Next Article in Journal
Targeting Molecular Mechanisms of Obesity- and Type 2 Diabetes Mellitus-Induced Skeletal Muscle Atrophy with Nerve Growth Factor
Previous Article in Journal
The Effect of Enteric-Derived Lipopolysaccharides on Obesity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endometriosis-Associated Ovarian Cancer: From Molecular Pathologies to Clinical Relevance

by
Sophie Charlotte Steinbuch
1,
Anne-Marie Lüß
1,
Stephanie Eltrop
1,
Martin Götte
1,2 and
Ludwig Kiesel
1,*
1
Department of Obstetrics and Gynecology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
2
Cells-in-Motion Interfaculty Centre (CiMIC), University of Münster, 48149 Münster, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(8), 4306; https://doi.org/10.3390/ijms25084306
Submission received: 25 March 2024 / Revised: 8 April 2024 / Accepted: 11 April 2024 / Published: 13 April 2024

Abstract

:
Endometriosis is a chronic condition affecting reproductive-aged women, characterized by the growth of ectopic endometrial tissue. Despite being benign, endometriosis is associated with an increased risk of certain cancers, including endometriosis-associated ovarian cancer (EAOC). Ovarian cancer is rare, but more common in women with endometriosis, particularly endometrioid and clear-cell carcinomas. Factors such as hormonal imbalance, reproductive history, environmental exposures, and genetic predisposition contribute to the malignant transformation of endometriosis. Thus, understanding potential risk factors causing malignancy is crucial. Over the past few decades, various genetic mutations, microRNAs, as well as tumor microenvironmental factors have been identified, impacting pathways like PI3K/AKT/mTOR, DNA repair mechanisms, oxidative stress, and inflammation. Thus, this review aims to summarize molecular studies involved in EAOC pathogenesis as potential therapeutic targets. However, further research is needed to better understand the molecular and environmental factors driving EAOC development, to target the susceptibility of endometriotic lesions to malignant progression, and to identify effective therapeutic strategies.

1. Introduction

Endometriosis is a common, benign chronic disease in women of reproductive age, where endometrial tissue grows outside the uterine cavity in the entire abdominal and pelvic cavity. Since this ectopic tissue underlies the hormonal cycle with estrogen-induced proliferation, symptoms are associated with the menstrual cycle (dysmenorrhea, hypermenorrhea, menorrhagia) but also depend upon the localization (dysuria, dyspareunia, dyschezia). In addition to chronic pain, endometriosis is associated with infertility and an elevated risk for certain cancers including endometriosis-associated ovarian cancer (EAOC) [1,2].

1.1. Diagnosis

Generally, endometriosis is diagnosed based on typical signs and symptoms according to the European Society of Human Reproduction and Embryology (https://www.eshre.eu, accessed on 8 April 2024) [3], the American College of Obstetricians and Gynecologists (https://www.acog.org, accessed on 8 April 2024), or the Asian Society of Endometriosis and Adenomyosis (http://endometriosis-adenomyosis.asia, accessed on 8 April 2024).
Diagnostic measures include a clinical examination and imaging (ultrasound or magnet resonance imaging) but can be extended with a diagnostic laparoscopy to confirm the histology [3]. Diagnosing endometriosis with architectural atypia is crucial as it may indicate a precursor lesion of ovarian cancer, prompting pathologists to carefully examine surgical specimens to identify patients with hyperplasia-type endometriosis who could be at increased risk of developing endometriosis-associated ovarian cancer (EAOC) [4]. To distinguish between EAOC and benign endometriotic cysts or borderline tumors, the Sampson and Scott diagnostic criteria are used, including (1) the presence of endometriosis and cancer within the same ovary, (2) exhibiting a comparable histological structure to endometrial stroma, (3) while excluding the possibility of a metastatic tumor affecting the ovary, and (4) histopathological proofing of the progression from benign endometriosis to malignancy [5,6].

1.2. Risk Factors

Overall, the risk for developing ovarian cancer is low, with a cumulative risk of under 1% [7]. However, women with endometriosis are more likely to develop ovarian cancer [8], especially the endometrioid and clear-cell histotype [9,10]. Hereby, ovarian clear cell carcinomas (OCCCs) are per-definition high-grade ovarian carcinomas with a poor prognosis in advanced stages, whereas endometrioid ovarian carcinomas (EnOCs) can manifest as either low- or high-grade tumors [11]. However, there is no difference in the histological profiles of EAOC and ovarian cancer arising in endometriosis, indicating a comparable etiopathological mechanism [12].
Certain risk factors like a larger number of ovulatory cycles (early menarche/late menopause) [13], less pregnancies (infertility, nulliparity) [14], and a shorter breastfeeding duration [15] contribute to the malignant transformation of endometriosis into ovarian cancer [16]. Generally, a hormonal imbalance with an estrogen excess and progesterone deficit promotes the development of EOAC [17]. Thus, hormonal and reproductive factors such as the use of oral contraceptives [18,19], tubal ligation [20], polycystic ovarian syndrome (reviewed by Throwba et al. [21]), recreational physical activity [22], hormone therapy [23], and obesity [24,25,26] affect the risk for developing endometriosis and ovarian cancer.
Furthermore, endometrial cysts are a significant risk factor for developing cancer and thus should be carefully monitored [27]. However, the sonographic differentiation between benign endometrioma (“chocolate cysts”) and early-stage ovarian cancer remains challenging [11].
Besides hyperestrogenism and endometrial cysts, oxidative stress and inflammation are discussed as contributors to malignant transformation [17,28]. Therefore, other risk factors include exposure to environmental toxins, including dioxins [29,30], phthalates [31], alcohol [32] and coffee consumption [33], and smoking [34], as well as improper nutritional habits [35,36]. Finally, genetic predisposition has a large influence on the transition from benign endometriosis to malign ovarian cancer.

2. Molecular Pathologies

Almost one hundred years ago, John A. Sampson first described ectopic endometrium-like tissue as the cause of ovarian carcinoma [5] and concluded that the “Metastatic or Embolic Endometriosis, [is] due to the Menstrual Dissemination of Endometrial Tissue into the Venous Circulation” [37]. During the subsequent century, this concept of retrograde menstruation [38], followed by the implantation of desquamated endometrial cells in the peritoneal cavity and the sequential transformation of those endometriotic lesions in the ovarian endometrium into ovarian cancer through atypical endometriosis was further examined [39]. However, only 2% of women with laparoscopically confirmed endometriosis develop ovarian cancer [40]. While the exact etiopathology is still unknown, both endometrioid tissue intrinsic and microenvironmental factors are discussed as causal factors for survival in the peritoneum and malignant transformation [39].
For instance, the increased occurrence of chromosomal abnormalities observed in ovarian endometriosis compared to extragonadal endometriosis implies that the ovarian stromal environment may influence the initiation of genetic alterations, potentially progressing to invasive cancer [41].

2.1. Genetic Mutations

Hereby, several genetic mutations were identified as key players in the malignant conversion of endometriosis and manifestation of EAOC (Figure 1). In general, it is widely known that p53 and K-ras mutations and microsatellite instability promote tumor induction and growth, which is also the case for the malignant transformation of endometriosis [42,43,44]. Further common driver mutations in endometriosis include ARID1A, PIK3CA, and PPP2R1A [45,46]. However, BRCA mutations, commonly observed in ovarian carcinomas, are predominantly associated with serous ovarian cancer and are thus less frequent in EAOC [45,47,48].

2.1.1. Chromatin Remodeling

Comparing shared genetics in uterine endometria, endometriosis, and EAOC, almost half of clear-cell carcinomas and endometrioid carcinomas harbored mutations in the tumor-suppressor gene ARID1A (AT-rich interactive domain-containing protein 1A) [46], encoding BAF250a—a main component of the ATP-dependent chromatin remodeling complex SWI/SNF involved in the transcriptional activation of chromatin-repressed genes [49]. The loss of BAF250a in tissues of patients with EAOC was associated with the elevated expression of the phosphorylated H2A histone family member X γH2AX (S139), involved in chromatin remodeling and the repair of DNA double-strand breaks [50], as well as a higher expression of the pro-apoptotic regulators BIM (Bcl-2 Interacting Mediator of cell death) and BAX (Bcl-2-associated X protein), but lower expression of the anti-apoptotic gene Bcl-2 [51]. These findings indicate an initiation of DNA damage response and apoptosis pathways, potentially as a response to genomic instability at an early stage in precancerous lesions [51]. Furthermore, ARIDA1A shares some common downstream targets with p53, including CDKN1A and SMAD3 [52]. Thus, the loss of ARIDA1A function leads to the dysregulation of p53-controlled genes [52].

2.1.2. PI3K/AKT/mTOR Pathway

Among the subset of clear-cell EAOC, somatic mutations of the PIK3CA gene, encoding a catalytic subunit of phosphatidylinositol-3 kinases (PI3K), occur often as an early event [53,54], frequently coexisting with the loss of ARID1A protein expression [55], and may have syngeneic effects [56]. Other early signatures found in ARID1A-deficient carcinomas include the activation of the RAC (Rho family)-alpha serine/threonine-protein kinase via the increased expression of AKT1 and phosphorylation (pAKT) [49,51]. Furthermore, the differential expression of components involved in the mechanistic target of rapamycin (mTOR) pathway seem to connect endometriosis and the development of ovarian cancer [57].
A less common mutation in EOAC and ovarian clear-cell carcinoma (OCCC), detected in approximately 16–19% of OCCC cases [58,59], affects the function of the oncogene PPP2R1A, encoding a regulatory subunit of serine/threonine phosphatase 2 (PP2A) [60], which is a negative regulator of cell growth [61].
Altogether, the PI3K/AKT/mTOR pathway is a major regulator of the cell cycle; thus, alterations in gene regulation due to mutations contribute to the development and progression of ovarian cancer (reviewed by Aziz et al. [62]) as well as in the transformation process from a healthy endometrium to endometriosis and EAOC (reviewed by Driva et al. [63]). In contrast, the activity of the phosphatase and tensin homolog (PTEN), the counterpart of the PI3K/AKT pathway, was reduced due to PTEN silencing in EAOC, thus alleviating the negative control of PTEN on cell growth and division [64,65,66].

2.1.3. Other Genetic Alterations

Overall, the development of endometriosis-associated ovarian cancer is associated with genetic aberrations in multiple pathways, promoting malignant transformation and cancer cell growth. Hereby, the genetic profile differs between benign ovaries and ovarian endometriosis from EAOC and ovarian cancer [67]. Er et al. identified several more mutated genes belonging to the Wnt pathway, the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway, the Notch signaling pathway, cell cycle control, and the mismatch repair system via targeted next-generation sequencing [68]. Of note, the Notch signaling pathway is also dysregulated in endometriosis and has been mechanistically linked to its pathogenesis [69,70]. Other sequencing approaches highlighted the genomic heterogeneity and the diversity of cancer-associated mutations in ovarian endometriotic and normal uterine endometrial epithelium samples from patients [71]. Hereby, shared mutations among cases of EAOC displayed higher median variant allele frequencies, suggesting positive selection for clones with these mutations [71]. Therefore, the future whole-genome sequencing of EAOCs will certainly detect more differential expressed genes, whose relevance needs to be confirmed in consecutive experimental studies.

2.2. Epigenetic Reprogramming

In addition to genetic mutations, epigenetic mechanisms are involved in the malignant transformation of endometriosis to EAOC. For instance, the transcriptional inactivation of the MLH1 gene, encoding a DNA mismatch repair (MMR) protein, by promoter hypermethylation was reported [72], leading to microsatellite instability and the accumulation of spontaneous mutations, thus promoting progression towards EAOC. Further differentially methylated genes were identified as potential candidates in facilitating the malignant transformation, including the RASSF2 gene [73], encoding the KRAS-specific effector protein Ras association domain-containing protein 2, as well as the RUNX3 gene [74], which encodes the tumor suppressing Runt-related transcription factor 3.

2.3. The Tumor Microenvironment

2.3.1. Estrogen Concentration

In addition to tumor-intrinsic genetic factors, the tumor microenvironment is an emerging hallmark of cancer [75] and thus shapes EAOCs. One major player in the malignant transformation to EAOC is the estrogen concentration in the surrounding milieu, either exogenously provided by estrogen replacement therapy [76,77] or endogenously produced by the ovaries [78]. Hereby, high estrogen levels promote the proliferation of endometriotic cells via binding to the non-classical, G-protein-coupled estrogen receptor 1, followed by the activation of various signaling pathways such as the PI3K/AKT/mTOR [79], as reviewed by Kozieł et Piastowska-Ciesielska [80]. Furthermore, an imbalance between the expression levels of the classical estrogen receptors ER-alpha and ER-beta, favoring ER-alpha dominance due to the overexpression of ER-alpha [81] or loss of the anti-cancerous ER-beta [82,83,84], contributes to (ovarian) carcinogenesis [85]. While supporting the role of ER-beta as a tumor suppressor via the inhibition of ER-alpha-induced cell proliferation [86], some researchers assume a contrary shift from ER-alpha to ER-beta signaling during the estrogen-dependent progression of endometriosis to EAOC [87,88]. Altogether, the role of estrogen signaling in EAOC is more complex, especially considering the fact that the signaling is affected by the nutritional status [89], oxidative stress [90], and surrounding cells [91], but that estrogen vice versa shapes the cellular metabolism [92], the epithelial to mesenchymal transition [93], angiogenesis [94], and viability and invasiveness [95].
Further, estrogen influences epigenetic processes by regulating the DNA methyltransferase 1 (DNMT1)-dependent hypermethylation of the runt-related transcription factor 3 (RUNX3) in the malignant transformation of ovarian endometriosis [96].

