis R, Tarnauskaite Z, Muir M, Fluteau A, Aregger M, McEwan A, Yuan W, Clarke M, Lambros MB, Paneesha S, Moss P, Chandrashekhar M, Angers S, Moffat J, Brunton VG, Hart T, de Bono J, Stankovic T, Jackson AP, & Durocher D (2018). CRISPR screens identify genomic ribonucleotides as a source of PARP-trapping lesions. Nature, 559, 285–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Zweemer RP, van Diest PJ, Verheijen RH, Ryan A, Gille JJ, Sijmons RH, Jacobs IJ, Menko FH, & Kenemans P (2000). Molecular evidence linking primary cancer of the fallopian tube to BRCA1 germline mutations. Gynecol Oncol, 76, 45–50. [DOI] [PubMed] [Google Scholar]

RESOURCES