2.3.2. microRNAs

Besides estrogen, microRNAs (miRNAs) are other important, post-transcriptional factors regulating the gene expression and thus are considered as “novel biomarkers” in endometriosis and EAOCs [97]. These small non-coding RNA molecules, typically consisting of 21 to 25 nucleotides, bind to complementary sequences in the messenger RNA and thus lead to RNA silencing by mRNA degradation or translational repression [98,99,100,101]. Over the past few decades, the complex dysregulation of microRNA expression has been observed in several human cancers [102], including differential expressed miRNAs in ovarian cancer [103,104], endometriosis [105,106], and EAOC [97,107].
In ovarian cancer, the miR-200 and lethal-7 families were the groups with the most significantly altered expression [108]. In this context, the let-7 family members were identified as cell cycle regulators as well as suppressors of oncogenes like the high-mobility group AT-hook 2 (HMGA-2), K-Ras, and Myc [108,109]. miRNA-200 is involved in inhibiting the epithelial-to-mesenchymal transition [110], while its downregulation induces tumor progression [111,112]. Notably, miR-200b also has a role in the pathogenesis of endometriosis, as it regulates the stem cell phenotype, proliferation, invasiveness, and growth of invasive protrusions of endometriotic cells by targeting ZEB1, ZEB2, and KLF4 [113,114].
Interestingly, Szubert et al. examined a decrease in the expression of miR-31-3p and miR-200b in cancerous lesions in contrast to normal ovarian tissue and endometriosis tissue [115]. miR-31 inhibits the tumor suppressor ARID1A, thus elevating the oncogenicity and stemness of head and neck squamous cell carcinoma [116]. Furthermore, miR-31 activates hypoxia-inducible factor under normoxic conditions by targeting the 3′untranslated region of the regulator FIH (factor-inhibiting hypoxia-inducible factor), leading to increased vascular endothelial growth factor (VEGF) production [117]. In turn, VEGF overexpression is associated with endometriosis [118] and the progression to EAOC [119]. Moreover, decreased levels of various other microRNAs such as miR-17-5p, miR-20a, miR-222, and miR-125a have been associated with angiogenesis in endometriosis through the regulation of runt-related transcription factor 1 (RUNX1), connective tissue growth factor (CTGF), thrombospondin-1 (TSP-1), or vascular endothelial growth factor-A (VEGF-A) expression [106,120,121].

2.3.3. Oxidative Stress

Recently, Marí-Alexandre et al. addressed the complex interaction between microRNAs and oxidative stress in a review, shedding light on their interplay during endometriosis, EAOC, and high-grade serous ovarian cancer (HGSOC) [39]. In short, microRNAs regulate the expression of reactive oxygen species (ROS)-producing or detoxifying enzymes, whereas oxidative stress induces the production of responsive microRNA via ROS-sensitive transcription factors [39]. However, oxidative stress itself also affects the gene expression of EAOC-related genes directly. For example, ARD1A expression is downregulated via oxidative stress, characterized by the enzymatic activity of manganese superoxide dismutase and by the formation of malondialdehyde due to ROS-dependent lipid degradation or peroxidation [122]. Mandai et al. even propose that persistent oxidative stress in endometriotic cysts is a causal contributor to their carcinogenic transformation [123]. A probable reason for these elevated ROS levels in the intra-cystic fluid within the ovarian endometrioma could result from the release of free iron during monthly bleeding via the Fenton reaction [39,124,125].

2.3.4. Inflammation

In addition to oxidative stress, inflammation promotes the carcinogenesis of EAOCs [123] by creating a pro-tumorigenic microenvironment characterized by DNA damage, tissue remodeling, immune suppression, angiogenesis, and epithelial–mesenchymal transition. In this context, several inflammatory cytokines [67], complement factors [126], inflammasome-related genes [127], as well as distinct immunologic and inflammatory signatures [91] were identified, indicating the relevance of the immune system in endometriotic cells and EAOCs. Recently, Linder et al. pointed out that genes related to the immune system are commonly mutated yet conserved between ovarian endometriosis and the subsequent development of ovarian carcinoma, suggesting that genetic alterations impacting immune response occur early and play a crucial role in these conditions [128].

2.3.5. Nutrient Availability

An emerging hallmark of cancer involves the tumor’s adaptation to the local nutrient availability through metabolic reprogramming [75,129]. On the one hand, endometriotic cells prefer aerobic glycolysis to generate energy, even in the presence of oxygen (called the “Warburg effect”) [130], facilitating their survival in extrauterine sites under oxidative and hypoxic stress [131]. On the other hand, recent studies demonstrated that oxidative phosphorylation (OXPHOS) is altered in ovarian cancer [132,133,134]. Overall, Dar et al. (2017) assume that ovarian cancer cells exhibit heterogeneity in energy metabolism by utilizing both glycolysis and OXPHOS to enhance their “cellular fitness” and chemoresistance as part of their survival strategy [135].
In general, cancer cell proliferation requires not only energy (ATP), but also the conversion of available nutrients into biomass [136]. Therefore, metabolite flexibility allows cancer cells to adapt to their environment [137], characterized by site-specific levels of glucose, lipids, and amino acids. However, subtypes of ovarian cancer display different metabolic preferences (as reviewed, for instance, in [133,138,139]), with no specific data available for EAOC.

2.4. The Complex and Heterogenous Nature of Cancer

To summarize, EAOC exemplifies the intricate and diverse landscape of cancer, marked by intra-tumoral, inter-tumoral, and inter-patient heterogeneity [128]. Within tumors, diverse cell populations coexist, exhibiting distinct genetic mutations, epigenetic alterations, and phenotypic characteristics. Inter-tumoral variability extends to differences between individual tumors, contributing to varied clinical presentations and responses to therapy. Moreover, the inter-patient tumor heterogeneity highlights the unique molecular signatures and disease trajectories observed among patients with EAOC. To conclude, this multifaceted nature of EAOC mirrors the complexity of understanding and effectively treating ovarian cancer, necessitating personalized approaches tailored to individual tumor characteristics. In particular, metastatic involvement in EAOC represents a complex clinical scenario associated with poor outcomes [140], thus requiring a multidisciplinary approach and targeted treatment strategies.

3. Therapeutic Strategies

3.1. Endometriosis

As a primary treatment, surgical removal is very common [11]—often through a laparoscopic approach—involving the ovaries (oophorectomy), fallopian tubes (salpingectomy), uterus (hysterectomy), and the surrounding tissue, as well as lymph nodes (lymphadenectomy). In patients with endometriosis-associated pain, surgical treatment combined with analgesics or hormone treatment (including combined hormonal contraceptives, progestogens, gonadotropin-releasing hormone (GnRH) agonist/antagonists, or aromatase inhibitors) is recommended (https://www.eshre.eu/Guideline/Endometriosis, accessed on 8 April 2024). Nevertheless, information is still lacking about the advantage of surgery (“unilateral salpingo-oophorectomy or cystectomy/partial ovarian excision”) compared to conservative surveillance on mortality from EAOC in patients with (asymptomatic) endometriosis [141].

3.2. Prevention of Cancer in Patients with Endometriosis

Younis states that, even if the likelihood of developing EAOC is nearly doubled in women with endometrioma, the overall lifetime risk remains minimal and that EAOC occurs more frequently in older women, with around one-third of EAOC cases diagnosed in the premenopausal stage (below 50 years), 2.10% cases in women under the age of 45, and only 0.017% of all cases in women below 40 years [142]. Thus, Younis suggests conservative management with a focus on transvaginal ultrasound to differentiate between benign, “homogenous cystic ‘ground glass’”-appearing endometrioma and EAOC [142]. Only EAOC-suspicious findings, usually presenting as a large, vascularized, papillose, unilateral cysts (>9 cm) with solid features, should be further characterized via T2-weighted magnetic resonance imaging [142,143].
Likewise, the ESHRE guideline concludes that “both diagnostic laparoscopy and imaging combined with empirical treatment (hormonal contraceptives or progestogens) can be considered in women suspected of endometriosis” [3]. Furthermore, the guideline development group highlights the importance of an individual case decision regarding long-term monitoring and empirical treatment based on specific risk factors [3,144]. In conclusion, the decision for the complete excision of all visible endometriotic lesions as a potential benefit to reduce the risk for developing EAOC should always be weighed against the risks of surgery like pain, morbidity, and the ovarian reserve [3]. Preserving the ovarian reserve is especially important in younger women due to reproductive benefits [145], but also due to the increased risk of coronary heart disease associated with premature hypoestrogenism [141,146]. Furthermore, hypoestrogenism leads to decreased bone mineral density, resulting in osteopenia and osteoporosis and subsequently an increased risk of pathological fractures [147].
Therefore, the use of combined oral contraceptives is discussed as an alternative early intervention in adolescents suffering from acute dysmenorrhea episodes—a potential sensitizer to chronic pelvic pain as well as an indicator for early-onset endometriosis [148]. In addition to alleviating the acute symptoms, reducing the amount of menstrual bleeding [149], and preventing the development of endometriosis [148], the long-term use of combined hormonal contraceptives decreases the risk of certain cancers, including ovarian cancer [150,151,152,153]. Overall, the long-term use of oral contraceptives might be a suitable preventor of EAOC by limiting disease progression while preserving future reproductive potential [148].

3.3. Treatment of Diagnosed Endometriosis-Associated Ovarian Cancer

A completely different therapeutic situation occurs in patients with confirmed endometriosis-associated ovarian carcinoma (Figure 2). Here, the therapy typically involves a multidisciplinary approach including surgery, chemotherapy, radiation therapy, targeted therapy, and/or hormonal therapy. However, the individual treatment plan depends on various factors such as the stage of cancer, the extent of spread, the patient’s overall health, and their fertility desires. Therefore, the therapy of patients with EAOC and other ovarian cancers differs from their histological subtype [154], namely, the high-grade serous ovarian carcinoma (HGSOC), low-grade serous carcinoma of the ovary (LGSOC), clear-cell ovarian carcinoma (CCOC), endometrioid ovarian carcinoma, mucinous carcinoma, and borderline ovarian tumors, as well as germ cell tumors and stromal tumors [155]. Nevertheless, primary ovarian cancer and EAOC with the same pathological type share a similar origin [12,156], and thus may result in comparable treatment strategies.

3.3.1. Surgical Treatment

Primary treatment for epithelial ovarian carcinoma and endometriosis-associated ovarian carcinoma typically involves initial debulking surgery followed by platinum-based chemotherapy [157]. Cytoreductive therapy in ovarian cancer usually aims for the complete resection of tumor masses, while the surgical management of EAOC may also involve removing endometriotic lesions along with any associated ovarian masses. Given the younger age of patients with EAOC, it is essential to prioritize preserving ovarian function whenever feasible, particularly in women who desire to have children.
In general, ovarian debulking surgery in endometriosis-associated ovarian cancer has a greater success rate than in non-EAOC patients [158]. However, a stage-matched analysis revealed no difference in the effectiveness of the standard cytoreductive surgery followed by taxane- and platinum-based chemotherapy in patients with EAOC and non-EAOC [8]. Therefore, the better prognosis of EAOC is more likely due to the early-stage, low-grade disease [8,159,160].

3.3.2. Chemotherapy

Besides surgery, adjuvant chemotherapy is a standard treatment option for ovarian cancer [161,162,163,164], including EAOC, particularly in advanced stages. Chemotherapeutic drugs commonly used in the treatment of ovarian cancer include platinum-based drugs like cisplatin and carboplatin, as well as taxanes such as paclitaxel [165,166,167,168,169]. However, the chemotherapy resistance of the ovarian clear-cell carcinoma subtype, also within EAOC, remains challenging and requires new therapeutic approaches.

3.3.3. Hormonal Therapies

Hormonotherapy is another adjuvant systemic treatment option in endometrioid ovarian cancer exhibiting hormonal receptor positivity [155,163]. Thus, the hormone-receptor status has an impact on the treatment [170], as is known for other gynecological tumors. For instance, high expression levels of progesterone receptor in endometrioid ovarian carcinoma were associated with a favorable outcome and thus could be potential targets for endocrine therapy [171]. On the contrary, the loss of estrogen receptor alpha immunoreactivity [172,173] or the high expression of estrogen receptor beta and gamma [174] decreased overall survival in ovarian cancer. Therefore, the tumor expression of the progesterone receptor and estrogen receptor are prognostic biomarkers in certain ovarian cancer subtypes [175] but could also have some predictive value as biomarkers predicting the response to endocrine therapies [176]. Several biomarker studies have been conducted in the past few years, using endocrine agents such as letrozole, tamoxifen, aromatase inhibitors, and fulvestrant in ovarian cancer, as reviewed by Langdon et al. [176]. In addition, estradiol–triazole analogs were developed using a molecular modeling approach to target several proteins in the epidermal growth factor receptor/mitogen-activated protein kinase pathway synergistically in ovarian cancer [177].

3.3.4. Targeted Therapies

To identify new therapeutic targets interfering with the underlying molecular pathology, several animal models were developed, including mice with a conditional doxycycline-induced Arid1a and/or Pten knockout [178], mice with endometrial autoimplantation and 7,12-Dimethylbenz[a]anthracene (DMBA) tumor induction [179].
In recent years, a functional precision oncology strategy was applied to identify patient specific drug regimens [154]. In this context, vascular endothelial growth factor (VEGF)/VEGFR pathway inhibitors such as bevacizumab [180,181,182] and poly(ADP-ribose) polymerase (PARP) inhibitors (like olaparib, niraparib, and rucaparib) are approved by the US Food and Drug Administration [183,184,185] and increasingly utilized in clinical settings for the treatment of ovarian cancer patients [186]. These anti-VEGF drugs interfere with tumor angiogenesis and normalize tumor vasculature [187], thus reducing tumor progression and enhancing drug susceptibility. Therefore, combining bevacizumab either with chemotherapy [180,181] or with PARP inhibitors [188,189,190] leads to synergistic effects. Interestingly, the differential expression of some microRNAs correlates to therapy response in ovarian cancer [191,192].
Especially patients with BRCA1/2-mutated tumors benefit from targeted therapies with PARP inhibitors due to the concept of synthetic lethality [193,194]: Tumor cells with a BRCA mutation lack the ability to repair DNA double-strand breaks via homologous recombination and are thus dependent on repairing DNA damage via the single-strand break (SSB) repair pathway [195]. By inhibiting the ribozyme PARP, mainly involved in SSB repair, tumor cells with BRCA mutations cannot repair DNA damage, leading to the accumulation of DNA damage and subsequently cell death without affecting normal cells [195].
To conclude, targeted therapies like PARP inhibitors are promising first-line treatment options in patients with BRCA-mutated ovarian cancer. However, further research and clinical trials in patients with EAOC are necessary to confirm the benefit of PARP inhibitors and bevacizumab in this clinical subgroup. In addition, targeted therapies using tyrosine-kinase inhibitors, MEK inhibitors, and PI3K/mTOR/Akt inhibitors are currently being tested in several ovarian cancer subtypes [155]. For example, the efficiency of the mTOR/AKT inhibitor temsirolimus in decreasing endometriotic cell proliferation was confirmed in a mouse model of deep-infiltrating endometriosis [196]. However, early clinical trials have shown that the efficacy of temsirolimus in treating ovarian cancer is limited [197], with no data available for patients with EAOC.

3.3.5. Immunotherapy

Immunotherapy is emerging as a promising approach in ovarian cancer treatment, with its role continually evolving. Checkpoint inhibitors targeting the programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) expression in EAOC are promising, as endometriosis-associated ovarian cancer expresses higher levels of PD-1/PD-L1 compared to benign lesions [198]. Additionally, the tumor mutation burden might predict the response to immunotherapy in EAOC [199], making it a potential biomarker for therapy susceptibility, as demonstrated in other tumors [200,201].
Using in vitro and in vivo models, Hsieh et al. revealed that the ARID1A-induced expression of HDAC6, resulting in IL-10 release and the subsequent M2 polarization of macrophages, is a new therapeutic target addressed by the HDAC inhibitor vorinostat [202].

4. Conclusions

In conclusion, the understanding of the molecular pathways and genetic aberrations involved in the development of endometriosis-associated ovarian cancer (EAOC) has significantly advanced, shedding light on potential therapeutic targets. Furthermore, the emerging role of the tumor microenvironment, estrogen signaling, oxidative stress, and inflammation highlights the complexity of EAOC pathogenesis, paving the way for novel therapeutic strategies including targeted therapies and immunotherapies.
In summary, while there may be similarities in the treatment approach for ovarian cancer and endometriosis-associated ovarian cancer, there are also important differences in terms of surgical considerations, chemotherapy regimens, and the potential role of hormonal therapy, reflecting the distinct etiology and characteristics of these diseases.

Author Contributions

Literature research, visualization, and writing—original draft preparation, S.C.S.; literature research—A.-M.L.; supervision and writing—review and editing, L.K.; review and editing—M.G. and S.E. All authors have read and agreed to the published version of the manuscript.

Funding

We acknowledge funding by the Open Access Publishing Fund of Münster University.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Buis, C.C.; van Leeuwen, F.E.; Mooij, T.M.; Burger, C.W.; OMEGA_ProjectGroup. Increased risk for ovarian cancer and borderline ovarian tumours in subfertile women with endometriosis. Hum. Reprod. 2013, 28, 3358–3369. [Google Scholar] [CrossRef] [PubMed]
  2. Al-Badawi, I.A.; Abu-Zaid, A.; Alomar, O.; Alsabban, M.; Alsehaimi, S.O.; Alqarni, S.M.S.; Alabdrabalamir, S.N.; Baradwan, S.; Al Baalharith, M.; AlOdaini, A.A.; et al. Association between Endometriosis and the Risk of Ovarian, Endometrial, Cervical, and Breast Cancer: A Population-Based Study from the U.S. National Inpatient Sample 2016–2019. Curr. Oncol. 2024, 31, 472–481. [Google Scholar] [CrossRef] [PubMed]
  3. Becker, C.M.; Bokor, A.; Heikinheimo, O.; Horne, A.; Jansen, F.; Kiesel, L.; King, K.; Kvaskoff, M.; Nap, A.; Petersen, K.; et al. ESHRE guideline: Endometriosis. Hum. Reprod. Open 2022, 2022, hoac009. [Google Scholar] [CrossRef]
  4. Ñiguez Sevilla, I.; Machado Linde, F.; Marín Sánchez, M.D.P.; Arense, J.J.; Torroba, A.; Nieto Díaz, A.; Sánchez Ferrer, M.L. Prognostic importance of atypical endometriosis with architectural hyperplasia versus cytologic atypia in endometriosis-associated ovarian cancer. J. Gynecol. Oncol. 2019, 30, e63. [Google Scholar] [CrossRef] [PubMed]
  5. Sampson, J. Endometrial carcinoma of the ovary, arising in endometrial tissue in that organ. Arch Surg. 1925, 10, 1–72. [Google Scholar] [CrossRef]
  6. Scott, R. Malignant changes in endometriosis. Obstet. Gynecol. 1953, 2, 283–289. Available online: https://journals.lww.com/greenjournal/citation/1953/09000/malignant_changes_in_endometriosis.8.aspx (accessed on 8 April 2024).
  7. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  8. Kim, H.S.; Kim, T.H.; Chung, H.H.; Song, Y.S. Risk and prognosis of ovarian cancer in women with endometriosis: A meta-analysis. Br. J. Cancer 2014, 110, 1878–1890. [Google Scholar] [CrossRef] [PubMed]
  9. Heidemann, L.N.; Hartwell, D.; Heidemann, C.H.; Jochumsen, K.M. The relation between endometriosis and ovarian cancer—A review. Acta Obstet. Gynecol. Scand. 2014, 93, 20–31. [Google Scholar] [CrossRef]
  10. Bergamini, A.; Mangili, G.; Ambrosi, A.; Taccagni, G.; Rabaiotti, E.; Bocciolone, L.; Candotti, G.; Cioffi, R.; Pella, F.; Sabetta, G.; et al. Endometriosis-Related Ovarian Cancers: Evidence for a Dichotomy in the Histogenesis of the Two Associated Histotypes. Diagnostics 2023, 13, 1425. [Google Scholar] [CrossRef]
  11. Samartzis, E.P.; Labidi-Galy, S.I.; Moschetta, M.; Uccello, M.; Kalaitzopoulos, D.R.; Perez-Fidalgo, J.A.; Boussios, S. Endometriosis-associated ovarian carcinomas: Insights into pathogenesis, diagnostics, and therapeutic targets-a narrative review. Ann. Transl. Med. 2020, 8, 1712. [Google Scholar] [CrossRef] [PubMed]
  12. Chiaffarino, F.; Cipriani, S.; Ricci, E.; Esposito, G.; Parazzini, F.; Vercellini, P. Histologic Subtypes in Endometriosis-Associated Ovarian Cancer and Ovarian Cancer Arising in Endometriosis: A Systematic Review and Meta-Analysis. Reprod. Sci. 2024; advance online publication. [Google Scholar] [CrossRef]
  13. Nnoaham, K.E.; Webster, P.; Kumbang, J.; Kennedy, S.H.; Zondervan, K.T. Is early age at menarche a risk factor for endometriosis? A systematic review and meta-analysis of case-control studies. Fertil. Steril. 2012, 98, 702–712.e6. [Google Scholar] [CrossRef] [PubMed]
  14. Stewart, L.M.; Holman, C.D.; Aboagye-Sarfo, P.; Finn, J.C.; Preen, D.B.; Hart, R. In vitro fertilization, endometriosis, nulliparity and ovarian cancer risk. Gynecol. Oncol. 2013, 128, 260–264. [Google Scholar] [CrossRef] [PubMed]
  15. Vercellini, P.; Bandini, V.; Viganò, P.; Di Stefano, G.; Merli, C.E.M.; Somigliana, E. Proposal for targeted, neo-evolutionary-oriented, secondary prevention of early-onset endometriosis and adenomyosis. Part I: Pathogenic aspects. Hum. Reprod. 2024, 39, 1–17. [Google Scholar] [CrossRef] [PubMed]
  16. Torng, L.P. Clinical implication for endometriosis associated with ovarian cancer. Gynecol. Minim. Invasive Ther. 2017, 6, 152–156. [Google Scholar] [CrossRef] [PubMed]
  17. Ness, R.B. Endometriosis and ovarian cancer: Thoughts on shared pathophysiology. Am. J. Obstet. Gynecol. 2003, 189, 280–294. [Google Scholar] [CrossRef] [PubMed]
  18. Huang, T.; Townsend, M.K.; Wentzensen, N.; Trabert, B.; White, E.; Arslan, A.A.; Weiderpass, E.; Buring, J.E.; Clendenen, T.V.; Giles, G.G.; et al. Reproductive and Hormonal Factors and Risk of Ovarian Cancer by Tumor Dominance: Results from the Ovarian Cancer Cohort Consortium (OC3). Cancer Epidemiol. Biomark. Prev. 2020, 29, 200–207. [Google Scholar] [CrossRef] [PubMed]
  19. Vercellini, P.; Eskenazi, B.; Consonni, D.; Somigliana, E.; Parazzini, F.; Abbiati, A.; Fedele, L. Oral contraceptives and risk of endometriosis: A systematic review and meta-analysis. Hum. Reprod. Update 2011, 17, 159–170. [Google Scholar] [CrossRef] [PubMed]
  20. Wang, C.; Liang, Z.; Liu, X.; Zhang, Q.; Li, S. The Association between Endometriosis, Tubal Ligation, Hysterectomy and Epithelial Ovarian Cancer: Meta-Analyses. Int. J. Environ. Res. Public Health 2016, 13, 1138. [Google Scholar] [CrossRef]
  21. Throwba, H.P.; Unnikrishnan, L.; Pangath, M.; Vasudevan, K.; Jayaraman, S.L.M.; Iyaswamy, A.; Palaniyandi, K.; Gnanasampanthapandian, D. The epigenetic correlation among ovarian cancer, endometriosis and PCOS: A review. Crit. Rev. Oncol. Hematol. 2022, 180, 103852. [Google Scholar] [CrossRef]
  22. Dhillon, P.K.; Holt, V.L. Recreational physical activity and endometrioma risk. Am. J. Epidemiol. 2003, 158, 156–164. [Google Scholar] [CrossRef] [PubMed]
  23. Mørch, L.S.; Løkkegaard, E.; Andreasen, A.H.; Krüger-Kjaer, S.; Lidegaard, O. Hormone therapy and ovarian cancer. JAMA 2009, 302, 298–305. [Google Scholar] [CrossRef] [PubMed]
  24. Yan, Y.S.; Qu, Z.; Lv, P.P.; Huang, H.F. Pediatric and adult obesity concerns in female health: A Mendelian randomization study. Endocrine 2022, 75, 400–408. [Google Scholar] [CrossRef] [PubMed]
  25. Wójtowicz, M.; Zdun, D.; Owczarek, A.J.; Skrzypulec-Plinta, V.; Olszanecka-Glinianowicz, M. Evaluation of adipokines concentrations in plasma, peritoneal, and endometrioma fluids in women operated on for ovarian endometriosis. Front. Endocrinol. 2023, 14, 1218980. [Google Scholar] [CrossRef] [PubMed]
  26. Olsen, C.M.; Green, A.C.; Whiteman, D.C.; Sadeghi, S.; Kolahdooz, F.; Webb, P.M. Obesity and the risk of epithelial ovarian cancer: A systematic review and meta-analysis. Eur. J. Cancer 2007, 43, 690–709. [Google Scholar] [CrossRef] [PubMed]
  27. Murakami, K.; Kotani, Y.; Shiro, R.; Takaya, H.; Nakai, H.; Matsumura, N. Endometriosis-associated ovarian cancer occurs early during follow-up of endometrial cysts. Int. J. Clin. Oncol. 2020, 25, 51–58. [Google Scholar] [CrossRef]
  28. Worley, M.J.; Welch, W.R.; Berkowitz, R.S.; Ng, S.W. Endometriosis-associated ovarian cancer: A review of pathogenesis. Int. J. Mol. Sci. 2013, 14, 5367–5379. [Google Scholar] [CrossRef] [PubMed]
  29. Rier, S.E.; Martin, D.C.; Bowman, R.E.; Dmowski, W.P.; Becker, J.L. Endometriosis in rhesus monkeys (Macaca mulatta) following chronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Fundam. Appl. Toxicol. Off. J. Soc. Toxicol. 1993, 21, 433–441. [Google Scholar] [CrossRef]
  30. Cummings, A.M.; Metcalf, J.L.; Birnbaum, L. Promotion of endometriosis by 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats and mice: Time-dose dependence and species comparison. Toxicol. Appl. Pharmacol. 1996, 138, 131–139. [Google Scholar] [CrossRef]
  31. Huang, P.C.; Tsai, E.M.; Li, W.F.; Liao, P.C.; Chung, M.C.; Wang, Y.H.; Wang, S.L. Association between phthalate exposure and glutathione S-transferase M1 polymorphism in adenomyosis, leiomyoma and endometriosis. Hum. Reprod. 2010, 25, 986–994. [Google Scholar] [CrossRef]
  32. L’Espérance, K.; Grundy, A.; Abrahamowicz, M.; Arseneau, J.; Gilbert, L.; Gotlieb, W.H.; Provencher, D.; Koushik, A. Alcohol intake and the risk of epithelial ovarian cancer. Cancer Causes Control CCC 2023, 34, 533–541. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, S.; Feng, S.; Du, F.; Zhang, K.; Shen, Y. Association of smoking, alcohol, and coffee consumption with the risk of ovarian cancer and prognosis: A mendelian randomization study. BMC Cancer 2023, 23, 256. [Google Scholar] [CrossRef]
  34. Sung, S.; Hong, Y.; Kim, B.G.; Choi, J.Y.; Kim, J.W.; Park, S.Y.; Kim, J.H.; Kim, Y.M.; Lee, J.M.; Kim, T.J.; et al. Stratifying the risk of ovarian cancer incidence by histologic subtypes in the Korean Epithelial Ovarian Cancer Study (Ko-EVE). Cancer Med. 2023, 12, 8742–8753. [Google Scholar] [CrossRef] [PubMed]
  35. Dunneram, Y.; Greenwood, D.C.; Cade, J.E. Diet, menopause and the risk of ovarian, endometrial and breast cancer. Proc. Nutr. Soc. 2019, 78, 438–448. [Google Scholar] [CrossRef] [PubMed]
  36. Chang, K.; Gunter, M.J.; Rauber, F.; Levy, R.B.; Huybrechts, I.; Kliemann, N.; Millett, C.; Vamos, E.P. Ultra-processed food consumption, cancer risk and cancer mortality: A large-scale prospective analysis within the UK Biobank. EclinicalMedicine 2023, 56, 101840. [Google Scholar] [CrossRef] [PubMed]
  37. Sampson, J.A. Metastatic or Embolic Endometriosis, due to the Menstrual Dissemination of Endometrial Tissue into the Venous Circulation. Am. J. Pathol. 1927, 3, 93–110.43. Available online: http://www.ncbi.nlm.nih.gov/pmc/articles/pmc1931779/ (accessed on 8 April 2024).
  38. Halme, J.; Hammond, M.G.; Hulka, J.F.; Raj, S.G.; Talbert, L.M. Retrograde menstruation in healthy women and in patients with endometriosis. Obstet. Gynecol. 1984, 64, 151–154. Available online: https://journals.lww.com/greenjournal/abstract/1984/08000/retrograde_menstruation_in_healthy_women_and_in.1.aspx (accessed on 8 April 2024).
  39. Marí-Alexandre, J.; Carcelén, A.P.; Agababyan, C.; Moreno-Manuel, A.; García-Oms, J.; Calabuig-Fariñas, S.; Gilabert-Estellés, J. Interplay Between MicroRNAs and Oxidative Stress in Ovarian Conditions with a Focus on Ovarian Cancer and Endometriosis. Int. J. Mol. Sci. 2019, 20, 5322. [Google Scholar] [CrossRef] [PubMed]
  40. Poole, E.M.; Lin, W.T.; Kvaskoff, M.; De Vivo, I.; Terry, K.L.; Missmer, S.A. Endometriosis and risk of ovarian and endometrial cancers in a large prospective cohort of U.S. nurses. Cancer Causes Control CCC 2017, 28, 437–445. [Google Scholar] [CrossRef]
  41. Körner, M.; Burckhardt, E.; Mazzucchelli, L. Higher frequency of chromosomal aberrations in ovarian endometriosis compared to extragonadal endometriosis: A possible link to endometrioid adenocarcinoma. Mod. Pathol. 2006, 19, 1615–1623. [Google Scholar] [CrossRef]
  42. Okuda, T.; Otsuka, J.; Sekizawa, A.; Saito, H.; Makino, R.; Kushima, M.; Farina, A.; Kuwano, Y.; Okai, T. p53 mutations and overexpression affect prognosis of ovarian endometrioid cancer but not clear cell cancer. Gynecol. Oncol. 2003, 88, 318–325. [Google Scholar] [CrossRef]
  43. Amemiya, S.; Sekizawa, A.; Otsuka, J.; Tachikawa, T.; Saito, H.; Okai, T. Malignant transformation of endometriosis and genetic alterations of K-ras and microsatellite instability. Int. J. Gynaecol. Obstet. 2004, 86, 371–376. [Google Scholar] [CrossRef] [PubMed]
  44. Otsuka, J.; Okuda, T.; Sekizawa, A.; Amemiya, S.; Saito, H.; Okai, T.; Kushima, M.; Tachikawa, T. K-ras mutation may promote carcinogenesis of endometriosis leading to ovarian clear cell carcinoma. Med. Electron Microsc. 2004, 37, 188–192. [Google Scholar] [CrossRef]
  45. Anglesio, M.S.; Papadopoulos, N.; Ayhan, A.; Nazeran, T.M.; Noë, M.; Horlings, H.M.; Lum, A.; Jones, S.; Senz, J.; Seckin, T.; et al. Cancer-Associated Mutations in Endometriosis without Cancer. N. Engl. J. Med. 2017, 376, 1835–1848. [Google Scholar] [CrossRef]
  46. Yachida, N.; Yoshihara, K.; Yamaguchi, M.; Suda, K.; Tamura, R.; Enomoto, T. How Does Endometriosis Lead to Ovarian Cancer? The Molecular Mechanism of Endometriosis-Associated Ovarian Cancer Development. Cancers 2021, 13, 1439. [Google Scholar] [CrossRef]
  47. Pennington, K.P.; Walsh, T.; Harrell, M.I.; Lee, M.K.; Pennil, C.C.; Rendi, M.H.; Thornton, A.; Norquist, B.M.; Casadei, S.; Nord, A.S.; et al. Germline and somatic mutations in homologous recombination genes predict platinum response and survival in ovarian, fallopian tube, and peritoneal carcinomas. Clin. Cancer Res. 2014, 20, 764–775. [Google Scholar] [CrossRef]
  48. Manchana, T.; Phoolcharoen, N.; Tantbirojn, P. BRCA mutation in high grade epithelial ovarian cancers. Gynecol. Oncol. Rep. 2019, 29, 102–105. [Google Scholar] [CrossRef] [PubMed]
  49. Wiegand, K.C.; Hennessy, B.T.; Leung, S.; Wang, Y.; Ju, Z.; McGahren, M.; Kalloger, S.E.; Finlayson, S.; Stemke-Hale, K.; Lu, Y.; et al. A functional proteogenomic analysis of endometrioid and clear cell carcinomas using reverse phase protein array and mutation analysis: Protein expression is histotype-specific and loss of ARID1A/BAF250a is associated with AKT phosphorylation. BMC Cancer 2014, 14, 120. [Google Scholar] [CrossRef] [PubMed]
  50. Lee, H.S.; Park, J.H.; Kim, S.J.; Kwon, S.J.; Kwon, J. A cooperative activation loop among SWI/SNF, gamma-H2AX and H3 acetylation for DNA double-strand break repair. EMBO J. 2010, 29, 1434–1445. [Google Scholar] [CrossRef]
  51. Chene, G.; Ouellet, V.; Rahimi, K.; Barres, V.; Caceres, K.; Meunier, L.; Cyr, L.; DeLadurantaye, M.; Provencher, D.; Mes Masson, A.M. DNA damage signaling and apoptosis in preinvasive tubal lesions of ovarian carcinoma. Int. J. Gynecol. Cancer 2015, 25, 761–769. [Google Scholar] [CrossRef]
  52. Guan, B.; Wang, T.L.; Shih, I. ARID1A, a factor that promotes formation of SWI/SNF-mediated chromatin remodeling, is a tumor suppressor in gynecologic cancers. Cancer Res. 2011, 71, 6718–6727. [Google Scholar] [CrossRef] [PubMed]
  53. Kuo, K.T.; Mao, T.L.; Jones, S.; Veras, E.; Ayhan, A.; Wang, T.L.; Glas, R.; Slamon, D.; Velculescu, V.E.; Kuman, R.J.; et al. Frequent activating mutations of PIK3CA in ovarian clear cell carcinoma. Am. J. Pathol. 2009, 174, 1597–1601. [Google Scholar] [CrossRef] [PubMed]
  54. Yamamoto, S.; Tsuda, H.; Takano, M.; Iwaya, K.; Tamai, S.; Matsubara, O. PIK3CA mutation is an early event in the development of endometriosis-associated ovarian clear cell adenocarcinoma. J. Pathol. 2011, 225, 189–194. [Google Scholar] [CrossRef] [PubMed]
  55. Yamamoto, S.; Tsuda, H.; Takano, M.; Tamai, S.; Matsubara, O. Loss of ARID1A protein expression occurs as an early event in ovarian clear-cell carcinoma development and frequently coexists with PIK3CA mutations. Mod. Pathol. 2012, 25, 615–624. [Google Scholar] [CrossRef] [PubMed]
  56. Huang, H.N.; Lin, M.C.; Huang, W.C.; Chiang, Y.C.; Kuo, K.T. Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations and ZNF217 amplification in ovarian clear cell carcinoma. Mod. Pathol. 2014, 27, 983–990. [Google Scholar] [CrossRef] [PubMed]
  57. Rogers-Broadway, K.R.; Kumar, J.; Sisu, C.; Wander, G.; Mazey, E.; Jeyaneethi, J.; Pados, G.; Tsolakidis, D.; Klonos, E.; Grunt, T.; et al. Differential expression of mTOR components in endometriosis and ovarian cancer: Effects of rapalogues and dual kinase inhibitors on mTORC1 and mTORC2 stoichiometry. Int. J. Mol. Med. 2019, 43, 47–56. [Google Scholar] [CrossRef]
  58. Shibuya, Y.; Tokunaga, H.; Saito, S.; Shimokawa, K.; Katsuoka, F.; Bin, L.; Kojima, K.; Nagasaki, M.; Yamamoto, M.; Yaegashi, N.; et al. Identification of somatic genetic alterations in ovarian clear cell carcinoma with next generation sequencing. Genes Chromosomes Cancer 2018, 57, 51–60. [Google Scholar] [CrossRef] [PubMed]
  59. Ackroyd, S.A.; Arguello, D.; Ramos, P.; Mahdi, H.; ElNaggar, A.; Winer, I.; Holloway, R.; Krivak, T.; Jones, N.; Turner, V.G.; et al. Molecular portraits of clear cell ovarian and endometrial carcinoma with comparison to clear cell renal cell carcinoma. Gynecol. Oncol. 2023, 169, 164–171. [Google Scholar] [CrossRef]
  60. Jones, S.; Wang, T.L.; Shih, I.; Mao, T.L.; Nakayama, K.; Roden, R.; Glas, R.; Slamon, D.; Diaz, L.A.; Vogelstein, B.; et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 2010, 330, 228–231. [Google Scholar] [CrossRef]
  61. Wera, S.; Hemmings, B.A. Serine/threonine protein phosphatases. Biochem. J. 1995, 311 Pt 1, 17–29. [Google Scholar] [CrossRef]
  62. Aziz, A.U.R.; Farid, S.; Qin, K.; Wang, H.; Liu, B. PIM Kinases and Their Relevance to the PI3K/AKT/mTOR Pathway in the Regulation of Ovarian Cancer. Biomolecules 2018, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  63. Driva, T.S.; Schatz, C.; Haybaeck, J. Endometriosis-Associated Ovarian Carcinomas: How PI3K/AKT/mTOR Pathway Affects Their Pathogenesis. Biomolecules 2023, 13, 1253. [Google Scholar] [CrossRef] [PubMed]
  64. Obata, K.; Hoshiai, H. Common genetic changes between endometriosis and ovarian cancer. Gynecol. Obstet. Investig. 2000, 50 (Suppl. S1), 39–43. [Google Scholar] [CrossRef] [PubMed]
  65. Sato, N.; Tsunoda, H.; Nishida, M.; Morishita, Y.; Takimoto, Y.; Kubo, T.; Noguchi, M. Loss of heterozygosity on 10q23.3 and mutation of the tumor suppressor gene PTEN in benign endometrial cyst of the ovary: Possible sequence progression from benign endometrial cyst to endometrioid carcinoma and clear cell carcinoma of the ovary. Cancer Res. 2000, 60, 7052–7056. Available online: https://aacrjournals.org/cancerres/article/60/24/7052/506957/Loss-of-Heterozygosity-on-10q23-3-and-Mutation-of (accessed on 8 April 2024). [PubMed]
  66. Martini, M.; Ciccarone, M.; Garganese, G.; Maggiore, C.; Evangelista, A.; Rahimi, S.; Zannoni, G.; Vittori, G.; Larocca, L.M. Possible involvement of hMLH1, p16(INK4a) and PTEN in the malignant transformation of endometriosis. Int. J. Cancer 2002, 102, 398–406. [Google Scholar] [CrossRef] [PubMed]
  67. Banz, C.; Ungethuem, U.; Kuban, R.J.; Diedrich, K.; Lengyel, E.; Hornung, D. The molecular signature of endometriosis-associated endometrioid ovarian cancer differs significantly from endometriosis-independent endometrioid ovarian cancer. Fertil. Steril. 2010, 94, 1212–1217. [Google Scholar] [CrossRef] [PubMed]
  68. Er, T.K.; Su, Y.F.; Wu, C.C.; Chen, C.C.; Wang, J.; Hsieh, T.H.; Herreros-Villanueva, M.; Chen, W.T.; Chen, Y.T.; Liu, T.C.; et al. Targeted next-generation sequencing for molecular diagnosis of endometriosis-associated ovarian cancer. J. Mol. Med. 2016, 94, 835–847. [Google Scholar] [CrossRef] [PubMed]
  69. Schüring, A.N.; Dahlhues, B.; Korte, A.; Kiesel, L.; Titze, U.; Heitkötter, B.; Ruckert, C.; Götte, M. The endometrial stem cell markers notch-1 and numb are associated with endometriosis. Reprod. Biomed. Online 2018, 36, 294–301. [Google Scholar] [CrossRef] [PubMed]
  70. Strauß, T.; Greve, B.; Gabriel, M.; Achmad, N.; Schwan, D.; Espinoza-Sanchez, N.A.; Laganà, A.S.; Kiesel, L.; Poutanen, M.; Götte, M.; et al. Impact of Musashi-1 and Musashi-2 Double Knockdown on Notch Signaling and the Pathogenesis of Endometriosis. Int. J. Mol. Sci. 2022, 23, 2851. [Google Scholar] [CrossRef]
  71. Suda, K.; Nakaoka, H.; Yoshihara, K.; Ishiguro, T.; Tamura, R.; Mori, Y.; Yamawaki, K.; Adachi, S.; Takahashi, T.; Kase, H.; et al. Clonal Expansion and Diversification of Cancer-Associated Mutations in Endometriosis and Normal Endometrium. Cell Rep. 2018, 24, 1777–1789. [Google Scholar] [CrossRef]
  72. Ren, F.; Wang, D.; Jiang, Y.; Ren, F. Epigenetic inactivation of hMLH1 in the malignant transformation of ovarian endometriosis. Arch. Gynecol. Obstet. 2012, 285, 215–221. [Google Scholar] [CrossRef] [PubMed]
  73. Ren, F.; Wang, D.B.; Li, T.; Chen, Y.H.; Li, Y. Identification of differentially methylated genes in the malignant transformation of ovarian endometriosis. J. Ovarian Res. 2014, 7, 73. [Google Scholar] [CrossRef] [PubMed]
  74. Guo, C.; Ren, F.; Wang, D.; Li, Y.; Liu, K.; Liu, S.; Chen, P. RUNX3 is inactivated by promoter hypermethylation in malignant transformation of ovarian endometriosis. Oncol. Rep. 2014, 32, 2580–2588. [Google Scholar] [CrossRef] [PubMed]
  75. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  76. Lavery, S.; Gillmer, M. Malignant transformation of residual endometriosis in women on unopposed oestrogen hormone replacement therapy. BJOG Int. J. Obstet. Gynaecol. 2001, 108, 1106–1107. [Google Scholar] [CrossRef] [PubMed]
  77. Collaborative Group on Epidemiological Studies of Ovarian Cancer; Beral, V.; Gaitskell, K.; Hermon, C.; Moser, K.; Reeves, G.; Peto, R. Menopausal hormone use and ovarian cancer risk: Individual participant meta-analysis of 52 epidemiological studies. Lancet 2015, 385, 1835–1842. [Google Scholar] [CrossRef] [PubMed]
  78. Xu, X.L.; Huang, Z.Y.; Yu, K.; Li, J.; Fu, X.W.; Deng, S.L. Estrogen Biosynthesis and Signal Transduction in Ovarian Disease. Front. Endocrinol. 2022, 13, 827032. [Google Scholar] [CrossRef]
  79. Revankar, C.M.; Cimino, D.F.; Sklar, L.A.; Arterburn, J.B.; Prossnitz, E.R. A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science 2005, 307, 1625–1630. [Google Scholar] [CrossRef] [PubMed]
  80. Kozieł, M.J.; Piastowska-Ciesielska, A.W. Estrogens, Estrogen Receptors and Tumor Microenvironment in Ovarian Cancer. Int. J. Mol. Sci. 2023, 24, 14673. [Google Scholar] [CrossRef]
  81. Fujimoto, J.; Alam, S.M.; Jahan, I.; Sato, E.; Sakaguchi, H.; Tamaya, T. Clinical implication of estrogen-related receptor (ERR) expression in ovarian cancers. J. Steroid Biochem. Mol. Biol. 2007, 104, 301–304. [Google Scholar] [CrossRef]
  82. Brandenberger, A.W.; Tee, M.K.; Jaffe, R.B. Estrogen receptor alpha (ER-alpha) and beta (ER-beta) mRNAs in normal ovary, ovarian serous cystadenocarcinoma and ovarian cancer cell lines: Down-regulation of ER-beta in neoplastic tissues. J. Clin. Endocrinol. Metab. 1998, 83, 1025–1028. [Google Scholar] [CrossRef] [PubMed]
  83. Pujol, P.; Rey, J.M.; Nirde, P.; Roger, P.; Gastaldi, M.; Laffargue, F.; Rochefort, H.; Maudelonde, T. Differential expression of estrogen receptor-alpha and -beta messenger RNAs as a potential marker of ovarian carcinogenesis. Cancer Res. 1998, 58, 5367–5373. Available online: https://aacrjournals.org/cancerres/article/58/23/5367/504774/Differential-Expression-of-Estrogen-Receptor-and (accessed on 8 April 2024).
  84. Rutherford, T.; Brown, W.D.; Sapi, E.; Aschkenazi, S.; Muñoz, A.; Mor, G. Absence of estrogen receptor-beta expression in metastatic ovarian cancer. Obstet. Gynecol. 2000, 96, 417–421. [Google Scholar] [CrossRef] [PubMed]
  85. Bardin, A.; Boulle, N.; Lazennec, G.; Vignon, F.; Pujol, P. Loss of ERbeta expression as a common step in estrogen-dependent tumor progression. Endocr.-Relat. Cancer 2004, 11, 537–551. [Google Scholar] [CrossRef]
  86. Bossard, C.; Busson, M.; Vindrieux, D.; Gaudin, F.; Machelon, V.; Brigitte, M.; Jacquard, C.; Pillon, A.; Balaguer, P.; Balabanian, K.; et al. Potential role of estrogen receptor beta as a tumor suppressor of epithelial ovarian cancer. PLoS ONE 2012, 7, e44787. [Google Scholar] [CrossRef]
  87. Trukhacheva, E.; Lin, Z.; Reierstad, S.; Cheng, Y.H.; Milad, M.; Bulun, S.E. Estrogen receptor (ER) beta regulates ERalpha expression in stromal cells derived from ovarian endometriosis. J. Clin. Endocrinol. Metab. 2009, 94, 615–622. [Google Scholar] [CrossRef]
  88. Andersen, C.L.; Boisen, M.M.; Sikora, M.J.; Ma, T.; Tseng, G.; Suryawanshi, S.; Vlad, A.; Elishaev, E.; Edwards, R.P.; Oesterreich, S. The Evolution of Estrogen Receptor Signaling in the Progression of Endometriosis to Endometriosis-Associated Ovarian Cancer. Horm. Cancer 2018, 9, 399–407. [Google Scholar] [CrossRef] [PubMed]
  89. Shimura, K.; Tarumi, Y.; Fujii, M.; Ogawa, K.; Maeda, E.; Tanaka, Y.; Okimura, H.; Kataoka, H.; Takaoka, O.; Ito, F.; et al. Low-Nutrient Environment-Induced Changes in Inflammation, Cell Proliferation, and PGC-1α Expression in Stromal Cells with Ovarian Endometriosis. Reprod. Sci. 2023, 30, 1094–1102. [Google Scholar] [CrossRef]
  90. Kobayashi, H.; Yamada, Y.; Kawahara, N.; Ogawa, K.; Yoshimoto, C. Integrating modern approaches to pathogenetic concepts of malignant transformation of endometriosis. Oncol. Rep. 2019, 41, 1729–1738. [Google Scholar] [CrossRef]
  91. Shin, S.; Chung, Y.J.; Moon, S.W.; Choi, E.J.; Kim, M.R.; Chung, Y.J.; Lee, S.H. Single-cell profiling identifies distinct hormonal, immunologic, and inflammatory signatures of endometriosis-constituting cells. J. Pathol. 2023, 261, 323–334. [Google Scholar] [CrossRef]
  92. Liu, G.; Sun, P.; Dong, B.; Sehouli, J. Key regulator of cellular metabolism, estrogen-related receptor α, a new therapeutic target in endocrine-related gynecological tumor. Cancer Manag. Res. 2018, 10, 6887–6895. [Google Scholar] [CrossRef] [PubMed]
  93. Park, S.H.; Cheung, L.W.; Wong, A.S.; Leung, P.C. Estrogen regulates Snail and Slug in the down-regulation of E-cadherin and induces metastatic potential of ovarian cancer cells through estrogen receptor alpha. Mol. Endocrinol. 2008, 22, 2085–2098. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, X.; Lin, Y.; Zheng, Y. Antitumor effects of aconitine in A2780 cells via estrogen receptor β mediated apoptosis, DNA damage and migration. Mol. Med. Rep. 2020, 22, 2318–2328. [Google Scholar] [CrossRef] [PubMed]
  95. Kozieł, M.J.; Habrowska-Górczyńska, D.E.; Urbanek, K.A.; Domińska, K.; Piastowska-Ciesielska, A.W.; Kowalska, K. Estrogen receptor α mediates alternariol-induced apoptosis and modulation of the invasiveness of ovarian cancer cells. Toxicol. Lett. 2023, 386, 9–19. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, D.; Guo, C.; Li, Y.; Zhou, M.; Wang, H.; Liu, J.; Chen, P. Oestrogen up-regulates DNMT1 and leads to the hypermethylation of RUNX3 in the malignant transformation of ovarian endometriosis. Reprod. Biomed. Online 2022, 44, 27–37. [Google Scholar] [CrossRef] [PubMed]
  97. Suryawanshi, S.; Vlad, A.M.; Lin, H.M.; Mantia-Smaldone, G.; Laskey, R.; Lee, M.; Lin, Y.; Donnellan, N.; Klein-Patel, M.; Lee, T.; et al. Plasma microRNAs as novel biomarkers for endometriosis and endometriosis-associated ovarian cancer. Clin. Cancer Res. 2013, 19, 1213–1224. [Google Scholar] [CrossRef] [PubMed]
  98. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef] [PubMed]
  99. Lagos-Quintana, M.; Rauhut, R.; Lendeckel, W.; Tuschl, T. Identification of novel genes coding for small expressed RNAs. Science 2001, 294, 853–858. [Google Scholar] [CrossRef]
  100. Lau, N.C.; Lim, L.P.; Weinstein, E.G.; Bartel, D.P. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science 2001, 294, 858–862. [Google Scholar] [CrossRef]
  101. Lee, R.C.; Ambros, V. An extensive class of small RNAs in Caenorhabditis elegans. Science 2001, 294, 862–864. [Google Scholar] [CrossRef]
  102. Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA expression profiles classify human cancers. Nature 2005, 435, 834–838. [Google Scholar] [CrossRef] [PubMed]
  103. Iorio, M.V.; Visone, R.; Di Leva, G.; Donati, V.; Petrocca, F.; Casalini, P.; Taccioli, C.; Volinia, S.; Liu, C.G.; Alder, H.; et al. MicroRNA signatures in human ovarian cancer. Cancer Res. 2007, 67, 8699–8707. [Google Scholar] [CrossRef]
  104. Resnick, K.E.; Alder, H.; Hagan, J.P.; Richardson, D.L.; Croce, C.M.; Cohn, D.E. The detection of differentially expressed microRNAs from the serum of ovarian cancer patients using a novel real-time PCR platform. Gynecol. Oncol. 2009, 112, 55–59. [Google Scholar] [CrossRef]
  105. Filigheddu, N.; Gregnanin, I.; Porporato, P.E.; Surico, D.; Perego, B.; Galli, L.; Patrignani, C.; Graziani, A.; Surico, N. Differential expression of microRNAs between eutopic and ectopic endometrium in ovarian endometriosis. J. Biomed. Biotechnol. 2010, 2010, 369549. [Google Scholar] [CrossRef]
  106. Teague, E.M.O.; VanderHoek, K.H.; VanderHoek, M.B.; Perry, N.; Wagaarachchi, P.; Robertson, S.A.; Print, C.G.; Hull, L.M. MicroRNA-regulated pathways associated with endometriosis. Mol. Endocrinol. 2009, 23, 265–275. [Google Scholar] [CrossRef] [PubMed]
  107. SiufiNeto, J.; Kho, R.M.; Siufi, D.F.; Baracat, E.C.; Anderson, K.S.; Abrão, M.S. Cellular, histologic, and molecular changes associated with endometriosis and ovarian cancer. J. Minim. Invasive Gynecol. 2014, 21, 55–63. [Google Scholar] [CrossRef] [PubMed]
  108. vanJaarsveld, M.T.; Helleman, J.; Berns, E.M.; Wiemer, E.A. MicroRNAs in ovarian cancer biology and therapy resistance. Int. J. Biochem. Cell Biol. 2010, 42, 1282–1290. [Google Scholar] [CrossRef]
  109. Büssing, I.; Slack, F.J.; Grosshans, H. let-7 microRNAs in development, stem cells and cancer. Trends Mol. Med. 2008, 14, 400–409. [Google Scholar] [CrossRef]
  110. Liu, Y.N.; Yin, J.J.; Abou-Kheir, W.; Hynes, P.G.; Casey, O.M.; Fang, L.; Yi, M.; Stephens, R.M.; Seng, V.; Sheppard-Tillman, H.; et al. MiR-1 and miR-200 inhibit EMT via Slug-dependent and tumorigenesis via Slug-independent mechanisms. Oncogene 2013, 32, 296–306. [Google Scholar] [CrossRef] [PubMed]
  111. Shimono, Y.; Zabala, M.; Cho, R.W.; Lobo, N.; Dalerba, P.; Qian, D.; Diehn, M.; Liu, H.; Panula, S.P.; Chiao, E.; et al. Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell 2009, 138, 592–603. [Google Scholar] [CrossRef]
  112. Olson, P.L.J.; Zhang, H.; Shai, A.; Chun, M.G.; Wang, Y.; Libutti, S.K.; Nakakura, E.K.; Golub, T.R.; Hanahan, D. MicroRNA dynamics in the stages of tumorigenesis correlate with hallmark capabilities of cancer. Genes Dev. 2009, 23, 2152–2165. [Google Scholar] [CrossRef] [PubMed]
  113. Eggers, J.C.; Martino, V.; Reinbold, R.; Schäfer, S.D.; Kiesel, L.; Starzinski-Powitz, A.; Schüring, A.N.; Kemper, B.; Greve, B.; Götte, M. microRNA miR-200b affects proliferation, invasiveness and stemness of endometriotic cells by targeting ZEB1, ZEB2 and KLF4. Reprod. Biomed. Online 2016, 32, 434–445. [Google Scholar] [CrossRef] [PubMed]
  114. Stejskalová, A.; Fincke, V.; Nowak, M.; Schmidt, Y.; Borrmann, K.; vonWahlde, M.K.; Schäfer, S.D.; Kiesel, L.; Greve, B.; Götte, M. Collagen I triggers directional migration, invasion and matrix remodeling of stroma cells in a 3D spheroid model of endometriosis. Sci. Rep. 2021, 11, 4115. [Google Scholar] [CrossRef] [PubMed]
  115. Szubert, M.; Nowak-Glück, A.; Domańska-Senderowska, D.; Szymańska, B.; Sowa, P.; Rycerz, A.; Wilczyński, J.R. miRNA Expression Profiles in Ovarian Endometriosis and Two Types of Ovarian Cancer-Endometriosis-Associated Ovarian Cancer and High-Grade Ovarian Cancer. Int. J. Mol. Sci. 2023, 24, 17470. [Google Scholar] [CrossRef] [PubMed]
  116. Lu, W.C.; Liu, C.J.; Tu, H.F.; Chung, Y.T.; Yang, C.C.; Kao, S.Y.; Chang, K.W.; Lin, S.C. miR-31 targets ARID1A and enhances the oncogenicity and stemness of head and neck squamous cell carcinoma. Oncotarget 2016, 7, 57254–57267. [Google Scholar] [CrossRef] [PubMed]
  117. Liu, C.J.; Tsai, M.M.; Hung, P.S.; Kao, S.Y.; Liu, T.Y.; Wu, K.J.; Chiou, S.H.; Lin, S.C.; Chang, K.W. miR-31 ablates expression of the HIF regulatory factor FIH to activate the HIF pathway in head and neck carcinoma. Cancer Res. 2010, 70, 1635–1644. [Google Scholar] [CrossRef] [PubMed]
  118. Muharam, R.; RahmalaFebri, R.; Mutia, K.; Iffanolida, P.A.; Maidarti, M.; Wiweko, B.; Hestiantoro, A. Down-Regulation of miR-93 Negatively Correlates with Overexpression of VEGFA and MMP3 in Endometriosis: A Cross-Sectional Study. Int. J. Fertil. Steril. 2023, 17, 28–33. [Google Scholar] [CrossRef] [PubMed]
  119. DelCarmen, M.G.; SmithSehdev, A.E.; Fader, A.N.; Zahurak, M.L.; Richardson, M.; Fruehauf, J.P.; Montz, F.J.; Bristow, R.E. Endometriosis-associated ovarian carcinoma: Differential expression of vascular endothelial growth factor and estrogen/progesterone receptors. Cancer 2003, 98, 1658–1663. [Google Scholar] [CrossRef]
  120. Fontana, L.; Pelosi, E.; Greco, P.; Racanicchi, S.; Testa, U.; Liuzzi, F.; Croce, C.M.; Brunetti, E.; Grignani, F.; Peschle, C. MicroRNAs 17-5p-20a-106a control monocytopoiesis through AML1 targeting and M-CSF receptor upregulation. Nat. Cell Biol. 2007, 9, 775–787. [Google Scholar] [CrossRef]
  121. Ramón, L.A.; Braza-Boïls, A.; Gilabert-Estellés, J.; Gilabert, J.; España, F.; Chirivella, M.; Estellés, A. microRNAs expression in endometriosis and their relation to angiogenic factors. Hum. Reprod. 2011, 26, 1082–1090. [Google Scholar] [CrossRef]
  122. Winarto, H.; Tan, M.I.; Sadikin, M.; Wanandi, S.I. ARID1A Expression is Down-Regulated by Oxidative Stress in Endometriosis and Endometriosis-Associated Ovarian Cancer. Transl. Oncogenomics 2017, 9, 1177272716689818. [Google Scholar] [CrossRef] [PubMed]
  123. Mandai, M.; Yamaguchi, K.; Matsumura, N.; Baba, T.; Konishi, I. Ovarian cancer in endometriosis: Molecular biology, pathology, and clinical management. Int. J. Clin. Oncol. 2009, 14, 383–391. [Google Scholar] [CrossRef] [PubMed]
  124. Rockfield, S.; Raffel, J.; Mehta, R.; Rehman, N.; Nanjundan, M. Iron overload and altered iron metabolism in ovarian cancer. Biol. Chem. 2017, 398, 995–1007. [Google Scholar] [CrossRef] [PubMed]
  125. Yamaguchi, K.; Mandai, M.; Toyokuni, S.; Hamanishi, J.; Higuchi, T.; Takakura, K.; Fujii, S. Contents of endometriotic cysts, especially the high concentration of free iron, are a possible cause of carcinogenesis in the cysts through the iron-induced persistent oxidative stress. Clin. Cancer Res. 2008, 14, 32–40. [Google Scholar] [CrossRef] [PubMed]
  126. Suryawanshi, S.; Huang, X.; Elishaev, E.; Budiu, R.A.; Zhang, L.; Kim, S.; Donnellan, N.; Mantia-Smaldone, G.; Ma, T.; Tseng, G.; et al. Complement pathway is frequently altered in endometriosis and endometriosis-associated ovarian cancer. Clin. Cancer Res. 2014, 20, 6163–6174. [Google Scholar] [CrossRef] [PubMed]
  127. Chang, C.M.; Wang, M.L.; Lu, K.H.; Yang, Y.P.; Juang, C.M.; Wang, P.H.; Hsu, R.J.; Yu, M.H.; Chang, C.C. Integrating the dysregulated inflammasome-based molecular functionome in the malignant transformation of endometriosis-associated ovarian carcinoma. Oncotarget 2017, 9, 3704–3726. [Google Scholar] [CrossRef] [PubMed]
  128. Linder, A.; Westbom-Fremer, S.; Mateoiu, C.; OlssonWidjaja, A.; Österlund, T.; Veerla, S.; Ståhlberg, A.; Ulfenborg, B.; Hedenfalk, I.; Sundfeldt, K. Genomic alterations in ovarian endometriosis and subsequently diagnosed ovarian carcinoma. Hum. Reprod. 2024; deae043, advance online publication. [Google Scholar] [CrossRef]
  129. De Berardinis, R.J.; Chandel, N.S. Fundamentals of cancer metabolism. Sci. Adv. 2016, 2, e1600200. [Google Scholar] [CrossRef]
  130. Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef] [PubMed]
  131. Kobayashi, H.; Shigetomi, H.; Imanaka, S. Nonhormonal therapy for endometriosis based on energy metabolism regulation. Reprod. Fertil. 2021, 2, C42–C57. [Google Scholar] [CrossRef]
  132. Kobayashi, H. Recent advances in understanding the metabolic plasticity of ovarian cancer: A systematic review. Heliyon 2022, 8, e11487. [Google Scholar] [CrossRef]
  133. Nantasupha, C.; Thonusin, C.; Charoenkwan, K.; Chattipakorn, S.; Chattipakorn, N. Metabolic reprogramming in epithelial ovarian cancer. Am. J. Transl. Res. 2021, 13, 9950–9973. Available online: http://www.ncbi.nlm.nih.gov/pmc/articles/pmc8507042/ (accessed on 8 April 2024). [PubMed]
  134. Wu, Y.; Zhang, X.; Wang, Z.; Zheng, W.; Cao, H.; Shen, W. Targeting oxidative phosphorylation as an approach for the treatment of ovarian cancer. Front. Oncol. 2022, 12, 971479. [Google Scholar] [CrossRef] [PubMed]
  135. Dar, S.; Chhina, J.; Mert, I.; Chitale, D.; Buekers, T.; Kaur, H.; Giri, S.; Munkarah, A.; Rattan, R. Bioenergetic Adaptations in Chemoresistant Ovarian Cancer Cells. Sci. Rep. 2017, 7, 8760. [Google Scholar] [CrossRef] [PubMed]
  136. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [PubMed]
  137. Kreuzaler, P.; Panina, Y.; Segal, J.; Yuneva, M. Adapt and conquer: Metabolic flexibility in cancer growth, invasion and evasion. Mol. Metab. 2020, 33, 83–101. [Google Scholar] [CrossRef] [PubMed]
  138. Wang, M.; Zhang, J.; Wu, Y. Tumor metabolism rewiring in epithelial ovarian cancer. J. Ovarian Res. 2023, 16, 108. [Google Scholar] [CrossRef] [PubMed]
  139. Zhang, Y.; Wang, Y.; Zhao, G.; Orsulic, S.; Matei, D. Metabolic dependencies and targets in ovarian cancer. Pharmacol. Ther. 2023, 245, 108413. [Google Scholar] [CrossRef] [PubMed]
  140. Zheng, Q.; Chen, S.; Chen, L.; Wang, J. Analysis of Clinical Characteristics and Prognostic Factors Related to EMs Correlation in Ovarian Cancer Patients. Altern. Ther. Health Med. 2024; AT9934, advance online publication. Available online: http://alternative-therapies.com/oa/index.html?fid=9934(accessed on 8 April 2024).
  141. Vercellini, P.; Viganò, P.; Buggio, L.; Makieva, S.; Scarfone, G.; Cribiù, F.M.; Parazzini, F.; Somigliana, E. Perimenopausal management of ovarian endometriosis and associated cancer risk: When is medical or surgical treatment indicated? Best Pract. Res. Clin. Obstet. Gynaecol. 2018, 51, 151–168. [Google Scholar] [CrossRef] [PubMed]
  142. Younis, J. Should Endometriosis-Associated Ovarian Cancer Alter the Management of Women with an Intact Endometrioma in the Reproductive Age? Reprod. Med. 2023, 4, 100–105. [Google Scholar] [CrossRef]
  143. Zhang, X.; Li, M.; Tang, Z.; Li, X.; Song, T. Differentiation between endometriosis-associated ovarian cancers and non-endometriosis-associated ovarian cancers based on magnetic resonance imaging. Br. J. Radiol. 2021, 94, 20201441. [Google Scholar] [CrossRef]
  144. Thomsen, L.H.; Schnack, T.H.; Buchardi, K.; Hummelshoj, L.; Missmer, S.A.; Forman, A.; Blaakaer, J. Risk factors of epithelial ovarian carcinomas among women with endometriosis: A systematic review. Acta Obstet. Gynecol. Scand. 2017, 96, 761–778. [Google Scholar] [CrossRef] [PubMed]
  145. Yılmaz-Hanege, B.; Güler Çekıç, S.; Ata, B. Endometrioma and ovarian reserve: Effects of endometriomata per se and its surgical treatment on the ovarian reserve. Facts Views Vis. ObGyn 2019, 11, 151–157. Available online: http://www.ncbi.nlm.nih.gov/pmc/articles/pmc6897522/ (accessed on 8 April 2024). [PubMed]
  146. Parker, W.H.; Broder, M.S.; Chang, E.; Feskanich, D.; Farquhar, C.; Liu, Z.; Shoupe, D.; Berek, J.S.; Hankinson, S.; Manson, J.E. Ovarian conservation at the time of hysterectomy and long-term health outcomes in the nurses’ health study. Obstet. Gynecol. 2009, 113, 1027–1037. [Google Scholar] [CrossRef] [PubMed]
  147. Meczekalski, B.; Podfigurna-Stopa, A.; Genazzani, A.R. Hypoestrogenism in young women and its influence on bone mass density. Gynecol. Endocrinol. 2010, 26, 652–657. [Google Scholar] [CrossRef] [PubMed]
  148. Vercellini, P.; Bandini, V.; Viganò, P.; Ambruoso, D.; Cetera, G.E.; Somigliana, E. Proposal for targeted, neo-evolutionary-oriented secondary prevention of early-onset endometriosis and adenomyosis. Part II: Medical interventions. Hum. Reprod. 2024, 39, 18–34. [Google Scholar] [CrossRef] [PubMed]
  149. Mansour, D.; Westhoff, C.; Kher, U.; Korver, T. Pooled analysis of two randomized, open-label studies comparing the effects of nomegestrol acetate/17β-estradiol and drospirenone/ethinyl estradiol on bleeding patterns in healthy women. Contraception 2017, 95, 390–397. [Google Scholar] [CrossRef] [PubMed]
  150. Modugno, F.; Ness, R.B.; Allen, G.O.; Schildkraut, J.M.; Davis, F.G.; Goodman, M.T. Oral contraceptive use, reproductive history, and risk of epithelial ovarian cancer in women with and without endometriosis. Am. J. Obstet. Gynecol. 2004, 191, 733–740. [Google Scholar] [CrossRef] [PubMed]
  151. Malvezzi, M.; Carioli, G.; Rodriguez, T.; Negri, E.; La Vecchia, C. Global trends and predictions in ovarian cancer mortality. Ann. Oncol. 2016, 27, 2017–2025. [Google Scholar] [CrossRef] [PubMed]
  152. Iversen, L.; Fielding, S.; Lidegaard, Ø.; Mørch, L.S.; Skovlund, C.W.; Hannaford, P.C. Association between contemporary hormonal contraception and ovarian cancer in women of reproductive age in Denmark: Prospective, nationwide cohort study. BMJ 2018, 362, k3609. [Google Scholar] [CrossRef]
  153. Dalmartello, M.; La Vecchia, C.; Bertuccio, P.; Boffetta, P.; Levi, F.; Negri, E.; Malvezzi, M. European cancer mortality predictions for the year 2022 with focus on ovarian cancer. Ann. Oncol. 2022, 33, 330–339. [Google Scholar] [CrossRef]
  154. Murumägi, A.; Ungureanu, D.; Khan, S.; Arjama, M.; Välimäki, K.; Ianevski, A.; Ianevski, P.; Bergström, R.; Dini, A.; Kanerva, A.; et al. Drug response profiles in patient-derived cancer cells across histological subtypes of ovarian cancer: Real-time therapy tailoring for a patient with low-grade serous carcinoma. Br. J. Cancer 2023, 128, 678–690. [Google Scholar] [CrossRef]
  155. Gil-Martin, M.; Pardo, B.; Barretina-Ginesta, M.P. Rare ovarian tumours. Other treatments for ovarian cancer. Eur. J. Cancer Suppl. 2020, 15, 96–103. [Google Scholar] [CrossRef] [PubMed]
  156. Wang, H.; Chen, C.; Wang, D.; Zhu, Y.; Chen, P. Correlation of clinicopathological and prognostic characteristics between endometriosis-associated and primary ovarian cancer. BMC Cancer 2023, 23, 1210. [Google Scholar] [CrossRef] [PubMed]
  157. Vergote, I.; Tropé, C.G.; Amant, F.; Kristensen, G.B.; Ehlen, T.; Johnson, N.; Verheijen, R.H.; van der Burg, M.E.; Lacave, A.J.; Panici, P.B.; et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N. Engl. J. Med. 2010, 363, 943–953. [Google Scholar] [CrossRef]
  158. Sananpanichkul, P.; Muangtan, S.; Suknikhom, W.; Bhamarapravatana, K.; Suwannarurk, K. Does Endometriosis Hinder Successful Ovarian Debulking Surgery? Asian Pac. J. Cancer Prev. APJCP 2018, 19, 509–512. [Google Scholar] [CrossRef] [PubMed]
  159. Erzen, M.; Rakar, S.; Klancnik, B.; Syrjänen, K. Endometriosis-associated ovarian carcinoma (EAOC): An entity distinct from other ovarian carcinomas as suggested by a nested case-control study. Gynecol. Oncol. 2001, 83, 100–108. [Google Scholar] [CrossRef]
  160. Li, Q.; Sun, Y.; Zhang, X.; Wang, L.; Wu, W.; Wu, M.; Meng, C.; Liu, G. Endometriosis-associated ovarian cancer is a single entity with distinct clinicopathological characteristics. Cancer Biol. Ther. 2019, 20, 1029–1034. [Google Scholar] [CrossRef]
  161. Trimbos, B.; Timmers, P.; Pecorelli, S.; Coens, C.; Ven, K.; van der Burg, M.; Casado, A. Surgical staging and treatment of early ovarian cancer: Long-term analysis from a randomized trial. J. Natl. Cancer Inst. 2010, 102, 982–987. [Google Scholar] [CrossRef] [PubMed]
  162. Trimbos, J.B.; Vergote, I.; Bolis, G.; Vermorken, J.B.; Mangioni, C.; Madronal, C.; Franchi, M.; Tateo, S.; Zanetta, G.; Scarfone, G.; et al. Impact of adjuvant chemotherapy and surgical staging in early-stage ovarian carcinoma: European Organisation for Research and Treatment of Cancer-Adjuvant ChemoTherapy in Ovarian Neoplasm trial. J. Natl. Cancer Inst. 2003, 95, 113–125. [Google Scholar] [CrossRef]
  163. Armstrong, D.K.; Alvarez, R.D.; Bakkum-Gamez, J.N.; Barroilhet, L.; Behbakht, K.; Berchuck, A.; Chen, L.M.; Cristea, M.; DeRosa, M.; Eisenhauer, E.L.; et al. Ovarian Cancer, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. JNCCN 2021, 19, 191–226. [Google Scholar] [CrossRef]
  164. Ledermann, J.A.; Matias-Guiu, X.; Amant, F.; Concin, N.; Davidson, B.; Fotopoulou, C.; González-Martin, A.; Gourley, C.; Leary, A.; Lorusso, D.; et al. ESGO-ESMO-ESP consensus conference recommendations on ovarian cancer: Pathology and molecular biology and early, advanced and recurrent disease. Ann. Oncol. 2024, 35, 248–266. [Google Scholar] [CrossRef] [PubMed]
  165. Adams, G.; Zekri, J.; Wong, H.; Walking, J.; Green, J.A. Platinum-based adjuvant chemotherapy for early-stage epithelial ovarian cancer: Single or combination chemotherapy? BJOG Int. J. Obstet. Gynaecol. 2010, 117, 1459–1467. [Google Scholar] [CrossRef] [PubMed]
  166. Bell, J.; Brady, M.F.; Young, R.C.; Lage, J.; Walker, J.L.; Look, K.Y.; Rose, G.S.; Spirtos, N.M.; Gynecologic Oncology Group. Randomized phase III trial of three versus six cycles of adjuvant carboplatin and paclitaxel in early stage epithelial ovarian carcinoma: A Gynecologic Oncology Group study. Gynecol. Oncol. 2006, 102, 432–439. [Google Scholar] [CrossRef] [PubMed]
  167. ICON-Collaborators. ICON2: Randomised trial of single-agent carboplatin against three-drug combination of CAP (cyclophosphamide, doxorubicin, and cisplatin) in women with ovarian cancer. Lancet 1998, 352, 1571–1576. [Google Scholar] [CrossRef]
  168. International Collaborative Ovarian Neoplasm Group. Paclitaxel plus carboplatin versus standard chemotherapy with either single-agent carboplatin or cyclophosphamide, doxorubicin, and cisplatin in women with ovarian cancer: The ICON3 randomised trial. Lancet 2002, 360, 505–515. [Google Scholar] [CrossRef] [PubMed]
  169. Muggia, F.M.; Braly, P.S.; Brady, M.F.; Sutton, G.; Niemann, T.H.; Lentz, S.L.; Alvarez, R.D.; Kucera, P.R.; Small, J.M. Phase III randomized study of cisplatin versus paclitaxel versus cisplatin and paclitaxel in patients with suboptimal stage III or IV ovarian cancer: A gynecologic oncology group study. J. Clin. Oncol. 2000, 18, 106–115. [Google Scholar] [CrossRef] [PubMed]
  170. Borella, F.; Fucina, S.; Mangherini, L.; Cosma, S.; Carosso, A.R.; Cusato, J.; Cassoni, P.; Bertero, L.; Katsaros, D.; Benedetto, C. Hormone Receptors and Epithelial Ovarian Cancer: Recent Advances in Biology and Treatment Options. Biomedicines 2023, 11, 2157. [Google Scholar] [CrossRef] [PubMed]
  171. Hollis, R.L.; Stanley, B.; Iida, Y.; Thomson, J.; Churchman, M.; Rye, T.; Mackean, M.; Nussey, F.; Gourley, C.; Herrington, C.S. Hormone receptor expression patterns define clinically meaningful subgroups of endometrioid ovarian carcinoma. Gynecol. Oncol. 2019, 155, 318–323. [Google Scholar] [CrossRef] [PubMed]
  172. Burges, A.; Brüning, A.; Dannenmann, C.; Blankenstein, T.; Jeschke, U.; Shabani, N.; Friese, K.; Mylonas, I. Prognostic significance of estrogen receptor alpha and beta expression in human serous carcinomas of the ovary. Arch. Gynecol. Obstet. 2010, 281, 511–517. [Google Scholar] [CrossRef]
  173. Halon, A.; Materna, V.; Drag-Zalesinska, M.; Nowak-Markwitz, E.; Gansukh, T.; Donizy, P.; Spaczynski, M.; Zabel, M.; Dietel, M.; Lage, H.; et al. Estrogen receptor alpha expression in ovarian cancer predicts longer overall survival. Pathol. Oncol. Res. POR 2011, 17, 511–518. [Google Scholar] [CrossRef]
  174. Schüler-Toprak, S.; Weber, F.; Skrzypczak, M.; Ortmann, O.; Treeck, O. Expression of estrogen-related receptors in ovarian cancer and impact on survival. J. Cancer Res. Clin. Oncol. 2021, 147, 2555–2567. [Google Scholar] [CrossRef]
  175. Sieh, W.; Köbel, M.; Longacre, T.A.; Bowtell, D.D.; deFazio, A.; Goodman, M.T.; Høgdall, E.; Deen, S.; Wentzensen, N.; Moysich, K.B.; et al. Hormone-receptor expression and ovarian cancer survival: An Ovarian Tumor Tissue Analysis consortium study. Lancet. Oncol. 2013, 14, 853–862. [Google Scholar] [CrossRef]
  176. Langdon, S.P.; Herrington, C.S.; Hollis, R.L.; Gourley, C. Estrogen Signaling and Its Potential as a Target for Therapy in Ovarian Cancer. Cancers 2020, 12, 1647. [Google Scholar] [CrossRef] [PubMed]
  177. Ostlund, T.; Alotaibi, F.; Kyeremateng, J.; Halaweish, H.; Kasten, A.; Iram, S.; Halaweish, F. Triazole-estradiol analogs: A potential cancer therapeutic targeting ovarian and colorectal cancer. Steroids 2022, 177, 108950. [Google Scholar] [CrossRef]
  178. Ono, M.; Miyamoto, T.; Asaka, R.; Uchikawa, J.; Ando, H.; Tanaka, Y.; Shinagawa, M.; Yokokawa, Y.; Asaka, S.; Wang, T.L.; et al. Establishment of a novel model of endometriosis-associated ovarian cancer by transplanting uterine tissue from Arid1a/Pten knockout mice. Sci. Rep. 2023, 13, 8348. [Google Scholar] [CrossRef]
  179. Wuyung, P.E.; Rahadiati, F.B.; Tjahjadi, H.; Salinah, S.; Kusmardi, K.; Kodariah, R.; Wiweko, B. Histopathology and ARID1A Expression in Endometriosis-Associated Ovarian Carcinoma (EAOC) Carcinogenesis Model with Endometrial Autoimplantation and DMBA Induction. Asian Pac. J. Cancer Prev. APJCP 2021, 22, 553–558. [Google Scholar] [CrossRef] [PubMed]
  180. Burger, R.A.; Brady, M.F.; Bookman, M.A.; Fleming, G.F.; Monk, B.J.; Huang, H.; Mannel, R.S.; Homesley, H.D.; Fowler, J.; Greer, B.E.; et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N. Engl. J. Med. 2011, 365, 2473–2483. [Google Scholar] [CrossRef] [PubMed]
  181. Perren, T.J.; Swart, A.M.; Pfisterer, J.; Ledermann, J.A.; Pujade-Lauraine, E.; Kristensen, G.; Carey, M.S.; Beale, P.; Cervantes, A.; Kurzeder, C.; et al. A phase 3 trial of bevacizumab in ovarian cancer. N. Engl. J. Med. 2011, 365, 2484–2496. [Google Scholar] [CrossRef]
  182. Tewari, K.S.; Burger, R.A.; Enserro, D.; Norquist, B.M.; Swisher, E.M.; Brady, M.F.; Bookman, M.A.; Fleming, G.F.; Huang, H.; Homesley, H.D.; et al. Final Overall Survival of a Randomized Trial of Bevacizumab for Primary Treatment of Ovarian Cancer. J. Clin. Oncol. 2019, 37, 2317–2328. [Google Scholar] [CrossRef]
  183. Moore, K.; Colombo, N.; Scambia, G.; Kim, B.G.; Oaknin, A.; Friedlander, M.; Lisyanskaya, A.; Floquet, A.; Leary, A.; Sonke, G.S.; et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N. Engl. J. Med. 2018, 379, 2495–2505. [Google Scholar] [CrossRef]
  184. González-Martín, A.; Pothuri, B.; Vergote, I.D.C.R.; Graybill, W.; Mirza, M.R.; McCormick, C.; Lorusso, D.; Hoskins, P.; Freyer, G.; Baumann, K.; et al. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2391–2402. [Google Scholar] [CrossRef] [PubMed]
  185. Coleman, R.L.; Oza, A.M.; Lorusso, D.; Aghajanian, C.; Oaknin, A.; Dean, A.; Colombo, N.; Weberpals, J.I.; Clamp, A.; Scambia, G.; et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 390, 1949–1961. [Google Scholar] [CrossRef] [PubMed]
  186. Hirschl, N.; Leveque, W.; Granitto, J.; Sammarco, V.; Fontillas, M.; Penson, R.T. PARP Inhibitors: Strategic Use and Optimal Management in Ovarian Cancer. Cancers 2024, 16, 932. [Google Scholar] [CrossRef] [PubMed]
  187. Jain, R. A new target for tumor therapy. N. Engl. J. Med. 2009, 360, 2669–2671. [Google Scholar] [CrossRef] [PubMed]
  188. Ray-Coquard, I.; Pautier, P.; Pignata, S.; Pérol, D.; González-Martín, A.; Berger, R.; Fujiwara, K.; Vergote, I.; Colombo, N.; Mäenpää, J.; et al. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2416–2428. [Google Scholar] [CrossRef] [PubMed]
  189. Hardesty, M.M.; Krivak, T.C.; Wright, G.S.; Hamilton, E.; Fleming, E.L.; Belotte, J.; Keeton, E.K.; Wang, P.; Gupta, D.; Clements, A.; et al. OVARIO phase II trial of combination niraparib plus bevacizumab maintenance therapy in advanced ovarian cancer following first-line platinum-based chemotherapy with bevacizumab. Gynecol. Oncol. 2022, 166, 219–229. [Google Scholar] [CrossRef]
  190. Roy, S.; Ghosh, J.; Ganguly, S.; Biswas, B.; Bhaumik, J. First-line Rucaparib Plus Bevacizumab Maintenance Completed One-Year in Germline BRCA1-Mutated Advanced Ovarian Cancer. Cureus 2022, 14, e32493. [Google Scholar] [CrossRef]
  191. Li, J.; Yue, H.; Li, W.; Zhu, G.; Zhu, T.; Chen, R.; Lu, X. Bevacizumab confers significant improvements in survival for ovarian cancer patients with low miR-25 expression and high miR-142 expression. J. Ovarian Res. 2021, 14, 166. [Google Scholar] [CrossRef] [PubMed]
  192. Halvorsen, A.R.; Kristensen, G.; Embleton, A.; Adusei, C.; Barretina-Ginesta, M.P.; Beale, P.; Helland, Å. Evaluation of Prognostic and Predictive Significance of Circulating MicroRNAs in Ovarian Cancer Patients. Dis. Markers 2017, 2017, 3098542. [Google Scholar] [CrossRef]
  193. Hartman, J.L.; Garvik, B.; Hartwell, L. Principles for the buffering of genetic variation. Science 2001, 291, 1001–1004. [Google Scholar] [CrossRef]
  194. Kaelin, W.G. The concept of synthetic lethality in the context of anticancer therapy. Nat. Rev. Cancer 2005, 5, 689–698. [Google Scholar] [CrossRef] [PubMed]
  195. Wu, Y.; Xu, S.; Cheng, S.; Yang, J.; Wang, Y. Clinical application of PARP inhibitors in ovarian cancer: From molecular mechanisms to the current status. J. Ovarian Res. 2023, 16, 6. [Google Scholar] [CrossRef] [PubMed]
  196. Leconte, M.; Nicco, C.; Ngô, C.; Chéreau, C.; Chouzenoux, S.; Marut, W.; Guibourdenche, J.; Arkwright, S.; Weill, B.; Chapron, C.; et al. The mTOR/AKT inhibitor temsirolimus prevents deep infiltrating endometriosis in mice. Am. J. Pathol. 2011, 179, 880–889. [Google Scholar] [CrossRef] [PubMed]
  197. Farley, J.H.; Brady, W.E.; O’Malley, D.; Fujiwara, K.; Yonemori, K.; Bonebrake, A.; Secord, A.A.; Stephan, J.M.; Walker, J.L.; Nam, J.H.; et al. A phase II evaluation of temsirolimus with carboplatin and paclitaxel followed by temsirolimus consolidation in clear cell ovarian cancer: An NRG oncology trial. Gynecol. Oncol. 2022, 167, 423–428. [Google Scholar] [CrossRef] [PubMed]
  198. Nero, C.; Romito, I.; Spadola, S.; Romito, A.; Turco, L.C.; Cosentino, F.; DeNinno, M.; Catena, U.; De Cicco Nardone, A.; Moroni, R.; et al. Infiltrating T lymphocytes and programmed cell death protein-1/programmed death-ligand 1 expression in endometriosis-associated ovarian cancer. Fertil. Steril. 2022, 117, 160–168. [Google Scholar] [CrossRef] [PubMed]
  199. Bi, F.; Chen, Y.; Yang, Q. Significance of tumor mutation burden combined with immune infiltrates in the progression and prognosis of ovarian cancer. Cancer Cell Int. 2020, 20, 373. [Google Scholar] [CrossRef]
  200. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef]
  201. Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef]
  202. Hsieh, T.H.; Hsu, C.Y.; Wu, C.W.; Wang, S.H.; Yeh, C.H.; Cheng, K.H.; Tsai, E.M. Vorinostat decrease M2 macrophage polarization through ARID1A6488delG/HDAC6/IL-10 signaling pathway in endometriosis-associated ovarian carcinoma. Biomed. Pharmacother. 2023, 161, 114500. [Google Scholar] [CrossRef]
Figure 1. Molecular factors involved in the malignant transformation of endometrioma to endometriosis-associated ovarian cancer (EAOC). Through retrograde menstruation, endometrial tissue grows outside the uterus in ectopic locations including the ovaries. Hereby, several genetic alterations, microRNAs, local estrogen levels, and oxidative stress contribute to the malignant transformation of benign endometrial lesions to tumor progression. Common genetic mutations accumulating during the growth process affect the function of the proto-oncogene K-Ras (Kirsten rat sarcoma virus), the tumor suppressor gene P53 (TP53), the catalytic subunit alpha (PIK3CA) of the phosphatidylinositol 3-kinase (PI3K), as well as negative regulators of the PI3K/AKT/mTOR pathway, including the phosphatase and tensin homolog (PTEN) and a regulatory subunit of protein phosphatase 2 (PP2A) (encoded by the PPP2R1A gene). Furthermore, the AT-rich interactive domain-containing protein 1A (ARID1A) is frequently mutated, leading to the altered function of the mammalian SWItch/Sucrose Non-Fermentable (mSWI/SNF) complex, a subfamily of ATP-dependent chromatin remodeling complexes. Altogether, those mutated genes are mainly responsible for DNA damage repair, the regulation of apoptosis, protein synthesis, cell cycle control, proliferation, survival, and growth. Therefore, other signaling pathways such as the Wnt pathway (e.g., Wnt protein, lipoprotein receptor-related protein (LRP), T-cell factor/lymphoid enhancer factor, beta-catenin, but also its destruction complex [axin, adenomatosis polyposis coli (APC), glycogen synthase kinase 3 (GSK3) and casein kinase 1α (CK1α)]) and Notch pathways (neurogenic locus notch homolog protein-induced liberation of N-formylmaleamic acid amidohydrolase) may also be related to the development of EAOC. Alongside tumor-intrinsic genetic factors, microRNAs (miRNAs) serve as crucial post-transcriptional regulators of gene expression, exhibiting complex dysregulation in various human cancers, with the differential expression of specific families like miR-200, let-7, and miR-31 influencing epithelial-to-mesenchymal transition, oncogenicity, and angiogenesis. Furthermore, estrogen promotes endometriotic cell proliferation through various signaling pathways, while an imbalance favoring estrogen receptor (ER)-alpha dominance or loss of ER-beta contributes to carcinogenesis via an estrogen-responsive element (ERE)- or transcription factor (TF) response element (TFRE)-mediated gene transcription. In addition, estradiol binds to the G protein-coupled estrogen receptor 1 (GPER1 or GPR30), leading to the activation of the cAMP/PKA (protein kinase A)/CREB (cAMP response element-binding protein) pathway. Finally, oxidative stress plays a pivotal role in shaping EAOC by inducing DNA damage, inflammation, and aberrant cellular signaling and disrupting antioxidant defenses. [Figure created with BioRender].
Figure 1. Molecular factors involved in the malignant transformation of endometrioma to endometriosis-associated ovarian cancer (EAOC). Through retrograde menstruation, endometrial tissue grows outside the uterus in ectopic locations including the ovaries. Hereby, several genetic alterations, microRNAs, local estrogen levels, and oxidative stress contribute to the malignant transformation of benign endometrial lesions to tumor progression. Common genetic mutations accumulating during the growth process affect the function of the proto-oncogene K-Ras (Kirsten rat sarcoma virus), the tumor suppressor gene P53 (TP53), the catalytic subunit alpha (PIK3CA) of the phosphatidylinositol 3-kinase (PI3K), as well as negative regulators of the PI3K/AKT/mTOR pathway, including the phosphatase and tensin homolog (PTEN) and a regulatory subunit of protein phosphatase 2 (PP2A) (encoded by the PPP2R1A gene). Furthermore, the AT-rich interactive domain-containing protein 1A (ARID1A) is frequently mutated, leading to the altered function of the mammalian SWItch/Sucrose Non-Fermentable (mSWI/SNF) complex, a subfamily of ATP-dependent chromatin remodeling complexes. Altogether, those mutated genes are mainly responsible for DNA damage repair, the regulation of apoptosis, protein synthesis, cell cycle control, proliferation, survival, and growth. Therefore, other signaling pathways such as the Wnt pathway (e.g., Wnt protein, lipoprotein receptor-related protein (LRP), T-cell factor/lymphoid enhancer factor, beta-catenin, but also its destruction complex [axin, adenomatosis polyposis coli (APC), glycogen synthase kinase 3 (GSK3) and casein kinase 1α (CK1α)]) and Notch pathways (neurogenic locus notch homolog protein-induced liberation of N-formylmaleamic acid amidohydrolase) may also be related to the development of EAOC. Alongside tumor-intrinsic genetic factors, microRNAs (miRNAs) serve as crucial post-transcriptional regulators of gene expression, exhibiting complex dysregulation in various human cancers, with the differential expression of specific families like miR-200, let-7, and miR-31 influencing epithelial-to-mesenchymal transition, oncogenicity, and angiogenesis. Furthermore, estrogen promotes endometriotic cell proliferation through various signaling pathways, while an imbalance favoring estrogen receptor (ER)-alpha dominance or loss of ER-beta contributes to carcinogenesis via an estrogen-responsive element (ERE)- or transcription factor (TF) response element (TFRE)-mediated gene transcription. In addition, estradiol binds to the G protein-coupled estrogen receptor 1 (GPER1 or GPR30), leading to the activation of the cAMP/PKA (protein kinase A)/CREB (cAMP response element-binding protein) pathway. Finally, oxidative stress plays a pivotal role in shaping EAOC by inducing DNA damage, inflammation, and aberrant cellular signaling and disrupting antioxidant defenses. [Figure created with BioRender].
Ijms 25 04306 g001
Figure 2. Diagnostic and therapeutic algorithm for endometriosis and endometriosis-associated ovarian cancer (EAOC). For further detailed information, see ESHRE guidelines.
Figure 2. Diagnostic and therapeutic algorithm for endometriosis and endometriosis-associated ovarian cancer (EAOC). For further detailed information, see ESHRE guidelines.
Ijms 25 04306 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Steinbuch, S.C.; Lüß, A.-M.; Eltrop, S.; Götte, M.; Kiesel, L. Endometriosis-Associated Ovarian Cancer: From Molecular Pathologies to Clinical Relevance. Int. J. Mol. Sci. 2024, 25, 4306. https://doi.org/10.3390/ijms25084306

AMA Style

Steinbuch SC, Lüß A-M, Eltrop S, Götte M, Kiesel L. Endometriosis-Associated Ovarian Cancer: From Molecular Pathologies to Clinical Relevance. International Journal of Molecular Sciences. 2024; 25(8):4306. https://doi.org/10.3390/ijms25084306

Chicago/Turabian Style

Steinbuch, Sophie Charlotte, Anne-Marie Lüß, Stephanie Eltrop, Martin Götte, and Ludwig Kiesel. 2024. "Endometriosis-Associated Ovarian Cancer: From Molecular Pathologies to Clinical Relevance" International Journal of Molecular Sciences 25, no. 8: 4306. https://doi.org/10.3390/ijms25084306

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

Article Metrics

Back to TopTop